1
|
Somaiah N, Tap W. MDM2-p53 in liposarcoma: The need for targeted therapies with novel mechanisms of action. Cancer Treat Rev 2024; 122:102668. [PMID: 38104352 DOI: 10.1016/j.ctrv.2023.102668] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Well-differentiated and dedifferentiated liposarcomas (WDLPS and DDLPS) are rare tumors that arise from lipocytes in soft tissue. There is a high unmet need in patients with these liposarcomas given poor outcomes, particularly for DDLPS. WDLPS and DDLPS share important genetic and histological characteristics - most notably, the amplification of the 2 genes MDM2 and CDK4. Both genes are considered oncogenes because of their ability to shut down tumor suppressor pathways. There are multiple therapeutic approaches that aim to target MDM2 and CDK4 activity for the purpose of restoring intrinsic tumor suppressor cellular response and terminating oncogenesis. However, current understanding of the molecular mechanisms involved in WDLPS and DDLPS pathology is limited. In recent years, significant efforts have been made to refine and implement targeted therapy for this patient population. The use of patient-derived cell and tumor xenograft models has been an important tool for recapitulating WDLPS and DDLPS biology. These models also offer valuable insights for drug development and drug combination studies. Here we offer a review of the current understanding of WDLPS and DDLPS biology and its therapeutic implications.
Collapse
Affiliation(s)
- Neeta Somaiah
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, United States.
| | - William Tap
- Sarcoma Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States.
| |
Collapse
|
2
|
Chen Y, Yin H, Sun J, Zhang G, Zhang Y, Zeng H. TrxR/Trx inhibitor butaselen ameliorates pulmonary fibrosis by suppressing NF-κB/TGF-β1/Smads signaling. Biomed Pharmacother 2023; 169:115822. [PMID: 37944440 DOI: 10.1016/j.biopha.2023.115822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
Pulmonary fibrosis is highly lethal with limited treatments. Butaselen (BS) is an inhibitor of thioredoxin reductase (TrxR)/thioredoxin (Trx) with anti-tumor activity. However, its impact on pulmonary fibrosis and the involved mechanisms remain unclear. Here, we demonstrate that BS is a potential drug for the treatment of pulmonary fibrosis. Specifically, BS can inhibit pulmonary fibrosis both in vitro and in vivo, with comparable efficacy and enhanced safety when compared with pirfenidone. BS and dexamethasone display a synergistic effect in inhibiting pulmonary fibrosis both in vitro and in vivo. Mechanistic studies reveal that BS can inhibit the TrxR activity during pulmonary fibrosis. RNA-sequencing analysis identifies that genes of ECM-related signaling pathways are notably affected by BS. BS can not only inhibit the activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and reduce pulmonary fibrosis-related inflammation, but also reduce NF-κB-activated transcriptional expression of transforming growth factor-β1 (TGF-β1), which leads to the inactivation of Smad2/Smad3 and decrease of collagen formation and fibrosis. Moreover, the knockdown of Trx1 with siRNA can also inhibit NF-κB/TGF-β1/Smads signaling. In conclusion, the TrxR/Trx inhibitor butaselen can suppress pulmonary fibrosis by inhibiting NF-κB/TGF-β1/Smads signaling.
Collapse
Affiliation(s)
- Yifan Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China; Cancer Center, Peking University Third Hospital, Beijing, China; Biobank, Peking University Third Hospital, Beijing, China
| | - Hanwei Yin
- Shanghai Yuanxi Medicine Corp, Shanghai, China
| | - Jing Sun
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Guozhou Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Ying Zhang
- Shanghai Yuanxi Medicine Corp, Shanghai, China
| | - Huihui Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| |
Collapse
|
3
|
Zhou Y, Zhang T, Wang S, Yang R, Jiao Z, Lu K, Li H, Jiang W, Zhang X. Targeting of HBP1/TIMP3 axis as a novel strategy against breast cancer. Pharmacol Res 2023; 194:106846. [PMID: 37414199 DOI: 10.1016/j.phrs.2023.106846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/30/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023]
Abstract
Malignant proliferation and metastasis are the main causes of breast cancer death. The transcription factor high mobility group (HMG) box-containing protein 1 (HBP1) is an important tumor suppressor whose deletion or mutation is closely related to the appearance of tumors. Here, we investigated the role of HBP1 in breast cancer suppression. HBP1 enhances the activity of the tissue inhibitors of metalloproteinases 3 (TIMP3) promoter, thereby increasing protein and mRNA levels of TIMP3. TIMP3 increases the phosphatase and tensin homolog (PTEN) protein level by inhibiting its degradation and acts as a metalloproteinase inhibitor to inhibit the protein levels of MMP2/9. In this study, we demonstrated that the HBP1/TIMP3 axis plays a crucial role in inhibiting the tumorigenesis of breast cancer. HBP1 deletion interferes with the regulation of the axis and induces the occurrence and malignant progression of breast cancer. In addition, the HBP1/TIMP3 axis promotes the sensitivity of breast cancer to radiation therapy and hormone therapy. Our study opens new perspectives on the treatment and prognosis of breast cancer.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Tongjia Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Shujie Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Ruixiang Yang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Zitao Jiao
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Kejia Lu
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Hui Li
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Wei Jiang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China
| | - Xiaowei Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, PR China.
| |
Collapse
|
4
|
Yang R, Zhou Y, Zhang T, Wang S, Wang J, Cheng Y, Li H, Jiang W, Yang Z, Zhang X. The transcription factor HBP1 promotes ferroptosis in tumor cells by regulating the UHRF1-CDO1 axis. PLoS Biol 2023; 21:e3001862. [PMID: 37406020 DOI: 10.1371/journal.pbio.3001862] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 05/31/2023] [Indexed: 07/07/2023] Open
Abstract
The induction of ferroptosis in tumor cells is one of the most important mechanisms by which tumor progression can be inhibited; however, the specific regulatory mechanism underlying ferroptosis remains unclear. In this study, we found that transcription factor HBP1 has a novel function of reducing the antioxidant capacity of tumor cells. We investigated the important role of HBP1 in ferroptosis. HBP1 down-regulates the protein levels of UHRF1 by inhibiting the expression of the UHRF1 gene at the transcriptional level. Reduced levels of UHRF1 have been shown to regulate the ferroptosis-related gene CDO1 by epigenetic mechanisms, thus up-regulating the level of CDO1 and increasing the sensitivity of hepatocellular carcinoma and cervical cancer cells to ferroptosis. On this basis, we constructed metal-polyphenol-network coated HBP1 nanoparticles by combining biological and nanotechnological. MPN-HBP1 nanoparticles entered tumor cells efficiently and innocuously, induced ferroptosis, and inhibited the malignant proliferation of tumors by regulating the HBP1-UHRF1-CDO1 axis. This study provides a new perspective for further research on the regulatory mechanism underlying ferroptosis and its potential role in tumor therapy.
Collapse
Affiliation(s)
- Ruixiang Yang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Yue Zhou
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Tongjia Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Shujie Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Jiyin Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Yuning Cheng
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Hui Li
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Wei Jiang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| | - Zhe Yang
- Department of pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Xiaowei Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, China
| |
Collapse
|
5
|
Wen L, Ren Q, Guo F, Du X, Yang H, Fu P, Ma L. Tubular aryl hydratocarbon receptor upregulates EZH2 to promote cellular senescence in cisplatin-induced acute kidney injury. Cell Death Dis 2023; 14:18. [PMID: 36635272 PMCID: PMC9837170 DOI: 10.1038/s41419-022-05492-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 01/13/2023]
Abstract
Acute kidney injury (AKI) is one of the serious clinical syndromes with high morbidity and mortality. Despite substantial progress in understanding the mechanism of AKI, no effective drug is available for treatment or prevention. In this study, we identified that a ligand-activated transcription factor aryl hydrocarbon receptor (AhR) was abnormally increased in the kidneys of cisplatin-induced AKI mice or tubular epithelial TCMK-1 cells. The AhR inhibition by BAY2416964 and tubular conditional deletion both alleviated cisplatin-induced kidney dysfunction and tubular injury. Notably, inhibition of AhR could improve cellular senescence of injured kidneys, which was indicated by senescence-associated β-galactosidase (SA-β-gal) activity, biomarker p53, p21, p16 expression, and secretory-associated secretory phenotype IL-1β, IL-6 and TNFα level. Mechanistically, the abnormal AhR expression was positively correlated with the increase of a methyltransferase EZH2, and AhR inhibition suppressed the EZH2 expression in cisplatin-injured kidneys. Furthermore, the result of ChIP assay displayed that EZH2 might indirectly interact with AhR promoter region by affecting H3K27me3. The direct recruitment between H3K27me3 and AhR promoter is higher in the kidneys of control than that of cisplatin-treated mice, suggesting EZH2 reversely influenced AhR expression through weakening H3K27me3 transcriptional inhibition on AhR promoter. The present study implicated that AhR and EZH2 have mutual regulation, which further accelerated tubular senescence in cisplatin-induced AKI. Notably, the crucial role of AhR is potential to become a promising target for AKI.
Collapse
Affiliation(s)
- Li Wen
- Kidney Research Institute, Department of Nephrology, West China Hospital of Sichuan University, Chengdu, 610041, China
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qian Ren
- Kidney Research Institute, Department of Nephrology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Fan Guo
- Kidney Research Institute, Department of Nephrology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Xiaoyan Du
- Department of Pharmacy, West China Hospital, Chengdu, 610041, China
| | - Hongliu Yang
- Kidney Research Institute, Department of Nephrology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Ping Fu
- Kidney Research Institute, Department of Nephrology, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Liang Ma
- Kidney Research Institute, Department of Nephrology, West China Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
6
|
Cheng Y, Yang R, Zhou Y, Wang J, Zhang T, Wang S, Li H, Jiang W, Zhang X. HBP1 inhibits the development of type 2 diabetes mellitus through transcriptional activation of the IGFBP1 gene. Aging (Albany NY) 2022; 14:8763-8782. [DOI: 10.18632/aging.204364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/21/2022] [Indexed: 11/22/2022]
Affiliation(s)
- Yuning Cheng
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Ruixiang Yang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Yue Zhou
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Jiyin Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Tongjia Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Shujie Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Hui Li
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Wei Jiang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Xiaowei Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, P. R. China
| |
Collapse
|
7
|
Traweek RS, Cope BM, Roland CL, Keung EZ, Nassif EF, Erstad DJ. Targeting the MDM2-p53 pathway in dedifferentiated liposarcoma. Front Oncol 2022; 12:1006959. [PMID: 36439412 PMCID: PMC9684653 DOI: 10.3389/fonc.2022.1006959] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/19/2022] [Indexed: 10/12/2023] Open
Abstract
Dedifferentiated liposarcoma (DDLPS) is an aggressive adipogenic cancer with poor prognosis. DDLPS tumors are only modestly sensitive to chemotherapy and radiation, and there is a need for more effective therapies. Genetically, DDLPS is characterized by a low tumor mutational burden and frequent chromosomal structural abnormalities including amplification of the 12q13-15 chromosomal region and the MDM2 gene, which are defining features of DDLPS. The MDM2 protein is an E3 ubiquitin ligase that targets the tumor suppressor, p53, for proteasomal degradation. MDM2 amplification or overexpression in human malignancies is associated with cell-cycle progression and worse prognosis. The MDM2-p53 interaction has thus garnered interest as a therapeutic target for DDLPS and other malignancies. MDM2 binds p53 via a hydrophobic protein interaction that is easily accessible with synthetic analogues. Multiple agents have been developed, including Nutlins such as RG7112 and small molecular inhibitors including SAR405838 and HDM201. Preclinical in vitro and animal models have shown promising results with MDM2 inhibition, resulting in robust p53 reactivation and cancer cell death. However, multiple early-phase clinical trials have failed to show a benefit with MDM2 pathway inhibition for DDLPS. Mechanisms of resistance are being elucidated, and novel inhibitors and combination therapies are currently under investigation. This review provides an overview of these strategies for targeting MDM2 in DDLPS.
Collapse
Affiliation(s)
- Raymond S. Traweek
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Brandon M. Cope
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Christina L. Roland
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Emily Z. Keung
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Elise F. Nassif
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Derek J. Erstad
- Division of Surgical Oncology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
8
|
Huang W, Hickson LJ, Eirin A, Kirkland JL, Lerman LO. Cellular senescence: the good, the bad and the unknown. Nat Rev Nephrol 2022; 18:611-627. [PMID: 35922662 PMCID: PMC9362342 DOI: 10.1038/s41581-022-00601-z] [Citation(s) in RCA: 522] [Impact Index Per Article: 174.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2022] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a ubiquitous process with roles in tissue remodelling, including wound repair and embryogenesis. However, prolonged senescence can be maladaptive, leading to cancer development and age-related diseases. Cellular senescence involves cell-cycle arrest and the release of inflammatory cytokines with autocrine, paracrine and endocrine activities. Senescent cells also exhibit morphological alterations, including flattened cell bodies, vacuolization and granularity in the cytoplasm and abnormal organelles. Several biomarkers of cellular senescence have been identified, including SA-βgal, p16 and p21; however, few markers have high sensitivity and specificity. In addition to driving ageing, senescence of immune and parenchymal cells contributes to the development of a variety of diseases and metabolic disorders. In the kidney, senescence might have beneficial roles during development and recovery from injury, but can also contribute to the progression of acute kidney injury and chronic kidney disease. Therapies that target senescence, including senolytic and senomorphic drugs, stem cell therapies and other interventions, have been shown to extend lifespan and reduce tissue injury in various animal models. Early clinical trials confirm that senotherapeutic approaches could be beneficial in human disease. However, larger clinical trials are needed to translate these approaches to patient care.
Collapse
Affiliation(s)
- Weijun Huang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - LaTonya J Hickson
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, FL, USA
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
9
|
Wang J, Yang R, Cheng Y, Zhou Y, Zhang T, Wang S, Li H, Jiang W, Zhang X. Methylation of HBP1 by PRMT1 promotes tumor progression by regulating actin cytoskeleton remodeling. Oncogenesis 2022; 11:45. [PMID: 35941115 PMCID: PMC9360041 DOI: 10.1038/s41389-022-00421-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/09/2022] Open
Abstract
HBP1 is a sequence-specific transcription factor which generally considered as a crucial growth inhibitor. Posttranslational modification of HBP1 is vital for its function. In this study, we demonstrate that HBP1 is methylated at R378 by PRMT1, which decreases HBP1 protein stability by promoting its ubiquitination and proteasome-mediated degradation. PRMT1-mediated methylation of HBP1 alleviates the repressive effects of HBP1 on tumor metastasis and growth. GSN is identified as a novel target gene of HBP1. Methylation of HBP1 promotes actin cytoskeleton remodeling, glycolysis and tumor progression by downregulating GSN (a vital actin-binding protein) levels. The methylated HBP1-GSN axis is associated with the clinical outcomes of cancer patients. This investigation elucidates the mechanism of how methylated HBP1 facilitates actin cytoskeleton remodeling, thus attenuates its tumor-suppressive function and promotes tumor progression. Targeting methylated HBP1-GSN axis may provide a therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Jiyin Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Ruixiang Yang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Yuning Cheng
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Yue Zhou
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Tongjia Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Shujie Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Hui Li
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Wei Jiang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Xiaowei Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China.
| |
Collapse
|
10
|
Cao Z, Cheng Y, Wang J, Liu Y, Yang R, Jiang W, Li H, Zhang X. HBP1-mediated transcriptional repression of AFP inhibits hepatoma progression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:118. [PMID: 33794968 PMCID: PMC8015059 DOI: 10.1186/s13046-021-01881-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/15/2021] [Indexed: 12/31/2022]
Abstract
Background Hepatoma is a common malignancy of the liver. The abnormal high expression of alpha-fetoprotein (AFP) is intimately associated with hepatoma progress, but the mechanism of transcriptional regulation and singularly activation of AFP gene in hepatoma is not clear. Methods The expression of transcription factor HBP1 and AFP and clinical significance were further analyzed in hepatoma tissues from the patients who received surgery or TACE and then monitored for relapse for up 10 years. HBP1-mediated transcriptional regulation of AFP was analyzed by Western blotting, Luciferase assay, Realtime-PCR, ChIP and EMSA. After verified the axis of HBP-AFP, its impact on hepatoma was measured by MTT, Transwell and FACS in hepatoma cells and by tumorigenesis in HBP1−/− mice. Results The relative expressions of HBP1 and AFP correlated with survival and prognosis in hepatoma patients. HBP1 repressed the expression of AFP gene by directly binding to the AFP gene promoter. Hepatitis B Virus (HBV)-encoded protein HBx promoted malignancy in hepatoma cells through binding to HBP1 directly. Icaritin, an active ingredient of Chinese herb epimedium, inhibited malignancy in hepatoma cells through enhancing HBP1 transrepression of AFP. The repression of AFP by HBP1 attenuated AFP effect on PTEN, MMP9 and caspase-3, thus inhibited proliferation and migration, and induced apoptosis in hepatoma cells. The deregulation of AFP by HBP1 contributed to hepatoma progression in mice. Conclusions Our data clarify the mechanism of HBP1 in inhibiting the expression of AFP and its suppression in malignancy of hepatoma cells, providing a more comprehensive theoretical basis and potential solutions for the diagnosis and treatment of hepatoma. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01881-2.
Collapse
Affiliation(s)
- Zhengyi Cao
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, People's Republic of China.,Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, 100029, People's Republic of China
| | - Yuning Cheng
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, People's Republic of China
| | - Jiyin Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, People's Republic of China
| | - Yujuan Liu
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, People's Republic of China
| | - Ruixiang Yang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, People's Republic of China
| | - Wei Jiang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, People's Republic of China
| | - Hui Li
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, People's Republic of China.
| | - Xiaowei Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, People's Republic of China.
| |
Collapse
|
11
|
Li H, Liu Y, Jiang W, Xue J, Cheng Y, Wang J, Yang R, Zhang X. Icaritin promotes apoptosis and inhibits proliferation by down-regulating AFP gene expression in hepatocellular carcinoma. BMC Cancer 2021; 21:318. [PMID: 33765973 PMCID: PMC7992931 DOI: 10.1186/s12885-021-08043-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/15/2021] [Indexed: 11/10/2022] Open
Abstract
Background Icaritin, an active ingredient of the Chinese herb Epimedium, plays an anti-tumor role in liver cancer by inhibiting the proliferation of hepatocellular cells and promoting their apoptosis. In China, phase II and a large phase III clinical trial of icaritin reagent for the treatment of hepatocellular cancer is under-going, but the specific mechanism of icaritin action was unclear. Alpha-fetoprotein (AFP), an oncofetal protein, produced in the healthy fetal liver and yolk sac. Intracellular AFP promoted cellular proliferation and inhibited cellular apoptosis in hepatocellular carcinoma (HCC). The study was aimed to investigate the effect of icaritin on HCC through p53/AFP pathway. Methods Real-time RT PCR and western blot were used to detect p53 and AFP expression levels in HCC cells treated with icaritin. The mechanism of icaritin affecting p53 expression was verified by ubiquitination experiment, and the binding activity of icaritin on p53 in AFP promoter region was verified by luciferase experiment. EdU, MTT and flow cytometry were used to determine whether icaritin affected HCC cellular proliferation and apoptosis through p53/ AFP pathway. Expression levels of p53 and AFP in xenograft mouse model were determined by western blotting. Results Our results showed icaritin inhibited AFP expression at mRNA and protein level. AFP was also identified as the target gene of the p53 transcription factor. Icaritin abrogated murine double minute (Mdm) 2-mediated p53 ubiquitination degradation to improve the stability of p53. Up-regulated p53 protein levels then transcriptionally inhibited the AFP promoter. Icaritin-mediated decrease of AFP through Mdm2/p53 pathways inhibited HCC cellular proliferation and promoted HCC cellular apoptosis. Conclusion Our findings revealed the mechanism of icaritin in promoting apoptosis and inhibiting proliferation in liver cancer cells. The regulatory mechanism of icaritin in AFP protein down-regulation provides a theoretical and experimental basis for further research into new drugs for the treatment of liver cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08043-9.
Collapse
Affiliation(s)
- Hui Li
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 100191, Beijing, People's Republic of China.
| | - Yujuan Liu
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 100191, Beijing, People's Republic of China
| | - Wei Jiang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 100191, Beijing, People's Republic of China
| | - Junhui Xue
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 100191, Beijing, People's Republic of China
| | - Yuning Cheng
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 100191, Beijing, People's Republic of China
| | - Jiyin Wang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 100191, Beijing, People's Republic of China
| | - Ruixiang Yang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 100191, Beijing, People's Republic of China
| | - Xiaowei Zhang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, 100191, Beijing, People's Republic of China
| |
Collapse
|
12
|
Soni P, Ghufran MS, Olakkaran S, Puttaswamygowda GH, Duddukuri GR, Kanade SR. Epigenetic alterations induced by aflatoxin B 1: An in vitro and in vivo approach with emphasis on enhancer of zeste homologue-2/p21 axis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 762:143175. [PMID: 33131875 DOI: 10.1016/j.scitotenv.2020.143175] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/21/2020] [Accepted: 10/14/2020] [Indexed: 06/11/2023]
Abstract
The potent environmental toxicant aflatoxin B1 (AFB1), is a group I carcinogen reported to induce the expression of many cancer associated proteins. Epigenetic alterations such as DNA methylation and histone modifications play vital role in AFB1-mediated carcinogenesis. These epigenetic modifications may result in the recruitment of specific proteins and transcription factors to the promoter region and regulate gene expression. Here we show that AFB1, at lower concentrations (100 and 1000 nM) induced proliferation in L-132 and HaCaT cells with activation of the Akt pathway, which ultimately steered abnormal proliferation and transmission of survival signals. We demonstrated a significant reduction in the expression of p21 with a remarkable increase in the expression of cyclin D1 that correlated with increased methylation of CpG dinucleotides in p21 proximal promoter, while cyclin D1 promoter remained unmethylated. The chromatin immunoprecipitation results revealed the enrichment of DNMT3a and H3K27me3 repressive marks on the p21 proximal promoter where EZH2 mediated H3K27me3 mark enhanced the binding of DNMT3a at the promoter and further contributed to the transcriptional inactivation. The overall study provided the novel information on the impact of AFB1 on p21 inactivation via EZH2 and promoter methylation which is known to be a vital process in proliferation. Furthermore, AFB1 induced the expression of EZH2 analogue protein E(z), cyclin D1 analogue cyclin D and decreased the expression of p21 analogue Dacapo in Drosophila melanogaster. Interestingly, the aggressiveness in their expression upon re-exposure in successive generations suggested first hand perspectives on multigenerational epigenetic memory.
Collapse
Affiliation(s)
- Priyanka Soni
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periye, Kasargod 671316, Kerala, India
| | - Md Sajid Ghufran
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periye, Kasargod 671316, Kerala, India
| | - Shilpa Olakkaran
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periye, Kasargod 671316, Kerala, India
| | | | - Govinda Rao Duddukuri
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periye, Kasargod 671316, Kerala, India
| | - Santosh R Kanade
- Department of Plant Science, School of Life Science, University of Hyderabad, Prof. C. R. Rao Road Gachibowli, Hyderabad 500046, India.
| |
Collapse
|
13
|
Ren B, Liu J, Wu K, Zhang J, Lv Y, Wang S, Liu L, Liu D. TNF-α-elicited miR-29b potentiates resistance to apoptosis in peripheral blood monocytes from patients with rheumatoid arthritis. Apoptosis 2020; 24:892-904. [PMID: 31473844 DOI: 10.1007/s10495-019-01567-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CD14-positive monocytes from patients with rheumatoid arthritis (RA) are more resistant to apoptosis, which promotes their persistence at the inflammatory site and thereby contributes crucially to immunopathology. We sought to elucidate one mechanism underlying this unique pathogenesis: resistance to apoptosis and the potential involvement of miR-29b in this process. CD14-positive peripheral blood monocytes (PBMs) from RA patients were observed to be resistant to spontaneous apoptosis compared to PBMs from healthy volunteers. Intriguingly, expression of miR-29b was significantly upregulated in PBMs from RA patients than those from healthy volunteers, and this upregulation was correlated with RA disease activity. Functionally, forced expression of the exogenous miR-29b in CD14-positive Ctrl PBMs conferred resistance to spontaneous apoptosis and Fas-induced death, thereafter enhancing the production of major proinflammatory cytokines in there cells. Following identification of the potential miR-29b target transcripts using bioinformatic algorithms, we showed that miR-29b could directly bind to the 3'-UTR of the high-mobility group box-containing protein 1 (HBP1) and inhibited its transcription in PBMs. Importantly, stable expression of the exogenous HBP1 in differentiated THP-1 monocytes effectively abolished miR-29b-elicited resistance to Fas-induced apoptosis. Finally, among patients with RA and good clinical responses to immunotherapy, expression levels of miR-29b were significantly compromised in those treated with infliximab (a TNF-α inhibitor) but not in those treated with tocilizumab (a humanized mAb against the IL-6 receptor), pointing to a potential association between miR-29b activation and TNF-α induction. The available data collectively suggest that TNF-α-elicited miR-29b potentiates resistance to apoptosis in PBMs from RA patients via inhibition of HBP1 signaling, and testing patients for miR-29b/HBP1 expression ratios may provide more accurate prognostic information and could influence the recommended course of immunotherapy.
Collapse
Affiliation(s)
- Baodi Ren
- Department of Rheumatology and Immunology, Xi'an Institute of Rheumatology, Xi'an No.5 Hospital, No. 112 XiGuanZhengJie, Lian Hu District, Xi'an, 710082, Shaanxi Province, China.,Department of Rheumatology and Immunology, Shaanxi Hospital of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Shaanxi University of Chinese Medicine, No. 112 XiGuanZhengJie, Lian Hu District, Xi'an, 710082, Shaanxi Province, China
| | - Jiayu Liu
- Department of Rheumatology and Immunology, Xi'an Institute of Rheumatology, Xi'an No.5 Hospital, No. 112 XiGuanZhengJie, Lian Hu District, Xi'an, 710082, Shaanxi Province, China.,Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, No.157 XiWu Road, Xincheng District, Xi'an, 710004, China
| | - Kunyi Wu
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, No.157 XiWu Road, Xincheng District, Xi'an, 710004, China
| | - Junli Zhang
- Department of Rheumatology and Immunology, Xi'an Institute of Rheumatology, Xi'an No.5 Hospital, No. 112 XiGuanZhengJie, Lian Hu District, Xi'an, 710082, Shaanxi Province, China.,Department of Rheumatology and Immunology, Shaanxi Hospital of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Shaanxi University of Chinese Medicine, No. 112 XiGuanZhengJie, Lian Hu District, Xi'an, 710082, Shaanxi Province, China
| | - Yanyan Lv
- Department of Rheumatology and Immunology, Xi'an Institute of Rheumatology, Xi'an No.5 Hospital, No. 112 XiGuanZhengJie, Lian Hu District, Xi'an, 710082, Shaanxi Province, China
| | - Suzhi Wang
- Department of Rheumatology and Immunology, Xi'an Institute of Rheumatology, Xi'an No.5 Hospital, No. 112 XiGuanZhengJie, Lian Hu District, Xi'an, 710082, Shaanxi Province, China
| | - Liping Liu
- Department of Rheumatology and Immunology, Xi'an Institute of Rheumatology, Xi'an No.5 Hospital, No. 112 XiGuanZhengJie, Lian Hu District, Xi'an, 710082, Shaanxi Province, China.,Department of Rheumatology and Immunology, Shaanxi Hospital of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Shaanxi University of Chinese Medicine, No. 112 XiGuanZhengJie, Lian Hu District, Xi'an, 710082, Shaanxi Province, China
| | - Dan Liu
- Department of Rheumatology and Immunology, Xi'an Institute of Rheumatology, Xi'an No.5 Hospital, No. 112 XiGuanZhengJie, Lian Hu District, Xi'an, 710082, Shaanxi Province, China. .,Department of Rheumatology and Immunology, Shaanxi Hospital of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Shaanxi University of Chinese Medicine, No. 112 XiGuanZhengJie, Lian Hu District, Xi'an, 710082, Shaanxi Province, China. .,Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, No.157 XiWu Road, Xincheng District, Xi'an, 710004, China.
| |
Collapse
|
14
|
Yang JQ, Wu M, Han FY, Sun YM, Zhang L, Liu HX. High risk HPV detection by RNAscope in situ hybridization combined with Cdc2 protein expression by immunohistochemistry for prognosis of oropharyngeal squamous cell carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:2192-2200. [PMID: 32922620 PMCID: PMC7476946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 06/29/2020] [Indexed: 06/11/2023]
Abstract
High risk human papillomavirus (HPV) infection is related to the development of head and neck squamous cell carcinoma (HNSCC). Oropharyngeal squamous cell carcinoma (OPSCC) is a common type of HNSCC, and its incidence has increased significantly in recent years. In this study, high risk HPV, the expression of P53, P21, and Cdc2 in OPSCC tissues was detected and the prognostic factors and clinical value of OPSCC were discussed. According to the WHO classification and diagnosis standard for head and neck tumors (2017 Edition), 49 OPSCC cases with complete clinical data were collected from Tangshan Head and Neck Disease Pathology Research Base from January 1, 2012 to December 31, 2018. The E6 and E7 mRNA of HPV 16 and HPV 18 were detected by RNAscope in situ hybridization. The expression of P53, P21, and Cdc2 protein was observed by SP immunohistochemical method and all cases were followed up for survival. Median survival time was analyzed by Kaplan-Meier method. The Log-rank test was used for single factor analysis and Cox regression model was used to analyze multiple prognostic factors. In 49 OPSCC cases the median age was 53 years; 14 were HPV-DNA positive (14/49, 28.6%) while 35 were negative (35/49, 71.4%). E6, E7 mRNA test showed that 20 cases (20/49, 40.8%) were positive for HPV-16. Among them 11 cases were positive for HPV-16 DNA. 2 cases were positive for HPV-18 mRNA (2/49, 4.08%). 27 cases were negative for mRNA16 and 18 (27/49, 55.1%). The prevalence of HPV was 68.8% (11/16) in the non-smoking group, which was higher than that of the smoking group (10/33, 33.3%), (χ2=5.463, P=0.019). There was no significant correlation between HPV detection and gender, age, drinking, tumor differentiation degree, and clinical stage (P > 0.05). The expression rates of P53, P21, and Cdc2 in OPSCC tissues were 63.3% (31/49), 65.3% (32/49), and 67.3% (33/49), respectively. There was no significant correlation between expression of all the three proteins and gender, age, HPV, smoking, drinking, tumor differentiation, and clinical stage (P > 0.05). Cox multifactor regression analysis showed that HPV (HR=0.275, 95% CI: 0.146-0.517), tumor differentiation (HR=1.751, 95% CI: 1.231-2.492), stage (HR=3.268, 95% CI: 1.758-6.074) and expression of Cdc2 protein (HR=1.804, 95% CI: 0.990-3.286) were related to the survival time of patients (P < 0.05). Our findings support that most of the HPV-positive OPSSC patients were non-smokers. The patients with negative HPV, low differentiation, late stage, and Cdc2 positive expression have poor prognosis and need to be followed up.
Collapse
Affiliation(s)
- Jun-Quan Yang
- Department of Radio-Chemotherapy Oncology, Tangshan People’s HospitalTangshan, P. R. China
| | - Meng Wu
- Department of Pathology, Division of Basic Medicine, Tangshan Vocational and Technical CollegeTangshan, P. R. China
| | - Feng-Yan Han
- Department of Pathology, Tangshan Union HospitalTangshan, P. R. China
| | - Yu-Man Sun
- Department of Pathology, Tangshan Union HospitalTangshan, P. R. China
| | - Ling Zhang
- Department of Pathology, Tangshan Union HospitalTangshan, P. R. China
| | - Hong-Xia Liu
- Department of Pathology, Tangshan Union HospitalTangshan, P. R. China
| |
Collapse
|
15
|
Fu T, Liang A, Liu Y. [Role of P21 in Resistance of Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:597-602. [PMID: 32434295 PMCID: PMC7406443 DOI: 10.3779/j.issn.1009-3419.2020.101.16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lung cancer is the most common malignant tumor in the world with the highest incidence of deaths. In recent years, the treatment of lung cancer has made a significant breakthrough. However, as the tumor progresses, lung cancer cells inevitably acquire resistance and the efficacy of the treatment are greatly reduced. P21 protein plays a dual role in tumors, which not only regulates the cell cycle, induces apoptosis, inhibits cell proliferation, but also protects cells against apoptosis and promotes tumor cell resistance. This article reviews the research on P21 and lung cancer resistance, to provide new ideas for individualized treatment of lung cancer and overcoming lung cancer resistance.
Collapse
Affiliation(s)
- Tian Fu
- Medical Molecular Diagnostics Key Laboratory of Guangdong, Dongguan 523808, China.,Department of Biochemistry and Molecular Biology and Department of Clinical Biochemistry in Guangdong Medical University, Dongguan 523808, China
| | - Ailing Liang
- Medical Molecular Diagnostics Key Laboratory of Guangdong, Dongguan 523808, China.,Department of Clinical Laboratory Biochemistry of Guangdong Medical University, Dongguan 523808, China
| | - Yongjun Liu
- Medical Molecular Diagnostics Key Laboratory of Guangdong, Dongguan 523808, China.,Department of Biochemistry and Molecular Biology and Department of Clinical Biochemistry in Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
16
|
Fan Z, Li L, Li X, Zhang M, Dou M, Zhao J, Cao J, Deng X, Zhang M, Li H, Suo Z. Anti-senescence role of heterozygous fumarate hydratase gene knockout in rat lung fibroblasts in vitro. Aging (Albany NY) 2020; 11:573-589. [PMID: 30668541 PMCID: PMC6366963 DOI: 10.18632/aging.101761] [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: 09/17/2018] [Accepted: 01/05/2019] [Indexed: 12/21/2022]
Abstract
Abnormalities in tricarboxylic acid (TCA) cycle function were related to a variety of pathological processes. Fumarate hydratase (FH) is a required enzyme in the TCA cycle. To explore the general influence of FH knockout, we isolated FH+/- rat and normal rat lung fibroblasts and cultured these cells in vitro. The isolated fibroblasts with the current method were rather homogeneous and were confirmed spindle in morphology, positive for vimentin and negative for α-SMA (α-smooth muscle actin). Sequencing of the PCR (polymerase chain reaction) products flanking the FH gene mutation verified the FH+/- status, and the FH gene and protein expression were confirmed to be reduced in the FH+/- cells. No sign of ageing for the FH+/- cells after 61 passages was observed, but the controls died out at this stage. Flow cytometry revealed increased S-phase and decreased G1/G0 proportions with significantly less early apoptosis in FH+/- cells compared to that in control cells. At the same time, increased glucose consumption, intracellular fumarate production and extracellular lactate secretion were verified in the FH+/- cells. Correspondingly, FH+/- cells showed a lower basal oxygen consumption rate (OCR) but a higher level of reactive oxygen species (ROS) production. Single cell cloning and cell line establishment were successfully performed with the FH+/- cells at the 84th passage. All the above results indicate an important role for FH+/- in the longevity or immortality of the FH+/- cells, in which increased p53 and TERT (telomerase reverse transcriptase) protein expression, decreased p21 and p16 protein expression and negative SA-β-Gal (senescence-associated beta-galactosidase) were verified along with metabolic reprogramming.
Collapse
Affiliation(s)
- Zhirui Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Lifeng Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xiaoli Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Meng Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Mengmeng Dou
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jing Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jing Cao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China.,Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xiaoming Deng
- Department of Chinese and Western Integrative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Huixiang Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Zhenhe Suo
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China.,Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Montebello, Oslo, Norway
| |
Collapse
|
17
|
EZH2 Overexpression in Multiple Myeloma: Prognostic Value, Correlation With Clinical Characteristics, and Possible Mechanisms. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 19:744-750. [DOI: 10.1016/j.clml.2019.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/18/2019] [Accepted: 08/22/2019] [Indexed: 12/11/2022]
|
18
|
Bai Y, Li S. Long noncoding RNA OIP5-AS1 aggravates cell proliferation, migration in gastric cancer by epigenetically silencing NLRP6 expression via binding EZH2. J Cell Biochem 2019; 121:353-362. [PMID: 31219209 DOI: 10.1002/jcb.29183] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/29/2019] [Indexed: 01/01/2023]
Abstract
The critical role of long noncoding RNAs (lncRNAs) in the development of multiple cancers has been revealed either functioning as a tumor initiator or a cancer suppressor. A widely recognized OIP5 antisense RNA 1 (lncRNA OIP5-AS1) has been validated to be an essential regulator of the tumorigenesis of various malignancies. Whereas, the potential role and the exact mechanism of lncRNA OIP5-AS1 by which OIP5-AS1 mediates gastric cancer (GC) progression remains vague. Therefore, first our work probed its expression levels in GC cell lines and related normal cells by real-time quantitative polymerase chain reaction. The heightened level of OIP5-AS1 was detected in GC cell lines. In terms of its cellular effects, we performed a series of functional experiments and as presented in the assays, the proliferative potential and motility was diminished. However, more apoptotic cells were induced with the introduction of OIP5-AS1 silencing. Meanwhile, higher Nod-like receptor pyrin domain-containing protein 6 (NLRP6) and enhancer of zeste homolog 2 (EZH2) expression in the GC cells was monitored. Besides, OIP5-AS1 was disclosed to locate mainly in the nucleus. In terms of mechanism, OIP5-AS1 directly bound to EZH2 and obstructed NLRP6 expression, speeding up GC progression.
Collapse
Affiliation(s)
- Yunlei Bai
- Department of GI Medicine, The First Hospital of Yulin, Yulin, Shaanxi, China
| | - Sheng Li
- Department of General Surgery, Yulin No. 2 Hospital, Yulin, Shaanxi, China
| |
Collapse
|
19
|
Bollaert E, de Rocca Serra A, Demoulin JB. The HMG box transcription factor HBP1: a cell cycle inhibitor at the crossroads of cancer signaling pathways. Cell Mol Life Sci 2019; 76:1529-1539. [PMID: 30683982 PMCID: PMC11105191 DOI: 10.1007/s00018-019-03012-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/20/2018] [Accepted: 01/15/2019] [Indexed: 12/19/2022]
Abstract
HMG box protein 1 (HBP1) is a transcription factor and a potent cell cycle inhibitor in normal and cancer cells. HBP1 activates or represses the expression of different cell cycle genes (such as CDKN2A, CDKN1A, and CCND1) through direct DNA binding, cofactor recruitment, chromatin remodeling, or neutralization of other transcription factors. Among these are LEF1, TCF4, and MYC in the WNT/beta-catenin pathway. HBP1 also contributes to oncogenic RAS-induced senescence and terminal cell differentiation. Collectively, these activities suggest a tumor suppressor function. However, HBP1 is not listed among frequently mutated cancer driver genes. Nevertheless, HBP1 expression is lower in several tumor types relative to matched normal tissues. Several micro-RNAs, such as miR-155, miR-17-92, and miR-29a, dampen HBP1 expression in cancer cells of various origins. The phosphatidylinositol-3 kinase (PI3K)/AKT pathway also inhibits HBP1 transcription by preventing FOXO binding to the HBP1 promoter. In addition, AKT directly phosphorylates HBP1, thereby inhibiting its transcriptional activity. Taken together, these findings place HBP1 at the center of a network of micro-RNAs and oncoproteins that control cell proliferation. In this review, we discuss our current understanding of HBP1 function in human physiology and diseases.
Collapse
Affiliation(s)
- Emeline Bollaert
- Université Catholique de Louvain, de Duve Institute, Avenue Hippocrate 75, 1200, Brussels, Belgium
| | - Audrey de Rocca Serra
- Université Catholique de Louvain, de Duve Institute, Avenue Hippocrate 75, 1200, Brussels, Belgium
| | - Jean-Baptiste Demoulin
- Université Catholique de Louvain, de Duve Institute, Avenue Hippocrate 75, 1200, Brussels, Belgium.
| |
Collapse
|
20
|
Cao Z, Xue J, Cheng Y, Wang J, Liu Y, Li H, Jiang W, Li G, Gui Y, Zhang X. MDM2 promotes genome instability by ubiquitinating the transcription factor HBP1. Oncogene 2019; 38:4835-4855. [PMID: 30816344 PMCID: PMC6756050 DOI: 10.1038/s41388-019-0761-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/09/2018] [Accepted: 02/10/2019] [Indexed: 12/26/2022]
Abstract
Genome instability is a common feature of tumor cells, and the persistent presence of genome instability is a potential mechanism of tumorigenesis. The E3 ubiquitin ligase MDM2 is intimately involved in genome instability, but its mechanisms are unclear. Our data demonstrated that the transcription factor HBP1 is a target of MDM2. MDM2 facilitates HBP1 proteasomal degradation by ubiquitinating HBP1, regardless of p53 status, thus attenuating the transcriptional inhibition of HBP1 in the expression of its target genes, such as the DNA methyltransferase DNMT1 and histone methyltransferase EZH2, which results in global DNA hypermethylation and histone hypermethylation and ultimately genome instability. The repression of HBP1 by MDM2 finally promotes cell growth and tumorigenesis. Next, we thoroughly explored the regulatory mechanism of the MDM2/HBP1 axis in DNA damage repair following ionizing radiation. Our data indicated that MDM2 overexpression-mediated repression of HBP1 delays DNA damage repair and causes cell death in a p53-independent manner. This investigation elucidated the mechanism of how MDM2 promotes genome instability and enhances tumorigenesis in the absence of p53, thus providing a theoretical and experimental basis for targeting MDM2 as a cancer therapy.
Collapse
Affiliation(s)
- Zhengyi Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Junhui Xue
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Yuning Cheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Jiyin Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Yujuan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Hui Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Wei Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Gang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Yaoting Gui
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, 518000, P. R. China
| | - Xiaowei Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing, 100191, P. R. China.
| |
Collapse
|
21
|
Peng X, Zha L, Chen A, Wang Z. HOXA5 is a tumor suppressor gene that is decreased in gastric cancer. Oncol Rep 2018; 40:1317-1329. [PMID: 30015922 PMCID: PMC6072397 DOI: 10.3892/or.2018.6537] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 05/31/2018] [Indexed: 12/15/2022] Open
Abstract
The abnormal expression of homeobox A5 (HOXA5) has been observed in breast and colon cancer; however, the clinical significance of HOXA5 in gastric cancer (GC) is not yet clear. In this study, we found that HOXA5 expression was decreased in GC tissues at the mRNA and protein level compared with paracancerous tissues using reverse transcription‑quantitative PCR (RT‑qPCR) and western blot analysis, respectively. Immunohistochemistry and Kaplan‑Meier survival analysis confirmed that the underexpression of HOXA5 was associated with GC progression and indicated a poor prognosis of patients with GC. Given that proliferation‑related genes may be potential target genes of HOXA5, we performed a series of experiments in vitro to examine the effects of HOXA5 on the proliferation of GC cells. A CCK‑8 assay, colony formation assay and flow cytometry revealed that HOXA5 inhibited the abnormal proliferation of GC cells, and this finding was further supported by a 5‑ethynyl‑2'‑deoxyuridine (EdU) assay. Further mechanistic investigation clarified that HOXA5 promoted the protein expression of p21 and inhibited the protein expression of c‑Myc and Ki67. Additionally, the use of nude mouse models also verified that HOXA5 suppressed the proliferation of GC cells in vivo. Collectively, the findings of this study demonstrate that HOXA5 acts as a tumor suppressor gene during the development and progresion of GC, possibly functioning by inhibiting the abnormal proliferation of cancer cells.
Collapse
Affiliation(s)
- Xudong Peng
- Gastrointestinal Surgical Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400000, P.R. China
| | - Lang Zha
- Gastrointestinal Surgical Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400000, P.R. China
| | - Anqi Chen
- Gastrointestinal Surgical Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400000, P.R. China
| | - Ziwei Wang
- Gastrointestinal Surgical Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400000, P.R. China
| |
Collapse
|
22
|
Su C, Cheng X, Li Y, Han Y, Song X, Yu D, Cao X, Liu Z. MiR-21 improves invasion and migration of drug-resistant lung adenocarcinoma cancer cell and transformation of EMT through targeting HBP1. Cancer Med 2018; 7:2485-2503. [PMID: 29663730 PMCID: PMC6010699 DOI: 10.1002/cam4.1294] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/22/2017] [Accepted: 11/26/2017] [Indexed: 12/12/2022] Open
Abstract
This study was aimed at the investigation of the effects of miR-21 on drug resistance, invasion, migration, and epithelial-mesenchymal transition (EMT) of lung adenocarcinoma cells and the related molecular mechanisms. Cell viability of A549 cell line was measured by MTT assay. Wound healing assay and transwell assay were, respectively, employed to examine cell migration and invasion abilities. The cells were transfected with miR-21 mimic or inhibitor using Lipofectamine 3000. The target relationship between miR-21 and HBP1 was confirmed by luciferase reporter gene assay. Western blot and qRT-PCR were used to examine the expression of HBP1 and EMT-related molecules. Compared with A549 cells, drug resistance of A549/PTX cells and A549/DDP cells were obviously stronger. A549/PTX cells and A549/DDP cells had stronger ability of migration and invasion compared with parental A549 cells. Meanwhile, EMT of A549/PTX and A549/DDP was significantly higher than that of A549 cells. MiR-21 promoted migration, invasion, and EMT of human lung adenocarcinoma cancer cells. Our experiment also verified the target relationship between miR-21 and HBP1. MiR-21 may affect migration and invasion ability of drug-resistant lung adenocarcinoma cells by targeting HBP1, therefore modulating EMT.
Collapse
Affiliation(s)
- Chongyu Su
- Department of Thoracic SurgeryBeijing Chest HospitalCapital Medical UniversityBeijing101149China
| | - Xu Cheng
- Department of Thoracic SurgeryBeijing Chest HospitalCapital Medical UniversityBeijing101149China
| | - Yunsong Li
- Department of Thoracic SurgeryBeijing Chest HospitalCapital Medical UniversityBeijing101149China
| | - Yi Han
- Department of Thoracic SurgeryBeijing Chest HospitalCapital Medical UniversityBeijing101149China
| | - Xiaoyun Song
- Department of Thoracic SurgeryBeijing Chest HospitalCapital Medical UniversityBeijing101149China
| | - Daping Yu
- Department of Thoracic SurgeryBeijing Chest HospitalCapital Medical UniversityBeijing101149China
| | - Xiaoqing Cao
- Department of Thoracic SurgeryBeijing Chest HospitalCapital Medical UniversityBeijing101149China
| | - Zhidong Liu
- Department of Thoracic SurgeryBeijing Chest HospitalCapital Medical UniversityBeijing101149China
| |
Collapse
|
23
|
Bollaert E, Johanns M, Herinckx G, de Rocca Serra A, Vandewalle VA, Havelange V, Rider MH, Vertommen D, Demoulin JB. HBP1 phosphorylation by AKT regulates its transcriptional activity and glioblastoma cell proliferation. Cell Signal 2018; 44:158-170. [PMID: 29355710 DOI: 10.1016/j.cellsig.2018.01.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/22/2017] [Accepted: 01/10/2018] [Indexed: 12/31/2022]
Abstract
The HMG-box protein 1 (HBP1) is a transcriptional regulator and a potential tumor suppressor that controls cell proliferation, differentiation and oncogene-mediated senescence. In a previous study, we showed that AKT activation through the PI3K/AKT/FOXO pathway represses HBP1 expression at the transcriptional level in human fibroblasts as well as in cancer cell lines. In the present study, we investigated whether AKT could also regulate HBP1 directly. First, AKT1 phosphorylated recombinant human HBP1 in vitro on three conserved sites, Ser380, Thr484 and Ser509. In living cells, we confirmed the phosphorylation of HBP1 on residues 380 and 509 using phospho-specific antibodies. HBP1 phosphorylation was induced by growth factors, such as EGF or IGF-1, which activated AKT. Conversely, it was blocked by treatment of cells with an AKT inhibitor (MK-2206) or by AKT knockdown. Next, we observed that HBP1 transcriptional activity was strongly modified by mutating its phosphorylation sites. The regulation of target genes such as DNMT1, P47phox, p16INK4A and cyclin D1 was also affected. HBP1 had previously been shown to limit glioma cell growth. Accordingly, HBP1 silencing by small-hairpin RNA increased human glioblastoma cell proliferation. Conversely, HBP1 overexpression decreased cell growth and foci formation. This effect was amplified by mutations that prevented phosphorylation by AKT, and blunted by mutations that mimicked phosphorylation. In conclusion, our results suggest that HBP1 phosphorylation by AKT blocks its functions as transcriptional regulator and tumor suppressor.
Collapse
Affiliation(s)
- Emeline Bollaert
- de Duve Institute, Université Catholique de Louvain (UCL), MEXP Unit, Avenue Hippocrate 75, Box B1.74.05, 1200 Brussels, Belgium
| | - Manuel Johanns
- de Duve Institute, Université Catholique de Louvain (UCL), PHOS Unit, Avenue Hippocrate 75, Box B1.74.05, 1200 Brussels, Belgium
| | - Gaëtan Herinckx
- de Duve Institute, Université Catholique de Louvain (UCL), PHOS Unit, Avenue Hippocrate 75, Box B1.74.05, 1200 Brussels, Belgium
| | - Audrey de Rocca Serra
- de Duve Institute, Université Catholique de Louvain (UCL), MEXP Unit, Avenue Hippocrate 75, Box B1.74.05, 1200 Brussels, Belgium
| | - Virginie A Vandewalle
- de Duve Institute, Université Catholique de Louvain (UCL), MEXP Unit, Avenue Hippocrate 75, Box B1.74.05, 1200 Brussels, Belgium
| | - Violaine Havelange
- de Duve Institute, Université Catholique de Louvain (UCL), MEXP Unit, Avenue Hippocrate 75, Box B1.74.05, 1200 Brussels, Belgium
| | - Mark H Rider
- de Duve Institute, Université Catholique de Louvain (UCL), PHOS Unit, Avenue Hippocrate 75, Box B1.74.05, 1200 Brussels, Belgium
| | - Didier Vertommen
- de Duve Institute, Université Catholique de Louvain (UCL), PHOS Unit, Avenue Hippocrate 75, Box B1.74.05, 1200 Brussels, Belgium
| | - Jean-Baptiste Demoulin
- de Duve Institute, Université Catholique de Louvain (UCL), MEXP Unit, Avenue Hippocrate 75, Box B1.74.05, 1200 Brussels, Belgium.
| |
Collapse
|
24
|
HMG-box transcription factor 1: a positive regulator of the G1/S transition through the Cyclin-CDK-CDKI molecular network in nasopharyngeal carcinoma. Cell Death Dis 2018; 9:100. [PMID: 29367693 PMCID: PMC5833394 DOI: 10.1038/s41419-017-0175-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 10/30/2017] [Accepted: 11/16/2017] [Indexed: 11/09/2022]
Abstract
HMG-box transcription factor 1 (HBP1) has been reported to be a tumor suppressor in diverse malignant carcinomas. However, our findings provide a conclusion that HBP1 plays a novel role in facilitating nasopharyngeal carcinoma (NPC) growth. The Kaplan-Meier analysis indicates that high expression HBP1 and low miR-29c expression both are negatively correlated with the overall survival rates of NPC patients. HBP1 knockdown inhibits cellular proliferation and growth, and arrested cells in G1 phase rather than affected cell apoptosis via flow cytometry (FCM) analysis. Mechanistically, HBP1 induces the expression of CCND1 and CCND3 levels by binding to their promoters, and binds to CDK4, CDK6 and p16INK4A promoters while not affects their expression levels. CCND1 and CCND3 promote CCND1-CDK4, CCND3-CDK6, and CDK2-CCNE1 complex formation, thus, E2F-1 and DP-1 are activated to accelerate the G1/S transition in the cell cycle. MiR-29c is down-regulated and correlated with NPC tumorigenesis and progression. Luciferase assays confirms that miR-29c binds to the 3' untranslated region (3'-UTR) of HBP1. Introduction of pre-miR-29c decreased HBP1 mRNA and protein levels. Therefore, the high endogenous HBP1 expression might be attributed to the low levels of endogenous miR-29c in NPC. In addition, HBP1 knockdown and miR-29c agomir administration both decrease xenograft growth in nude mice in vivo. It is firstly reported that HBP1 knockdown inhibited the proliferation and metastasis of NPC, which indicates that HBP1 functions as a non-tumor suppressor gene in NPC. This study provides a novel potential target for the prevention of and therapies for NPC.
Collapse
|
25
|
Chan CY, Yu P, Chang FT, Chen ZH, Lee MF, Huang CY. Transcription factor HMG box-containing protein 1 (HBP1) modulates mitotic clonal expansion (MCE) during adipocyte differentiation. J Cell Physiol 2017; 233:4205-4215. [PMID: 29030964 DOI: 10.1002/jcp.26237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 10/05/2017] [Indexed: 11/10/2022]
Abstract
Transcription factor HMG box-containing protein 1 (HBP1) has been found to be up-regulated in rat adipose tissue and differentiated preadipocyte; however, how HBP1 is involved in adipocyte formation remains unclear. In the present study, we demonstrated that under a standard differentiation protocol HBP1 expression fluctuates with down-regulation in the mitotic clonal expansion (MCE) stage followed by up-regulation in the terminal differentiation stage in both 3T3-L1 and MEF cell models. Also, HBP1 knockdown accelerated cell cycle progression in the MCE stage, but it impaired final adipogenesis. To gain further insight into the role of HBP1 in the MCE stage, we found that the HBP1 expression pattern is reciprocal to that of C/EBPβ, and ectopic expression of HBP1suppresses C/EBPβ expression. These data indicate that HBP1 functions as a negative regulator of MCE. In contrast, when HBP1 expression was gradually elevated along with a concomitant induction of C/EBPα at the end of the MCE, HBP1 knockdown leads to a significant reduction of C/EBPα expression, suggesting that HBP1-mediated C/EBPα expression may be needed for the termination of the cell cycle at the end of MCE for terminal differentiation. All told, our findings show that HBP1 is a key transcription factor in the already complicated regulatory cascade during adipocyte differentiation.
Collapse
Affiliation(s)
- Chien-Yi Chan
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Ping Yu
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Feng-Tzu Chang
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Zih-Hua Chen
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Ming-Fen Lee
- Department of Nutrition and Health Sciences, Chang Jung Christian University, Tainan, Taiwan
| | - Chun-Yin Huang
- Department of Nutrition, China Medical University, Taichung, Taiwan.,Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
26
|
Cao S, Shen D, Wang Y, Li L, Zhou L, Wang Y. Potential malignant transformation in the gastric mucosa of immunodeficient mice with persistent Mycoplasma penetrans infection. PLoS One 2017; 12:e0180514. [PMID: 28692662 PMCID: PMC5503272 DOI: 10.1371/journal.pone.0180514] [Citation(s) in RCA: 8] [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: 05/03/2017] [Accepted: 06/17/2017] [Indexed: 12/11/2022] Open
Abstract
Mycoplasma infection has been reported in immunocompromised cancer patients; nevertheless, it is not clear if persistent Mycoplasma infection could facilitate the proliferation of cancer cells in immunocompromised organisms. The aim of this study was to examine the relationship between persistent Mycoplasma infection and malignant transformation in an immunodeficient host model. Immunodeficient mouse model was established using cyclophosphamide and mice gastric mucosal cells were infected with Mycoplasma penetrans (Mpe). After 18 weeks, mice were sacrificed and gastric mucosal Mpe infected cells were identified by fluorescence in situ hybridization (FISH). Moreover, pathological and ultrastructural changes in mice gastric mucosa were evaluated and the expression of multiple proto-oncogenes was examined by Western blot. Our data show that Mpe infection was detected in the blood of immunodeficient mice and Mpe persistent infection in mice gastric mucosa was confirmed by FISH. There were pathological and ultrastructural malignant transformation occurred in the gastric mucosa of infected mice compared to control mice. Mpe infected mice showed lower expression of p53 and p21 and higher H-ras expression compared to the control group. Moreover, expression of NF-κB p65 subunit increased in Mpe infected mice, similar to the TNF-α expression. Bax expression in gastric mucosa of Mpe infected mice was lower while Bcl-2 expression was higher than in the uninfected control group. Collectively these data demonstrate that persistent Mpe infection is associated with aberrant expression of multiple proto-oncogenes in gastric mucosa of immunodeficient mice which potentially facilitate the malignant transformation.
Collapse
Affiliation(s)
- Shuyan Cao
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dandan Shen
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, Shanghai, China
| | - Yadong Wang
- Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linxi Li
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Liping Zhou
- Department of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuxue Wang
- Department of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, China
- * E-mail:
| |
Collapse
|
27
|
Zhang F, Peng H. LncRNA-ANCR regulates the cell growth of osteosarcoma by interacting with EZH2 and affecting the expression of p21 and p27. J Orthop Surg Res 2017; 12:103. [PMID: 28679390 PMCID: PMC5499053 DOI: 10.1186/s13018-017-0599-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 06/17/2017] [Indexed: 01/15/2023] Open
Abstract
Background Osteosarcoma (OS) is one of the most common malignant tumors developed in the bone. EZH2 has been found to play pivotal roles in the development of various cancers. LncRNA-ANCR (anti-differentiation ncRNA) has been reported to interact with EZH2 and regulated osteoblast differentiation. Our study aimed to investigate the effect of lncRNA-ANCR on the tumorigenesis of osteosarcoma and explore the underlying molecular mechanism. Methods RT-PCR was performed to detect the messenger RNA (mRNA) levels of lncRNA-ANCR, EZH2, p21, and p27 in OS tissues and cell lines. The cell proliferation, transwell invasion, and migration assays were conducted to evaluate the influence of lncRNA-ANCR depletion on the growth of OS cells. RNA pull-down assay was carried out to detect the interaction between lncRNA-ANCR and EZH2. Correlation between the expression of lncRNA-ANCR and the expression of EZH2 were analyzed by cross-tabulation. Results LncRNA-ANCR is highly expressed in both OS tissues and cell lines. Reduced expression of lncRNA-ANCR inhibited the cell proliferation, invasion, and migration of OS cells. The cell apoptosis rate was also increased with the overexpression of lncRNA-ANCR. Mechanistically, downregulation of lncRNA-ANCR reduced the mRNA level of EZH2 and increased the expression of p21 and p27 at both mRNA and protein levels. LncRNA-ANCR interacted with EZH2 and their expression abundance was positively correlated in OS patients. Conclusion LncRNA-ANCR inhibited the cell proliferation, migration, and invasion of OS cells possibly through interacting with EZH2 and regulating the expression of p21 and p27.
Collapse
Affiliation(s)
- Fei Zhang
- Department of Orthopaedics Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Hao Peng
- Department of Orthopaedics Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
28
|
Cheng J, Su M, Jin Y, Xi Q, Deng Y, Chen J, Wang W, Chen Y, Chen L, Shi N, Mao G. Upregulation of SENP3/SMT3IP1 promotes epithelial ovarian cancer progression and forecasts poor prognosis. Tumour Biol 2017; 39:1010428317694543. [PMID: 28351334 DOI: 10.1177/1010428317694543] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
As a crucial member of the small ubiquitin-like modifier system, SUMO-specific protease 3, was identified to be essential for cell proliferation and ribosomal RNA processing. Recent studies showed that SUMO-specific protease 3 was elevated in ovarian cancer compared to normal tissue samples. However, the connection between SUMO-specific protease 3-specific expression and clinicopathological parameters of epithelial ovarian cancer, as well as the physiologically potential role of SUMO-specific protease 3 in epithelial ovarian cancer remained unclear. In this study, an analysis of 124 paraffin-embedded slices by immunohistochemistry indicated that SUMO-specific protease 3 expression was positively correlated with the International Federation of Gynecology and Obstetrics stages (p = 0.025), tumor grade (p = 0.004), and lymph node metastasis (p = 0.001) and was also a critical prognostic factor for the overall survival of epithelial ovarian cancer patients, as revealed by Kaplan-Meier curve analysis. Knockdown of SUMO-specific protease 3 weakened the proliferation, migration, and invasion capability of ovarian cancer cells, down-regulated the expression of Proliferating Cell Nuclear Antigen, Forkhead Box C2, and N-cadherin, and resulted in upregulation of p21 and E-cadherin. Consistent with our results, SUMO-specific protease 3 had been verified to promote cell proliferation, metastasis, and tumorigenesis in multiple malignant cancers, which was a redox-sensitive molecule mediating the epithelial-mesenchymal transition. Collectively, our findings for the first time specifically supported that SUMO-specific protease 3 might play an important role in the regulation of epithelial ovarian cancer progression and could serve as a potential biomarker for prognosis as well as provide a promising therapeutic target against epithelial ovarian cancer.
Collapse
Affiliation(s)
- Jialin Cheng
- 1 Department of Oncology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| | - Min Su
- 2 Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| | - Yunfeng Jin
- 2 Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| | - Qinghua Xi
- 2 Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| | - Yan Deng
- 2 Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| | - Jie Chen
- 1 Department of Oncology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China.,3 Department of Oncology, Jiangyin People's Hospital, Wuxi, People's Republic of China
| | - Wei Wang
- 2 Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| | - Yannan Chen
- 2 Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| | - Lingling Chen
- 4 Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| | - Nannan Shi
- 5 Department of Radiology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| | - Guoxin Mao
- 1 Department of Oncology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| |
Collapse
|
29
|
Wang S, Cao Z, Xue J, Li H, Jiang W, Cheng Y, Li G, Zhang X. A positive feedback loop between Pim-1 kinase and HBP1 transcription factor contributes to hydrogen peroxide-induced premature senescence and apoptosis. J Biol Chem 2017; 292:8207-8222. [PMID: 28348080 DOI: 10.1074/jbc.m116.768101] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 03/16/2017] [Indexed: 12/31/2022] Open
Abstract
Oxidative stress can induce cell dysfunction and lead to a broad range of degenerative alterations, including carcinogenesis, aging, and other oxidative stress-related conditions. To avoid undergoing carcinogenesis in response to oxidative stress, cells trigger a succession of checkpoint responses, including premature senescence and apoptosis. Increasing evidence indicates that H2O2, an important cause of oxidative stress, functions as an important physiological regulator of intracellular signaling pathways that participate in regulation of cell premature senescence and apoptosis. However, the precise mechanisms underlying this process remain to be studied extensively. In this study, we describe the importance of Pim-1 kinase in this checkpoint response to oxidative stress. Pim-1 binds to and phosphorylates the transcription factor high mobility group box transcription factor 1 (HBP1), activating it. H2O2 enhances the interaction between Pim-1 and HBP1 and promotes HBP1 accumulation. In turn, HBP1 rapidly and selectively up-regulates Pim-1 expression in H2O2-stimulated cells, thereby creating a Pim-1-HBP1 positive feedback loop that regulates H2O2-induced premature senescence and apoptosis. Furthermore, the Pim-1-HBP1 positive feedback loop exerts its effect by regulating the senescence markers DNMT1 and p16 and the apoptosis marker Bax. The Pim-1-HBP1 axis thus constitutes a novel checkpoint pathway critical for the inhibition of tumorigenesis.
Collapse
Affiliation(s)
- Shuya Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, China
| | - Zhengyi Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, China
| | - Junhui Xue
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, China
| | - Hui Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, China
| | - Wei Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, China
| | - Yuning Cheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, China
| | - Gang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, China
| | - Xiaowei Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|