1
|
Li Y, Lin Y, Chen Z, Ji W, Liu H. Deficiency of ATF2 retards senescence induced by replication stress and pamidronate in mouse jaw bone marrow stem cells. Cell Signal 2025; 127:111579. [PMID: 39733927 DOI: 10.1016/j.cellsig.2024.111579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 12/31/2024]
Abstract
The aging process is associated with a loss of bone mass and an accumulation of senescent cells, which is under epigenetic control. Morphological and molecular analysis revealed a notable reduction in bone mass and alveolar crest height in aged mice, accompanied by increased levels of senescent mouse jaw bone marrow stem cells (mJBMSCs). To investigate whether specific transcription factors are involved, assay for transposase-accessible chromatin with sequencing (ATAC-seq) was performed on mJBMSCs isolated from 2-, 4-, 8-, and 20-month-old mice. In 20-month-old mJBMSCs, increased chromatin accessibility was observed alongside elevated expression of activating transcription factor 2 (ATF2) in both cells and alveolar bone. Silencing Atf2 in mJBMSCs failed to reverse physiological aging, but delayed replication stress and pamidronate (PAM) induced senescence. The analysis of ATAC-seq and RNA sequencing indicated that the differentially expressed genes upregulated by PAM but downregulated by ATF2 deficiency were related to some key biological processes, including negative regulation of cell proliferation, inflammatory response, adipogenesis, and cellular senescence. The dual-luciferase assay was conducted to demonstrate that ATF2 enhances Cdkn2a transcription by binding to its promoter region. Our findings suggest significant chromatin alterations in aged mJBMSCs, positioning ATF2 as a potential target for combating externally induced senescence.
Collapse
Affiliation(s)
- Yuanyuan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yuxiu Lin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Cariology and Endodontics, School of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zhi Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Cariology and Endodontics, School of Stomatology, Wuhan University, Wuhan 430079, China
| | - Wei Ji
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral Implantology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Huan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
2
|
Deng Z, Fan T, Xiao C, Tian H, Zheng Y, Li C, He J. TGF-β signaling in health, disease, and therapeutics. Signal Transduct Target Ther 2024; 9:61. [PMID: 38514615 PMCID: PMC10958066 DOI: 10.1038/s41392-024-01764-w] [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: 12/07/2022] [Revised: 08/31/2023] [Accepted: 01/31/2024] [Indexed: 03/23/2024] Open
Abstract
Transforming growth factor (TGF)-β is a multifunctional cytokine expressed by almost every tissue and cell type. The signal transduction of TGF-β can stimulate diverse cellular responses and is particularly critical to embryonic development, wound healing, tissue homeostasis, and immune homeostasis in health. The dysfunction of TGF-β can play key roles in many diseases, and numerous targeted therapies have been developed to rectify its pathogenic activity. In the past decades, a large number of studies on TGF-β signaling have been carried out, covering a broad spectrum of topics in health, disease, and therapeutics. Thus, a comprehensive overview of TGF-β signaling is required for a general picture of the studies in this field. In this review, we retrace the research history of TGF-β and introduce the molecular mechanisms regarding its biosynthesis, activation, and signal transduction. We also provide deep insights into the functions of TGF-β signaling in physiological conditions as well as in pathological processes. TGF-β-targeting therapies which have brought fresh hope to the treatment of relevant diseases are highlighted. Through the summary of previous knowledge and recent updates, this review aims to provide a systematic understanding of TGF-β signaling and to attract more attention and interest to this research area.
Collapse
Affiliation(s)
- Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
3
|
Shang S, Yang C, Chen F, Xiang RS, Zhang H, Dai SY, Liu J, Lv XX, Zhang C, Liu XT, Zhang Q, Lu SB, Song JW, Yu JJ, Zhou JC, Zhang XW, Cui B, Li PP, Zhu ST, Zhang HZ, Hua F. ID1 expressing macrophages support cancer cell stemness and limit CD8 + T cell infiltration in colorectal cancer. Nat Commun 2023; 14:7661. [PMID: 37996458 PMCID: PMC10667515 DOI: 10.1038/s41467-023-43548-w] [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: 03/13/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023] Open
Abstract
Elimination of cancer stem cells (CSCs) and reinvigoration of antitumor immunity remain unmet challenges for cancer therapy. Tumor-associated macrophages (TAMs) constitute the prominant population of immune cells in tumor tissues, contributing to the formation of CSC niches and a suppressive immune microenvironment. Here, we report that high expression of inhibitor of differentiation 1 (ID1) in TAMs correlates with poor outcome in patients with colorectal cancer (CRC). ID1 expressing macrophages maintain cancer stemness and impede CD8+ T cell infiltration. Mechanistically, ID1 interacts with STAT1 to induce its cytoplasmic distribution and inhibits STAT1-mediated SerpinB2 and CCL4 transcription, two secretory factors responsible for cancer stemness inhibition and CD8+ T cell recruitment. Reducing ID1 expression ameliorates CRC progression and enhances tumor sensitivity to immunotherapy and chemotherapy. Collectively, our study highlights the pivotal role of ID1 in controlling the protumor phenotype of TAMs and paves the way for therapeutic targeting of ID1 in CRC.
Collapse
Affiliation(s)
- Shuang Shang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Chen Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Fei Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Ren-Shen Xiang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China
| | - Huan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Shu-Yuan Dai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Jing Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Xiao-Xi Lv
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Cheng Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Department of Pharmacy, China-Japan Friendship Hospital, 100029, Beijing, P. R. China
| | - Xiao-Tong Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Qi Zhang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China
| | - Shuai-Bing Lu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China
| | - Jia-Wei Song
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Jiao-Jiao Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Ji-Chao Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Xiao-Wei Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Bing Cui
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Ping-Ping Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Sheng-Tao Zhu
- Beijing Digestive Diseases Center, Beijing Friendship Hospital, 100050, Beijing, P. R. China
- Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases, Beijing Friendship Hospital, 100050, Beijing, P. R. China
| | - Hai-Zeng Zhang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China.
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China.
| | - Fang Hua
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China.
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China.
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China.
| |
Collapse
|
4
|
Endo T. Postnatal skeletal muscle myogenesis governed by signal transduction networks: MAPKs and PI3K-Akt control multiple steps. Biochem Biophys Res Commun 2023; 682:223-243. [PMID: 37826946 DOI: 10.1016/j.bbrc.2023.09.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/06/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023]
Abstract
Skeletal muscle myogenesis represents one of the most intensively and extensively examined systems of cell differentiation, tissue formation, and regeneration. Muscle regeneration provides an in vivo model system of postnatal myogenesis. It comprises multiple steps including muscle stem cell (or satellite cell) quiescence, activation, migration, myogenic determination, myoblast proliferation, myocyte differentiation, myofiber maturation, and hypertrophy. A variety of extracellular signaling and subsequent intracellular signal transduction pathways or networks govern the individual steps of postnatal myogenesis. Among them, MAPK pathways (the ERK, JNK, p38 MAPK, and ERK5 pathways) and PI3K-Akt signaling regulate multiple steps of myogenesis. Ca2+, cytokine, and Wnt signaling also participate in several myogenesis steps. These signaling pathways often control cell cycle regulatory proteins or the muscle-specific MyoD family and the MEF2 family of transcription factors. This article comprehensively reviews molecular mechanisms of the individual steps of postnatal skeletal muscle myogenesis by focusing on signal transduction pathways or networks. Nevertheless, no or only a partial signaling molecules or pathways have been identified in some responses during myogenesis. The elucidation of these unidentified signaling molecules and pathways leads to an extensive understanding of the molecular mechanisms of myogenesis.
Collapse
Affiliation(s)
- Takeshi Endo
- Department of Biology, Graduate School of Science, Chiba University, Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan.
| |
Collapse
|
5
|
Lin Y, Zhan M, Xu B. Exportin XPO7 acts as an oncogenic factor in prostate cancer via upregulation of TCF3. J Cancer Res Clin Oncol 2023; 149:7663-7677. [PMID: 37000263 DOI: 10.1007/s00432-023-04705-2] [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: 01/08/2023] [Accepted: 03/17/2023] [Indexed: 04/01/2023]
Abstract
PURPOSE As a nuclear transport protein, XPO7 has been observed to show abnormal expression in various types of human cancers. However, the role of XPO7 in PCa remains elusive. METHODS Here, in this study, immunohistochemistry and bioinformatics were used to determine the expression pattern and prognostic significance of XPO7. To investigate the functions of XPO7 in vitro and in vivo, we knocked down XPO7 in PCa cell lines and established xenograft mice models. Then, we used multiple experiments to determine the cell proliferation, migration, invasion, cell cycle and EMT in PCa cells after XPO7 modulation. Mechanistically, we conducted RNA-seq and identified the regulating effect of XPO7 on cell cycle-related and PI3K-AKT pathways. Furthermore, we assessed the regulating correlation between XPO7 and TCF3 and verified by a series of rescue experiments. RESULTS We found a higher XPO7 expression in prostate cancer tissues and predicted a poorer prognosis of prostate cancer. Then, we further revealed that the ectopic expression of XPO7 in PCa cells facilitated cells proliferation, migration, cell cycle progression and EMT in vitro and promoted tumor growth in vivo. Mechanistically, we conducted RNA-seq and identified the regulating effect of XPO7 on cell cycle-related and PI3K-AKT pathways. Furthermore, a significantly positive correlation was discovered between the expression of XPO7 and TCF3. In addition, XPO7 may regulate PCa through mediating TCF3 expression. TCF3 depletion could alleviate the influence of XPO7 overexpression on malignant phenotypes of PCa cells. CONCLUSIONS These findings indicate that XPO7 promotes PCa initiation and progression and that targeting XPO7 might be therapeutically beneficial to patients with PCa.
Collapse
Affiliation(s)
- Yu Lin
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ming Zhan
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Bin Xu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
6
|
Tanaka M, Homme M, Teramura Y, Kumegawa K, Yamazaki Y, Yamashita K, Osato M, Maruyama R, Nakamura T. HEY1-NCOA2 expression modulates chondrogenic differentiation and induces mesenchymal chondrosarcoma in mice. JCI Insight 2023; 8:160279. [PMID: 37212282 DOI: 10.1172/jci.insight.160279] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/12/2023] [Indexed: 05/23/2023] Open
Abstract
Mesenchymal chondrosarcoma affects adolescents and young adults, and most cases usually have the HEY1::NCOA2 fusion gene. However, the functional role of HEY1-NCOA2 in the development and progression of mesenchymal chondrosarcoma remains largely unknown. This study aimed to clarify the functional role of HEY1-NCOA2 in transformation of the cell of origin and induction of typical biphasic morphology of mesenchymal chondrosarcoma. We generated a mouse model for mesenchymal chondrosarcoma by introducing HEY1-NCOA2 into mouse embryonic superficial zone (eSZ) followed by subcutaneous transplantation into nude mice. HEY1-NCOA2 expression in eSZ cells successfully induced subcutaneous tumors in 68.9% of recipients, showing biphasic morphologies and expression of Sox9, a master regulator of chondrogenic differentiation. ChIP sequencing analyses indicated frequent interaction between HEY1-NCOA2 binding peaks and active enhancers. Runx2, which is important for differentiation and proliferation of the chondrocytic lineage, is invariably expressed in mouse mesenchymal chondrosarcoma, and interaction between HEY1-NCOA2 and Runx2 is observed using NCOA2 C-terminal domains. Although Runx2 knockout resulted in significant delay in tumor onset, it also induced aggressive growth of immature small round cells. Runx3, which is also expressed in mesenchymal chondrosarcoma and interacts with HEY1-NCOA2, replaced the DNA-binding property of Runx2 only in part. Treatment with the HDAC inhibitor panobinostat suppressed tumor growth both in vitro and in vivo, abrogating expression of genes downstream of HEY1-NCOA2 and Runx2. In conclusion, HEY1::NCOA2 expression modulates the transcriptional program in chondrogenic differentiation, affecting cartilage-specific transcription factor functions.
Collapse
Affiliation(s)
- Miwa Tanaka
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Experimental Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
- Project for Cancer Epigenomics, The Cancer Institute, and
| | - Mizuki Homme
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yasuyo Teramura
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kohei Kumegawa
- Project for Cancer Epigenomics, The Cancer Institute, and
| | - Yukari Yamazaki
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Experimental Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Kyoko Yamashita
- Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Motomi Osato
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Reo Maruyama
- Project for Cancer Epigenomics, The Cancer Institute, and
| | - Takuro Nakamura
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Experimental Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
7
|
Pankow A, Sun XH. The divergence between T cell and innate lymphoid cell fates controlled by E and Id proteins. Front Immunol 2022; 13:960444. [PMID: 36032069 PMCID: PMC9399370 DOI: 10.3389/fimmu.2022.960444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/13/2022] [Indexed: 11/18/2022] Open
Abstract
T cells develop in the thymus from lymphoid primed multipotent progenitors or common lymphoid progenitors into αβ and γδ subsets. The basic helix-loop-helix transcription factors, E proteins, play pivotal roles at multiple stages from T cell commitment to maturation. Inhibitors of E proteins, Id2 and Id3, also regulate T cell development while promoting ILC differentiation. Recent findings suggest that the thymus can also produce innate lymphoid cells (ILCs). In this review, we present current findings that suggest the balance between E and Id proteins is likely to be critical for controlling the bifurcation of T cell and ILC fates at early stages of T cell development.
Collapse
Affiliation(s)
- Aneta Pankow
- Program in Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Xiao-Hong Sun
- Program in Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- *Correspondence: Xiao-Hong Sun,
| |
Collapse
|
8
|
Singh S, Sarkar T, Jakubison B, Gadomski S, Spradlin A, Gudmundsson KO, Keller JR. Inhibitor of DNA binding proteins revealed as orchestrators of steady state, stress and malignant hematopoiesis. Front Immunol 2022; 13:934624. [PMID: 35990659 PMCID: PMC9389078 DOI: 10.3389/fimmu.2022.934624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/12/2022] [Indexed: 11/24/2022] Open
Abstract
Adult mammalian hematopoiesis is a dynamic cellular process that provides a continuous supply of myeloid, lymphoid, erythroid/megakaryocyte cells for host survival. This process is sustained by regulating hematopoietic stem cells (HSCs) quiescence, proliferation and activation under homeostasis and stress, and regulating the proliferation and differentiation of downstream multipotent progenitor (MPP) and more committed progenitor cells. Inhibitor of DNA binding (ID) proteins are small helix-loop-helix (HLH) proteins that lack a basic (b) DNA binding domain present in other family members, and function as dominant-negative regulators of other bHLH proteins (E proteins) by inhibiting their transcriptional activity. ID proteins are required for normal T cell, B cell, NK and innate lymphoid cells, dendritic cell, and myeloid cell differentiation and development. However, recent evidence suggests that ID proteins are important regulators of normal and leukemic hematopoietic stem and progenitor cells (HSPCs). This chapter will review our current understanding of the function of ID proteins in HSPC development and highlight future areas of scientific investigation.
Collapse
Affiliation(s)
- Shweta Singh
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
| | - Tanmoy Sarkar
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
| | - Brad Jakubison
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Stephen Gadomski
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
| | - Andrew Spradlin
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
| | - Kristbjorn O. Gudmundsson
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Jonathan R. Keller
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
- *Correspondence: Jonathan R. Keller,
| |
Collapse
|
9
|
Aubrey M, Warburg ZJ, Murre C. Helix-Loop-Helix Proteins in Adaptive Immune Development. Front Immunol 2022; 13:881656. [PMID: 35634342 PMCID: PMC9134016 DOI: 10.3389/fimmu.2022.881656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
The E/ID protein axis is instrumental for defining the developmental progression and functions of hematopoietic cells. The E proteins are dimeric transcription factors that activate gene expression programs and coordinate changes in chromatin organization. Id proteins are antagonists of E protein activity. Relative levels of E/Id proteins are modulated throughout hematopoietic development to enable the progression of hematopoietic stem cells into multiple adaptive and innate immune lineages including natural killer cells, B cells and T cells. In early progenitors, the E proteins promote commitment to the T and B cell lineages by orchestrating lineage specific programs of gene expression and regulating VDJ recombination of antigen receptor loci. In mature B cells, the E/Id protein axis functions to promote class switch recombination and somatic hypermutation. E protein activity further regulates differentiation into distinct CD4+ and CD8+ T cells subsets and instructs mature T cell immune responses. In this review, we discuss how the E/Id proteins define the adaptive immune system lineages, focusing on their role in directing developmental gene programs.
Collapse
Affiliation(s)
| | | | - Cornelis Murre
- Division of Biological Sciences, Section of Molecular Biology, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
10
|
Veiga DFT, Tremblay M, Gerby B, Herblot S, Haman A, Gendron P, Lemieux S, Zúñiga-Pflücker JC, Hébert J, Cohen JP, Hoang T. Monoallelic Heb/Tcf12 Deletion Reduces the Requirement for NOTCH1 Hyperactivation in T-Cell Acute Lymphoblastic Leukemia. Front Immunol 2022; 13:867443. [PMID: 35401501 PMCID: PMC8987207 DOI: 10.3389/fimmu.2022.867443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/28/2022] [Indexed: 12/21/2022] Open
Abstract
Early T-cell development is precisely controlled by E proteins, that indistinguishably include HEB/TCF12 and E2A/TCF3 transcription factors, together with NOTCH1 and pre-T cell receptor (TCR) signalling. Importantly, perturbations of early T-cell regulatory networks are implicated in leukemogenesis. NOTCH1 gain of function mutations invariably lead to T-cell acute lymphoblastic leukemia (T-ALL), whereas inhibition of E proteins accelerates leukemogenesis. Thus, NOTCH1, pre-TCR, E2A and HEB functions are intertwined, but how these pathways contribute individually or synergistically to leukemogenesis remain to be documented. To directly address these questions, we leveraged Cd3e-deficient mice in which pre-TCR signaling and progression through β-selection is abrogated to dissect and decouple the roles of pre-TCR, NOTCH1, E2A and HEB in SCL/TAL1-induced T-ALL, via the use of Notch1 gain of function transgenic (Notch1ICtg) and Tcf12+/- or Tcf3+/- heterozygote mice. As a result, we now provide evidence that both HEB and E2A restrain cell proliferation at the β-selection checkpoint while the clonal expansion of SCL-LMO1-induced pre-leukemic stem cells in T-ALL is uniquely dependent on Tcf12 gene dosage. At the molecular level, HEB protein levels are decreased via proteasomal degradation at the leukemic stage, pointing to a reversible loss of function mechanism. Moreover, in SCL-LMO1-induced T-ALL, loss of one Tcf12 allele is sufficient to bypass pre-TCR signaling which is required for Notch1 gain of function mutations and for progression to T-ALL. In contrast, Tcf12 monoallelic deletion does not accelerate Notch1IC-induced T-ALL, indicating that Tcf12 and Notch1 operate in the same pathway. Finally, we identify a tumor suppressor gene set downstream of HEB, exhibiting significantly lower expression levels in pediatric T-ALL compared to B-ALL and brain cancer samples, the three most frequent pediatric cancers. In summary, our results indicate a tumor suppressor function of HEB/TCF12 in T-ALL to mitigate cell proliferation controlled by NOTCH1 in pre-leukemic stem cells and prevent NOTCH1-driven progression to T-ALL.
Collapse
Affiliation(s)
- Diogo F. T. Veiga
- Department of Pharmacology and Physiology, Université de Montréal, Institute for Research in Immunology and Cancer, QC, Canada
- Department of Translational Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Mathieu Tremblay
- Department of Pharmacology and Physiology, Université de Montréal, Institute for Research in Immunology and Cancer, QC, Canada
| | - Bastien Gerby
- Department of Pharmacology and Physiology, Université de Montréal, Institute for Research in Immunology and Cancer, QC, Canada
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-1037, Université Toulouse III Paul Sabatier (UPS), Toulouse, France
| | - Sabine Herblot
- Department of Pharmacology and Physiology, Université de Montréal, Institute for Research in Immunology and Cancer, QC, Canada
- Unité de recherche en hémato-oncologie Charles-Bruneau, Centre de Recherche du CHU Sainte-Justine, Montréal, Canada
| | - André Haman
- Department of Pharmacology and Physiology, Université de Montréal, Institute for Research in Immunology and Cancer, QC, Canada
| | - Patrick Gendron
- Department of Pharmacology and Physiology, Université de Montréal, Institute for Research in Immunology and Cancer, QC, Canada
| | - Sébastien Lemieux
- Department of Pharmacology and Physiology, Université de Montréal, Institute for Research in Immunology and Cancer, QC, Canada
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | | | - Josée Hébert
- Department of Pharmacology and Physiology, Université de Montréal, Institute for Research in Immunology and Cancer, QC, Canada
- Institut universitaire d’hémato-oncologie et de thérapie cellulaire, Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Quebec Leukemia Cell Bank, Centre de recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Joseph Paul Cohen
- Department of Computer Science and Operations Research, Université de Montréal, Montreal, QC, Canada
- Université de Montréal, Montreal, QC, Canada
| | - Trang Hoang
- Department of Pharmacology and Physiology, Université de Montréal, Institute for Research in Immunology and Cancer, QC, Canada
- *Correspondence: Trang Hoang,
| |
Collapse
|
11
|
Kyei F. Initial Assessment of Id Protein Antagonist (aHLH) shows Anti-proliferative Activity on HEK293T, LS174T and HCT116 Cells. SCIENTIFIC AFRICAN 2022. [DOI: 10.1016/j.sciaf.2022.e01099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
12
|
Kiyokawa H, Yamaoka A, Matsuoka C, Tokuhara T, Abe T, Morimoto M. Airway basal stem cells reutilize the embryonic proliferation regulator, Tgfβ-Id2 axis, for tissue regeneration. Dev Cell 2021; 56:1917-1929.e9. [PMID: 34129836 DOI: 10.1016/j.devcel.2021.05.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/28/2021] [Accepted: 05/19/2021] [Indexed: 12/15/2022]
Abstract
During development, quiescent airway basal stem cells are derived from proliferative primordial progenitors through the cell-cycle slowdown. In contrast, basal cells contribute to adult tissue regeneration by shifting from slow cycling to proliferating and subsequently back to slow cycling. Although sustained proliferation results in tumorigenesis, the molecular mechanisms regulating these transitions remain unknown. Using temporal single-cell transcriptomics of developing murine airway progenitors and genetic validation experiments, we found that TGF-β signaling decelerated cell cycle by inhibiting Id2 and contributed to slow-cycling basal cell specification during development. In adult tissue regeneration, reduced TGF-β signaling restored Id2 expression and initiated regeneration. Id2 overexpression and Tgfbr2 knockout enhanced epithelial proliferation; however, persistent Id2 expression drove basal cell hyperplasia that resembled a precancerous state. Together, the TGF-β-Id2 axis commonly regulates the proliferation transitions in basal cells during development and regeneration, and its fine-tuning is critical for normal regeneration while avoiding basal cell hyperplasia.
Collapse
Affiliation(s)
- Hirofumi Kiyokawa
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Akira Yamaoka
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Chisa Matsuoka
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Tomoko Tokuhara
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Mitsuru Morimoto
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan.
| |
Collapse
|
13
|
Ha CT, Cheng CY, Zheng MY, Hsu TH, Miao CC, Lee CJ, Wang HD, Pan ST, Chou YT. ID4 predicts poor prognosis and promotes BDNF-mediated oncogenesis of colorectal cancer. Carcinogenesis 2021; 42:951-960. [PMID: 33993270 DOI: 10.1093/carcin/bgab037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/19/2021] [Accepted: 05/06/2021] [Indexed: 11/14/2022] Open
Abstract
Inhibitors of DNA binding and cell differentiation (ID) proteins regulate cellular differentiation and tumor progression. Whether ID family proteins serve as a linkage between pathological differentiation and cancer stemness in colorectal cancer is largely unknown. Here, the expression of ID4, but not other ID family proteins, was enriched in LGR5-high colon cancer stem cells. Its high expression was associated with poor pathological differentiation of colorectal tumors and shorter survival in patients. Knockdown of ID4 inhibited the growth and dissemination of colon cancer cells, while enhancing chemosensitivity. Through gene expression profiling analysis, brain-derived neurotrophic factor (BDNF) was identified as a downstream target of ID4 expression in colorectal cancer. BDNF knockdown decreased the growth and migration of colon cancer cells, and its expression enhanced dissemination, anoikis resistance and chemoresistance. ID4 silencing attenuated the epithelial-to-mesenchymal transition pattern in colon cancer cells. Gene cluster analysis revealed that ID4 and BDNF expression was clustered with mesenchymal markers and distant from epithelial genes. BDNF silencing decreased the expression of mesenchymal markers Vimentin, CDH2 and SNAI1. These findings demonstrated that ID4-BDNF signaling regulates colorectal cancer survival, with the potential to serve as a prognostic marker in colorectal cancer.
Collapse
Affiliation(s)
- Cam-Thu Ha
- Institute of Biotechnology, National Tsing Hua University, Hsinchu,Taiwan
| | | | - Ming-Yi Zheng
- Institute of Biotechnology, National Tsing Hua University, Hsinchu,Taiwan
| | - Tang-Hui Hsu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu,Taiwan
| | - Chia-Cheng Miao
- Institute of Biotechnology, National Tsing Hua University, Hsinchu,Taiwan
| | - Chang-Jung Lee
- Institute of Biotechnology, National Tsing Hua University, Hsinchu,Taiwan
| | - Horng-Dar Wang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu,Taiwan
| | - Shien-Tung Pan
- Department of Pathology, China Medical University Hsinchu Hospital, Hsinchu County, Taiwan
| | - Yu-Ting Chou
- Institute of Biotechnology, National Tsing Hua University, Hsinchu,Taiwan
| |
Collapse
|
14
|
Mechanical stimulation induced osteogenic differentiation of BMSCs through TWIST/E2A/p21 axis. Biosci Rep 2021; 40:222707. [PMID: 32309849 PMCID: PMC7199451 DOI: 10.1042/bsr20193876] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/02/2020] [Accepted: 04/16/2020] [Indexed: 12/11/2022] Open
Abstract
The relationship between mechanical force and alveolar bone remodeling is an important issue in orthodontics because tooth movement is dependent on the response of bone tissue to the mechanical force induced by the appliances used. Mechanical cyclical stretch plays an essential role in the cell osteogenic differentiation involved in bone remodeling. However, the underlying mechanisms are unclear, particularly the molecular pathways regulated by mechanical stimulation. In the present study, we reported a dynamic change of p21 level in response to mechanical cyclical stretch, and shRNA-p21 in bone marrow mesenchymal stem cells (BMSCs) induced osteogenic differentiation. The mechanism was mediated through TWIST/E2A/p21 axis. These results supported the mechanical stimulation-induced osteogenic differentiation is negatively regulated by p21.
Collapse
|
15
|
Ha TC, Stahlhut M, Rothe M, Paul G, Dziadek V, Morgan M, Brugman M, Fehse B, Kustikova O, Schambach A, Baum C. Multiple Genes Surrounding Bcl-xL, a Common Retroviral Insertion Site, Can Influence Hematopoiesis Individually or in Concert. Hum Gene Ther 2020; 32:458-472. [PMID: 33012194 DOI: 10.1089/hum.2019.344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Retroviral insertional mutagenesis (RIM) is both a relevant risk in gene therapy and a powerful tool for identifying genes that enhance the competitiveness of repopulating hematopoietic stem and progenitor cells (HSPCs). However, focusing only on the gene closest to the retroviral vector insertion site (RVIS) may underestimate the effects of RIM, as dysregulation of distal and/or multiple genes by a single insertion event was reported in several studies. As a proof of concept, we examined the common insertion site (CIS) Bcl-xL, which revealed seven genes located within ±150 kb from the RVIS for our study. We confirmed that Bcl-xL enhanced the competitiveness of HSPCs, whereas the Bcl-xL neighbor Id1 hindered HSPC long-term repopulation. This negative influence of Id1 could be counteracted by co-expressing Bcl-xL. Interestingly, >90% of early reconstituted myeloid cells were found to originate from transduced HSPCs upon simultaneous overexpression of Bcl-xL and Id1, which implies that Bcl-xL and Id1 can collaborate to rapidly replenish the myeloid compartment under stress conditions. To directly compare the competitiveness of HSPCs conveyed by multiple transgenes, we developed a multiple competitor competitive repopulation (MCCR) assay to simultaneously screen effects on HSPC repopulating capacity in a single mouse. The MCCR assay revealed that multiple genes within a CIS can have positive or negative impact on hematopoiesis. Furthermore, these data highlight the importance of studying multiple genes located within the proximity of an insertion site to understand complex biological effects, especially as the number of gene therapy patients increases.
Collapse
Affiliation(s)
- Teng-Cheong Ha
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover, Germany.,Hannover Biomedical Research School, Hannover, Germany
| | - Maike Stahlhut
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Michael Rothe
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover, Germany
| | - Gabi Paul
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Violetta Dziadek
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover, Germany
| | - Martijn Brugman
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center (UKE) Hamburg-Eppendorf, Hamburg, Germany
| | - Olga Kustikova
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover, Germany.,Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher Baum
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
16
|
Singh K, Baird M, Fischer R, Chaitankar V, Seifuddin F, Chen YC, Tunc I, Waterman CM, Pirooznia M. Misregulation of ELK1, AP1, and E12 Transcription Factor Networks Is Associated with Melanoma Progression. Cancers (Basel) 2020; 12:E458. [PMID: 32079144 PMCID: PMC7072154 DOI: 10.3390/cancers12020458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 01/17/2023] Open
Abstract
Melanoma is among the most malignant cutaneous cancers and when metastasized results in dramatically high mortality. Despite advances in high-throughput gene expression profiling in cancer transcriptomic studies, our understanding of mechanisms driving melanoma progression is still limited. We present here an in-depth bioinformatic analysis of the melanoma RNAseq, chromatin immunoprecipitation (ChIP)seq, and single-cell (sc)RNA seq data to understand cancer progression. Specifically, we have performed a consensus network analysis of RNA-seq data from clinically re-grouped melanoma samples to identify gene co-expression networks that are conserved in early (stage 1) and late (stage 4/invasive) stage melanoma. Overlaying the fold-change information on co-expression networks revealed several coordinately up or down-regulated subnetworks that may play a critical role in melanoma progression. Furthermore, by incorporating histone lysine-27 acetylation information and highly expressed genes identified from the single-cell RNA data from melanoma patient samples, we present a comprehensive list of pathways, putative protein-protein interactions (PPIs) and transcription factor (TF) networks that are driving cancer progression. From this analysis, we have identified Elk1, AP1 and E12 TF networks that coordinately change expression in late melanoma when compared to early melanoma, implicating these TFs in melanoma progression. Additionally, the sumoylation-associated interactome is upregulated in invasive melanoma. Together, this bioinformatic analysis potentially implicates a combination of TF networks and PPIs in melanoma progression, which if confirmed in the experimental systems, could be used as targets for drug intervention in melanoma.
Collapse
Affiliation(s)
- Komudi Singh
- Bioinformatics and Computational Biology Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.S.); (V.C.); (F.S.); (Y.-C.C.); (I.T.)
| | - Michelle Baird
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (M.B.); (R.F.); (C.M.W.)
| | - Robert Fischer
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (M.B.); (R.F.); (C.M.W.)
| | - Vijender Chaitankar
- Bioinformatics and Computational Biology Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.S.); (V.C.); (F.S.); (Y.-C.C.); (I.T.)
| | - Fayaz Seifuddin
- Bioinformatics and Computational Biology Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.S.); (V.C.); (F.S.); (Y.-C.C.); (I.T.)
| | - Yun-Ching Chen
- Bioinformatics and Computational Biology Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.S.); (V.C.); (F.S.); (Y.-C.C.); (I.T.)
| | - Ilker Tunc
- Bioinformatics and Computational Biology Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.S.); (V.C.); (F.S.); (Y.-C.C.); (I.T.)
| | - Clare M. Waterman
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (M.B.); (R.F.); (C.M.W.)
| | - Mehdi Pirooznia
- Bioinformatics and Computational Biology Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.S.); (V.C.); (F.S.); (Y.-C.C.); (I.T.)
| |
Collapse
|
17
|
Boen JRA, Gevaert AB, De Keulenaer GW, Van Craenenbroeck EM, Segers VFM. The role of endothelial miRNAs in myocardial biology and disease. J Mol Cell Cardiol 2019; 138:75-87. [PMID: 31756323 DOI: 10.1016/j.yjmcc.2019.11.151] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 01/08/2023]
Abstract
The myocardium is a highly structured pluricellular tissue which is governed by an intricate network of intercellular communication. Endothelial cells are the most abundant cell type in the myocardium and exert crucial roles in both healthy myocardium and during myocardial disease. In the last decade, microRNAs have emerged as new actors in the regulation of cellular function in almost every cell type. Here, we review recent evidence on the regulatory function of different microRNAs expressed in endothelial cells, also called endothelial microRNAs, in healthy and diseased myocardium. Endothelial microRNA emerged as modulators of angiogenesis in the myocardium, they are implicated in the paracrine role of endothelial cells in regulating cardiac contractility and homeostasis, and interfere in the crosstalk between endothelial cells and cardiomyocytes.
Collapse
Affiliation(s)
- Jente R A Boen
- Research group Cardiovascular Diseases, GENCOR Department, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.
| | - Andreas B Gevaert
- Research group Cardiovascular Diseases, GENCOR Department, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Department of Cardiology, Antwerp University Hospital (UZA), Wilrijkstraat 10, Edegem, Belgium.
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Department of Cardiology, ZNA Middelheim Hospital, Lindendreef 1, 2020 Antwerp, Belgium.
| | - Emeline M Van Craenenbroeck
- Research group Cardiovascular Diseases, GENCOR Department, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Department of Cardiology, Antwerp University Hospital (UZA), Wilrijkstraat 10, Edegem, Belgium.
| | - Vincent F M Segers
- Department of Cardiology, Antwerp University Hospital (UZA), Wilrijkstraat 10, Edegem, Belgium; Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.
| |
Collapse
|
18
|
Solorzano-Vargas RS, Bjerknes M, Wu SV, Wang J, Stelzner M, Dunn JCY, Dhawan S, Cheng H, Georgia S, Martín MG. The cellular regulators PTEN and BMI1 help mediate NEUROGENIN-3-induced cell cycle arrest. J Biol Chem 2019; 294:15182-15192. [PMID: 31341016 DOI: 10.1074/jbc.ra119.008926] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/25/2019] [Indexed: 11/06/2022] Open
Abstract
Neurogenin-3 (NEUROG3) is a helix-loop-helix (HLH) transcription factor involved in the production of endocrine cells in the intestine and pancreas of humans and mice. However, the human NEUROG3 loss-of-function phenotype differs subtly from that in mice, but the reason for this difference remains poorly understood. Because NEUROG3 expression precedes exit of the cell cycle and the expression of endocrine cell markers during differentiation, we investigated the effect of lentivirus-mediated overexpression of the human NEUROG3 gene on the cell cycle of BON4 cells and various human nonendocrine cell lines. NEUROG3 overexpression induced a reversible cell cycle exit, whereas expression of a neuronal lineage homolog, NEUROG1, had no such effect. In endocrine lineage cells, the cellular quiescence induced by short-term NEUROG3 expression required cyclin-dependent kinase inhibitor 1A (CDKN1A)/p21CIP1 expression. Expression of endocrine differentiation markers required sustained NEUROG3 expression in the quiescent, but not in the senescent, state. Inhibition of the phosphatase and tensin homolog (PTEN) pathway reversed quiescence by inducing cyclin-dependent kinase 2 (CDK2) and reducing p21CIP1 and NEUROG3 protein levels in BON4 cells and human enteroids. We discovered that NEUROG3 expression stimulates expression of CDKN2a/p16INK4a and BMI1 proto-oncogene polycomb ring finger (BMI1), with the latter limiting expression of the former, delaying the onset of CDKN2a/p16INK4a -driven cellular senescence. Furthermore, NEUROG3 bound to the promoters of both CDKN1a/p21CIP1 and BMI1 genes, and BMI1 attenuated NEUROG3 binding to the CDKN1a/p21CIP1 promoter. Our findings reveal how human NEUROG3 integrates inputs from multiple signaling pathways and thereby mediates cell cycle exit at the onset of differentiation.
Collapse
Affiliation(s)
- R Sergio Solorzano-Vargas
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Matthew Bjerknes
- Department of Medicine, Medical Sciences Building, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - S Vincent Wu
- Veterans Affairs Greater Los Angeles Healthcare System, and Department of Medicine, University of California, Los Angeles, Los Angeles, California 90073
| | - Jiafang Wang
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Matthias Stelzner
- Division of General Surgery, Department of Surgery, University of California, Los Angeles, Los Angeles, California 90095
| | - James C Y Dunn
- Division of Pediatric Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California 94305
| | - Sangeeta Dhawan
- Department of Translational Research and Cellular Therapeutics, City of Hope, Duarte, California 91010
| | - Hazel Cheng
- Department of Medicine, Medical Sciences Building, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Senta Georgia
- Department of Pediatrics, Division of Endocrinology, Children's Hospital of Los Angeles, University of Southern California, Los Angeles, Los Angeles, California 90027
| | - Martín G Martín
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| |
Collapse
|
19
|
Manzo G. Similarities Between Embryo Development and Cancer Process Suggest New Strategies for Research and Therapy of Tumors: A New Point of View. Front Cell Dev Biol 2019; 7:20. [PMID: 30899759 PMCID: PMC6416183 DOI: 10.3389/fcell.2019.00020] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/05/2019] [Indexed: 12/25/2022] Open
Abstract
Here, I propose that cancer stem cells (CSCs) would be equivalent to para-embryonic stem cells (p-ESCs), derived from adult cells de-re-programmed to a ground state. p-ESCs would differ from ESCs by the absence of genomic homeostasis. A p-ESC would constitute the cancer cell of origin (i-CSC or CSC0), capable of generating an initial tumor, corresponding to a pre-implantation blastocyst. In a niche with proper signals, it would engraft as a primary tumor, corresponding to a post-implantation blastocyst. i-CSC progeny would form primary pluripotent and slow self-renewing CSCs (CSC1s), blocked in an undifferentiated state, corresponding to epiblast cells; CSC1s would be tumor-initiating cells (TICs). CSC1s would generate secondary CSCs (CSC2s), corresponding to hypoblast cells; CSC2s would be tumor growth cells (TGCs). CSC1s/CSC2s would generate tertiary CSCs (CSC3s), with a mesenchymal phenotype; CSC3s would be tumor migrating cells (TMCs), corresponding to mesodermal precursors at primitive streak. CSC3s with more favorable conditions (normoxia), by asymmetrical division, would differentiate into cancer progenitor cells (CPCs), and these into cancer differentiated cells (CDCs), thus generating a defined cell hierarchy and tumor progression, mimicking somito-histo-organogenesis. CSC3s with less favorable conditions (hypoxia) would delaminate and migrate as quiescent circulating micro-metastases, mimicking mesenchymal cells in gastrula morphogenetic movements. In metastatic niches, these CSC3s would install and remain dormant in the presence of epithelial/mesenchymal transition (EMT) signals and hypoxia. But, in the presence of mesenchymal/epithelial transition (MET) signals and normoxia, they would revert to self-renewing CSC1s, reproducing the same cell hierarchy of the primary tumor as macro-metastases. Further similarities between ontogenesis and oncogenesis involving crucial factors, such as ID, HSP70, HLA-G, CD44, LIF, and STAT3, are strongly evident at molecular, physiological and immunological levels. Much experimental data about these factors led to considering the cancer process as ectopic rudimentary ontogenesis, where CSCs have privileged immunological conditions. These would consent to CSC development in an adverse environment, just like an embryo, which is tolerated, accepted and favored by the maternal organism in spite of its paternal semi-allogeneicity. From all these considerations, novel research directions, potential innovative tumor therapy and prophylaxis strategies might, theoretically, result.
Collapse
Affiliation(s)
- Giovanni Manzo
- General Pathology, “La Sapienza” University of Rome, Retired, Botrugno, Italy
| |
Collapse
|
20
|
Abstract
In this review from Murre, the evolution of HLH genes, the structures of HLH domains, and the elaborate activities of HLH proteins in multicellular life are discussed. Helix–loop–helix (HLH) proteins are dimeric transcription factors that control lineage- and developmental-specific gene programs. Genes encoding for HLH proteins arose in unicellular organisms >600 million years ago and then duplicated and diversified from ancestral genes across the metazoan and plant kingdoms to establish multicellularity. Hundreds of HLH proteins have been identified with diverse functions in a wide variety of cell types. HLH proteins orchestrate lineage specification, commitment, self-renewal, proliferation, differentiation, and homing. HLH proteins also regulate circadian clocks, protect against hypoxic stress, promote antigen receptor locus assembly, and program transdifferentiation. HLH proteins deposit or erase epigenetic marks, activate noncoding transcription, and sequester chromatin remodelers across the chromatin landscape to dictate enhancer–promoter communication and somatic recombination. Here the evolution of HLH genes, the structures of HLH domains, and the elaborate activities of HLH proteins in multicellular life are discussed.
Collapse
Affiliation(s)
- Cornelis Murre
- Division of Biological Sciences, University of California at San Diego, La Jolla, California 92903, USA
| |
Collapse
|
21
|
Up-regulation of Transcription Factor 3 Is Correlated With Poor Prognosis in Cervical Carcinoma. Int J Gynecol Cancer 2018; 27:1422-1430. [PMID: 28604457 DOI: 10.1097/igc.0000000000001032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVES Transcription factor 3 (TCF3, or E2A) is a multifunctional bHLH (basic helix loop helix) transcription factor. The role of TCF3 expression in cancer and the multiple cell signaling pathways that regulate or are influenced by TCF3 are unclear. Therefore, the expression level of TCF3 in patients with cervical squamous cell carcinoma (CSCC) is discussed in this study. METHODS Total RNA was extracted using real-time quantitative reverse transcription-polymerase chain reaction. Western blotting was applied to confirm the results. Immunohistochemistry was used to characterize the expression patterns of TCF3 in CSCC specimens. The close relationship between the expression levels of TCF3 and the 5-year overall survival time was described by survival curves. The association between TCF3 expression and clinicopathological characteristics of 119 CSCC patients was analyzed by Chi-square, Fisher exact test, and Cox regression analysis. TCF3 was overexpressed or inhibited by plasmid transfection, and the proliferation, invasion, and migration of cells were detected using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), wound healing, and Transwell assays. RESULTS The expression of TCF3 was higher in CSCC tissues than in nonmalignant cervical tissues. Messenger RNA (mRNA) and protein in patient tissues were increased compared with nonmalignant cervical tissues. Moreover, the level of expression in early-stage disease was higher than in the advanced stage. From FIGO (International Federation of Gynecology and Obstetrics) stages I to IV, immunohistochemistry staining intensity gradually increased. A high level of expression was closely related to clinical stages. The expression of TCF3 was negatively correlated with overall survival time. TCF3 can promote HeLa cell growth, invasion, and migration in vitro. CONCLUSIONS Based on our results, TCF3 is clearly associated with the progression of CSCC. This is the first time that it has been reported that TCF3 can act as a tumor promoter in cervical cancer and thus might be of great significance in the prognosis of CSCC.
Collapse
|
22
|
Repression of TCF3/E2A contributes to Hodgkin lymphomagenesis. Oncotarget 2018; 7:36854-36864. [PMID: 27166193 PMCID: PMC5095044 DOI: 10.18632/oncotarget.9210] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 04/25/2016] [Indexed: 01/12/2023] Open
Abstract
Although Hodgkin and Reed-Sternberg (HRS) cells of classical Hodgkin lymphoma (cHL) derived from germinal or post germinal B cells, they have lost the B cell phenotype in the process of lymphomagenesis. The phenomenon can be at least partially explained by repression of B-cell-specific transcription factors including TCF3, early B-cell factor 1 (EBF1), SPI1/PU.1, and FOXO1, which are down-regulated by genetic and epigenetic mechanisms. The unique phenotype has been suspected to be advantageous for survival of HRS cells. Ectopic expression of some of these transcription factors (EBF1, PU.1, FOXO1) indeed impaired survival of cHL cells. Here we show that forced expression of TCF3 causes cell death and cell cycle arrest in cHL cell lines. Mechanistically, TCF3 overexpression modulated expression of multiple pro-apoptotic genes including BIK, APAF1, FASLG, BOK, and TNFRSF10A/DR4. We conclude that TCF3 inactivation contributes not only to extinguishing of B cell phenotype but also to cHL oncogenesis.
Collapse
|
23
|
Pfurr S, Chu YH, Bohrer C, Greulich F, Beattie R, Mammadzada K, Hils M, Arnold SJ, Taylor V, Schachtrup K, Uhlenhaut NH, Schachtrup C. The E2A splice variant E47 regulates the differentiation of projection neurons via p57(KIP2) during cortical development. Development 2017; 144:3917-3931. [PMID: 28939666 DOI: 10.1242/dev.145698] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 09/11/2017] [Indexed: 02/01/2023]
Abstract
During corticogenesis, distinct classes of neurons are born from progenitor cells located in the ventricular and subventricular zones, from where they migrate towards the pial surface to assemble into highly organized layer-specific circuits. However, the precise and coordinated transcriptional network activity defining neuronal identity is still not understood. Here, we show that genetic depletion of the basic helix-loop-helix (bHLH) transcription factor E2A splice variant E47 increased the number of Tbr1-positive deep layer and Satb2-positive upper layer neurons at E14.5, while depletion of the alternatively spliced E12 variant did not affect layer-specific neurogenesis. While ChIP-Seq identified a big overlap for E12- and E47-specific binding sites in embryonic NSCs, including sites at the cyclin-dependent kinase inhibitor (CDKI) Cdkn1c gene locus, RNA-Seq revealed a unique transcriptional regulation by each splice variant. E47 activated the expression of the CDKI Cdkn1c through binding to a distal enhancer. Finally, overexpression of E47 in embryonic NSCs in vitro impaired neurite outgrowth, and overexpression of E47 in vivo by in utero electroporation disturbed proper layer-specific neurogenesis and upregulated p57(KIP2) expression. Overall, this study identifies E2A target genes in embryonic NSCs and demonstrates that E47 regulates neuronal differentiation via p57(KIP2).
Collapse
Affiliation(s)
- Sabrina Pfurr
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany.,Faculty of Biology, University of Freiburg, Freiburg 79104, Germany
| | - Yu-Hsuan Chu
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany.,Faculty of Biology, University of Freiburg, Freiburg 79104, Germany
| | - Christian Bohrer
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany.,Faculty of Biology, University of Freiburg, Freiburg 79104, Germany
| | - Franziska Greulich
- Helmholtz Diabetes Center (HDC) and German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Robert Beattie
- Department of Biomedicine, Embryology and Stem Cell Biology, University of Basel, Basel 4058, Switzerland
| | - Könül Mammadzada
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany.,Faculty of Biology, University of Freiburg, Freiburg 79104, Germany
| | - Miriam Hils
- Faculty of Biology, University of Freiburg, Freiburg 79104, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Sebastian J Arnold
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany.,BIOSS Centre of Biological Signalling Studies, Albert-Ludwigs-University, Freiburg 79104, Germany
| | - Verdon Taylor
- Department of Biomedicine, Embryology and Stem Cell Biology, University of Basel, Basel 4058, Switzerland
| | - Kristina Schachtrup
- Faculty of Biology, University of Freiburg, Freiburg 79104, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - N Henriette Uhlenhaut
- Helmholtz Diabetes Center (HDC) and German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Christian Schachtrup
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| |
Collapse
|
24
|
The sound of tumor cell-microenvironment communication - composed by the Cancer Cluster Salzburg research network. Cell Commun Signal 2017; 15:20. [PMID: 28577565 PMCID: PMC5457625 DOI: 10.1186/s12964-017-0176-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 05/25/2017] [Indexed: 11/14/2022] Open
|
25
|
Liu P, Li X, Song F, Li P, Wei J, Yan Q, Xu X, Yang J, Li C, Fu X. Testosterone promotes tube formation of endothelial cells isolated from veins via activation of Smad1 protein. Mol Cell Endocrinol 2017; 446:21-31. [PMID: 28167128 DOI: 10.1016/j.mce.2017.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/11/2017] [Accepted: 02/02/2017] [Indexed: 01/03/2023]
Abstract
Testosterone (T) deficiency is positively correlated with the increased incidence of cardiovascular disease. However, the effects of T on vascular endothelial cells remain obscure. Tube formation capacity is critical for vascular regeneration/repair and Smad1 plays an important role in these events. In this study, we investigated the effects of T on Smad1 activation and tube formation of cultured human umbilical endothelial cells (HUVECs). Our results showed that T rapidly increased endothelial Smad1 phosphorylation. This effect was mimicked by cell-impermeable T-BSA conjugates and was not altered by transcriptional inhibitor actinomycin D or translational inhibitor cycloheximide. T-induced Smad1 phosphorylation was blocked by ERK1/2 and c-Src inhibitors or their specific siRNAs, while it was reinforced by ERK1/2 or c-Src overexpression. Indeed, T rapidly activated ERK1/2 and c-Src signalings and c-Src was confirmed as the upstream of ERK1/2. Moreover, caveolae disruptor methyl-β-cyclodextrin (β-MCD) blocked Smad1 activation induced by T. The association of caveolin-1 with androgen receptor (AR) or c-Src was detected by immunoprecipitation and it was significantly increased by rapid T stimulation. Furthermore, fractional analysis showed that AR and c-Src were expressed in caveolae-enriched membrane fractions. T promoted tube formation of HUVECs, which was blocked by c-Src and ERK1/2 inhibitors or by the knockdown of Smad1. In conclusion, T increased tube formation of endothelial cells isolated from veins by stimulating Smad1 phosphorylation in a nongenomic manner, which was mediated by signals from AR/c-Src located in caveolae to ERK1/2 cascade. These findings may shed new light on the relevance of T to its vascular functions.
Collapse
Affiliation(s)
- Pei Liu
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou City, Guangdong Province, 510630, China
| | - Xiaosa Li
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Fuhu Song
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou City, Guangdong Province, 510630, China
| | - Ping Li
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Jinzhi Wei
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Qing Yan
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Xingyan Xu
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Jun Yang
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Chuanxiang Li
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou City, Guangdong Province, 510630, China.
| | - Xiaodong Fu
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
26
|
Shen X, Yuan J, Zhang M, Li W, Ni B, Wu Y, Jiang L, Fan W, Tian Z. The increased expression of TCF3 is correlated with poor prognosis in Chinese patients with nasopharyngeal carcinoma. Clin Otolaryngol 2017; 42:824-830. [PMID: 28107608 DOI: 10.1111/coa.12834] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2016] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Regulatory factors controlling stem cell identity and self-renewal are often active in aggressive cancers and are thought to promote cancer growth and progression. B-cell-specific transcription factor 3 (TCF3/E2A) is a member of the T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription factor family that is central to regulating epidermal and embryonic stem cell identity. It has been reported that TCF3 was connected with the development and progression of a number of human cancers. In this study, we aimed to identify the expression of TCF3 in human nasopharyngeal carcinoma (NPC) and evaluate its clinical significance. DESIGN To investigate the expression of TCF3 in NPC and its relationship to prognosis. SETTING An in vitro study. MAIN OUTCOME MEASURES We analysed the expression of TCF3 in NPC and in non-tumourous nasopharyngeal tissues by quantitative RT-PCR and Western blotting. The expression patterns of TCF3 in 117 archived paraffin-embedded NPC specimens were characterised by immunohistochemistry, and the correlation between the TCF3 protein expression and the clinicopathological features of NPC was analysed. RESULTS We observed that TCF3 had a higher expression in NPC than in non-tumourous nasopharyngeal tissues of 117 archived paraffin-embedded NPC specimens, and 80 (68.4%) biopsy tissues revealed high levels of TCF3 expression. Furthermore, statistical analyses demonstrated that the increased expression of TCF3 was closely related to clinical stage, locoregional recurrence and distant metastasis of NPC. NPC patients with high levels of TCF3 expression had a shorter survival time, whereas patients with lower levels of TCF3 expression survived longer. Moreover, multivariate analysis suggested that the upregulation of TCF3 was a critical prognostic factor for NPC. CONCLUSIONS Our observations suggest, for the first time, that TCF3 is significantly associated with the development and progression of NPC, which can be used as an important prognostic marker for patients with NPC and may be an effective target for the treatment of NPC.
Collapse
Affiliation(s)
- X Shen
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China
| | - J Yuan
- Institute of Immunology PLA, Third Military Medical University, Chongqing, China
| | - M Zhang
- Institute of Immunology PLA, Third Military Medical University, Chongqing, China
| | - W Li
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - B Ni
- Institute of Immunology PLA, Third Military Medical University, Chongqing, China
| | - Y Wu
- Institute of Immunology PLA, Third Military Medical University, Chongqing, China
| | - L Jiang
- Department of Infectious Diseases, Southwestern Hospital, Third Military Medical University, Chongqing, China
| | - W Fan
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China
| | - Z Tian
- Institute of Immunology PLA, Third Military Medical University, Chongqing, China
| |
Collapse
|
27
|
Abstract
Inhibitors of DNA binding and cell differentiation (Id) proteins are members of the large family of the helix-loop-helix (HLH) transcription factors, but they lack any DNA-binding motif. During development, the Id proteins play a key role in the regulation of cell-cycle progression and cell differentiation by modulating different cell-cycle regulators both by direct and indirect mechanisms. Several Id-protein interacting partners have been identified thus far, which belong to structurally and functionally unrelated families, including, among others, the class I and II bHLH transcription factors, the retinoblastoma protein and related pocket proteins, the paired-box transcription factors, and the S5a subunit of the 26 S proteasome. Although the HLH domain of the Id proteins is involved in most of their protein-protein interaction events, additional motifs located in their N-terminal and C-terminal regions are required for the recognition of diverse protein partners. The ability of the Id proteins to interact with structurally different proteins is likely to arise from their conformational flexibility: indeed, these proteins contain intrinsically disordered regions that, in the case of the HLH region, undergo folding upon self- or heteroassociation. Besides their crucial role for cell-fate determination and cell-cycle progression during development, other important cellular events have been related to the Id-protein expression in a number of pathologies. Dysregulated Id-protein expression has been associated with tumor growth, vascularization, invasiveness, metastasis, chemoresistance and stemness, as well as with various developmental defects and diseases. Herein we provide an overview on the structural properties, mode of action, biological function and therapeutic potential of these regulatory proteins.
Collapse
Affiliation(s)
- Cornelia Roschger
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria
| | - Chiara Cabrele
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria.
| |
Collapse
|
28
|
Roschger C, Cabrele C. The Id-protein family in developmental and cancer-associated pathways. Cell Commun Signal 2017; 15:7. [PMID: 28122577 PMCID: PMC5267474 DOI: 10.1186/s12964-016-0161-y] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/29/2016] [Indexed: 01/15/2023] Open
Abstract
Inhibitors of DNA binding and cell differentiation (Id) proteins are members of the large family of the helix-loop-helix (HLH) transcription factors, but they lack any DNA-binding motif. During development, the Id proteins play a key role in the regulation of cell-cycle progression and cell differentiation by modulating different cell-cycle regulators both by direct and indirect mechanisms. Several Id-protein interacting partners have been identified thus far, which belong to structurally and functionally unrelated families, including, among others, the class I and II bHLH transcription factors, the retinoblastoma protein and related pocket proteins, the paired-box transcription factors, and the S5a subunit of the 26 S proteasome. Although the HLH domain of the Id proteins is involved in most of their protein-protein interaction events, additional motifs located in their N-terminal and C-terminal regions are required for the recognition of diverse protein partners. The ability of the Id proteins to interact with structurally different proteins is likely to arise from their conformational flexibility: indeed, these proteins contain intrinsically disordered regions that, in the case of the HLH region, undergo folding upon self- or heteroassociation. Besides their crucial role for cell-fate determination and cell-cycle progression during development, other important cellular events have been related to the Id-protein expression in a number of pathologies. Dysregulated Id-protein expression has been associated with tumor growth, vascularization, invasiveness, metastasis, chemoresistance and stemness, as well as with various developmental defects and diseases. Herein we provide an overview on the structural properties, mode of action, biological function and therapeutic potential of these regulatory proteins.
Collapse
Affiliation(s)
- Cornelia Roschger
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria
| | - Chiara Cabrele
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria.
| |
Collapse
|
29
|
Han C, Liao X, Qin W, Yu L, Liu X, Chen G, Liu Z, Lu S, Chen Z, Su H, Zhu G, Lu Z, Liu Z, Qin X, Gui Y, Mo Z, Li L, Peng T. EGFR and SYNE2 are associated with p21 expression and SYNE2 variants predict post-operative clinical outcomes in HBV-related hepatocellular carcinoma. Sci Rep 2016; 6:31237. [PMID: 27502069 PMCID: PMC4977508 DOI: 10.1038/srep31237] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 07/15/2016] [Indexed: 02/08/2023] Open
Abstract
This study was to explore the association between gene variants and p21 expression and investigate the TP53-independent p21 regulation in hepatitis B virus (HBV) related hepatocellular carcinoma (HCC) patients from Guangxi by genome-wide association study. 426 HBV-related HCC patients were enrolled. Results showed that, after quality control, a total of 21,643 SNPs were identified in 107 p21 positive and 298 p21 negative patients. The variants of epidermal growth factor receptor (EGFR; rs2227983 and rs6950826) and spectrin repeat containing, nuclear envelope 2 (SYNE2; rs8010699, rs4027405 and rs1890908) were associated with p21 expression. Moreover the haplotype block (rs2227983 and rs6950826, r2 = 0.378) in EGFR and the haplotype block in SYNE2 (rs8010699 was in strong LD with rs4027405 and rs1890908 (r2 = 0.91 and 0.70, respectively)) were identified, and the haplotype A-G of EGFR and haplotype G-A-A of SYNE2 were significantly associated with p21 expression (P < 0.01). rs4027405 and rs1890908 were significantly associated with overall survival, and patients with AG/GG genotypes of SYNE2 gene had a worse overall survival (P = 0.001, P = 0.002). Our findings indicate that variants of EGFR and SYNE2 play an important role in p21 regulation and are associated with the clinical outcome of HBV-related HCC in a TP53-indenpdent manner.
Collapse
Affiliation(s)
- Chuangye Han
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Wei Qin
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Long Yu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Xiaoguang Liu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Gang Chen
- Department of Pathology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Zhengtao Liu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Sicong Lu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Zhiwei Chen
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Hao Su
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Guangzhi Zhu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Zili Lu
- Department of Pathology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Zhiming Liu
- Department of General Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Xue Qin
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Ying Gui
- Department of Clinical laboratory center, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Lequn Li
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Tao Peng
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| |
Collapse
|
30
|
Augeri DJ, Langenfeld E, Castle M, Gilleran JA, Langenfeld J. Inhibition of BMP and of TGFβ receptors downregulates expression of XIAP and TAK1 leading to lung cancer cell death. Mol Cancer 2016; 15:27. [PMID: 27048361 PMCID: PMC4822253 DOI: 10.1186/s12943-016-0511-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 03/23/2016] [Indexed: 02/07/2023] Open
Abstract
Background Bone morphogenetic proteins (BMP) are embryonic proteins that are part of the transforming growth factor (TGFβ) superfamily, which are aberrantly expressed in many carcinomas. Inhibition of BMP receptors with small molecule inhibitors decreases growth and induces death of lung cancer cells, which involves the downregulation of Id1 and Id3 by a Smad dependent mechanism. Developmentally, BMP and TGFβ signaling utilizes Smad-1/5 independent mechanisms to stabilize the expression of X-linked inhibitor of apoptosis protein (XIAP) and activate TGFβ activated kinase 1 (TAK1), which are known to be potent inhibitors of apoptosis. The role of BMP signaling in regulating XIAP and TAK1 in cancer cells is poorly understood. Furthermore, the interaction between the BMP and TGFβ signaling cascades in regulating the activation of TAK1 in cancer cells has not been elucidated. Methods Feedback regulation between the BMP and TGFβ signaling pathways and their regulation of XIAP, TAK1, and Id1 were examined in lung cancer cells utilizing siRNA and inhibitors targeting BMP type I receptors, inhibitors of BMP and TGFβ type I receptors, and an inhibitor of BMP and TGFβ type I and type II receptors. Results We show that upon inhibition of BMP signaling in lung cancer cells, the TGFβ signaling cascade is activated. Both the BMP and TGFβ pathways activate TAK1, which then increases the expression of Id1. Inhibition of TGFβ signaling increased Id1 expression except when BMP signaling is suppressed, which then causes a dose-related decrease in the expression of Id1. Inhibition of both BMP and TGFβ signaling enhances the downregulation of TAK1. Our data also suggests that the blockade of the BMP type II receptor enhances the downregulation XIAP, which is important in decreasing the activity of TAK1. Knockdown studies demonstrate that both XIAP and TAK1 regulate the survival of lung cancer cells. Conclusions This paper highlights that targeting the BMP and TGFβ type I and type II receptors causes a downregulation of XIAP, TAK1, and Id1 leading to cell death of lung cancer cells. Small molecule inhibitors targeting the BMP and TGFβ receptors represents a potential novel means to treat cancer patients. Electronic supplementary material The online version of this article (doi:10.1186/s12943-016-0511-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dave J Augeri
- Rutgers Translational Sciences, Department of Medicinal Chemistry, School of Pharmacy, New Brunswick, NJ, USA
| | - Elaine Langenfeld
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, MEB 536, One Robert Wood Johnson Place, P.O. Box 19, New Brunswick, NJ, 08903-0019, USA
| | - Monica Castle
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, MEB 536, One Robert Wood Johnson Place, P.O. Box 19, New Brunswick, NJ, 08903-0019, USA
| | - John A Gilleran
- Rutgers Translational Sciences, Department of Medicinal Chemistry, School of Pharmacy, New Brunswick, NJ, USA
| | - John Langenfeld
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, MEB 536, One Robert Wood Johnson Place, P.O. Box 19, New Brunswick, NJ, 08903-0019, USA.
| |
Collapse
|
31
|
The Wnt/β-catenin signaling/Id2 cascade mediates the effects of hypoxia on the hierarchy of colorectal-cancer stem cells. Sci Rep 2016; 6:22966. [PMID: 26965643 PMCID: PMC4786801 DOI: 10.1038/srep22966] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 02/22/2016] [Indexed: 01/03/2023] Open
Abstract
Hypoxia, a feature common to most solid tumors, is known to regulate many aspects of tumorigenesis. Recently, it was suggested that hypoxia increased the size of the cancer stem-cell (CSC) subpopulations and promoted the acquisition of a CSC-like phenotype. However, candidate hypoxia-regulated mediators specifically relevant to the stemness-related functions of colorectal CSCs have not been examined in detail. In the present study, we showed that hypoxia specifically promoted the self-renewal potential of CSCs. Through various in vitro studies, we found that hypoxia-induced Wnt/β-catenin signaling increased the occurrence of CSC-like phenotypes and the level of Id2 expression in colorectal-cancer cells. Importantly, the levels of hypoxia-induced CSC-sphere formation and Id2 expression were successfully attenuated by treatment with a Wnt/β-catenin-signaling inhibitor. We further demonstrated, for the first time, that the degree of hypoxia-induced CSC-sphere formation (CD44(+) subpopulation) in vitro and of tumor metastasis/dissemination in vivo were markedly suppressed by knocking down Id2 expression. Taken together, these data suggested that Wnt/β-catenin signaling mediated the hypoxia-induced self-renewal potential of colorectal-cancer CSCs through reactivating Id2 expression.
Collapse
|
32
|
Wang LH, Baker NE. E Proteins and ID Proteins: Helix-Loop-Helix Partners in Development and Disease. Dev Cell 2016; 35:269-80. [PMID: 26555048 DOI: 10.1016/j.devcel.2015.10.019] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 10/18/2015] [Accepted: 10/23/2015] [Indexed: 01/12/2023]
Abstract
The basic Helix-Loop-Helix (bHLH) proteins represent a well-known class of transcriptional regulators. Many bHLH proteins act as heterodimers with members of a class of ubiquitous partners, the E proteins. A widely expressed class of inhibitory heterodimer partners-the Inhibitor of DNA-binding (ID) proteins-also exists. Genetic and molecular analyses in humans and in knockout mice implicate E proteins and ID proteins in a wide variety of diseases, belying the notion that they are non-specific partner proteins. Here, we explore relationships of E proteins and ID proteins to a variety of disease processes and highlight gaps in knowledge of disease mechanisms.
Collapse
Affiliation(s)
- Lan-Hsin Wang
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Nicholas E Baker
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| |
Collapse
|
33
|
Sullivan JM, Havrda MC, Kettenbach AN, Paolella BR, Zhang Z, Gerber SA, Israel MA. Phosphorylation Regulates Id2 Degradation and Mediates the Proliferation of Neural Precursor Cells. Stem Cells 2016; 34:1321-31. [PMID: 26756672 DOI: 10.1002/stem.2291] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/23/2015] [Accepted: 12/07/2015] [Indexed: 01/07/2023]
Abstract
Inhibitor of DNA binding proteins (Id1-Id4) function to inhibit differentiation and promote proliferation of many different cell types. Among the Id family members, Id2 has been most extensively studied in the central nervous system (CNS). Id2 contributes to cultured neural precursor cell (NPC) proliferation as well as to the proliferation of CNS tumors such as glioblastoma that are likely to arise from NPC-like cells. We identified three phosphorylation sites near the N-terminus of Id2 in NPCs. To interrogate the importance of Id2 phosphorylation, Id2(-/-) NPCs were modified to express wild type (WT) Id2 or an Id2 mutant protein that could not be phosphorylated at the identified sites. We observed that NPCs expressing this mutant lacking phosphorylation near the N-terminus had higher steady-state levels of Id2 when compared to NPCs expressing WT Id2. This elevated level was the result of a longer half-life and reduced proteasome-mediated degradation. Moreover, NPCs expressing constitutively de-phosphorylated Id2 proliferated more rapidly than NPCs expressing WT Id2, a finding consistent with the well-characterized function of Id2 in driving proliferation. Observing that phosphorylation of Id2 modulates the degradation of this important cell-cycle regulator, we sought to identify a phosphatase that would stabilize Id2 enhancing its activity in NPCs and extended our analysis to include human glioblastoma-derived stem cells (GSCs). We found that expression of the phosphatase PP2A altered Id2 levels. Our findings suggest that inhibition of PP2A may be a novel strategy to regulate the proliferation of normal NPCs and malignant GSCs by decreasing Id2 levels. Stem Cells 2016;34:1321-1331.
Collapse
Affiliation(s)
- Jaclyn M Sullivan
- Pharmacology and Toxicology, Norris Cotton Cancer Center, One Medical Center Drive, Lebanon, NH, 03756.,Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Matthew C Havrda
- Pharmacology and Toxicology, Norris Cotton Cancer Center, One Medical Center Drive, Lebanon, NH, 03756.,Department of Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Arminja N Kettenbach
- Pharmacology and Toxicology, Norris Cotton Cancer Center, One Medical Center Drive, Lebanon, NH, 03756.,Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Brenton R Paolella
- Pharmacology and Toxicology, Norris Cotton Cancer Center, One Medical Center Drive, Lebanon, NH, 03756.,Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Zhonghua Zhang
- Pharmacology and Toxicology, Norris Cotton Cancer Center, One Medical Center Drive, Lebanon, NH, 03756.,Department of Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Scott A Gerber
- Pharmacology and Toxicology, Norris Cotton Cancer Center, One Medical Center Drive, Lebanon, NH, 03756.,Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA.,Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Mark A Israel
- Pharmacology and Toxicology, Norris Cotton Cancer Center, One Medical Center Drive, Lebanon, NH, 03756.,Department of Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA.,Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|
34
|
Qiu L, Wu J, Pan C, Tan X, Lin J, Liu R, Chen S, Geng R, Huang W. Downregulation of CDC27 inhibits the proliferation of colorectal cancer cells via the accumulation of p21Cip1/Waf1. Cell Death Dis 2016; 7:e2074. [PMID: 26821069 PMCID: PMC4816181 DOI: 10.1038/cddis.2015.402] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/01/2015] [Accepted: 12/03/2015] [Indexed: 12/14/2022]
Abstract
Dysregulated cell cycle progression has a critical role in tumorigenesis. Cell division cycle 27 (CDC27) is a core subunit of the anaphase-promoting complex/cyclosome, although the specific role of CDC27 in cancer remains unknown. In our study, we explored the biological and clinical significance of CDC27 in colorectal cancer (CRC) growth and progression and investigated the underlying molecular mechanisms. Results showed that CDC27 expression is significantly correlated with tumor progression and poor patient survival. Functional assays demonstrated that overexpression of CDC27 promoted proliferation in DLD1 cells, whereas knockdown of CDC27 in HCT116 cells inhibited proliferation both in vitro and in vivo. Further mechanistic investigation showed that CDC27 downregulation resulted in G1/S phase transition arrest via the significant accumulation of p21 in HCT116 cells, and the upregulation of CDC27 promoted G1/S phase transition via the attenuation of p21 in DLD1 cells. Furthermore, we also demonstrated that CDC27 regulated inhibitor of DNA binding 1 (ID1) protein expression in DLD1 and HCT116 cells, and rescue assays revealed that CDC27 regulated p21 expression through modulating ID1 expression. Taken together, our results indicate that CDC27 contributes to CRC cell proliferation via the modulation of ID1-mediated p21 regulation, which offers a novel approach to the inhibition of tumor growth. Indeed, these findings provide new perspectives for the future study of CDC27 as a target for CRC treatment.
Collapse
Affiliation(s)
- L Qiu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - J Wu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - C Pan
- Medical Oncology, Sichuan Cancer Hospital and Institute, Second People's Hospital of Sichuan Province, Chengdu, China
| | - X Tan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - J Lin
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - R Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - S Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - R Geng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - W Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
35
|
Wu CC, Kruse F, Vasudevarao MD, Junker JP, Zebrowski DC, Fischer K, Noël ES, Grün D, Berezikov E, Engel FB, van Oudenaarden A, Weidinger G, Bakkers J. Spatially Resolved Genome-wide Transcriptional Profiling Identifies BMP Signaling as Essential Regulator of Zebrafish Cardiomyocyte Regeneration. Dev Cell 2015; 36:36-49. [PMID: 26748692 DOI: 10.1016/j.devcel.2015.12.010] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 10/14/2015] [Accepted: 11/20/2015] [Indexed: 12/21/2022]
Abstract
In contrast to mammals, zebrafish regenerate heart injuries via proliferation of cardiomyocytes located near the wound border. To identify regulators of cardiomyocyte proliferation, we used spatially resolved RNA sequencing (tomo-seq) and generated a high-resolution genome-wide atlas of gene expression in the regenerating zebrafish heart. Interestingly, we identified two wound border zones with distinct expression profiles, including the re-expression of embryonic cardiac genes and targets of bone morphogenetic protein (BMP) signaling. Endogenous BMP signaling has been reported to be detrimental to mammalian cardiac repair. In contrast, we find that genetic or chemical inhibition of BMP signaling in zebrafish reduces cardiomyocyte dedifferentiation and proliferation, ultimately compromising myocardial regeneration, while bmp2b overexpression is sufficient to enhance it. Our results provide a resource for further studies on the molecular regulation of cardiac regeneration and reveal intriguing differential cellular responses of cardiomyocytes to a conserved signaling pathway in regenerative versus non-regenerative hearts.
Collapse
Affiliation(s)
- Chi-Chung Wu
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Fabian Kruse
- Hubrecht Institute, University Medical Centre Utrecht, Uppsalaan 8, 3584 CT Utrecht, the Netherlands
| | | | - Jan Philipp Junker
- Hubrecht Institute, University Medical Centre Utrecht, Uppsalaan 8, 3584 CT Utrecht, the Netherlands
| | - David C Zebrowski
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstr 8-10, 91054 Erlangen, Germany
| | - Kristin Fischer
- Institute of Clinical Genetics, Technische Universität Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Emily S Noël
- Hubrecht Institute, University Medical Centre Utrecht, Uppsalaan 8, 3584 CT Utrecht, the Netherlands
| | - Dominic Grün
- Hubrecht Institute, University Medical Centre Utrecht, Uppsalaan 8, 3584 CT Utrecht, the Netherlands
| | - Eugene Berezikov
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands; Skolkovo Institute of Science and Technology (Skoltech), Novaya Street 100, Skolkovo, Moscow Region 143025, Russia
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstr 8-10, 91054 Erlangen, Germany
| | - Alexander van Oudenaarden
- Hubrecht Institute, University Medical Centre Utrecht, Uppsalaan 8, 3584 CT Utrecht, the Netherlands
| | - Gilbert Weidinger
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany.
| | - Jeroen Bakkers
- Hubrecht Institute, University Medical Centre Utrecht, Uppsalaan 8, 3584 CT Utrecht, the Netherlands; Medical Physiology, University Medical Centre Utrecht, Yalelaan 50, 3584 CM Utrecht, the Netherlands.
| |
Collapse
|
36
|
Kaplan JL, Marshall MA, C. McSkimming C, Harmon DB, Garmey JC, Oldham SN, Hallowell P, McNamara CA. Adipocyte progenitor cells initiate monocyte chemoattractant protein-1-mediated macrophage accumulation in visceral adipose tissue. Mol Metab 2015; 4:779-94. [PMID: 26629403 PMCID: PMC4632113 DOI: 10.1016/j.molmet.2015.07.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 07/27/2015] [Accepted: 07/30/2015] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE Macrophages are important producers of obesity-induced MCP-1; however, initial obesity-induced increases in MCP-1 production precede M1 macrophage accumulation in visceral adipose tissue (VAT). The initial cellular source of obesity-induced MCP-1 in vivo is currently unknown. Preliminary reports based on in vitro studies of preadipocyte cell lines and adherent stroma-vascular fraction cells suggest that resident stromal cells express MCP-1. In the past several years, elegant methods of identifying adipocyte progenitor cells (AdPCs) have become available, making it possible to study these cells in vivo. We have previously published that global deletion of transcription factor Inhibitor of Differentiation 3 (Id3) attenuates high fat diet-induced obesity, but it is unclear if Id3 plays a role in diet-induced MCP-1 production. We sought to determine the initial cellular source of MCP-1 and identify molecular regulators mediating MCP-1 production. METHODS Id3 (+/+) and Id3 (-/-) mice were fed either a standard chow or HFD for varying lengths of time. Flow cytometry, semi-quantitative real-time PCR, ELISAs and adoptive transfers were used to assess the importance of AdPCs during diet-induced obesity. Flow cytometry was also performed on a cohort of 14 patients undergoing bariatric surgery. RESULTS Flow cytometry identified committed CD45(-)CD31 (-) Ter119(-)CD29(+)CD34(+)Sca-1(+)CD24(-) adipocyte progenitor cells as producers of high levels of MCP-1 in VAT. High-fat diet increased AdPC numbers, an effect dependent on Id3. Loss of Id3 increased p21(Cip1) levels and attenuated AdPC proliferation, resulting in reduced MCP-1 and M1 macrophage accumulation in VAT, compared to Id3 (+/+) littermate controls. AdPC rescue by adoptive transfer of 50,000 Id3 (+/+) AdPCs into Id3 (-/-) recipient mice increased MCP-1 levels and M1 macrophage number in VAT. Additionally, flow cytometry identified MCP-1-producing CD45(-)CD31(-)CD34(+)CD44(+)CD90(+) AdPCs in human omental and subcutaneous adipose tissue, with a higher percentage in omental adipose. Furthermore, high surface expression of CD44 marked abundant MCP-1 producers, only in visceral adipose tissue. CONCLUSIONS This study provides the first in vivo evidence, to our knowledge, that committed AdPCs in VAT are the initial source of obesity-induced MCP-1 and identifies the helix-loop-helix transcription factor Id3 as a critical regulator of p21(Cip1) expression, AdPC proliferation, MCP-1 expression and M1 macrophage accumulation in VAT. Inhibition of Id3 and AdPC expansion, as well as CD44 expression in human AdPCs, may serve as unique therapeutic targets for the regulation of adipose tissue inflammation.
Collapse
Affiliation(s)
- Jennifer L. Kaplan
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
| | - Melissa A. Marshall
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Chantel C. McSkimming
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Daniel B. Harmon
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
- Department of Biochemistry, Molecular Biology, and Genetics, University of Virginia, Charlottesville, VA, United States
| | - James C. Garmey
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Stephanie N. Oldham
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Peter Hallowell
- Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Coleen A. McNamara
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
- Department of Medicine, Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA, United States
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
37
|
Patel D, Chinaranagari S, Chaudhary J. Basic helix loop helix (bHLH) transcription factor 3 (TCF3, E2A) is regulated by androgens in prostate cancer cells. Am J Cancer Res 2015; 5:3407-3421. [PMID: 26807321 PMCID: PMC4697687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/06/2015] [Indexed: 06/05/2023] Open
Abstract
TCF3 (E2A) is a multifunctional basic helix loop helix (bHLH) transcription factor that is over-expressed in prostate cancer (PCa) as compared to normal prostate and that it acts as a tumor promoter in PCa. Given the diverse biological pathways regulated/influenced by TCF3, little is known about the mechanisms that regulate its expression. TCF3 expression in androgen sensitive LNCaP and insensitive C81 PCa cell lines was determined following treatments with androgen receptor (AR) agonist R1881 and antagonist Casodex. In silico analysis was used to discover putative Androgen Response Elements (ARE) in the TCF3 promoter/intron region. Chromatin Immunoprecipitation (ChIP) with AR antibody and luciferase reporter assays on the above mentioned cell lines was used to confirm AR biding and AR dependent transcriptional activity respectively. The results were confirmed by demonstrating TCF3 expression in LNCaP PCa xenograft models. The results suggested that TCF3 transcript increased in response to R1881 in LNCaP cells but was constitutively expressed in C-81 cell lines. The promoter/Intron region of the TCF3 gene was predicted to contain two putative ARE sites ARE1 and ARE2. ChIP after treatment of LNCaP and C81 cells with R1881 and Casodex showed that the ARE1 and ARE2 were bound by AR in LNCaP cells only in the presence of R1881, whereas C81 cells showed constitutive AR binding. Similar results were observed in luciferase reporter assays indicating that TCF3 is activated by AR in LNCaP cell lines whereas it is independent of androgens in C81 cell line. Luciferase reporter assays also confirmed that ARE1 alone drives androgen dependent transcription. TCF3 expression was only observed in castration resistant LNCaP xenografts in castrated mice. In conclusion, we demonstrate that in PCa androgen receptor regulates the expression of TCF3 which is mediated in part via a consensus androgen response element. The shift in TCF3 expression from androgen regulated to androgen independent during prostate cancer progression, together with lack of expression in normal prostate may provide mechanistic basis underlying the transition of androgen receptor from a tumor suppressor to an oncogene in prostate cancer.
Collapse
Affiliation(s)
- Divya Patel
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University Atlanta, GA, USA
| | - Swathi Chinaranagari
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University Atlanta, GA, USA
| | - Jaideep Chaudhary
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University Atlanta, GA, USA
| |
Collapse
|
38
|
Liu G, Wu D, Liang X, Yue H, Cui Y. Mechanisms and in vitro effects of cepharanthine hydrochloride: Classification analysis of the drug-induced differentially-expressed genes of human nasopharyngeal carcinoma cells. Oncol Rep 2015; 34:2002-10. [PMID: 26260412 DOI: 10.3892/or.2015.4193] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 06/11/2015] [Indexed: 11/06/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is the most commonly diagnosed head and neck malignancy and is prevalent worldwide. Previous studies have demonstrated the antitumor properties of cepharanthine hydrochloride (CH) in several human cancer cells. However, the action of CH in NPC cells has yet to be determined. In the present study, we investigated the effects of CH in human NPC cell lines including CNE-1 and CNE-2 on cell growth and apoptosis in vitro. Using MTT and ATP-tumor chemosensitivity assays it was found that CH inhibited cell viability. Additionally, flow cytometric and analysis electron microscopy revealed the inhibition of cell cycle progression and reduction of apoptosis, respectively, in human NPC cell lines including CNE-1 and CNE-2 in vitro. To identify the potential action mechanisms of CH, the cDNA microarray analysis results were confirmed by quantitative PCR analysis using a number of genes, including CDKN1A/P21, NR4A1/TR3 and DAXX. In total, 138 upregulated and 63 downregulated genes in CNE-2 cells were treated with CH. According to their biological function, the genes were classified as: i) cell cycle-related genes; ii) DNA repair‑related genes; iii) apoptosis-related genes and iv) nuclear factor-κB (NF-κB) transcription factors signal pathways. The results of the present study showed that CH is a potential therapeutic agent against human NPC, and provide rational explanations and a scientific basis for the study of the development of CH in the treatment of NPC.
Collapse
Affiliation(s)
- Guanjun Liu
- Department of Medical Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Dongmei Wu
- Department of Medical Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xinqiang Liang
- Department of Medical Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Huifen Yue
- Department of Medical Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Ying Cui
- Department of Medical Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
39
|
Hypoxia accelerates vascular repair of endothelial colony-forming cells on ischemic injury via STAT3-BCL3 axis. Stem Cell Res Ther 2015. [PMID: 26219963 PMCID: PMC4522108 DOI: 10.1186/s13287-015-0128-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Introduction Endothelial colony-forming cells (ECFCs) significantly improve tissue repair by providing regeneration potential within injured cardiovascular tissue. However, ECFC transplantation into ischemic tissue exhibits limited therapeutic efficacy due to poor engraftment in vivo. We established an adequate ex vivo expansion protocol and identified novel modulators that enhance functional bioactivities of ECFCs. Methods To augment the regenerative potential of ECFCs, functional bioactivities of hypoxia-preconditioned ECFCs (hypo-ECFCs) were examined. Results Phosphorylations of the JAK2/STAT3 pathway and clonogenic proliferation were enhanced by short-term ECFC culturing under hypoxia, whereas siRNA-targeting of STAT3 significantly reduced these activities. Expression of BCL3, a target molecule of STAT3, was increased in hypo-ECFCs. Moreover, siRNA inhibition of BCL3 markedly reduced survival of ECFCs during hypoxic stress in vitro and ischemic stress in vivo. In a hindlimb ischemia model of ischemia, hypo-ECFC transplantation enhanced blood flow ratio, capillary density, transplanted cell proliferation and survival, and angiogenic cytokine secretion at ischemic sites. Conclusions Hypoxia preconditioning facilitates functional bioactivities of ECFCs by mediating regulation of the STAT3-BCL3 axis. Thus, a hypoxic preconditioned ex vivo expansion protocol triggers expansion and functional bioactivities of ECFCs via modulation of the hypoxia-induced STAT3-BCL3 axis, suggesting that hypo-ECFCs offer a therapeutic strategy for accelerated neovasculogenesis in ischemic diseases.
Collapse
|
40
|
Hirano T, Murakami T, Ono H, Sakurai A, Tominaga T, Takahashi T, Nagai K, Doi T, Abe H. A Novel Interaction between FLICE-Associated Huge Protein (FLASH) and E2A Regulates Cell Proliferation and Cellular Senescence via Tumor Necrosis Factor (TNF)-Alpha-p21WAF1/CIP1 Axis. PLoS One 2015. [PMID: 26208142 PMCID: PMC4514670 DOI: 10.1371/journal.pone.0133205] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Dysregulation of the cell proliferation has been implicated in the pathophysiology of a number of diseases. Cellular senescence limits proliferation of cancer cells, preventing tumorigenesis and restricting tissue damage. However, the role of cellular senescence in proliferative nephritis has not been determined. The proliferative peak in experimental rat nephritis coincided with a peak in E2A expression in the glomeruli. Meanwhile, E12 (an E2A-encoded transcription factor) did not promote proliferation of Mesangial cells (MCs) by itself. We identified caspase-8-binding protein FLICE-associated huge protein (FLASH) as a novel E2A-binding partner by using a yeast two-hybrid screening. Knockdown of FLASH suppressed proliferation of MCs. This inhibitory effect was partially reversed by the knockdown of E2A. In addition, the knockdown of FLASH induced cyclin-dependent kinase inhibitor p21WAF1/CIP1 (p21) expression, but did not affect p53 expression. Furthermore, overexpression of E12 and E47 induced p21, but not p53 in MCs, in the absence of FLASH. We also demonstrated that E2A and p21 expression at the peak of proliferation was followed by significant induction of FLASH in mesangial areas in rat proliferative glomerulonephritis. Moreover, we revealed that FLASH negatively regulates cellular senescence via the interaction with E12. We also demonstrated that FLASH is involved in the TNF-α-induced p21 expressions. These results suggest that the functional interaction of E2A and FLASH play an important role in cell proliferation and cellular senescence via regulation of p21 expression in experimental glomerulonephritis.
Collapse
Affiliation(s)
- Takahiro Hirano
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Taichi Murakami
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Hiroyuki Ono
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Akiko Sakurai
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Tatsuya Tominaga
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Toshikazu Takahashi
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Kojiro Nagai
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Toshio Doi
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Hideharu Abe
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
- * E-mail:
| |
Collapse
|
41
|
The basic helix-loop-helix transcription factor E47 reprograms human pancreatic cancer cells to a quiescent acinar state with reduced tumorigenic potential. Pancreas 2015; 44:718-27. [PMID: 25894862 PMCID: PMC4464938 DOI: 10.1097/mpa.0000000000000328] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Pancreatic ductal adenocarcinoma (PDA) initiates from quiescent acinar cells that attain a Kras mutation, lose signaling from basic helix-loop-helix (bHLH) transcription factors, undergo acinar-ductal metaplasia, and rapidly acquire increased growth potential. We queried whether PDA cells can be reprogrammed to revert to their original quiescent acinar cell state by shifting key transcription programs. METHODS Human PDA cell lines were engineered to express an inducible form of the bHLH protein E47. Gene expression, growth, and functional studies were investigated using microarray, quantitative polymerase chain reaction, immunoblots, immunohistochemistry, small interfering RNA, chromatin immunoprecipitation analyses, and cell transplantation into mice. RESULTS In human PDA cells, E47 activity triggers stable G0/G1 arrest, which requires the cyclin-dependent kinase inhibitor p21 and the stress response protein TP53INP1. Concurrently, E47 induces high level expression of acinar digestive enzymes and feed forward activation of the acinar maturation network regulated by the bHLH factor MIST1. Moreover, induction of E47 in human PDA cells in vitro is sufficient to inhibit tumorigenesis. CONCLUSIONS Human PDA cells retain a high degree of plasticity, which can be exploited to induce a quiescent acinar cell state with reduced tumorigenic potential. Moreover, bHLH activity is a critical node coordinately regulating human PDA cell growth versus cell fate.
Collapse
|
42
|
Sharma P, Chinaranagari S, Chaudhary J. Inhibitor of differentiation 4 (ID4) acts as an inhibitor of ID-1, -2 and -3 and promotes basic helix loop helix (bHLH) E47 DNA binding and transcriptional activity. Biochimie 2015; 112:139-50. [PMID: 25778840 DOI: 10.1016/j.biochi.2015.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 03/05/2015] [Indexed: 01/15/2023]
Abstract
The four known ID proteins (ID1-4, Inhibitor of Differentiation) share a homologous helix loop helix (HLH) domain and act as dominant negative regulators of basic-HLH transcription factors. ID proteins also interact with many non-bHLH proteins in complex networks. The expression of ID proteins is increasingly observed in many cancers. Whereas ID-1, ID-2 and ID-3, are generally considered as tumor promoters, ID4 on the contrary has emerged as a tumor suppressor. In this study we demonstrate that ID4 heterodimerizes with ID-1, -2 and -3 and promote bHLH DNA binding, essentially acting as an inhibitor of inhibitors of differentiation proteins. Interaction of ID4 was observed with ID1, ID2 and ID3 that was dependent on intact HLH domain of ID4. Interaction with bHLH protein E47 required almost 3 fold higher concentration of ID4 as compared to ID1. Furthermore, inhibition of E47 DNA binding by ID1 was restored by ID4 in an EMSA binding assay. ID4 and ID1 were also colocalized in prostate cancer cell line LNCaP. The alpha helix forming alanine stretch N-terminal, unique to HLH ID4 domain was required for optimum interaction. Ectopic expression of ID4 in DU145 prostate cancer line promoted E47 dependent expression of CDKNI p21. Thus counteracting the biological activities of ID-1, -2 and -3 by forming inactive heterodimers appears to be a novel mechanism of action of ID4. These results could have far reaching consequences in developing strategies to target ID proteins for cancer therapy and understanding biologically relevant ID-interactions.
Collapse
Affiliation(s)
- Pankaj Sharma
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - Swathi Chinaranagari
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - Jaideep Chaudhary
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA.
| |
Collapse
|
43
|
Fischer B, Azim K, Hurtado-Chong A, Ramelli S, Fernández M, Raineteau O. E-proteins orchestrate the progression of neural stem cell differentiation in the postnatal forebrain. Neural Dev 2014; 9:23. [PMID: 25352248 PMCID: PMC4274746 DOI: 10.1186/1749-8104-9-23] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/08/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neural stem cell (NSC) differentiation is a complex multistep process that persists in specific regions of the postnatal forebrain and requires tight regulation throughout life. The transcriptional control of NSC proliferation and specification involves Class II (proneural) and Class V (Id1-4) basic helix-loop-helix (bHLH) proteins. In this study, we analyzed the pattern of expression of their dimerization partners, Class I bHLH proteins (E-proteins), and explored their putative role in orchestrating postnatal subventricular zone (SVZ) neurogenesis. RESULTS Overexpression of a dominant-negative form of the E-protein E47 (dnE47) confirmed a crucial role for bHLH transcriptional networks in postnatal neurogenesis by dramatically blocking SVZ NSC differentiation. In situ hybridization was used in combination with RT-qPCR to measure and compare the level of expression of E-protein transcripts (E2-2, E2A, and HEB) in the neonatal and adult SVZ as well as in magnetic affinity cell sorted progenitor cells and neuroblasts. Our results evidence that E-protein transcripts, in particular E2-2 and E2A, are enriched in the postnatal SVZ with expression levels increasing as cells engage towards neuronal differentiation. To investigate the role of E-proteins in orchestrating lineage progression, both in vitro and in vivo gain-of-function and loss-of-function experiments were performed for individual E-proteins. Overexpression of E2-2 and E2A promoted SVZ neurogenesis by enhancing not only radial glial cell differentiation but also cell cycle exit of their progeny. Conversely, knock-down by shRNA electroporation resulted in opposite effects. Manipulation of E-proteins and/or Ascl1 in SVZ NSC cultures indicated that those effects were Ascl1 dependent, although they could not solely be attributed to an Ascl1-induced switch from promoting cell proliferation to triggering cell cycle arrest and differentiation. CONCLUSIONS In contrast to former concepts, suggesting ubiquitous expression and subsidiary function for E-proteins to foster postnatal neurogenesis, this work unveils E-proteins as being active players in the orchestration of postnatal SVZ neurogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | - Olivier Raineteau
- Brain Research Institute, ETH Zurich/University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
44
|
Yang J, Li X, Morrell NW. Id proteins in the vasculature: from molecular biology to cardiopulmonary medicine. Cardiovasc Res 2014; 104:388-98. [PMID: 25274246 DOI: 10.1093/cvr/cvu215] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The inhibitors of differentiation (Id) proteins belong to the helix-loop-helix group of transcription factors and regulate cell differentiation and proliferation. Recent studies have reported that Id proteins play important roles in cardiogenesis and formation of the vasculature. We have also demonstrated that heritable pulmonary arterial hypertension (HPAH) patients have dysregulated Id gene expression in pulmonary artery smooth muscle cells. The interaction between bone morphogenetic proteins and other growth factors or cytokines regulates Id gene expression, which impacts on pulmonary vascular cell differentiation and proliferation. Exploration of the roles of Id proteins in vascular remodelling that occurs in PAH and atherosclerosis might provide new insights into the molecular basis of these diseases. In addition, current progress in identification of the interactors of Id proteins will further the understanding of the function of Ids in vascular cells and enable the identification of novel targets for therapy in PAH and other cardiovascular diseases.
Collapse
Affiliation(s)
- Jun Yang
- Department of Cell Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 DongdanSantiao, Beijing 100005, China
| | - Xiaohui Li
- Department of Pharmacology, School of Pharmaceutical Science, Central South University, Changsha, China
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Level 5, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| |
Collapse
|
45
|
Patel D, Knowell AE, Korang-Yeboah M, Sharma P, Joshi J, Glymph S, Chinaranagari S, Nagappan P, Palaniappan R, Bowen NJ, Chaudhary J. Inhibitor of differentiation 4 (ID4) inactivation promotes de novo steroidogenesis and castration-resistant prostate cancer. Mol Endocrinol 2014; 28:1239-53. [PMID: 24921661 DOI: 10.1210/me.2014-1100] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Prostate cancer (PCa) is the most commonly diagnosed cancer in men in the Western world. The transition of androgen-dependent PCa to castration-resistant (CRPC) is a major clinical manifestation during disease progression and presents a therapeutic challenge. Our studies have shown that genetic ablation of inhibitor of differentiation 4 (Id4), a dominant-negative helix loop helix protein, in mice results in prostatic intraepithelial neoplasia lesions and decreased Nkx3.1 expression without the loss of androgen receptor (Ar) expression. ID4 is also epigenetically silenced in the majority of PCa. However, the clinical relevance and molecular pathways altered by ID4 inactivation in PCa are not known. This study investigates the effect of loss of ID4 in PCa cell lines on tumorigenicity and addresses the underlying mechanism. Stable silencing of ID4 in LNCaP cells (L-ID4) resulted in increased proliferation, migration, invasion, and anchorage-independent growth. An increase in the rate of tumor growth, weight, and volume was observed in L-ID4 xenografts compared with that in the LNCaP cells transfected with nonspecific short hairpin RNA (L+ns) in noncastrated mice. Interestingly, tumors were also observed in castrated mice, suggesting that loss of ID4 promotes CRPC. RNA sequence analysis revealed a gene signature mimicking that of constitutively active AR in L-ID4, which was consistent with gain of de novo steroidogenesis. Prostate-specific antigen expression as a result of persistent AR activation was observed in L-ID4 cells but not in L+ns cells. The results demonstrate that ID4 acts as a tumor suppressor in PCa, and its loss, frequently observed in PCa, promotes CRPC through constitutive AR activation.
Collapse
Affiliation(s)
- Divya Patel
- Center for Cancer Research and Therapeutic Development (D.P., A.E.K., P.S., J.J., S.G., S.C., P.N., N.J.B., J.C.), Clark Atlanta University, Atlanta, Georgia 30314; and College of Pharmacy (M.K.-Y., R.P.), Mercer University, Atlanta, Georgia 30341
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Lai X, Liao J, Lin W, Huang C, Li J, Lin J, Chen Q, Ye Y. Inhibitor of DNA-binding protein 1 knockdown arrests the growth of colorectal cancer cells and suppresses hepatic metastasis in vivo. Oncol Rep 2014; 32:79-88. [PMID: 24804700 DOI: 10.3892/or.2014.3172] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 02/24/2014] [Indexed: 11/06/2022] Open
Abstract
Inhibitor of DNA-binding protein 1 (ID1) is commonly abnormally overexpressed in colorectal cancer (CRC); yet, the functional significance of ID1 in the growth and invasive properties of CRC cells remains largely unclear. The present study investigated the effects of ID1 downregulation on the cell growth and metastatic features of CRC. Using lentiviral shRNA infection, stable ID1-knockdown (KD) HCT116 and SW620 cells, human metastatic CRC cell lines, were created. In vitro, the migration/invasion capacity of the ID1-KD CRC cells was assessed by a wound healing assay. The activities of MMP2 and MMP-9 were measured by gelatin zymography. The expression of CXC chemokine receptor 4 (CXCR4), PCNA and survivin were determined by immunoblot analysis and qRT-PCR. The effects of ID1 knockdown on tumor growth and hepatic metastasis were demonstrated by a xenograft study in mice. The results showed evident decreases in proliferation, migration and invasion and an increased apoptosis rate in the ID1-KD CRC cells. Similarly, ID1 knockdown significantly decreased mRNA and protein levels of PCNA, survivin, CXCR4, MMP2 and MMP9. Overexpression of CXCR4 antagonized the negative effect on the migration and invasion abilities of the ID1-KD cells. As compared with the control, ID1 knockdown prevented tumor growth and profoundly suppressed hepatic metastasis in vivo. The present study demonstrated the significance of ID1 in colon cancer progression, and its effect on tumor invasiveness and metastatic properties may be partly dependent on CXCR4.
Collapse
Affiliation(s)
- Xiaolan Lai
- Graduate School of Education, Fujian Medical University, Fuzhou, Fujian 350108, P.R. China
| | - Jinrong Liao
- Graduate School of Education, Fujian Medical University, Fuzhou, Fujian 350108, P.R. China
| | - Wansong Lin
- Laboratory of Immuno-Oncology, Fujian Provincial Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Chuanzhong Huang
- Laboratory of Immuno-Oncology, Fujian Provincial Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Jieyu Li
- Laboratory of Immuno-Oncology, Fujian Provincial Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Jizhen Lin
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Qiang Chen
- Graduate School of Education, Fujian Medical University, Fuzhou, Fujian 350108, P.R. China
| | - Yunbin Ye
- Graduate School of Education, Fujian Medical University, Fuzhou, Fujian 350108, P.R. China
| |
Collapse
|
47
|
The 31-kDa caspase-generated cleavage product of p130Cas antagonizes the action of MyoD during myogenesis. Biochem Biophys Res Commun 2014; 444:509-13. [PMID: 24472550 DOI: 10.1016/j.bbrc.2014.01.085] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 01/18/2014] [Indexed: 11/23/2022]
Abstract
Myogenesis is regulated by the basic helix-loop-helix (bHLH) myogenic regulatory factor MyoD, which induces muscle-specific gene expression by binding to the E-box sequence as a heterodimer with ubiquitous bHLH E2A (E12/E47) proteins. Here, we report that a 31-kDa caspase-generated cleavage product of Crk-associated substrate (p130Cas), herein called 31-kDa, is downregulated during muscle cell differentiation. 31-kDa contains a helix-loop-helix (HLH) domain that shows greater sequence homology with Id (inhibitor of DNA binding) proteins than with bHLH proteins. This HLH domain, lacking the basic region required for DNA binding, mediated the direct interaction of 31-kDa with MyoD. Overexpression of 31-kDa in C3H10T1/2 cells inhibited not only the transcriptional activation of p21(Waf1/Cip1) and E-box-dependent muscle-specific genes by MyoD and/or E2A but also MyoD-induced myosin heavy chain expression and myogenic conversion. In sum, our results suggest a role for 31-kDa as a negative regulator of MyoD in the muscle differentiation program.
Collapse
|
48
|
Huang A, Zhao H, Quan Y, Jin R, Feng B, Zheng M. E2A predicts prognosis of colorectal cancer patients and regulates cancer cell growth by targeting miR-320a. PLoS One 2014; 9:e85201. [PMID: 24454819 PMCID: PMC3890311 DOI: 10.1371/journal.pone.0085201] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 11/25/2013] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Transcriptional factor E2A is crucial for the normal development and differentiation of B and T lymphocytes. Dysregulation of E2A leads to leukemia and tumorigenesis of some solid tumors. The expression and clinical significance of E2A as well as its role in colorectal cancer (CRC) are still unknown. This study aims to assess E2A expression in CRC tissues, evaluate its prognosis value, and investigate its role in colon cancer cell growth. METHODS E2A expression in CRC tissues and normal mucosa was detected by immunohistochemical staining; Kaplan-Meier survival curve and Cox regression model were used to evaluate the prognostic value of E2A. Lentivirus was used to construct E2A stably knocked-down cells. MTT assay was employed to detect cell proliferation change; cell cycle was analyzed by flow cytometry; and chromatin immunoprecipitation (ChIP) assay was used to validate the predicted binding target of E2A. RESULTS Expression of E2A was lower in CRC tissues than normal mucosa; low E2A expression correlated with advanced TNM stage and larger tumor size, and predicted poor prognosis of CRC patients. E2A knockdown resulted in increased cell proliferation rate and cell cycle acceleration. ChIP assay showed miR-320a was a direct target of E2A and upregulation of miR-320a in E2A downregulated cells could reverse cell proliferation and cell cycle changes caused by E2A deficiency. CONCLUSIONS E2A is an independent prognostic factor for CRC patients and targets miR-320a to regulate cell proliferation of colon cancer cells.
Collapse
Affiliation(s)
- Ao Huang
- Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Shanghai Institute of Digestive Surgery, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Shanghai, China
| | - Hongchao Zhao
- Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Shanghai Institute of Digestive Surgery, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Shanghai, China
| | - Yingjun Quan
- Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Shanghai Institute of Digestive Surgery, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Shanghai, China
| | - Runsen Jin
- Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Shanghai Institute of Digestive Surgery, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Shanghai, China
| | - Bo Feng
- Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Shanghai Institute of Digestive Surgery, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Shanghai, China
| | - Minhua Zheng
- Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Shanghai Institute of Digestive Surgery, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Shanghai, China
- * E-mail:
| |
Collapse
|
49
|
Abstract
The family of inhibitor of differentiation (Id) proteins is a group of evolutionarily conserved molecules, which play important regulatory roles in organisms ranging from Drosophila to humans. Id proteins are small polypeptides harboring a helix-loop-helix (HLH) motif, which are best known to mediate dimerization with other basic HLH proteins, primarily E proteins. Because Id proteins do not possess the basic amino acids adjacent to the HLH motif necessary for DNA binding, Id proteins inhibit the function of E protein homodimers, as well as heterodimers between E proteins and tissue-specific bHLH proteins. However, Id proteins have also been shown to have E protein-independent functions. The Id genes are broadly but differentially expressed in a variety of cell types. Transcription of the Id genes is controlled by transcription factors such as C/EBPβ and Egr as well as by signaling pathways triggered by different stimuli, which include bone morphogenic proteins, cytokines, and ligands of T cell receptors. In general, Id proteins are capable of inhibiting the differentiation of progenitors of different cell types, promoting cell-cycle progression, delaying cellular senescence, and facilitating cell migration. These properties of Id proteins enable them to play significant roles in stem cell maintenance, vasculogenesis, tumorigenesis and metastasis, the development of the immune system, and energy metabolism. In this review, we intend to highlight the current understanding of the function of Id proteins and discuss gaps in our knowledge about the mechanisms whereby Id proteins exert their diverse effects in multiple cellular processes.
Collapse
Affiliation(s)
- Flora Ling
- Immunobiology Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Bin Kang
- Immunobiology Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Xiao-Hong Sun
- Immunobiology Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA.
| |
Collapse
|
50
|
Lee SH, Lee JH, Yoo SY, Hur J, Kim HS, Kwon SM. Hypoxia Inhibits Cellular Senescence to Restore the Therapeutic Potential of Old Human Endothelial Progenitor Cells via the Hypoxia-Inducible Factor-1α–TWIST-p21 Axis. Arterioscler Thromb Vasc Biol 2013; 33:2407-14. [DOI: 10.1161/atvbaha.113.301931] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Sang Hun Lee
- From the Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea (S.H.L., J.H.L., S.Y.Y., S.M.K.); Immunoregulatory Therapeutics Group in Brain Busan 21 Project (S.H.L., J.H.L., S.Y.Y., S.M.K.), Department of Medical Biolotechnology (S.H.L.), Soonchunhyang University College of Medicine, Seoul, Korea; National Research Laboratory for Cardiovascular Stem Cell Niche, Innovative Research
| | - Jun Hee Lee
- From the Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea (S.H.L., J.H.L., S.Y.Y., S.M.K.); Immunoregulatory Therapeutics Group in Brain Busan 21 Project (S.H.L., J.H.L., S.Y.Y., S.M.K.), Department of Medical Biolotechnology (S.H.L.), Soonchunhyang University College of Medicine, Seoul, Korea; National Research Laboratory for Cardiovascular Stem Cell Niche, Innovative Research
| | - So Young Yoo
- From the Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea (S.H.L., J.H.L., S.Y.Y., S.M.K.); Immunoregulatory Therapeutics Group in Brain Busan 21 Project (S.H.L., J.H.L., S.Y.Y., S.M.K.), Department of Medical Biolotechnology (S.H.L.), Soonchunhyang University College of Medicine, Seoul, Korea; National Research Laboratory for Cardiovascular Stem Cell Niche, Innovative Research
| | - Jin Hur
- From the Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea (S.H.L., J.H.L., S.Y.Y., S.M.K.); Immunoregulatory Therapeutics Group in Brain Busan 21 Project (S.H.L., J.H.L., S.Y.Y., S.M.K.), Department of Medical Biolotechnology (S.H.L.), Soonchunhyang University College of Medicine, Seoul, Korea; National Research Laboratory for Cardiovascular Stem Cell Niche, Innovative Research
| | - Hyo-Soo Kim
- From the Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea (S.H.L., J.H.L., S.Y.Y., S.M.K.); Immunoregulatory Therapeutics Group in Brain Busan 21 Project (S.H.L., J.H.L., S.Y.Y., S.M.K.), Department of Medical Biolotechnology (S.H.L.), Soonchunhyang University College of Medicine, Seoul, Korea; National Research Laboratory for Cardiovascular Stem Cell Niche, Innovative Research
| | - Sang Mo Kwon
- From the Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea (S.H.L., J.H.L., S.Y.Y., S.M.K.); Immunoregulatory Therapeutics Group in Brain Busan 21 Project (S.H.L., J.H.L., S.Y.Y., S.M.K.), Department of Medical Biolotechnology (S.H.L.), Soonchunhyang University College of Medicine, Seoul, Korea; National Research Laboratory for Cardiovascular Stem Cell Niche, Innovative Research
| |
Collapse
|