1
|
Benegas P, Ziegler B, Dieminger V, Bengió R, Zapata P, Larripa I, Ferri C. Expression of genes potentially involved in loss of response in patients with chronic myeloid leukemia. Gene 2024; 896:148047. [PMID: 38042214 DOI: 10.1016/j.gene.2023.148047] [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: 10/28/2023] [Accepted: 11/28/2023] [Indexed: 12/04/2023]
Abstract
Chronic Myeloid Leukemia (CML) is a hematological malignancy characterized by the presence of the BCR::ABL1 fusion gene, which leads to uncontrolled cell growth and survival. Tyrosine kinase inhibitors (TKIs) have revolutionized the treatment of CML, but a significant proportion of patients develop resistance or lose response to these drugs. Understanding the molecular mechanisms underlying treatment response and resistance is crucial for improving patient outcomes. This study aimed to analyze the expression patterns of genes involved in treatment response and resistance in CML patients receiving TKI therapy. The expression levels of MET, FOXO3, p15, p16, HCK, and FYN genes were examined in CML patients and compared to healthy donors. Gene expression levels were compared between optimal responders (OR) and resistant patients (R) vs. healthy donors (HD). The MET and FOXO3 OR group showed significant differences compared with the HD, (p < 0.0001) and (p = 0.0003), respectively. p15 expression showed significant differences between OR and HD groups (p = 0.0078), while no significant differences were found in p16 expression between the HD groups. FYN showed a statistically significant difference between R vs. HD (p = 0.0157). The results of HCK expression analysis revealed significant differences between OR and HD (p = 0.0041) and between R and HD (p = 0.0026). When we analyzed OR patients with undetectable BCR::ABL1 transcripts, a greater expression of HCK was observed in the R group. These findings suggest that monitoring the expression levels of MET and FOXO3 genes could be valuable in predicting treatment response and relapse in CML patients. Our study provides important insights into the potential use of gene expression analysis as a tool for predicting treatment response and guiding treatment decisions in CML patients. This knowledge may ultimately contribute to the development of personalized treatment strategies to improve patient outcomes in CML.
Collapse
Affiliation(s)
- Paula Benegas
- Universidad Nacional de Misiones, Facultad de Ciencias Exactas Químicas y Naturales, Instituto de Biotecnología Misiones "Dra. María Ebe Reca" (INBIOMIS), Laboratorio de Biotecnología Molecular (BIOTECMOL), Misiones, Argentina; Consejo Nacional de Invetigaciones Cientìficas y Tècnicas (CONICET), Buenos Aires, Argentina
| | - Betiana Ziegler
- Universidad Nacional de Misiones, Facultad de Ciencias Exactas Químicas y Naturales, Instituto de Biotecnología Misiones "Dra. María Ebe Reca" (INBIOMIS), Laboratorio de Biotecnología Molecular (BIOTECMOL), Misiones, Argentina; Laboratorio de Genética Hematológica, IMEX, CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Victoria Dieminger
- Universidad Nacional de Misiones, Facultad de Ciencias Exactas Químicas y Naturales, Instituto de Biotecnología Misiones "Dra. María Ebe Reca" (INBIOMIS), Laboratorio de Biotecnología Molecular (BIOTECMOL), Misiones, Argentina
| | - Raquel Bengió
- Departamento de Hemato-oncología, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Pedro Zapata
- Universidad Nacional de Misiones, Facultad de Ciencias Exactas Químicas y Naturales, Instituto de Biotecnología Misiones "Dra. María Ebe Reca" (INBIOMIS), Laboratorio de Biotecnología Molecular (BIOTECMOL), Misiones, Argentina; Consejo Nacional de Invetigaciones Cientìficas y Tècnicas (CONICET), Buenos Aires, Argentina
| | - Irene Larripa
- Laboratorio de Genética Hematológica, IMEX, CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Cristian Ferri
- Universidad Nacional de Misiones, Facultad de Ciencias Exactas Químicas y Naturales, Instituto de Biotecnología Misiones "Dra. María Ebe Reca" (INBIOMIS), Laboratorio de Biotecnología Molecular (BIOTECMOL), Misiones, Argentina.
| |
Collapse
|
2
|
Georget M, Defois A, Guiho R, Bon N, Allain S, Boyer C, Halgand B, Waast D, Grimandi G, Fouasson-Chailloux A, Guicheux J, Vinatier C. Development of a DNA damage-induced senescence model in osteoarthritic chondrocytes. Aging (Albany NY) 2023; 15:8576-8593. [PMID: 37659108 PMCID: PMC10522398 DOI: 10.18632/aging.204881] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 06/28/2023] [Indexed: 09/04/2023]
Abstract
Senescent cells (SnCs) have been described to accumulate in osteoarthritis (OA) joint tissues in response to injury, thereby participating in OA development and progression. However, clinical therapeutic approaches targeting SnCs using senolysis, although promising in preclinical OA models, have not yet proven their efficacy in patients with knee OA. This pitfall may be due to the lack of understanding of the mechanisms underlying chondrocyte senescence. Therefore, our study aimed to generate models of chondrocyte senescence. This study used etoposide, to induce DNA damage-related senescence or chronic exposure to IL-1β to entail inflammation-related senescence in human OA chondrocytes. Several hallmarks of cellular senescence, such as cell cycle arrest, expression of cyclin-dependent kinase inhibitors, DNA damages, and senescence-associated secretory profile were evaluated. Chronic exposure to IL-1β induces only partial expression of senescence markers and does not allow us to conclude on its ability to induce senescence in chondrocytes. On the other hand, etoposide treatment reliably induces DNA damage-related senescence in human articular chondrocytes evidenced by loss of proliferative capacity, DNA damage accumulation, and expression of some SASP components. Etoposide-induced senescence model may help investigate the initiation of cellular senescence in chondrocytes, and provide a useful model to develop therapeutic approaches to target senescence in OA.
Collapse
Affiliation(s)
- Mélina Georget
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton RMeS, UMR 1229, Nantes F-44000, France
| | - Anaïs Defois
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton RMeS, UMR 1229, Nantes F-44000, France
| | - Romain Guiho
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton RMeS, UMR 1229, Nantes F-44000, France
| | - Nina Bon
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton RMeS, UMR 1229, Nantes F-44000, France
| | - Sophie Allain
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton RMeS, UMR 1229, Nantes F-44000, France
| | - Cécile Boyer
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton RMeS, UMR 1229, Nantes F-44000, France
| | - Boris Halgand
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton RMeS, UMR 1229, Nantes F-44000, France
| | - Denis Waast
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton RMeS, UMR 1229, Nantes F-44000, France
| | - Gaël Grimandi
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton RMeS, UMR 1229, Nantes F-44000, France
| | - Alban Fouasson-Chailloux
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton RMeS, UMR 1229, Nantes F-44000, France
| | - Jérôme Guicheux
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton RMeS, UMR 1229, Nantes F-44000, France
| | - Claire Vinatier
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton RMeS, UMR 1229, Nantes F-44000, France
| |
Collapse
|
3
|
RUNX1 overexpression triggers TGF-β signaling to upregulate p15 and thereby blocks early hematopoiesis by inducing cell cycle arrest. Stem Cell Res 2022; 60:102694. [DOI: 10.1016/j.scr.2022.102694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 01/14/2022] [Accepted: 01/25/2022] [Indexed: 11/19/2022] Open
|
4
|
Scruggs AM, Koh HB, Tripathi P, Leeper NJ, White ES, Huang SK. Loss of CDKN2B Promotes Fibrosis via Increased Fibroblast Differentiation Rather Than Proliferation. Am J Respir Cell Mol Biol 2019; 59:200-214. [PMID: 29420051 DOI: 10.1165/rcmb.2017-0298oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease characterized by excessive scarring and fibroblast activation. We previously showed that fibroblasts from patients with IPF are hypermethylated at the CDKN2B gene locus, resulting in decreased CDKN2B expression. Here, we examine how diminished CDKN2B expression in normal and IPF fibroblasts affect fibroblast function, and how loss of CDKN2B contributes to IPF pathogenesis. We first confirmed that protein expression of CDKN2B was diminished in IPF lungs in situ. Loss of CDKN2B was especially notable in regions of increased myofibroblasts and fibroblastic foci. The degree of CDKN2B hypermethylation was particularly elevated in patients with radiographic honeycombing, a marker of more advanced fibrosis, and increased DNA methylation correlated with decreased expression. Although CDKN2B is traditionally considered a cell cycle inhibitor, loss of CDKN2B did not result in an increase in fibroblast proliferation, but instead was associated with an increase in myofibroblast differentiation. An increase in myofibroblast differentiation was not observed when CDKN2A was silenced. Loss of CDKN2B was associated with an increase in the transcription factors serum response factor and myocardin-related transcription factor A, and overexpression of CDKN2B in IPF fibroblasts inhibited myofibroblast differentiation. Finally, decreased CDKN2B expression was noted in fibroblasts from a murine model of fibrosis, and Cdkn2b-/- mice developed greater histologic fibrosis after bleomycin injury. These findings identify a novel function for CDKN2B that differs from its conventional designation as a cell cycle inhibitor and demonstrate the importance of this protein in pulmonary fibrosis.
Collapse
Affiliation(s)
- Anne M Scruggs
- 1 Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan; and
| | - Hailey B Koh
- 1 Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan; and
| | - Priya Tripathi
- 1 Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan; and
| | - Nicholas J Leeper
- 2 Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Palo Alto, California
| | - Eric S White
- 1 Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan; and
| | - Steven K Huang
- 1 Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan; and
| |
Collapse
|
5
|
Guinn ZP, Petro TM. IFN-γ synergism with poly I:C reduces growth of murine and human cancer cells with simultaneous changes in cell cycle and immune checkpoint proteins. Cancer Lett 2018; 438:1-9. [PMID: 30205169 DOI: 10.1016/j.canlet.2018.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/13/2018] [Accepted: 09/06/2018] [Indexed: 01/05/2023]
Abstract
Previously, we reported that IFN-γ and poly I:C, a TLR3 Pattern Recognition Receptor (PRR) agonist, reduces growth of and induces Cleaved-Caspase-3, ISG54 and p27Kip in B16 melanoma cells. Here, analysis of IFN-γ/PRR synergism was expanded with UM-SCC1 and UM-SCC38 human squamous carcinoma cells and other PRR agonists. As in B16 cells, poly I:C plus IFN-γ synergism reduced UM-SCC1 and UM-SCC38 growth, and no more than 24 h was needed for significant growth reduction. IFN-γ synergism to stem B16 growth also occurred with TLR7, TLR9, TLR4, and STING agonists, but not TLR2 agonist. IFN-γ synergized with TLR3 and TLR4 agonists reducing UM-SCC1 growth, and with TLR7 and TLR3 agonists reducing UM-SCC38 growth. IFN-γ plus poly I:C, which had the most pronounced effect, decreased cyclin-D1, increased G1 cell cycle arrest, and increased Cleaved caspase-3 in B16 cells, as well as RAW264.7, a virus-transformed murine macrophage cell line. Finally, IFN-γ plus poly I:C modulated total but not cell surface expression of immune checkpoint protein PD-L1, as well as cell cycle checkpoint proteins in B16 cells. Thus IFN-γ plus poly I:C, and other PRR agonists, may well be effective adjuvants to cancer immunotherapy against several tumor cell types.
Collapse
Affiliation(s)
- Zachary P Guinn
- School of Biological Sciences, University of Nebraska-Lincoln, USA
| | - Thomas M Petro
- Nebraska Center for Virology, University of Nebraska-Lincoln, USA; Department of Oral Biology, University of Nebraska Medical Center, USA.
| |
Collapse
|
6
|
Zheng Q, Fan H, Meng Z, Yuan L, Liu C, Peng Y, Zhao W, Wang L, Li J, Feng J. Histone demethylase KDM2B promotes triple negative breast cancer proliferation by suppressing p15INK4B, p16INK4A, and p57KIP2 transcription. Acta Biochim Biophys Sin (Shanghai) 2018; 50:897-904. [PMID: 30060056 DOI: 10.1093/abbs/gmy084] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Indexed: 02/01/2023] Open
Abstract
H3K4me3 and H3K36me2 histone demethylase KDM2B is an epigenetic regulatory factor involved in cell proliferation in numerous cells including breast cancer cells, however, the regulatory mechanism of KDM2B in cell proliferation of breast cancer cells, specifically in triple negative breast cancer (TNBC), remains largely unknown. In this study, we showed that higher expression level of KDM2B was associated with poor prognosis in TNBC. Using cell proliferation assay, we found that KDM2B promoted TNBC cell proliferation by suppressing the transcription of the cell cycle inhibitors p15INK4B, p16INK4A, and p57KIP2. Chromatin immunoprecipitation assay results showed that KDM2B bound to the promoters of these genes and thereby reduced the H3K4me3 and H3K36me2 levels, leading to the suppression of gene transcription in a histone demethylation activity-dependent manner. Silencing of p15INK4B, p16INK4A, and p57KIP2 in TNBC cells was shown to restore the promoting effect of KDM2B on TNBC cell proliferation. The present study reveals a novel cell regulatory mechanism through which KDM2B promotes TNBC cell proliferation by binding to the promoters of p15INK4B, p16INK4A, and p57KIP2, which reduces H3K4me3 and H3K36me2 levels to suppress gene transcription.
Collapse
Affiliation(s)
- Qingping Zheng
- The Third School of Clinical Medicine, Southern Medical University, Guangdong Province, Guangzhou, China
- Department of Laboratory Medicine & Central Laboratory, Southern Medical University Affiliated Fengxian Hospital, Shanghai, China
| | - Hongjia Fan
- The Third School of Clinical Medicine, Southern Medical University, Guangdong Province, Guangzhou, China
- Department of Laboratory Medicine & Central Laboratory, Southern Medical University Affiliated Fengxian Hospital, Shanghai, China
| | - Zhenzhen Meng
- Department of Laboratory Medicine & Central Laboratory, Jinzhou Medical University Affiliated Fengxian Hospital, Shanghai, China
| | - Lin Yuan
- Department of Laboratory Medicine & Central Laboratory, Southern Medical University Affiliated Fengxian Hospital, Shanghai, China
| | - Cuicui Liu
- Department of Laboratory Medicine & Central Laboratory, Southern Medical University Affiliated Fengxian Hospital, Shanghai, China
- Shanghai University of Medicine & Health Sciences, Affiliated Sixth People’s Hospital South Campus, Shanghai, China
| | - You Peng
- Department of Laboratory Medicine & Central Laboratory, Southern Medical University Affiliated Fengxian Hospital, Shanghai, China
| | - Weiwei Zhao
- Department of Laboratory Medicine & Central Laboratory, Southern Medical University Affiliated Fengxian Hospital, Shanghai, China
| | - Lulu Wang
- Department of Laboratory Medicine & Central Laboratory, Southern Medical University Affiliated Fengxian Hospital, Shanghai, China
| | - Jing Li
- Department of Laboratory Medicine & Central Laboratory, Southern Medical University Affiliated Fengxian Hospital, Shanghai, China
- Joint Research Center for Precision Medicine, Shanghai Jiao Tong University & Affiliated Sixth People’s Hospital South Campus, Shanghai, China
| | - Jing Feng
- The Third School of Clinical Medicine, Southern Medical University, Guangdong Province, Guangzhou, China
- Department of Laboratory Medicine & Central Laboratory, Southern Medical University Affiliated Fengxian Hospital, Shanghai, China
- Shanghai University of Medicine & Health Sciences, Affiliated Sixth People’s Hospital South Campus, Shanghai, China
| |
Collapse
|
7
|
Duguang L, Jin H, Xiaowei Q, Peng X, Xiaodong W, Zhennan L, Jianjun Q, Jie Y. The involvement of lncRNAs in the development and progression of pancreatic cancer. Cancer Biol Ther 2017; 18:927-936. [PMID: 29053398 PMCID: PMC5718823 DOI: 10.1080/15384047.2017.1385682] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/09/2017] [Accepted: 09/24/2017] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer is one of the most malignant tumors that are difficult to diagnose at its early stage and there is no effective therapy. Recent studies uncovered that many non-protein-coding RNAs including the class of long noncoding RNAs (lncRNAs) are differentially expressed in various types of tumors and they are potent regulators of tumor progression and metastasis. LncRNA can mediate tumor initiation, proliferation, migration and metastasis through modulating epigenetic modification, alternative splicing, transcription, and protein translation. In this review, we discuss the molecular mechanism of lncRNAs in the involvement of tumor growth, survival, epithelial-mesenchymal transition, tumor microenvironment, cancer stem cells and chemoresistance in pancreatic ductal adenocarcinoma (PDAC).
Collapse
Affiliation(s)
- Li Duguang
- The Second Clinical College of Dalian Medical University, 9 Western District, Lvshun South Road, Dalian, Liaoning, P. R. China
| | - He Jin
- Medical college of Yangzhou University, Yangzhou, Jiangsu, P. R. China
| | - Qian Xiaowei
- Medical college of Yangzhou University, Yangzhou, Jiangsu, P. R. China
| | - Xu Peng
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Clinical medical college of Yangzhou University, Yangzhou, Jiangsu, P. R. China
| | - Wang Xiaodong
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Clinical medical college of Yangzhou University, Yangzhou, Jiangsu, P. R. China
| | - Li Zhennan
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Clinical medical college of Yangzhou University, Yangzhou, Jiangsu, P. R. China
| | - Qian Jianjun
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Clinical medical college of Yangzhou University, Yangzhou, Jiangsu, P. R. China
| | - Yao Jie
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Clinical medical college of Yangzhou University, Yangzhou, Jiangsu, P. R. China
| |
Collapse
|
8
|
Ma Z, Huang H, Wang J, Zhou Y, Pu F, Zhao Q, Peng P, Hui B, Ji H, Wang K. Long non-coding RNA SNHG15 inhibits P15 and KLF2 expression to promote pancreatic cancer proliferation through EZH2-mediated H3K27me3. Oncotarget 2017; 8:84153-84167. [PMID: 29137412 PMCID: PMC5663584 DOI: 10.18632/oncotarget.20359] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/29/2017] [Indexed: 01/17/2023] Open
Abstract
Long non-coding RNA (lncRNA) is emerging as an critical regulator in multiple cancers, including pancreatic cancer (PC). Recently, lncRNA SNHG15 was found to be up-regulated in gastric cancer and hepatocellular carcinoma, exerting oncogenic effects. Nevertheless, the biological function and regulatory mechanism of SNHG15 remain unclear in pancreatic cancer (PC). In this study, we reported that SNHG15 expression was also upregulated in PC tissues, and its overexpression was remarkably associated with tumor size, tumor node metastasis (TNM) stage and lymph node metastasis in patients with PC. SNHG15 knockdown inhibited proliferative capacities and suppressed apoptotic rate of PC cells in vitro, and impaired in-vivo tumorigenicity. Additionally, RNA immunoprecipitation (RIP) assays showed that SNHG15 epigenetically repressed the P15 and Kruppel-like factor 2 (KLF2) expression via binding to enhancer of zeste homolog 2 (EZH2), and chromatin immunoprecipitation assays (CHIP) assays demonstrated that EZH2 was capable of binding to promoter regions of P15 and KLF2 to induce histone H3 lysine 27 trimethylation (H3K27me3). Furthermore, rescue experiments indicated that SNHG15 oncogenic function partially involved P15 and KLF2 repression. Consistently, an inverse correlation between the expression of SNHG15 and traget genes were found in PC tissues. Our results reported that SNHG15 could act as an oncogene in PC, revealing its potential value as a biomarker for early detection and individualized therapy.
Collapse
Affiliation(s)
- Zhonghua Ma
- The Second Clinical Medical College of Nanjing Medical University, Nanjing 210000, Jiangsu, People's Republic of China.,Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu, People's Republic of China
| | - Hesuyuan Huang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu, People's Republic of China.,Department of General Surgery, Second Affiliated Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu, People's Republic of China
| | - Jirong Wang
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu, People's Republic of China
| | - Yan Zhou
- Department of Oncology, The Affiliated Yixing Hospital of Jiangsu University, Wuxi 214200, Jiangsu, People's Republic of China
| | - Fuxing Pu
- Department of Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu, People's Republic of China
| | - Qinghong Zhao
- Department of General Surgery, Second Affiliated Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu, People's Republic of China
| | - Peng Peng
- Department of Oncology, Second Hospital of Nanjing, Nanjing 210000, Jiangsu, People's Republic of China
| | - Bingqing Hui
- The Second Clinical Medical College of Nanjing Medical University, Nanjing 210000, Jiangsu, People's Republic of China.,Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu, People's Republic of China
| | - Hao Ji
- The Second Clinical Medical College of Nanjing Medical University, Nanjing 210000, Jiangsu, People's Republic of China.,Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu, People's Republic of China
| | - Keming Wang
- The Second Clinical Medical College of Nanjing Medical University, Nanjing 210000, Jiangsu, People's Republic of China.,Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu, People's Republic of China
| |
Collapse
|
9
|
Qin C, Ren L, Ji M, Lv S, Wei Y, Zhu D, Lin Q, Xu P, Chang W, Xu J. CDKL1 promotes tumor proliferation and invasion in colorectal cancer. Onco Targets Ther 2017; 10:1613-1624. [PMID: 28352193 PMCID: PMC5360398 DOI: 10.2147/ott.s133014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND CDKL1 is a member of the cell division cycle 2 (CDC2)-related serine threonine protein kinase family and is overexpressed in malignant tumors such as melanoma, breast cancer, and gastric cancer. OBJECTIVE This study aimed to evaluate whether CDKL1 can serve as a potential molecular target for colorectal cancer therapy. MATERIALS AND METHODS Expression of CDKL1 in colorectal cancer tissues and cell lines was measured by immunohistochemistry and Western blot, respectively. To investigate the role of CDKL1 in colorectal cancer, CDKL1-small hairpin RNA-expressing lentivirus was constructed and infected into HCT116 and Caco2 cells. The effects of RNA interference (RNAi)-mediated CDKL1 downregulation on cell proliferation and invasion were assessed by CCK-8, colony formation, transwell, and tumorigenicity assays in nude mice. The effects of CDKL1 downregulation on cell cycle and apoptosis were analyzed by flow cytometry. Furthermore, microarray method and data analysis elucidated the molecular mechanisms underlying the phenomenon. RESULTS CDKL1 protein was overexpressed in colorectal cancer tissues compared with paired normal tissues. Knockdown of CDKL1 in HCT116 and Caco2 significantly inhibited cell growth, colony formation ability, tumor invasion, and G1-S phase transition of the cell cycle. The knockdown of CDKL1 stimulated the upregulation of p15 and retinoblastoma protein. CONCLUSION CDKL1 plays a vital role in tumor proliferation and invasion in colorectal cancer in vitro and in vivo and, thus, may be considered as a valuable target for therapeutic intervention.
Collapse
Affiliation(s)
- Chunzhi Qin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai
| | - Li Ren
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai
| | - Meiling Ji
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai
| | - Shixu Lv
- Department of Surgical Oncology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Ye Wei
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai
| | - Dexiang Zhu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai
| | - Qi Lin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai
| | - Pingping Xu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai
| | - Wenju Chang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai
| | - Jianmin Xu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai
| |
Collapse
|
10
|
Lian Y, Wang J, Feng J, Ding J, Ma Z, Li J, Peng P, De W, Wang K. Long non-coding RNA IRAIN suppresses apoptosis and promotes proliferation by binding to LSD1 and EZH2 in pancreatic cancer. Tumour Biol 2016; 37:14929-14937. [PMID: 27644252 DOI: 10.1007/s13277-016-5380-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/09/2016] [Indexed: 12/23/2022] Open
Abstract
Long non-coding RNA (lncRNA) modulates gene expression, while lncRNA dysregulation is associated with human cancer. Furthermore, while recent studies have shown that lncRNA IRAIN plays an important role in other malignancies, the role of IRAIN in pancreatic cancer (PC) progression remains unclear. In this study, we found that upregulation of lncRNA IRAIN was significantly correlated with tumor size, TNM stage, and lymph node metastasis in a cohort of 37 PC patients. In vitro experiments showed that knockdown of IRAIN by small interfering RNA (siRNA) significantly induced cell apoptosis and inhibited cell proliferation in both BxPC-3 and PANC-1 cells. Further mechanism study showed that, by binding to histone demethylase lysine-specific demethylase 1 (LSD1), an enhancer of zeste homolog 2 (EZH2), IRAIN reduced PC tumor cell apoptosis and induced growth arrest by silencing the expression of Kruppel-like factor 2 (KLF2) and P15. Moreover, IRAIN expression was inversely correlated with that of KLF2 and P15 in PC tissues. To our knowledge, this is the first report elucidating the role and mechanism of IRAIN in PC progression.
Collapse
Affiliation(s)
- Yifan Lian
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210000, Jiangsu, People's Republic of China
| | - Juan Wang
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210000, Jiangsu, People's Republic of China
| | - Jing Feng
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, People's Republic of China
| | - Jie Ding
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210000, Jiangsu, People's Republic of China
| | - Zhonghua Ma
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210000, Jiangsu, People's Republic of China
| | - Juan Li
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210000, Jiangsu, People's Republic of China
| | - Peng Peng
- Department of Oncology, The Second Hospital of Nanjing, Nanjing, 210000, Jiangsu, People's Republic of China
| | - Wei De
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 210000, Jiangsu, People's Republic of China
| | - Keming Wang
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210000, Jiangsu, People's Republic of China.
| |
Collapse
|
11
|
Wei B, Yang S, Zhang B, Feng Y. Clinicopathological significance of p15 promoter hypermethylation in multiple myeloma: a meta-analysis. Onco Targets Ther 2016; 9:4015-22. [PMID: 27445492 PMCID: PMC4936815 DOI: 10.2147/ott.s102733] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Published studies reported that loss of function of the p15INK4B gene is caused by hypermethylation; however, whether or not the inactivation is associated with the incidence and clinical significance of multiple myeloma (MM) remains unclear. In this study, we performed a meta-analysis to quantitatively determine the effects of p15 hypermethylation on the incidence of MM. The related research articles in English and Chinese languages were evaluated. The data were extracted and assessed independently. The pooled data were analyzed and odds ratios were calculated and summarized. Sixteen eligible studies were selected for final analysis. We demonstrated that p15 hypermethylation is significantly higher in MM than that in normal bone marrow, as well as monoclonal gammopathy of undetermined significance. However, aberrant p15 hypermethylation was not significantly higher in advanced MM than that in early-stage MM. The results of this study reveal that p15 hypermethylation is correlated with an increased risk in the progression of monoclonal gammopathy of undetermined significance to MM. p15 hypermethylation, which induces the loss of function of the p15 gene, plays a critical role in the early tumorigenesis of MM and serves as a reputable diagnostic marker and potential drug target.
Collapse
Affiliation(s)
- Bing Wei
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Shuhua Yang
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Bo Zhang
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yong Feng
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
12
|
Rapamycin restores p14, p15 and p57 expression and inhibits the mTOR/p70S6K pathway in acute lymphoblastic leukemia cells. Int J Hematol 2015; 102:558-68. [DOI: 10.1007/s12185-015-1858-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 08/11/2015] [Accepted: 08/12/2015] [Indexed: 12/23/2022]
|
13
|
Role of HOXA9 in leukemia: dysregulation, cofactors and essential targets. Oncogene 2015; 35:1090-8. [PMID: 26028034 DOI: 10.1038/onc.2015.174] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/24/2015] [Accepted: 04/14/2015] [Indexed: 02/08/2023]
Abstract
HOXA9 is a homeodomain-containing transcription factor that has an important role in hematopoietic stem cell expansion and is commonly deregulated in acute leukemias. A variety of upstream genetic alterations in acute myeloid leukemia lead to overexpression of HOXA9, which is a strong predictor of poor prognosis. In many cases, HOXA9 has been shown to be necessary for maintaining leukemic transformation; however, the molecular mechanisms through which it promotes leukemogenesis remain elusive. Recent work has established that HOXA9 regulates downstream gene expression through binding at promoter distal enhancers along with a subset of cell-specific cofactor and collaborator proteins. Increasing efforts are being made to identify both the critical cofactors and target genes required for maintaining transformation in HOXA9-overexpressing leukemias. With continued advances in understanding HOXA9-mediated transformation, there is a wealth of opportunity for developing novel therapeutics that would be applicable for greater than 50% of AML with overexpression of HOXA9.
Collapse
|
14
|
Humeniuk R, Koller R, Bies J, Aplan P, Wolff L. Brief report: Loss of p15Ink4b accelerates development of myeloid neoplasms in Nup98-HoxD13 transgenic mice. Stem Cells 2014; 32:1361-6. [PMID: 24449168 DOI: 10.1002/stem.1635] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 11/13/2013] [Indexed: 12/26/2022]
Abstract
Homeostasis of hematopoietic stem and progenitor cells is a tightly regulated process. The disturbance of the balance in the hematopoietic progenitor pool can result in favorable conditions for development of diseases such as myelodysplastic syndromes and leukemia. It has been shown recently that mice lacking p15Ink4b have skewed differentiation of common myeloid progenitors toward the myeloid lineage at the expense of erythroid progenitors. The lack of p15INK4B expression in human leukemic blasts has been linked to poor prognosis and increased risk of myelodysplastic syndromes transformation to acute myeloid leukemia. However, the role of p15Ink4b in disease development is just beginning to be elucidated. This study examines the collaboration of the loss of p15Ink4b with Nup98-HoxD13 translocation in the development of hematological malignancies in a mouse model. Here, we report that loss of p15Ink4b collaborates with Nup98-HoxD13 transgene in the development of predominantly myeloid neoplasms, namely acute myeloid leukemia, myeloproliferative disease, and myelodysplastic syndromes. This mouse model could be a very valuable tool for studying p15Ink4b function in tumorigenesis as well as preclinical drug testing.
Collapse
Affiliation(s)
- Rita Humeniuk
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | |
Collapse
|
15
|
Bodoor K, Haddad Y, Alkhateeb A, Al-Abbadi A, Dowairi M, Magableh A, Bsoul N, Ghabkari A. DNA hypermethylation of cell cycle (p15 and p16) and apoptotic (p14, p53, DAPK and TMS1) genes in peripheral blood of leukemia patients. Asian Pac J Cancer Prev 2014; 15:75-84. [PMID: 24528084 DOI: 10.7314/apjcp.2014.15.1.75] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Aberrant DNA methylation of tumor suppressor genes has been reported in all major types of leukemia with potential involvement in the inactivation of regulatory cell cycle and apoptosis genes. However, most of the previous reports did not show the extent of concurrent methylation of multiple genes in the four leukemia types. Here, we analyzed six key genes (p14, p15, p16, p53, DAPK and TMS1) for DNA methylation using methylation specific PCR to analyze peripheral blood of 78 leukemia patients (24 CML, 25 CLL, 12 AML, and 17 ALL) and 24 healthy volunteers. In CML, methylation was detected for p15 (11%), p16 (9%), p53 (23%) and DAPK (23%), in CLL, p14 (25%), p15 (19%), p16 (12%), p53 (17%) and DAPK (36%), in AML, p14 (8%), p15 (45%), p53 (9%) and DAPK (17%) and in ALL, p15 (14%), p16 (8%), and p53 (8%). This study highlighted an essential role of DAPK methylation in chronic leukemia in contrast to p15 methylation in the acute cases, whereas TMS1 hypermethylation was absent in all cases. Furthermore, hypermethylation of multiple genes per patient was observed, with obvious selectiveness in the 9p21 chromosomal region genes (p14, p15 and p16). Interestingly, methylation of p15 increased the risk of methylation in p53, and vice versa, by five folds (p=0.03) indicating possible synergistic epigenetic disruption of different phases of the cell cycle or between the cell cycle and apoptosis. The investigation of multiple relationships between methylated genes might shed light on tumor specific inactivation of the cell cycle and apoptotic pathways.
Collapse
Affiliation(s)
- Khaldon Bodoor
- Department of Biology, Jordan University of Science and Technology, Irbid, Jordan E-mail :
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Honda H, Nagamachi A, Inaba T. -7/7q- syndrome in myeloid-lineage hematopoietic malignancies: attempts to understand this complex disease entity. Oncogene 2014; 34:2413-25. [PMID: 24998854 DOI: 10.1038/onc.2014.196] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 05/27/2014] [Accepted: 06/03/2014] [Indexed: 01/19/2023]
Abstract
The recurrence of chromosomal abnormalities in a specific subtype of cancer strongly suggests that dysregulated gene expression in the corresponding region has a critical role in disease pathogenesis. -7/7q-, defined as the entire loss of chromosome 7 and partial deletion of its long arm, is among the most frequently observed chromosomal aberrations in myeloid-lineage hematopoietic malignancies such as myelodysplastic syndrome and acute myeloid leukemia, particularly in patients treated with cytotoxic agents and/or irradiation. Tremendous efforts have been made to clarify the molecular mechanisms underlying the disease development, and several possible candidate genes have been cloned. However, the study is still underway, and the entire nature of this syndrome is not completely understood. In this review, we focus on the attempts to identify commonly deleted regions in patients with -7/7q-; isolate the candidate genes responsible for disease development, cooperative genes and the factors affecting disease prognosis; and determine effective and potent therapeutic approaches. We also refer to the possibility that the accumulation of multiple gene haploinsufficiency, rather than the loss of a single tumor suppressor gene, may contribute to the development of diseases with large chromosomal deletions such as -7/7q-.
Collapse
Affiliation(s)
- H Honda
- Department of Disease Model, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - A Nagamachi
- Department of Molecular Oncology and Leukemia Program Project, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - T Inaba
- Department of Molecular Oncology and Leukemia Program Project, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
17
|
C/EBPα is an essential collaborator in Hoxa9/Meis1-mediated leukemogenesis. Proc Natl Acad Sci U S A 2014; 111:9899-904. [PMID: 24958854 DOI: 10.1073/pnas.1402238111] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Homeobox A9 (HOXA9) is a homeodomain-containing transcription factor that plays a key role in hematopoietic stem cell expansion and is commonly deregulated in human acute leukemias. A variety of upstream genetic alterations in acute myeloid leukemia (AML) lead to overexpression of HOXA9, almost always in association with overexpression of its cofactor meis homeobox 1 (MEIS1) . A wide range of data suggests that HOXA9 and MEIS1 play a synergistic causative role in AML, although the molecular mechanisms leading to transformation by HOXA9 and MEIS1 remain elusive. In this study, we identify CCAAT/enhancer binding protein alpha (C/EBPα) as a critical collaborator required for Hoxa9/Meis1-mediated leukemogenesis. We show that C/EBPα is required for the proliferation of Hoxa9/Meis1-transformed cells in culture and that loss of C/EBPα greatly improves survival in both primary and secondary murine models of Hoxa9/Meis1-induced leukemia. Over 50% of Hoxa9 genome-wide binding sites are cobound by C/EBPα, which coregulates a number of downstream target genes involved in the regulation of cell proliferation and differentiation. Finally, we show that Hoxa9 represses the locus of the cyclin-dependent kinase inhibitors Cdkn2a/b in concert with C/EBPα to overcome a block in G1 cell cycle progression. Together, our results suggest a previously unidentified role for C/EBPα in maintaining the proliferation required for Hoxa9/Meis1-mediated leukemogenesis.
Collapse
|