1
|
Yang Z, Jiang S, Lu C, Ji T, Yang W, Li T, Lv J, Hu W, Yang Y, Jin Z. SOX11: friend or foe in tumor prevention and carcinogenesis? Ther Adv Med Oncol 2019; 11:1758835919853449. [PMID: 31210798 PMCID: PMC6547177 DOI: 10.1177/1758835919853449] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 04/26/2019] [Indexed: 12/12/2022] Open
Abstract
Sex-determining region Y-related high-mobility-group box transcription factor 11 (SOX11) is an essential member of the SOX transcription factors and has been highlighted as an important regulator in embryogenesis. SOX11 studies have only recently shifted focus from its role in embryogenesis and development to its function in disease. In particular, the role of SOX11 in carcinogenesis has become of major interest in the field. SOX11 expression is elevated in a wide variety of tumors. In many cancers, dysfunctional expression of SOX11 has been correlated with increased cancer cell survival, inhibited cell differentiation, and tumor progression through the induction of metastasis and angiogenesis. Nevertheless, in a limited number of malignancies, SOX11 has also been identified to function as a tumor suppressor. Herein, we review the correlation between the expression of SOX11 and tumor behaviors. We also summarize the mechanisms underlying the regulation of SOX11 expression and activity in pathological conditions. In particular, we focus on the pathological processes of cancer targeted by SOX11 and discuss whether SOX11 is protective or detrimental during tumor progression. Moreover, SOX11 is highlighted as a clinical biomarker for the diagnosis and prognosis of various human cancer. The information reviewed here should assist in future experimental designs and emphasize the potential of SOX11 as a therapeutic target for cancer.
Collapse
Affiliation(s)
- Zhi Yang
- Department of Cardiovascular Surgery, Xijing
Hospital, The Fourth Military Medical University, Xi’an, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth
Military Medical University, Xi’an, China
| | - Chenxi Lu
- Key Laboratory of Resource Biology and
Biotechnology in Western China, Ministry of Education, Faculty of Life
Sciences, Northwest University, Xi’an, China
| | - Ting Ji
- Key Laboratory of Resource Biology and
Biotechnology in Western China, Ministry of Education, Faculty of Life
Sciences, Northwest University, Xi’an, China
| | - Wenwen Yang
- Key Laboratory of Resource Biology and
Biotechnology in Western China, Ministry of Education, Faculty of Life
Sciences, Northwest University, Xi’an, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth
Military Medical University, Xi’an, China
| | - Jianjun Lv
- Department of Biomedical Engineering, The Fourth
Military Medical University, Xi’an, China
| | - Wei Hu
- Department of Immunology, The Fourth Military
Medical University, Xi’an, China
| | - Yang Yang
- Key Laboratory of Resource Biology and
Biotechnology in Western China, Ministry of Education, Faculty of Life
Sciences, Northwest University, Xi’an, China
| | - Zhenxiao Jin
- Department of Cardiovascular Surgery, Xijing
Hospital, The Fourth Military Medical University, 127 Changle West Road,
Xi’an 710032, China
| |
Collapse
|
2
|
Blachly JS, Byrd JC, Grever M. Cyclin-dependent kinase inhibitors for the treatment of chronic lymphocytic leukemia. Semin Oncol 2016; 43:265-73. [DOI: 10.1053/j.seminoncol.2016.02.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
3
|
Lu TX, Li JY, Xu W. The role of SOX11 in mantle cell lymphoma. Leuk Res 2013; 37:1412-9. [DOI: 10.1016/j.leukres.2013.07.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 07/26/2013] [Accepted: 07/27/2013] [Indexed: 12/14/2022]
|
4
|
Abstract
Abstract As the rational application of targeted therapies in cancer supplants traditional cytotoxic chemotherapy, there is an ever-greater need for a thorough understanding of the complex machinery of the cell and an application of this knowledge to the development of novel therapeutics and combinations of agents. Here, we review the current state of knowledge of the class of targeted agents known as cyclin-dependent kinase (CDK) inhibitors, with a focus on chronic lymphocytic leukemia (CLL). Flavopiridol (alvocidib) is the best studied of the CDK inhibitors, producing a dramatic cytotoxic effect in vitro and in vivo, with the principal limiting factor of acute tumor lysis. Unfortunately, flavopiridol has a narrow therapeutic window and is relatively non-selective with several off-target (i.e. non-CDK) effects, which prompted development of the second-generation CDK inhibitor dinaciclib. Dinaciclib appears to be both more potent and selective than flavopiridol, with at least an order of magnitude greater therapeutic index, and is currently in phase III clinical trials. In additional to flavopiridol and dinaciclib, we also review the current status of other members of this class, and provide commentary as to the future direction of combination therapy including CDK inhibitors.
Collapse
|
5
|
Ju Z, Zhang J, Gao Y, Cheng T. Telomere dysfunction and cell cycle checkpoints in hematopoietic stem cell aging. Int J Hematol 2011; 94:33-43. [PMID: 21671044 DOI: 10.1007/s12185-011-0882-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 05/22/2011] [Accepted: 05/23/2011] [Indexed: 12/13/2022]
Abstract
Stem cells are believed to be closely associated with tissue degeneration during aging. Studies of human genetic diseases and gene-targeted animal models have provided evidence that functional decline of telomeres and deregulation of cell cycle checkpoints contribute to the aging process of tissue stem cells. Telomere dysfunction can induce DNA damage response via key cell cycle checkpoints, leading to cellular senescence or apoptosis depending on the tissue type and developmental stage of a specific stem cell compartment. Telomerase mutation and telomere shortening have been observed in a variety of hematological disorders, such as dyskeratosis congenital, aplastic anemia, myelodysplastic syndromes and leukemia, in which the hematopoietic stem cells (HSC) are a major target during the pathogenesis. Moreover, telomere dysfunction is able to induce both cell-intrinsic checkpoints and environmental factors limiting the self-renewal capacity and differentiation potential of HSCs. Crucial components in the cascade of DNA damage response, including ataxia telangiectasia mutated, CHK2, p53, p21 and p16/p19(ARF), play important roles in HSC maintenance and self-renewal in the scenarios of both sufficient telomere reserve and dysfunctional telomere. Therefore, a further understanding of the molecular mechanisms underlying HSC aging may help identity new therapeutic targets for stem cell-based regenerative medicine.
Collapse
Affiliation(s)
- Zhenyu Ju
- School of Medicine, Hangzhou Normal University, Hangzhou, China.
| | - Junling Zhang
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science, Tianjin, China
| | - Yingdai Gao
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China. .,Department of Radiation Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
SOX11 expression correlates to promoter methylation and regulates tumor growth in hematopoietic malignancies. Mol Cancer 2010; 9:187. [PMID: 20624318 PMCID: PMC2913986 DOI: 10.1186/1476-4598-9-187] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Accepted: 07/12/2010] [Indexed: 12/31/2022] Open
Abstract
Background The transcription factor SOX11 plays an important role in embryonic development of the central nervous system (CNS) and is expressed in the adult immature neuron but is normally not expressed in any other adult tissue. It has recently been reported to be implicated in various malignant neoplasms, including several lymphoproliferative diseases, by its specific expression and in some cases correlation to prognosis. SOX11 has been shown to prevent gliomagenesis in vivo but the causes and consequences of aberrant expression of SOX11 outside the CNS remain unexplained. Results We now show the first function of SOX11 in lymphoproliferative diseases, by demonstrating in vitro its direct involvement in growth regulation, as assessed by siRNA-mediated silencing and ectopic overexpression in hematopoietic malignancies. Gene Chip analysis identified cell cycle regulatory pathways, including Rb-E2F, to be associated with SOX11-induced growth reduction. Furthermore, promoter analysis revealed that SOX11 is silenced through DNA methylation in B cell lymphomas, suggesting that its regulation is epigenetically controlled. Conclusions The data show that SOX11 is not a bystander but an active and central regulator of cellular growth, as both siRNA-mediated knock-down and ectopic overexpression of SOX11 resulted in altered proliferation. Thus, these data demonstrate a tumor suppressor function for SOX11 in hematopoietic malignancies and revealed a potential epigenetic regulation of this developmentally involved gene.
Collapse
|
7
|
Viatour P, Somervaille TC, Venkatasubrahmanyam S, Kogan S, McLaughlin ME, Weissman IL, Butte AJ, Passegué E, Sage J. Hematopoietic stem cell quiescence is maintained by compound contributions of the retinoblastoma gene family. Cell Stem Cell 2008; 3:416-28. [PMID: 18940733 PMCID: PMC2646421 DOI: 10.1016/j.stem.2008.07.009] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Revised: 04/22/2008] [Accepted: 07/16/2008] [Indexed: 12/12/2022]
Abstract
Individual members of the retinoblastoma (Rb) tumor suppressor gene family serve critical roles in the control of cellular proliferation and differentiation, but the extent of their contributions is masked by redundant and compensatory mechanisms. Here we employed a conditional knockout strategy to simultaneously inactivate all three members, Rb, p107, and p130, in adult hematopoietic stem cells (HSCs). Rb family triple knockout (TKO) mice develop a cell-intrinsic myeloproliferation that originates from hyperproliferative early hematopoietic progenitors and is accompanied by increased apoptosis in lymphoid progenitor populations. Loss of quiescence in the TKO HSC pool is associated with an expansion of these mutant stem cells but also with an enhanced mobilization and an impaired reconstitution potential upon transplantation. The presence of a single p107 allele is sufficient to largely rescue these defects. Thus, Rb family members collectively maintain HSC quiescence and the balance between lymphoid and myeloid cell fates in the hematopoietic system.
Collapse
Affiliation(s)
- Patrick Viatour
- Department of Pediatrics, Stanford Medical School, Stanford, CA 94305
- Department of Genetics, Stanford Medical School, Stanford, CA 94305
- Department of Medical Chemistry, University of Liège, Liège, Belgium
| | | | - Shivkumar Venkatasubrahmanyam
- Department of Pediatrics, Stanford Medical School, Stanford, CA 94305
- Department of Medicine, Stanford Medical School, Stanford, CA 94305
| | - Scott Kogan
- Department of Laboratory Medicine, UCSF Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
| | | | | | - Atul J. Butte
- Department of Pediatrics, Stanford Medical School, Stanford, CA 94305
- Department of Medicine, Stanford Medical School, Stanford, CA 94305
| | - Emmanuelle Passegué
- Institute for Regenerative Medicine, UCSF Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
| | - Julien Sage
- Department of Pediatrics, Stanford Medical School, Stanford, CA 94305
- Department of Genetics, Stanford Medical School, Stanford, CA 94305
| |
Collapse
|
8
|
Abstract
The retinoblastoma (RB) tumour suppressor gene is functionally inactivated in a broad range of paediatric and adult cancers, and a plethora of cellular functions and partners have been identified for the RB protein. Data from human tumours and studies from mouse models indicate that loss of RB function contributes to both cancer initiation and progression. However, we still do not know the identity of the cell types in which RB normally prevents cancer initiation in vivo, and the specific functions of RB that suppress distinct aspects of the tumorigenic process are poorly understood.
Collapse
Affiliation(s)
- Deborah L Burkhart
- Cancer Biology Program, Stanford University School of Medicine, Stanford, California 94305, USA
| | | |
Collapse
|