1
|
Gautam P, Ciuta I, Teif VB, Sinha SK. Predicting p53-dependent cell transitions from thermodynamic models. J Chem Phys 2024; 161:135101. [PMID: 39356070 DOI: 10.1063/5.0225166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024] Open
Abstract
A cell's fate involves transitions among its various states, each defined by a distinct gene expression profile governed by the topology of gene regulatory networks, which are affected by 3D genome organization. Here, we develop thermodynamic models to determine the fate of a malignant cell as governed by the tumor suppressor p53 signaling network, taking into account long-range chromatin interactions in the mean-field approximation. The tumor suppressor p53 responds to stress by selectively triggering one of the potential transcription programs that influence many layers of cell signaling. These range from p53 phosphorylation to modulation of its DNA binding affinity, phase separation phenomena, and internal connectivity among cell fate genes. We use the minimum free energy of the system as a fundamental property of biological networks that influences the connection between the gene network topology and the state of the cell. We constructed models based on network topology and equilibrium thermodynamics. Our modeling shows that the binding of phosphorylated p53 to promoters of target genes can have properties of a first order phase transition. We apply our model to cancer cell lines ranging from breast cancer (MCF-7), colon cancer (HCT116), and leukemia (K562), with each one characterized by a specific network topology that determines the cell fate. Our results clarify the biological relevance of these mechanisms and suggest that they represent flexible network designs for switching between developmental decisions.
Collapse
Affiliation(s)
- Pankaj Gautam
- Theoretical and Computational Biophysical Chemistry Group, Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Isabella Ciuta
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| | - Vladimir B Teif
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| | - Sudipta Kumar Sinha
- Theoretical and Computational Biophysical Chemistry Group, Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| |
Collapse
|
2
|
Lei L, Lu Q, Ma G, Li T, Deng J, Li W. P53 protein and the diseases in central nervous system. Front Genet 2023; 13:1051395. [PMID: 36712862 PMCID: PMC9880595 DOI: 10.3389/fgene.2022.1051395] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/08/2022] [Indexed: 01/11/2023] Open
Abstract
P53 protein is the product of P53 gene, which is a well acknowledged tumor suppressor gene. The function of P53 and the relevant mechanisms of anti-neoplasm have raised the interest of researchers since many years ago. It is demonstrated that P53 is a basic cell cycle regulator and a strong inhibitor for versatile cancers in humans. However, most research focuses on other organs and systems instead of the central nervous system (CNS). In fact, in recent years, more and more studies have been suggesting that P53 plays a significant role in multiple CNS tumors and other diseases and disorders such as cerebral stroke and neurodegenerative diseases. In this work, we mainly reviewed the P53's relationship with CNS tumors, cerebral stroke and neurodegenerative diseases, together with the relevant mechanisms, aiming to summarize the research achievements and providing new insight to the future study on diseases in CNS.
Collapse
Affiliation(s)
- Li Lei
- The Affiliated Hospital of Kunming University of Science and Technology, The Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Qixiong Lu
- The Affiliated Hospital of Kunming University of Science and Technology, The Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Guifang Ma
- Department of Ear, Nose and Throat (ENT) and Head and Neck (HN) Surgery, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Tao Li
- The Affiliated Hospital of Kunming University of Science and Technology, The Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Jiahong Deng
- Department of Ear, Nose and Throat (ENT) and Head and Neck (HN) Surgery, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China,*Correspondence: Jiahong Deng, ; Weijia Li,
| | - Weijia Li
- The Affiliated Hospital of Kunming University of Science and Technology, The Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China,*Correspondence: Jiahong Deng, ; Weijia Li,
| |
Collapse
|
3
|
Lopez KE, Bouchier-Hayes L. Lethal and Non-Lethal Functions of Caspases in the DNA Damage Response. Cells 2022; 11:cells11121887. [PMID: 35741016 PMCID: PMC9221191 DOI: 10.3390/cells11121887] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/12/2022] Open
Abstract
Members of the caspase family are well known for their roles in the initiation and execution of cell death. Due to their function in the removal of damaged cells that could otherwise become malignant, caspases are important players in the DNA damage response (DDR), a network of pathways that prevent genomic instability. However, emerging evidence of caspases positively or negatively impacting the accumulation of DNA damage in the absence of cell death demonstrates that caspases play a role in the DDR that is independent of their role in apoptosis. This review highlights the apoptotic and non-apoptotic roles of caspases in the DDR and how they can impact genomic stability and cancer treatment.
Collapse
Affiliation(s)
- Karla E. Lopez
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- William T. Shearer Center for Human Immunobiology, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Lisa Bouchier-Hayes
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- William T. Shearer Center for Human Immunobiology, Texas Children’s Hospital, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
4
|
Wang H, Yang L, Liu M, Luo J. Protein post-translational modifications in the regulation of cancer hallmarks. Cancer Gene Ther 2022; 30:529-547. [PMID: 35393571 DOI: 10.1038/s41417-022-00464-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 02/28/2022] [Accepted: 03/18/2022] [Indexed: 12/12/2022]
Abstract
Posttranslational modifications (PTMs) of proteins, the major mechanism of protein function regulation, play important roles in regulating a variety of cellular physiological and pathological processes. Although the classical PTMs, such as phosphorylation, acetylation, ubiquitination and methylation, have been well studied, the emergence of many new modifications, such as succinylation, hydroxybutyrylation, and lactylation, introduces a new layer to protein regulation, leaving much more to be explored and wide application prospects. In this review, we will provide a broad overview of the significant roles of PTMs in regulating human cancer hallmarks through selecting a diverse set of examples, and update the current advances in the therapeutic implications of these PTMs in human cancer.
Collapse
Affiliation(s)
- Haiying Wang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China.
| | - Liqian Yang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Minghui Liu
- Department of Medical Genetics, Center for Medical Genetics, Peking University Health Science Center, 100191, Beijing, China
| | - Jianyuan Luo
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China. .,Department of Medical Genetics, Center for Medical Genetics, Peking University Health Science Center, 100191, Beijing, China.
| |
Collapse
|
5
|
Zhou W, Sha Y, Zeng J, Zhang X, Zhang A, Ge X. Computational Systems Pharmacology, Molecular Docking and Experiments Reveal the Protective Mechanism of Li-Da-Qian Mixture in the Treatment of Glomerulonephritis. J Inflamm Res 2021; 14:6939-6958. [PMID: 34949932 PMCID: PMC8689049 DOI: 10.2147/jir.s338055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/22/2021] [Indexed: 01/05/2023] Open
Abstract
Background Glomerulonephritis is a common urinary system disease among children. Growing evidence suggests that traditional Chinese medicine has potential in treating glomerulonephritis, such as Li-Da-Qian mixture. Although its anti-glomerulonephritis and alleviating hematuria effects have been reported, the exact mechanism of Li-Da-Qian mixture devoting to glomerulonephritis remains unexplored. It was necessary to explore the mechanism of Li-Da-Qian mixture against glomerulonephritis using modern technology, such as Chinese medicine database and molecular biological experiments. Methods Online databases were used to look up ingredients and predict targets of Li-Da-Qian mixture against glomerulonephritis. The intersecting targets of Li-Da-Qian mixture and glomerulonephritis were selected for enrichment analysis. Cytoscape software was applied to establish network and MCODE analysis. Molecular docking was used for the primary validation. Furthermore, we examined the function of the core compounds analyzed from Li-Da-Qian mixture to rescue LPS-induced inflammation in vivo and vitro. We also explored whether the core compounds can alleviate TGFβ1-induced renal fibrosis in mouse proximal tubular cells. Results Network pharmacological analysis of Li-Da-Qian evaluated 20 active ingredients including baicalein, luteolin and quercetin. A total of 113 key targets were screened, including IL6, VEGFA, TP53, EGF, MMP2, etc, and they were enriched in AGE-RAGE signaling pathway in diabetic complications, TNF and IL-17 signaling pathways. Moreover, the core ingredients succeeded in binding to the main targets via molecular docking, further identifying the anti-glomerulonephritis effects and improvement of vascular injury. Western blotting and qPCR also suggested that baicalein and luteolin can improve inflammation and restore disturbance of mesangial cells or kidney induced by LPS. In addition, baicalein and luteolin inhibited renal fibrosis in vitro.
Collapse
Affiliation(s)
- Wei Zhou
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Yugen Sha
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Jingxia Zeng
- Pediatric Intensive Care Unit, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Xiaoyue Zhang
- Department of Chinese Medicine, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, People's Republic of China
| | - Aihua Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Xuhua Ge
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China.,Pediatric Intensive Care Unit, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
6
|
Liu Y, Xu Y, Jiang M, Chen W, Zhu X. Significant value of XRCC2 and XRCC9 expression in the prognosis of human ovarian carcinoma. J Cancer 2021; 12:6254-6264. [PMID: 34539898 PMCID: PMC8425215 DOI: 10.7150/jca.59273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 08/15/2021] [Indexed: 01/12/2023] Open
Abstract
Background: The x-ray repair cross-complementing (XRCC) family is essential in DNA repair processes. The predictive roles of XRCCs remain unclear in ovarian carcinomas. Therefore, detecting the relationship between XRCCs expression and ovarian carcinomas prognosis is increasingly pivotal. Methods: Using the "Kaplan-Meier (KM) plotter" database, progression-free survival (PFS) and overall survival (OS) were utilized to evaluate the prognosis of XRCCs mRNA expression in ovarian carcinoma patients with clinical outcomes. Then, mRNA level and protein levels of XRCCs were assessed in normal ovarian cells and ovarian carcinoma cell lines by real-time qPCR, Western blotting and immunofluorescence analysis. Additionally, expression of the XRCCs protein in tissues from ovarian carcinomas and normal ovary was identified by immunohistochemical staining. Results: Higher mRNA levels of XRCC2 and XRCC9 predicted longer PFS and OS in all women with ovarian malignance, while elevated XRCC4 mRNA levels were linked to poor PFS and OS in all ovarian cancer patients. Elevated mRNA of XRCC2 was also correlated with better PFS in patients with serous ovarian carcinomas, and better PFS and OS in grade III and stage III+IV ovarian carcinomas patients. What's more, highly expressed levels of XRCC9 mRNA were also linked to favorable PFS and OS in patients with serous, grade III and stage III+IV ovarian carcinomas. Nevertheless, elevated mRNA expression of XRCC4 was linked to worse PFS and OS for patients with serous, grade III as well as all stages of ovarian malignance. Additionally, when compared to ovarian carcinoma cell lines, elevated mRNA and protein levels of XRCC2 and XRCC9 were detected in normal ovarian cells. Consistently, higher staining of XRCC2 and XRCC9 was also detected in normal ovarian cells than that in ovarian cancer cells. Then, higher staining levels of XRCC2 and XRCC9 were discovered in healthy control tissues than that in ovarian carcinoma tissues. Meanwhile, XRCC4 was identified to be overexpressed in tissues of ovarian malignance as compared to normal control tissues. However, XRCC4 mRNA and protein levels were lower in ovarian cancer cells than that in normal cell line. Conclusion: Elevated XRCC2 and XRCC9 expression levels were observed in normal ovarian cells and tissues than that in ovarian malignance cells and tissues, and exhibited better prognostic value especially in patients with serous, poor differentiated and late stage, suggesting that XRCC2 and XRCC9 may be potent prognostic markers in ovarian cancer patients and can guide personalized surveillance for ovarian malignance.
Collapse
Affiliation(s)
- Yi Liu
- Department of obstetrics and gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Yichi Xu
- Department of obstetrics and gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengying Jiang
- Department of obstetrics and gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weinan Chen
- Department of obstetrics and gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xueqiong Zhu
- Department of obstetrics and gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
7
|
Mdm2 phosphorylation by Akt regulates the p53 response to oxidative stress to promote cell proliferation and tumorigenesis. Proc Natl Acad Sci U S A 2021; 118:2003193118. [PMID: 33468664 DOI: 10.1073/pnas.2003193118] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
We have shown previously that phosphorylation of Mdm2 by ATM and c-Abl regulates Mdm2-p53 signaling and alters the effects of DNA damage in mice, including bone marrow failure and tumorigenesis induced by ionizing radiation. Here, we examine the physiological effects of Mdm2 phosphorylation by Akt, another DNA damage effector kinase. Surprisingly, Akt phosphorylation of Mdm2 does not alter the p53-mediated effects of ionizing radiation in cells or mice but regulates the p53 response to oxidative stress. Akt phosphorylation of Mdm2 serine residue 183 increases nuclear Mdm2 stability, decreases p53 levels, and prevents senescence in primary cells exposed to reactive oxidative species (ROS). Using multiple mouse models of ROS-induced cancer, we show that Mdm2 phosphorylation by Akt reduces senescence to promote KrasG12D-driven lung cancers and carcinogen-induced papilloma and hepatocellular carcinomas. Collectively, we document a unique physiologic role for Akt-Mdm2-p53 signaling in regulating cell growth and tumorigenesis in response to oxidative stress.
Collapse
|
8
|
Ghaznavi H, Shirvaliloo M, Zarebkohan A, Shams Z, Radnia F, Bahmanpour Z, Sargazi S, Saravani R, Shirvalilou S, Shahraki O, Shahraki S, Nazarlou Z, Sheervalilou R. An Updated Review on Implications of Autophagy and Apoptosis in Tumorigenesis: Possible Alterations in Autophagy through Engineered Nanomaterials and Their Importance in Cancer Therapy. Mol Pharmacol 2021; 100:119-143. [PMID: 33990406 DOI: 10.1124/molpharm.121.000234] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/19/2021] [Indexed: 12/17/2022] Open
Abstract
Most commonly recognized as a catabolic pathway, autophagy is a perplexing mechanism through which a living cell can free itself of excess cytoplasmic components, i.e., organelles, by means of certain membranous vesicles or lysosomes filled with degrading enzymes. Upon exposure to external insult or internal stimuli, the cell might opt to activate such a pathway, through which it can gain control over the maintenance of intracellular components and thus sustain homeostasis by intercepting the formation of unnecessary structures or eliminating the already present dysfunctional or inutile organelles. Despite such appropriateness, autophagy might also be considered a frailty for the cell, as it has been said to have a rather complicated role in tumorigenesis. A merit in the early stages of tumor formation, autophagy appears to be salutary because of its tumor-suppressing effects. In fact, several investigations on tumorigenesis have reported diminished levels of autophagic activity in tumor cells, which might result in transition to malignancy. On the contrary, autophagy has been suggested to be a seemingly favorable mechanism to progressed malignancies, as it contributes to survival of such cells. Based on the recent literature, this mechanism might also be activated upon the entry of engineered nanomaterials inside a cell, supposedly protecting the host from foreign materials. Accordingly, there is a good chance that therapeutic interventions for modulating autophagy in malignant cells using nanoparticles may sensitize cancerous cells to certain treatment modalities, e.g., radiotherapy. In this review, we will discuss the signaling pathways involved in autophagy and the significance of the mechanism itself in apoptosis and tumorigenesis while shedding light on possible alterations in autophagy through engineered nanomaterials and their potential therapeutic applications in cancer. SIGNIFICANCE STATEMENT: Autophagy has been said to have a complicated role in tumorigenesis. In the early stages of tumor formation, autophagy appears to be salutary because of its tumor-suppressing effects. On the contrary, autophagy has been suggested to be a favorable mechanism to progressed malignancies. This mechanism might be affected upon the entry of nanomaterials inside a cell. Accordingly, therapeutic interventions for modulating autophagy using nanoparticles may sensitize cancerous cells to certain therapies.
Collapse
Affiliation(s)
- Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Milad Shirvaliloo
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Amir Zarebkohan
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Zinat Shams
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Fatemeh Radnia
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Zahra Bahmanpour
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Saman Sargazi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Ramin Saravani
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Sakine Shirvalilou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Omolbanin Shahraki
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Sheida Shahraki
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Ziba Nazarlou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Roghayeh Sheervalilou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| |
Collapse
|
9
|
Qiao Y, Agboola OS, Hu X, Wu Y, Lei L. Tumorigenic and Immunogenic Properties of Induced Pluripotent Stem Cells: a Promising Cancer Vaccine. Stem Cell Rev Rep 2020; 16:1049-1061. [PMID: 32939647 PMCID: PMC7494249 DOI: 10.1007/s12015-020-10042-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2020] [Indexed: 02/06/2023]
Abstract
Induced pluripotent stem cells (iPSCs) are mainly characterized by their unlimited proliferation abilities and potential to develop into almost any cell type. The creation of this technology has been of great interest to many scientific fields, especially regenerative biology. However, concerns about the safety of iPSC application in transplantation have arisen due to the tumorigenic and immunogenic properties of iPSCs. This review will briefly introduce the developing history of somatic reprogramming and applications of iPSC technology in regenerative medicine. In addition, the review will highlight two challenges to the efficient usage of iPSCs and the underlying mechanisms of these challenges. Finally, the review will discuss the expanding application of iPSC technology in cancer immunotherapy as a potential cancer vaccine and its advantages in auxiliary treatment compared with oncofetal antigen-based and embryonic stem cell (ESC)-based vaccines.
Collapse
Affiliation(s)
- Yu Qiao
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Harbin, Heilongjiang Province, 150081, People's Republic of China
| | - Oluwafemi Solomon Agboola
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Harbin, Heilongjiang Province, 150081, People's Republic of China
| | - Xinglin Hu
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Harbin, Heilongjiang Province, 150081, People's Republic of China
| | - Yanshuang Wu
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Harbin, Heilongjiang Province, 150081, People's Republic of China
| | - Lei Lei
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Harbin, Heilongjiang Province, 150081, People's Republic of China.
- Key laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China.
| |
Collapse
|
10
|
Cai S, Wang J, Zeng W, Cheng X, Liu L, Li W. Lysine-specific histone demethylase 1B (LSD2/KDM1B) represses p53 expression to promote proliferation and inhibit apoptosis in colorectal cancer through LSD2-mediated H3K4me2 demethylation. Aging (Albany NY) 2020; 12:14990-15001. [PMID: 32726297 PMCID: PMC7425427 DOI: 10.18632/aging.103558] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/04/2020] [Indexed: 11/25/2022]
Abstract
Epigenetic alterations have been reported to play critical roles in the development of colorectal cancer (CRC). However, the biological function of the lysine-specific histone demethylase 1B (LSD2/KDM1B) in CRC is not well understood. Therefore, we investigated the characteristics of LSD2 in CRC. We observed significant upregulation of LSD2 in CRC tissue compared to that in normal colorectal tissue. LSD2 promotes CRC cell proliferation and inhibits cell apoptosis through cell cycle regulation, promoting CRC progression both in vitro and in vivo. We found that LSD2 performs these functions by inhibiting the p53-p21-Rb pathway. Finally, we found that LSD2 directly binds to p53 and represses p53 expression via H3K4me2 demethylation at the p53 promoter. Our results revealed that LSD2 acts as an oncogene by binding and inhibiting p53 activity in CRC. Thus, LSD2 may be a new molecular target for CRC treatment.
Collapse
Affiliation(s)
- Shaoxin Cai
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
- Department of Surgical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Jinsi Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
- Department of Surgical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Wei Zeng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
- Department of Surgical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Xuefei Cheng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
- Department of Surgical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Lihang Liu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
- Department of Surgical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Weihua Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
- Department of Surgical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| |
Collapse
|
11
|
Shan L, Yu J, He Z, Chen S, Liu M, Ding H, Xu L, Zhao J, Yang A, Jiang H. Defining relative mutational difficulty to understand cancer formation. Cell Discov 2020; 6:48. [PMID: 32704382 PMCID: PMC7371891 DOI: 10.1038/s41421-020-0177-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 05/06/2020] [Indexed: 12/18/2022] Open
Abstract
Most mutations in human cancer are low-frequency missense mutations, whose functional status remains hard to predict. Here, we show that depending on the type of nucleotide change and the surrounding sequences, the tendency to generate each type of nucleotide mutations varies greatly, even by several hundred folds. Therefore, a cancer-promoting mutation may appear only in a small number of cancer cases, if the underlying nucleotide change is too difficult to generate. We propose a method that integrates both the original mutation counts and their relative mutational difficulty. Using this method, we can accurately predict the functionality of hundreds of low-frequency missense mutations in p53, PTEN, and INK4A. Many loss-of-function p53 mutations with dominant negative effects were identified, and the functional importance of several regions in p53 structure were highlighted by this analysis. Our study not only established relative mutational difficulties for different types of mutations in human cancer, but also showed that by incorporating such a parameter, we can bring new angles to understanding cancer formation.
Collapse
Affiliation(s)
- Lin Shan
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Jiao Yu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Zhengjin He
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Shishuang Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Mingxian Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Hongyu Ding
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Liang Xu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Jie Zhao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Ailing Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Hai Jiang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
12
|
Magnussen HM, Ahmed SF, Sibbet GJ, Hristova VA, Nomura K, Hock AK, Archibald LJ, Jamieson AG, Fushman D, Vousden KH, Weissman AM, Huang DT. Structural basis for DNA damage-induced phosphoregulation of MDM2 RING domain. Nat Commun 2020; 11:2094. [PMID: 32350255 PMCID: PMC7190642 DOI: 10.1038/s41467-020-15783-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/27/2020] [Indexed: 01/30/2023] Open
Abstract
Phosphorylation of MDM2 by ATM upon DNA damage is an important mechanism for deregulating MDM2, thereby leading to p53 activation. ATM phosphorylates multiple residues near the RING domain of MDM2, but the underlying molecular basis for deregulation remains elusive. Here we show that Ser429 phosphorylation selectively enhances the ubiquitin ligase activity of MDM2 homodimer but not MDM2-MDMX heterodimer. A crystal structure of phospho-Ser429 (pS429)-MDM2 bound to E2-ubiquitin reveals a unique 310-helical feature present in MDM2 homodimer that allows pS429 to stabilize the closed E2-ubiquitin conformation and thereby enhancing ubiquitin transfer. In cells Ser429 phosphorylation increases MDM2 autoubiquitination and degradation upon DNA damage, whereas S429A substitution protects MDM2 from auto-degradation. Our results demonstrate that Ser429 phosphorylation serves as a switch to boost the activity of MDM2 homodimer and promote its self-destruction to enable rapid p53 stabilization and resolve a long-standing controversy surrounding MDM2 auto-degradation in response to DNA damage.
Collapse
Affiliation(s)
- Helge M Magnussen
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Syed F Ahmed
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Gary J Sibbet
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Ventzislava A Hristova
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Koji Nomura
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Andreas K Hock
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- AstraZeneca, AstraZeneca R&D, Innovative Medicines, Discovery Sciences, Darwin (Building 310), Cambridge Science Park, Milton Road, Cambridge, CB4 0WG, UK
| | - Lewis J Archibald
- School of Chemistry, University of Glasgow, Joseph Black Building, G12 8QQ, Glasgow, UK
| | - Andrew G Jamieson
- School of Chemistry, University of Glasgow, Joseph Black Building, G12 8QQ, Glasgow, UK
| | - David Fushman
- Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Organization, University of Maryland, College Park, MD, 20742, USA
| | | | - Allan M Weissman
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Danny T Huang
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK.
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK.
| |
Collapse
|
13
|
Hensel N, Kubinski S, Claus P. The Need for SMN-Independent Treatments of Spinal Muscular Atrophy (SMA) to Complement SMN-Enhancing Drugs. Front Neurol 2020; 11:45. [PMID: 32117013 PMCID: PMC7009174 DOI: 10.3389/fneur.2020.00045] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 01/13/2020] [Indexed: 12/25/2022] Open
Abstract
Spinal Muscular Atrophy (SMA) is monogenic motoneuron disease caused by low levels of the Survival of Motoneuron protein (SMN). Recently, two different drugs were approved for the treatment of the disease. The antisense oligonucleotide Nusinersen/Spinraza® and the gene replacement therapy Onasemnogene Abeparvovec/Zolgensma® both enhance SMN levels. These treatments result in impressive benefits for the patients. However, there is a significant number of non-responders and an intervention delay has a strong negative impact on the efficacy. Obviously, later stages of motoneuron degeneration cannot be reversed by SMN-restoration. Therefore, complementary, SMN-independent strategies are needed which are able to address such SMN-irreversible degenerative processes. Those are defined as pathological alterations which are not reversed by SMN-restoration for a given dose and intervention delay. It is crucial to tailor SMN-independent approaches to the novel clinical situation with SMN-restoring treatments. On the molecular level, such SMN-irreversible changes become manifest in altered signaling modules as described by molecular systems biology. Based on our current knowledge about altered signaling, we introduce a network approach for an informed decision for the most potent SMN-independent treatment targets. Finally, we present recommendations for the identification of novel treatments which can be combined with SMN-restoring drugs.
Collapse
Affiliation(s)
- Niko Hensel
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany.,Center of Systems Neuroscience (ZSN), Hannover, Germany
| | - Sabrina Kubinski
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany.,Center of Systems Neuroscience (ZSN), Hannover, Germany
| | - Peter Claus
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany.,Center of Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
14
|
Liu L, Cao J, Zhao J, Li X, Suo Z, Li H. PDHA1 Gene Knockout In Human Esophageal Squamous Cancer Cells Resulted In Greater Warburg Effect And Aggressive Features In Vitro And In Vivo. Onco Targets Ther 2019; 12:9899-9913. [PMID: 31819487 PMCID: PMC6874154 DOI: 10.2147/ott.s226851] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 10/31/2019] [Indexed: 12/24/2022] Open
Abstract
Background One of the remarkable metabolic characteristics of cancer cells is that they prefer glycolysis rather than oxidative phosphorylation (OXPHOS). Pyruvate dehydrogenase E1 alpha subunit (PDHA1) is an important prerequisite for OXPHOS. Our previous studies have shown that low level of PDHA1 protein expression in esophageal squamous cell cancer (ESCC) was correlated with poor prognosis. However, the effect of PDHA1 inhibition on metabolism and biological behavior of esophageal cancer cells remains unclear. Methods And Results In this study, a KYSE450 PDHA1 knockout (KO) cell line of esophageal cancer was established by CRISPR/Cas9 technology. Then, the glycose metabolism, cell proliferation and migration abilities, chemotherapeutic tolerance and angiogenesis of the PDHA1 KO cells were investigated in vitro and in vivo. In the PDHA1 KO cells, the glycolysis and the consumption of glucose and glutamine were significantly enhanced, while the OXPHOS was significantly suppressed, implying Warburg effect in the PDHA1 KO cells. Furthermore, it was also proved in vitro experiments that the PDHA1 KO cell obtained proliferation advantage, as well as significantly greater chemotherapy tolerance and migration ability. Xenograft experiments discovered not only larger tumors but also increased angiogenesis in the PDHA1 KO cell group. Conclusion Inhibition of PDHA1 gene expression in human ESCC leads to metabolic reprogramming of Warburg effect and increased malignancies. Targeting ESCC metabolic reprogramming may become a potential therapeutic target.
Collapse
Affiliation(s)
- Lan Liu
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China.,Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China.,Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Montebello, Oslo, Norway
| | - Jing Cao
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Jing Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Xiangyu Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Zhenhe Suo
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Montebello, Oslo, Norway
| | - Huixiang Li
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China.,Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| |
Collapse
|
15
|
Abstract
Pluripotent stem cells (PSCs) are capable of unlimited self-renewal in culture and differentiation into all functional cell types in the body, and thus hold great promise for regenerative medicine. To achieve their clinical potential, it is critical for PSCs to maintain genomic stability during the extended proliferation. The critical tumor suppressor p53 is required to maintain genomic stability of mammalian cells. In response to DNA damage or oncogenic stress, p53 plays multiple roles in maintaining genomic stability of somatic cells by inducing cell cycle arrest, apoptosis, and senescence to prevent the passage of genetic mutations to the daughter cells. p53 is also required to maintain the genomic stability of PSCs. However, in response to the genotoxic stresses, a primary role of p53 in PSCs is to induce the differentiation of PSCs and inhibit pluripotency, providing mechanisms to maintain the genomic stability of the self-renewing PSCs. In addition, the roles of p53 in cellular metabolism might also contribute to genomic stability of PSCs by limiting oxidative stress. In summary, the elucidation of the roles of p53 in PSCs will be a prerequisite for developing safe PSC-based cell therapy.
Collapse
|
16
|
Zheng FM, Chen WB, Qin T, Lv LN, Feng B, Lu YL, Li ZQ, Wang XC, Tao LJ, Li HW, Li SY. ACOX1 destabilizes p73 to suppress intrinsic apoptosis pathway and regulates sensitivity to doxorubicin in lymphoma cells. BMB Rep 2019. [PMID: 31401980 PMCID: PMC6774419 DOI: 10.5483/bmbrep.2019.52.9.094] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Lymphoma is one of the most curable types of cancer. However, drug resistance is the main challenge faced in lymphoma treatment. Peroxisomal acyl-CoA oxidase 1 (ACOX1) is the rate-limiting enzyme in fatty acid β-oxidation. Deregulation of ACOX1 has been linked to peroxisomal disorders and carcinogenesis in the liver. Currently, there is no information about the function of ACOX1 in lymphoma. In this study, we found that upregulation of ACOX1 promoted proliferation in lymphoma cells, while downregulation of ACOX1 inhibited proliferation and induced apoptosis. Additionally, over-expression of ACOX1 increased resistance to doxorubicin, while suppression of ACOX1 expression markedly potentiated doxorubicin-induced apoptosis. Interestingly, downregulation of ACOX1 promoted mitochondrial location of Bad, reduced mitochondrial membrane potential and provoked apoptosis by activating caspase-9 and caspase-3 related apoptotic pathway. Overexpression of ACOX1 alleviated doxorubicin-induced activation of caspase-9 and caspase-3 and decrease of mitochondrial membrane potential. Importantly, downregulation of ACOX1 increased p73, but not p53, expression. p73 expression was critical for apoptosis induction induced by ACOX1 downregulation. Also, overexpression of ACOX1 significantly reduced stability of p73 protein thereby reducing p73 expression. Thus, our study indicated that suppression of ACOX1 could be a novel and effective approach for treatment of lymphoma.
Collapse
Affiliation(s)
- Fei-Meng Zheng
- Department of Medical Oncology, The Eastern Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Wang-Bing Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Qin
- Department of Medical Oncology, Sun Yat-sen Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Li-Na Lv
- Department of Hematology, Wuming Hospital of Guangxi Medical University, Naning 530199, China
| | - Bi Feng
- Department of Medical Oncology, The Eastern Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yan-Ling Lu
- Department of Medical Oncology, The Eastern Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zuo-Quan Li
- Department of Ultrasound, Guancheng Hospital, Dongguan 523009, China
| | - Xiao-Chao Wang
- Department of Hematology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
| | - Li-Ju Tao
- Department of Hematology, Wuming Hospital of Guangxi Medical University, Naning 530199, China
| | - Hong-Wen Li
- Department of Hematology, Wuming Hospital of Guangxi Medical University, Naning 530199, China
| | - Shu-You Li
- Department of Medical Oncology, Wuming Hospital of Guangxi Medical University, Naning 530199, China
| |
Collapse
|
17
|
Liu Y, Tavana O, Gu W. p53 modifications: exquisite decorations of the powerful guardian. J Mol Cell Biol 2019; 11:564-577. [PMID: 31282934 PMCID: PMC6736412 DOI: 10.1093/jmcb/mjz060] [Citation(s) in RCA: 250] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/23/2019] [Accepted: 06/03/2019] [Indexed: 02/05/2023] Open
Abstract
The last 40 years have witnessed how p53 rose from a viral binding protein to a central factor in both stress responses and tumor suppression. The exquisite regulation of p53 functions is of vital importance for cell fate decisions. Among the multiple layers of mechanisms controlling p53 function, posttranslational modifications (PTMs) represent an efficient and precise way. Major p53 PTMs include phosphorylation, ubiquitination, acetylation, and methylation. Meanwhile, other PTMs like sumoylation, neddylation, O-GlcNAcylation, adenosine diphosphate (ADP)-ribosylation, hydroxylation, and β-hydroxybutyrylation are also shown to play various roles in p53 regulation. By independent action or interaction, PTMs affect p53 stability, conformation, localization, and binding partners. Deregulation of the PTM-related pathway is among the major causes of p53-associated developmental disorders or diseases, especially in cancers. This review focuses on the roles of different p53 modification types and shows how these modifications are orchestrated to produce various outcomes by modulating p53 activities or targeted to treat different diseases caused by p53 dysregulation.
Collapse
Affiliation(s)
- Yanqing Liu
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Omid Tavana
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Wei Gu
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.,Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.,Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
18
|
Li X, Wu L, Zopp M, Kopelov S, Du W. p53-TP53-Induced Glycolysis Regulator Mediated Glycolytic Suppression Attenuates DNA Damage and Genomic Instability in Fanconi Anemia Hematopoietic Stem Cells. Stem Cells 2019; 37:937-947. [PMID: 30977208 PMCID: PMC6599562 DOI: 10.1002/stem.3015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/24/2019] [Accepted: 03/31/2019] [Indexed: 01/31/2023]
Abstract
Emerging evidence has shown that resting quiescent hematopoietic stem cells (HSCs) prefer to utilize anaerobic glycolysis rather than mitochondrial respiration for energy production. Compelling evidence has also revealed that altered metabolic energetics in HSCs underlies the onset of certain blood diseases; however, the mechanisms responsible for energetic reprogramming remain elusive. We recently found that Fanconi anemia (FA) HSCs in their resting state are more dependent on mitochondrial respiration for energy metabolism than on glycolysis. In the present study, we investigated the role of deficient glycolysis in FA HSC maintenance. We observed significantly reduced glucose consumption, lactate production, and ATP production in HSCs but not in the less primitive multipotent progenitors or restricted hematopoietic progenitors of Fanca−/− and Fancc−/− mice compared with that of wild‐type mice, which was associated with an overactivated p53 and TP53‐induced glycolysis regulator, the TIGAR‐mediated metabolic axis. We utilized Fanca−/− HSCs deficient for p53 to show that the p53‐TIGAR axis suppressed glycolysis in FA HSCs, leading to enhanced pentose phosphate pathway and cellular antioxidant function and, consequently, reduced DNA damage and attenuated HSC exhaustion. Furthermore, by using Fanca−/− HSCs carrying the separation‐of‐function mutant p53R172P transgene that selectively impairs the p53 function in apoptosis but not cell‐cycle control, we demonstrated that the cell‐cycle function of p53 was not required for glycolytic suppression in FA HSCs. Finally, ectopic expression of the glycolytic rate‐limiting enzyme PFKFB3 specifically antagonized p53‐TIGAR‐mediated metabolic reprogramming in FA HSCs. Together, our results suggest that p53‐TIGAR metabolic axis‐mediated glycolytic suppression may play a compensatory role in attenuating DNA damage and proliferative exhaustion in FA HSCs. stem cells2019;37:937–947
Collapse
Affiliation(s)
- Xue Li
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, West Virginia, USA.,Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, People's Republic of China
| | - Limei Wu
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, West Virginia, USA
| | - Morgan Zopp
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, West Virginia, USA
| | - Shaina Kopelov
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, West Virginia, USA
| | - Wei Du
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, West Virginia, USA.,Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program, West Virginia University Cancer Institute, Morgantown, West Virginia, USA
| |
Collapse
|
19
|
Converging Mechanisms of p53 Activation Drive Motor Neuron Degeneration in Spinal Muscular Atrophy. Cell Rep 2019; 21:3767-3780. [PMID: 29281826 DOI: 10.1016/j.celrep.2017.12.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/09/2017] [Accepted: 11/30/2017] [Indexed: 11/20/2022] Open
Abstract
The hallmark of spinal muscular atrophy (SMA), an inherited disease caused by ubiquitous deficiency in the SMN protein, is the selective degeneration of subsets of spinal motor neurons. Here, we show that cell-autonomous activation of p53 occurs in vulnerable but not resistant motor neurons of SMA mice at pre-symptomatic stages. Moreover, pharmacological or genetic inhibition of p53 prevents motor neuron death, demonstrating that induction of p53 signaling drives neurodegeneration. At late disease stages, however, nuclear accumulation of p53 extends to resistant motor neurons and spinal interneurons but is not associated with cell death. Importantly, we identify phosphorylation of serine 18 as a specific post-translational modification of p53 that exclusively marks vulnerable SMA motor neurons and provide evidence that amino-terminal phosphorylation of p53 is required for the neurodegenerative process. Our findings indicate that distinct events induced by SMN deficiency converge on p53 to trigger selective death of vulnerable SMA motor neurons.
Collapse
|
20
|
Mutant p53 Sequestration of the MDM2 Acidic Domain Inhibits E3 Ligase Activity. Mol Cell Biol 2019; 39:MCB.00375-18. [PMID: 30455251 DOI: 10.1128/mcb.00375-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 11/13/2018] [Indexed: 12/15/2022] Open
Abstract
Missense p53 mutants often accumulate in tumors and drive progression through gain of function. MDM2 efficiently degrades wild-type p53 but fails to degrade mutant p53 in tumor cells. Previous studies revealed that mutant p53 inhibits MDM2 autoubiquitination, suggesting that the interaction inhibits MDM2 E3 activity. Recent work showed that MDM2 E3 activity is stimulated by intramolecular interaction between the RING and acidic domains. Here, we show that in the mutant p53-MDM2 complex, the mutant p53 core domain binds to the MDM2 acidic domain with significantly higher avidity than wild-type p53. The mutant p53-MDM2 complex is deficient in catalyzing ubiquitin release from the activated E2 conjugating enzyme. An MDM2 construct with extra copies of the acidic domain is resistant to inhibition by mutant p53 and efficiently promotes mutant p53 ubiquitination and degradation. The results suggest that mutant p53 interferes with the intramolecular autoactivation mechanism of MDM2, contributing to reduced ubiquitination and increased accumulation in tumor cells.
Collapse
|
21
|
Yogosawa S, Yoshida K. Tumor suppressive role for kinases phosphorylating p53 in DNA damage-induced apoptosis. Cancer Sci 2018; 109:3376-3382. [PMID: 30191640 PMCID: PMC6215896 DOI: 10.1111/cas.13792] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/23/2018] [Accepted: 09/02/2018] [Indexed: 12/20/2022] Open
Abstract
Tumor suppressor p53 plays an important role in cancer prevention. Under normal conditions, p53 is maintained at a low level. However, in response to various cellular stresses, p53 is stabilized and activated, which, in turn, initiates DNA repair, cell-cycle arrest, senescence and apoptosis. Post-translational modifications of p53 including phosphorylation, ubiquitination, and acetylation at multiple sites are important to regulate its activation and subsequent transcriptional gene expression. Particularly, phosphorylation of p53 plays a critical role in modulating its activation to induce apoptosis in cancer cells. In this context, previous studies show that several serine/threonine kinases regulate p53 phosphorylation and downstream gene expression. The molecular basis by which p53 and its kinases induce apoptosis for cancer prevention has been extensively studied. However, the relationship between p53 phosphorylation and its kinases and how the activity of kinases is controlled are still largely unclear; hence, they need to be investigated. In this review, we discuss various roles for p53 phosphorylation and its responsible kinases to induce apoptosis and a new therapeutic approach in a broad range of cancers.
Collapse
Affiliation(s)
- Satomi Yogosawa
- Department of Biochemistry, Jikei University School of Medicine, Tokyo, Japan
| | - Kiyotsugu Yoshida
- Department of Biochemistry, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
22
|
Malvezzi H, Viana BG, Dobo C, Filippi RZ, Podgaec S, Piccinato CA. Depleted lamin B1: a possible marker of the involvement of senescence in endometriosis? Arch Gynecol Obstet 2018; 297:977-984. [PMID: 29417283 DOI: 10.1007/s00404-018-4691-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 01/25/2018] [Indexed: 02/06/2023]
Abstract
PROPOSE Endometriosis is a benign disease characterized by implantation and the growth of endometrial tissue outside the uterine cavity and it shares similarities with cancer. Lamin B1, p16 and p21 play a role on cell cycle regulation, development, cell repair and its activities are related to cancers. Considering the similarities between endometriosis and cancer, the aim of the present cross-sectional study is to detect p16, p21 and Lamin B1 in the ectopic endometrium of patients with endometriosis (n = 8) with eutopic (n = 8) and control endometrium (n = 8) and relate them to the maintenance and development of endometriosis. METHODS Biopsies were obtained from both eutopic and ectopic, from deep infiltrating lesions, endometrium frozen and used for immunofluorescent (p16) or immunohistochemistry procedures (p16, p21, lamin B1). RESULTS Detected higher lamin B1 in the eutopic endometrium when compared with ectopic endometrium, with no differences between endometriosis tissue with control endometrium. Similar presence of p16 in all groups of patients and no p21 detection was observed. CONCLUSION We observed reduced detection of lamin B1 in the ectopic endometrium raising the possibility that the presence of senescent cells might be contributing to the maintenance and progression of endometriosis by apoptosis resistance and peritoneal stress inherent of the disease.
Collapse
Affiliation(s)
- Helena Malvezzi
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi, SP, 05652-900, Brazil
| | - Bruno Gallani Viana
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi, SP, 05652-900, Brazil
| | - Cristine Dobo
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi, SP, 05652-900, Brazil
| | - Renee Zon Filippi
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi, SP, 05652-900, Brazil
| | - Sérgio Podgaec
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi, SP, 05652-900, Brazil
| | - Carla Azevedo Piccinato
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi, SP, 05652-900, Brazil.
| |
Collapse
|
23
|
Relevance of the p53-MDM2 axis to aging. Cell Death Differ 2017; 25:169-179. [PMID: 29192902 DOI: 10.1038/cdd.2017.187] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/19/2017] [Accepted: 09/21/2017] [Indexed: 12/13/2022] Open
Abstract
In response to varying stress signals, the p53 tumor suppressor is able to promote repair, survival, or elimination of damaged cells - processes that have great relevance to organismal aging. Although the link between p53 and cancer is well established, the contribution of p53 to the aging process is less clear. Delineating how p53 regulates distinct aging hallmarks such as cellular senescence, genomic instability, mitochondrial dysfunction, and altered metabolic pathways will be critical. Mouse models have further revealed the centrality and complexity of the p53 network in aging processes. While naturally aged mice have linked longevity with declining p53 function, some accelerated aging mice present with chronic p53 activation, whose phenotypes can be rescued upon p53 deficiency. Further, direct modulation of the p53-MDM2 axis has correlated elevated p53 activity with either early aging or with delayed-onset aging. We speculate that p53-mediated aging phenotypes in these mice must have (1) stably active p53 due to MDM2 dysregulation or chronic stress or (2) shifted p53 outcomes. Pinpointing which p53 stressors, modifications, and outcomes drive aging processes will provide further insights into our understanding of the human aging process and could have implications for both cancer and aging therapeutics.
Collapse
|
24
|
Carr MI, Jones SN. Regulation of the Mdm2-p53 signaling axis in the DNA damage response and tumorigenesis. Transl Cancer Res 2016; 5:707-724. [PMID: 28690977 PMCID: PMC5501481 DOI: 10.21037/tcr.2016.11.75] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The p53 tumor suppressor acts as a guardian of the genome in mammalian cells undergoing DNA double strand breaks induced by a various forms of cell stress, including inappropriate growth signals or ionizing radiation. Following damage, p53 protein levels become greatly elevated in cells and p53 functions primarily as a transcription factor to regulate the expression a wide variety of genes that coordinate this DNA damage response. In cells undergoing high amounts of DNA damage, p53 can promote apoptosis, whereas in cells undergoing less damage, p53 promotes senescence or transient cell growth arrest and the expression of genes involved in DNA repair, depending upon the cell type and level of damage. Failure of the damaged cell to undergo growth arrest or apoptosis, or to respond to the DNA damage by other p53-coordinated mechanisms, can lead to inappropriate cell growth and tumorigenesis. In cells that have successfully responded to genetic damage, the amount of p53 present in the cell must return to basal levels in order for the cell to resume normal growth and function. Although regulation of p53 levels and function is coordinated by many proteins, it is now widely accepted that the master regulator of p53 is Mdm2. In this review, we discuss the role(s) of p53 in the DNA damage response and in tumor suppression, and how post-translational modification of Mdm2 regulates the Mdm2-p53 signaling axis to govern p53 activities in the cell.
Collapse
Affiliation(s)
- Michael I Carr
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Stephen N Jones
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| |
Collapse
|
25
|
Tong KI, Ota K, Komuro A, Ueda T, Ito A, Anne Koch C, Okada H. Attenuated DNA damage repair delays therapy-related myeloid neoplasms in a mouse model. Cell Death Dis 2016; 7:e2401. [PMID: 27711078 PMCID: PMC5133969 DOI: 10.1038/cddis.2016.298] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 07/31/2016] [Accepted: 08/22/2016] [Indexed: 12/18/2022]
Abstract
Therapy-related cancers are potentially fatal late life complications for patients who received radio- or chemotherapy. So far, the mouse model showing reduction or delay of these diseases has not been described. We found that the disruption of Aplf in mice moderately attenuated DNA damage repair and, unexpectedly, impeded myeloid neoplasms after exposure to ionizing radiation (IR). Irradiated mutant mice showed higher rates of p53-dependent cell death, fewer chromosomal translocations, and a delay in malignancy-induce;/– mice. Depletion of APLF in non-tumorigenic human cells also markedly reduced the risk of radiation-induced chromosomal aberrations. We therefore conclude that proficient DNA damage repair may promote chromosomal aberrations in normal tissues after irradiation and induce malignant evolution, thus illustrating the potential benefit in sensitizing p53 function by manipulating DNA repair efficiency in cancer patients undergoing genotoxic therapies.
Collapse
Affiliation(s)
- Kit I Tong
- The Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada M5G 2M9
| | - Kazushige Ota
- Department of Biochemistry, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Osaka, Japan
| | - Akiyoshi Komuro
- Department of Biochemistry, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Osaka, Japan
| | - Takeshi Ueda
- Department of Biochemistry, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Osaka, Japan
| | - Akihiko Ito
- Department of Pathology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - C Anne Koch
- Radiation Medicine Program, Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada M5G 2M9.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada M5G 2M9
| | - Hitoshi Okada
- The Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada M5G 2M9.,Department of Biochemistry, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Osaka, Japan.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada M5G 2M9.,Anti-Aging Center, Kindai University, Higashi-Osaka, Osaka 577-8502, Japan
| |
Collapse
|
26
|
Mdm2 Phosphorylation Regulates Its Stability and Has Contrasting Effects on Oncogene and Radiation-Induced Tumorigenesis. Cell Rep 2016; 16:2618-2629. [PMID: 27568562 DOI: 10.1016/j.celrep.2016.08.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 06/17/2016] [Accepted: 08/03/2016] [Indexed: 11/20/2022] Open
Abstract
ATM phosphorylation of Mdm2-S394 is required for robust p53 stabilization and activation in DNA-damaged cells. We have now utilized Mdm2(S394A) knockin mice to determine that phosphorylation of Mdm2-S394 regulates p53 activity and the DNA damage response in lymphatic tissues in vivo by modulating Mdm2 stability. Mdm2-S394 phosphorylation delays lymphomagenesis in Eμ-myc transgenic mice, and preventing Mdm2-S394 phosphorylation obviates the need for p53 mutation in Myc-driven tumorigenesis. However, irradiated Mdm2(S394A) mice also have increased hematopoietic stem and progenitor cell functions, and we observed decreased lymphomagenesis in sub-lethally irradiated Mdm2(S394A) mice. These findings document contrasting effects of ATM-Mdm2 signaling on p53 tumor suppression and reveal that destabilizing Mdm2 by promoting its phosphorylation by ATM would be effective in treating oncogene-induced malignancies, while inhibiting Mdm2-S394 phosphorylation during radiation exposure or chemotherapy would ameliorate bone marrow failure and prevent the development of secondary hematological malignancies.
Collapse
|
27
|
Di Domenico EG, Toma L, Bordignon V, Trento E, D'Agosto G, Cordiali-Fei P, Ensoli F. Activation of DNA Damage Response Induced by the Kaposi's Sarcoma-Associated Herpes Virus. Int J Mol Sci 2016; 17:ijms17060854. [PMID: 27258263 PMCID: PMC4926388 DOI: 10.3390/ijms17060854] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 05/23/2016] [Accepted: 05/27/2016] [Indexed: 01/24/2023] Open
Abstract
The human herpes virus 8 (HHV-8), also known as Kaposi sarcoma-associated herpes virus (KSHV), can infect endothelial cells often leading to cell transformation and to the development of tumors, namely Kaposi’s sarcoma (KS), primary effusion lymphoma (PEL), and the plasmablastic variant of multicentric Castleman’s disease. KSHV is prevalent in areas such as sub-Saharan Africa and the Mediterranean region presenting distinct genotypes, which appear to be associated with differences in disease manifestation, according to geographical areas. In infected cells, KSHV persists in a latent episomal form. However, in a limited number of cells, it undergoes spontaneous lytic reactivation to ensure the production of new virions. During both the latent and the lytic cycle, KSHV is programmed to express genes which selectively modulate the DNA damage response (DDR) through the activation of the ataxia telangiectasia mutated (ATM) pathway and by phosphorylating factors associated with the DDR, including the major tumor suppressor protein p53 tumor suppressor p53. This review will focus on the interplay between the KSHV and the DDR response pathway throughout the viral lifecycle, exploring the putative molecular mechanism/s that may contribute to malignant transformation of host cells.
Collapse
Affiliation(s)
- Enea Gino Di Domenico
- Clinical Pathology and Microbiology Department, San Gallicano Institute, IRCCS, Rome 00144, Italy.
| | - Luigi Toma
- Infectious Disease Consultant, San Gallicano Institute, IRCCS, Rome 00144, Italy.
| | - Valentina Bordignon
- Clinical Pathology and Microbiology Department, San Gallicano Institute, IRCCS, Rome 00144, Italy.
| | - Elisabetta Trento
- Clinical Pathology and Microbiology Department, San Gallicano Institute, IRCCS, Rome 00144, Italy.
| | - Giovanna D'Agosto
- Clinical Pathology and Microbiology Department, San Gallicano Institute, IRCCS, Rome 00144, Italy.
| | - Paola Cordiali-Fei
- Clinical Pathology and Microbiology Department, San Gallicano Institute, IRCCS, Rome 00144, Italy.
| | - Fabrizio Ensoli
- Clinical Pathology and Microbiology Department, San Gallicano Institute, IRCCS, Rome 00144, Italy.
| |
Collapse
|
28
|
Luo H, Cowen L, Yu G, Jiang W, Tang Y. SMG7 is a critical regulator of p53 stability and function in DNA damage stress response. Cell Discov 2016; 2:15042. [PMID: 27462439 PMCID: PMC4860962 DOI: 10.1038/celldisc.2015.42] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 11/04/2015] [Indexed: 12/16/2022] Open
Abstract
The p53 tumor suppressor functions as a transcription factor and plays a pivotal role in regulation of cellular response to DNA damage by activating various genes including those involved in cell cycle arrest. p53 stability is essential for its function during stress response; however, the molecular mechanism for DNA damage-induced stabilization of p53 is not fully understood. In our present study, we have identified SMG7 (suppressor with morphological defects in genitalia 7), also known as EST1C, as a novel p53-binding protein. SMG7 is an mRNA surveillance factor implicated in degradation of p53 mRNA-containing nonsense mutations, yet it is completely unknown whether SMG7 regulates p53 function. Here, we show that SMG7 has a crucial role in p53-mediated response to genotoxic stress by regulating p53 stability. Using somatic gene knockout, we found that deletion of SMG7 abrogates DNA damage-induced p53 stabilization, although it exhibits minimal effect on the basal levels of p53. Importantly, loss of SMG7 impairs p53-mediated activation of p21 and cell cycle arrest following DNA damage. Pharmacological inhibition of Mdm2, a major E3 ubiquitin ligase for p53, restored p53 stability in gamma-irradiated SMG7-deficient cells. Furthermore, SMG7 physically interacts with Mdm2 and promotes ATM-mediated inhibitory phosphorylation of Mdm2 following ionizing radiation. Therefore, our present data demonstrate that SMG7 is critical for p53 function in DNA damage response, and reveal the SMG7-mediated phosphorylation of Mdm2 as a previously unknown mechanism for p53 regulation.
Collapse
Affiliation(s)
- Hongwei Luo
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY, USA
| | - Lauren Cowen
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY, USA
| | - Guowu Yu
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY, USA
| | - Wenguo Jiang
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY, USA
| | - Yi Tang
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY, USA
| |
Collapse
|
29
|
Specks J, Nieto-Soler M, Lopez-Contreras AJ, Fernandez-Capetillo O. Modeling the study of DNA damage responses in mice. Methods Mol Biol 2015; 1267:413-37. [PMID: 25636482 DOI: 10.1007/978-1-4939-2297-0_21] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Damaged DNA has a profound impact on mammalian health and overall survival. In addition to being the source of mutations that initiate cancer, the accumulation of toxic amounts of DNA damage can cause severe developmental diseases and accelerate aging. Therefore, understanding how cells respond to DNA damage has become one of the most intense areas of biomedical research in the recent years. However, whereas most mechanistic studies derive from in vitro or in cellulo work, the impact of a given mutation on a living organism is largely unpredictable. For instance, why BRCA1 mutations preferentially lead to breast cancer whereas mutations compromising mismatch repair drive colon cancer is still not understood. In this context, evaluating the specific physiological impact of mutations that compromise genome integrity has become crucial for a better dimensioning of our knowledge. We here describe the various technologies that can be used for modeling mutations in mice and provide a review of the genes and pathways that have been modeled so far in the context of DNA damage responses.
Collapse
Affiliation(s)
- Julia Specks
- Genomic Instability Group, Spanish National Cancer Research Center (CNIO), C/Melchor Fernandez Almagro, 3, E-28029, Madrid, Spain
| | | | | | | |
Collapse
|
30
|
Soriano‐Cantón R, Perez‐Villalba A, Morante‐Redolat JM, Marqués‐Torrejón MÁ, Pallás M, Pérez‐Sánchez F, Fariñas I. Regulation of the p19(Arf)/p53 pathway by histone acetylation underlies neural stem cell behavior in senescence-prone SAMP8 mice. Aging Cell 2015; 14:453-62. [PMID: 25728253 PMCID: PMC4406674 DOI: 10.1111/acel.12328] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2015] [Indexed: 01/24/2023] Open
Abstract
Brain aging is associated with increased neurodegeneration and reduced neurogenesis. B1/neural stem cells (B1-NSCs) of the mouse subependymal zone (SEZ) support the ongoing production of olfactory bulb interneurons, but their neurogenic potential is progressively reduced as mice age. Although age-related changes in B1-NSCs may result from increased expression of tumor suppressor proteins, accumulation of DNA damage, metabolic alterations, and microenvironmental or systemic changes, the ultimate causes remain unclear. Senescence-accelerated-prone mice (SAMP8) relative to senescence-accelerated-resistant mice (SAMR1) exhibit signs of hastened senescence and can be used as a model for the study of aging. We have found that the B1-NSC compartment is transiently expanded in young SAMP8 relative to SAMR1 mice, resulting in disturbed cytoarchitecture of the SEZ, B1-NSC hyperproliferation, and higher yields of primary neurospheres. These unusual features are, however, accompanied by premature loss of B1-NSCs. Moreover, SAMP8 neurospheres lack self-renewal and enter p53-dependent senescence after only two passages. Interestingly, in vitro senescence of SAMP8 cells could be prevented by inhibition of histone acetyltransferases and mimicked in SAMR1 cells by inhibition of histone deacetylases (HDAC). Our data indicate that expression of the tumor suppressor p19, but not of p16, is increased in SAMP8 neurospheres, as well as in SAMR1 neurospheres upon HDAC inhibition, and suggest that the SAMP8 phenotype may, at least in part, be due to changes in chromatin status. Interestingly, acute HDAC inhibition in vivo resulted in changes in the SEZ of SAMR1 mice that resembled those found in young SAMP8 mice.
Collapse
Affiliation(s)
- Raúl Soriano‐Cantón
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED) Universidad de Valencia Burjassot 46100 Spain
- Departamento de Biología Celular Universidad de Valencia Burjassot 46100Spain
| | - Ana Perez‐Villalba
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED) Universidad de Valencia Burjassot 46100 Spain
- Departamento de Biología Celular Universidad de Valencia Burjassot 46100Spain
| | - José Manuel Morante‐Redolat
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED) Universidad de Valencia Burjassot 46100 Spain
- Departamento de Biología Celular Universidad de Valencia Burjassot 46100Spain
| | - María Ángeles Marqués‐Torrejón
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED) Universidad de Valencia Burjassot 46100 Spain
- Departamento de Biología Celular Universidad de Valencia Burjassot 46100Spain
| | - Mercé Pallás
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED) Universidad de Valencia Burjassot 46100 Spain
- Departamento de Farmacología y Química Terapéutica Instituto de Biomedicina de la Universidad de Barcelona Barcelona 08028Spain
| | - Francisco Pérez‐Sánchez
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED) Universidad de Valencia Burjassot 46100 Spain
- Departamento de Biología Celular Universidad de Valencia Burjassot 46100Spain
| | - Isabel Fariñas
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED) Universidad de Valencia Burjassot 46100 Spain
- Departamento de Biología Celular Universidad de Valencia Burjassot 46100Spain
| |
Collapse
|
31
|
de Bruin C, Mericq V, Andrew SF, van Duyvenvoorde HA, Verkaik NS, Losekoot M, Porollo A, Garcia H, Kuang Y, Hanson D, Clayton P, van Gent DC, Wit JM, Hwa V, Dauber A. An XRCC4 splice mutation associated with severe short stature, gonadal failure, and early-onset metabolic syndrome. J Clin Endocrinol Metab 2015; 100:E789-98. [PMID: 25742519 PMCID: PMC4422886 DOI: 10.1210/jc.2015-1098] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CONTEXT Severe short stature can be caused by defects in numerous biological processes including defects in IGF-1 signaling, centromere function, cell cycle control, and DNA damage repair. Many syndromic causes of short stature are associated with medical comorbidities including hypogonadism and microcephaly. OBJECTIVE To identify an underlying genetic etiology in two siblings with severe short stature and gonadal failure. DESIGN Clinical phenotyping, genetic analysis, complemented by in vitro functional studies of the candidate gene. SETTING An academic pediatric endocrinology clinic. PATIENTS OR OTHER PARTICIPANTS Two adult siblings (male patient [P1] and female patient 2 [P2]) presented with a history of severe postnatal growth failure (adult heights: P1, -6.8 SD score; P2, -4 SD score), microcephaly, primary gonadal failure, and early-onset metabolic syndrome in late adolescence. In addition, P2 developed a malignant gastrointestinal stromal tumor at age 28. INTERVENTION(S) Single nucleotide polymorphism microarray and exome sequencing. RESULTS Combined microarray analysis and whole exome sequencing of the two affected siblings and one unaffected sister identified a homozygous variant in XRCC4 as the probable candidate variant. Sanger sequencing and mRNA studies revealed a splice variant resulting in an in-frame deletion of 23 amino acids. Primary fibroblasts (P1) showed a DNA damage repair defect. CONCLUSIONS In this study we have identified a novel pathogenic variant in XRCC4, a gene that plays a critical role in non-homologous end-joining DNA repair. This finding expands the spectrum of DNA damage repair syndromes to include XRCC4 deficiency causing severe postnatal growth failure, microcephaly, gonadal failure, metabolic syndrome, and possibly tumor predisposition.
Collapse
Affiliation(s)
- Christiaan de Bruin
- Cincinnati Center for Growth Disorders (C.d.B., S.F.A., V.H., A.D.), Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229; Institute of Maternal and Child Research (V.M.), Faculty of Medicine, University of Chile, 226-3 Santiago, Chile; Laboratory for Diagnostic Genome Analysis (H.A.v.D., M.L.), Department of Clinical Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; Department of Genetics (N.S.V., D.C.v.G.), Erasmus MC, 3015 CE Rotterdam, The Netherlands; Center for Autoimmune Genomics and Etiology (A.P.), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229; Pediatrics Division (H.G.), Faculty of Medicine, Pontificia Universidad Catolica de Chile Santiago, 340 Santiago, Chile; Division of Developmental Biology (Y.K.), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229; Institute of Human Development (D.H., P.C.), University of Manchester and Manchester Academic Health Sciences Centre, Manchester M13 9PL, United Kingdom; and Department of Pediatrics (J.M.W.), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Priami C, De Michele G, Cotelli F, Cellerino A, Giorgio M, Pelicci PG, Migliaccio E. Modelling the p53/p66Shc Aging Pathway in the Shortest Living Vertebrate Nothobranchius Furzeri. Aging Dis 2015; 6:95-108. [PMID: 25821638 DOI: 10.14336/ad.2014.0228] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 02/28/2014] [Indexed: 12/23/2022] Open
Abstract
Oxidative stress induced by reactive oxygen species (ROS) increases during lifespan and is involved in aging processes. The p66Shc adaptor protein is a master regulator of oxidative stress response in mammals. Ablation of p66Shc enhances oxidative stress resistance both in vitro and in vivo. Most importantly, it has been demonstrated that its deletion retards aging in mice. Recently, new insights in the molecular mechanisms involving p66Shc and the p53 tumor suppressor genes were given: a specific p66Shc/p53 transcriptional regulation pathway was uncovered as determinant in oxidative stress response and, likely, in aging. p53, in a p66Shc-dependent manner, negatively downregulates the expression of 200 genes which are involved in the G2/M transition of mitotic cell cycle and are downregulated during physiological aging. p66Shc modulates the response of p53 by activating a p53 isoform (p44/p53, also named Delta40p53). Based on these latest results, several developments are expected in the future, as the generation of animal models to study aging and the evaluation of the use of the p53/p66Shc target genes as biomarkers in aging related diseases. The aim of this review is to investigate the conservation of the p66Shc and p53 role in oxidative stress between fish and mammals. We propose to approach this study trough a new model organism, the annual fish Nothobranchius furzeri, that has been demonstrated to develop typical signs of aging, like in mammals, including senescence, neurodegeneration, metabolic disorders and cancer.
Collapse
Affiliation(s)
- Chiara Priami
- 1European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy. ; 3Dipartimento di Bioscienze, University of Milan, Italy
| | - Giulia De Michele
- 1European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy
| | | | | | - Marco Giorgio
- 1European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy
| | - Pier Giuseppe Pelicci
- 1European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy. ; 2Dipartimento di Medicina, Chirurgia e Odontoiatria, University of Milan, Italy
| | - Enrica Migliaccio
- 1European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy
| |
Collapse
|
33
|
Inhibition of p53 deSUMOylation exacerbates puromycin aminonucleoside-induced apoptosis in podocytes. Int J Mol Sci 2014; 15:21314-30. [PMID: 25411797 PMCID: PMC4264227 DOI: 10.3390/ijms151121314] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 11/04/2014] [Accepted: 11/05/2014] [Indexed: 11/17/2022] Open
Abstract
Apoptosis is a major cause of reduced podocyte numbers, which leads to proteinuria and/or glomerulosclerosis. Emerging evidence has indicated that deSUMOylation, a dynamic post-translational modification that reverses SUMOylation, is involved in the apoptosis of Burkitt’s lymphoma cells and cardiomyocytes; however, the impact of deSUMOylation on podocyte apoptosis remains unexplored. The p53 protein plays a major role in the pathogenesis of podocyte apoptosis, and p53 can be SUMOylated. Therefore, in the present study, we evaluated the effect of p53 deSUMOylation, which is regulated by sentrin/SUMO-specific protease 1 (SENP1), on podocyte apoptosis. Our results showed that SENP1 deficiency significantly increases puromycin aminonucleoside (PAN)-induced podocyte apoptosis. Moreover, SENP1 knockdown results in the accumulation of SUMOylated p53 protein and the increased expression of the p53 target pro-apoptotic genes, BAX, Noxa and PUMA, in podocytes during PAN stimulation. Thus, SENP1 may be essential for preventing podocyte apoptosis, at least partly through regulating the functions of p53 protein via deSUMOylation. The regulation of deSUMOylation may provide a novel strategy for the treatment of glomerular disorders that involve podocyte apoptosis.
Collapse
|
34
|
Abstract
The ubiquitin proteasome pathway is critical in restraining the activities of the p53 tumor suppressor. This review by Pant and Lozano focuses on ubiquitination as a mechanism for regulating p53 stability and function and reviews current findings from in vivo models that evaluate the importance of the ubiquitin proteasome system in regulating p53. The ubiquitin proteasome pathway is critical in restraining the activities of the p53 tumor suppressor. Numerous E3 and E4 ligases regulate p53 levels. Additionally, deubquitinating enzymes that modify p53 directly or indirectly also impact p53 function. When alterations of these proteins result in increased p53 activity, cells arrest in the cell cycle, senesce, or apoptose. On the other hand, alterations that result in decreased p53 levels yield tumor-prone phenotypes. This review focuses on the physiological relevance of these important regulators of p53 and their therapeutic implications.
Collapse
Affiliation(s)
- Vinod Pant
- Department of Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Guillermina Lozano
- Department of Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
35
|
Multifunctional role of ATM/Tel1 kinase in genome stability: from the DNA damage response to telomere maintenance. BIOMED RESEARCH INTERNATIONAL 2014; 2014:787404. [PMID: 25247188 PMCID: PMC4163350 DOI: 10.1155/2014/787404] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 07/28/2014] [Accepted: 08/07/2014] [Indexed: 12/19/2022]
Abstract
The mammalian protein kinase ataxia telangiectasia mutated (ATM) is a key regulator of the DNA double-strand-break response and belongs to the evolutionary conserved phosphatidylinositol-3-kinase-related protein kinases. ATM deficiency causes ataxia telangiectasia (AT), a genetic disorder that is characterized by premature aging, cerebellar neuropathy, immunodeficiency, and predisposition to cancer. AT cells show defects in the DNA damage-response pathway, cell-cycle control, and telomere maintenance and length regulation. Likewise, in Saccharomyces cerevisiae, haploid strains defective in the TEL1 gene, the ATM ortholog, show chromosomal aberrations and short telomeres. In this review, we outline the complex role of ATM/Tel1 in maintaining genomic stability through its control of numerous aspects of cellular survival. In particular, we describe how ATM/Tel1 participates in the signal transduction pathways elicited by DNA damage and in telomere homeostasis and its importance as a barrier to cancer development.
Collapse
|
36
|
Fan Y, Yin S, Hao Y, Yang J, Zhang H, Sun C, Ma M, Chang Q, Xi JJ. miR-19b promotes tumor growth and metastasis via targeting TP53. RNA (NEW YORK, N.Y.) 2014; 20:765-72. [PMID: 24742936 PMCID: PMC4024631 DOI: 10.1261/rna.043026.113] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Tumor suppressor TP53 (or p53) is one of the most important regulators in numerous physiological and pathological processes. Recently, the miRNA-mediated post-transcription regulation of p53 has been studied. However, systematic studies of miRNA targeting sites within the p53 gene are still a challenging task. Here, we developed a dual-color assay capable of identifying miRNA targeting sites in a certain gene, specifically p53, in a simple, direct, and robust manner. Results showed that p53 was a direct and critical target of miR-19b, but not miR-19a, regardless of sequence similarity. Overexpression of miR-19b observed in human cancer cells can diminish p53 protein levels and, subsequently, downstream components such as Bax and p21. This miR-19b-mediated p53 reduction was shown to promote cell cycle, cell migration or invasion, and repress senescence and apoptosis in vitro. Further investigation revealed that miR-19b controls tumor growth and metastasis in vivo. Therefore, it is possible that miR-19b antagomirs or sponges could be developed as therapeutic agents against tumor development.
Collapse
Affiliation(s)
- Yu Fan
- Biomedical Engineering Department, College of Engineering
| | - Shenyi Yin
- Biomedical Engineering Department, College of Engineering
| | - Yang Hao
- Biomedical Engineering Department, College of Engineering
| | - Junyu Yang
- Biomedical Engineering Department, College of Engineering
| | - Hanshuo Zhang
- Biomedical Engineering Department, College of Engineering
| | - Changhong Sun
- Biomedical Engineering Department, College of Engineering
| | - Ming Ma
- Biomedical Engineering Department, College of Engineering
| | - Qing Chang
- Biomedical Engineering Department, College of Engineering
- Corresponding authorsE-mail E-mail
| | - Jianzhong Jeff Xi
- Biomedical Engineering Department, College of Engineering
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Molecular Medicine, Peking University, Beijing, 100871, China
- Corresponding authorsE-mail E-mail
| |
Collapse
|
37
|
Abstract
The RING domain ubiquitin E3 ligase MDM2 is a key regulator of p53 degradation and a mediator of signals that stabilize p53. The current understanding of the mechanisms by which MDM2 posttranslational modifications and protein binding cause p53 stabilization remains incomplete. Here we present evidence that the MDM2 central acidic region is critical for activating RING domain E3 ligase activity. A 30-amino-acid minimal region of the acidic domain binds to the RING domain through intramolecular interactions and stimulates the catalytic function of the RING domain in promoting ubiquitin release from charged E2. The minimal activation sequence is also the binding site for the ARF tumor suppressor, which inhibits ubiquitination of p53. The acidic domain-RING domain intramolecular interaction is modulated by ATM-mediated phosphorylation near the RING domain or by binding of ARF. These results suggest that MDM2 phosphorylation and association with protein regulators share a mechanism in inhibiting the E3 ligase function and stabilizing p53 and suggest that targeting the MDM2 autoactivation mechanism may be useful for therapeutic modulation of p53 levels.
Collapse
|
38
|
Nguyen TA, Menendez D, Resnick MA, Anderson CW. Mutant TP53 posttranslational modifications: challenges and opportunities. Hum Mutat 2014; 35:738-55. [PMID: 24395704 DOI: 10.1002/humu.22506] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 01/02/2014] [Indexed: 12/13/2022]
Abstract
The wild-type (WT) human p53 (TP53) tumor suppressor can be posttranslationally modified at over 60 of its 393 residues. These modifications contribute to changes in TP53 stability and in its activity as a transcription factor in response to a wide variety of intrinsic and extrinsic stresses in part through regulation of protein-protein and protein-DNA interactions. The TP53 gene frequently is mutated in cancers, and in contrast to most other tumor suppressors, the mutations are mostly missense often resulting in the accumulation of mutant (MUT) protein, which may have novel or altered functions. Most MUT TP53s can be posttranslationally modified at the same residues as in WT TP53. Strikingly, however, codons for modified residues are rarely mutated in human tumors, suggesting that TP53 modifications are not essential for tumor suppression activity. Nevertheless, these modifications might alter MUT TP53 activity and contribute to a gain-of-function leading to increased metastasis and tumor progression. Furthermore, many of the signal transduction pathways that result in TP53 modifications are altered or disrupted in cancers. Understanding the signaling pathways that result in TP53 modification and the functions of these modifications in both WT TP53 and its many MUT forms may contribute to more effective cancer therapies.
Collapse
Affiliation(s)
- Thuy-Ai Nguyen
- Chromosome Stability Section, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | | | | | | |
Collapse
|
39
|
Deletion of individual Ku subunits in mice causes an NHEJ-independent phenotype potentially by altering apurinic/apyrimidinic site repair. PLoS One 2014; 9:e86358. [PMID: 24466051 PMCID: PMC3900520 DOI: 10.1371/journal.pone.0086358] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 12/07/2013] [Indexed: 01/25/2023] Open
Abstract
Ku70 and Ku80 form a heterodimer called Ku that forms a holoenzyme with DNA dependent-protein kinase catalytic subunit (DNA-PKCS) to repair DNA double strand breaks (DSBs) through the nonhomologous end joining (NHEJ) pathway. As expected mutating these genes in mice caused a similar DSB repair-defective phenotype. However, ku70-/- cells and ku80-/- cells also appeared to have a defect in base excision repair (BER). BER corrects base lesions, apurinic/apyrimidinic (AP) sites and single stand breaks (SSBs) utilizing a variety of proteins including glycosylases, AP endonuclease 1 (APE1) and DNA Polymerase β (Pol β). In addition, deleting Ku70 was not equivalent to deleting Ku80 in cells and mice. Therefore, we hypothesized that free Ku70 (not bound to Ku80) and/or free Ku80 (not bound to Ku70) possessed activity that influenced BER. To further test this hypothesis we performed two general sets of experiments. The first set showed that deleting either Ku70 or Ku80 caused an NHEJ-independent defect. We found ku80-/- mice had a shorter life span than dna-pkcs-/- mice demonstrating a phenotype that was greater than deleting the holoenzyme. We also found Ku70-deletion induced a p53 response that reduced the level of small mutations in the brain suggesting defective BER. We further confirmed that Ku80-deletion impaired BER via a mechanism that was not epistatic to Pol β. The second set of experiments showed that free Ku70 and free Ku80 could influence BER. We observed that deletion of either Ku70 or Ku80, but not both, increased sensitivity of cells to CRT0044876 (CRT), an agent that interferes with APE1. In addition, free Ku70 and free Ku80 bound to AP sites and in the case of Ku70 inhibited APE1 activity. These observations support a novel role for free Ku70 and free Ku80 in altering BER.
Collapse
|
40
|
Garcia PB, Attardi LD. Illuminating p53 function in cancer with genetically engineered mouse models. Semin Cell Dev Biol 2014; 27:74-85. [PMID: 24394915 DOI: 10.1016/j.semcdb.2013.12.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/06/2013] [Accepted: 12/23/2013] [Indexed: 01/23/2023]
Abstract
The key role of the p53 protein in tumor suppression is highlighted by its frequent mutation in human cancers and by the completely penetrant cancer predisposition of p53 null mice. Beyond providing definitive evidence for the critical function of p53 in tumor suppression, genetically engineered mouse models have offered numerous additional insights into p53 function. p53 knock-in mice expressing tumor-derived p53 mutants have revealed that these mutants display gain-of-function activities that actively promote carcinogenesis. The generation of p53 knock-in mutants with alterations in different domains of p53 has helped further elucidate the cellular and biochemical activities of p53 that are most fundamental for tumor suppression. In addition, modulation of p53 post-translational modification (PTM) status by generating p53 knock-in mouse strains with mutations in p53 PTM sites has revealed a subtlety and complexity to p53 regulation. Analyses of mouse models perturbing upstream regulators of p53 have solidified the notion that the p53 pathway can be compromised by means other than direct p53 mutation. Finally, switchable p53 models that allow p53 reactivation in tumors have helped evaluate the potential of p53 restoration therapy for cancer treatment. Collectively, mouse models have greatly enhanced our understanding of physiological p53 function and will continue to provide new biological and clinical insights in future investigations.
Collapse
Affiliation(s)
- Patty B Garcia
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Laura D Attardi
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
41
|
Requirement for phosphorylation of P53 at Ser312 in suppression of chemical carcinogenesis. Sci Rep 2013; 3:3105. [PMID: 24173284 PMCID: PMC3813944 DOI: 10.1038/srep03105] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 10/11/2013] [Indexed: 12/23/2022] Open
Abstract
The p53 tumour suppressor is activated in response to a wide variety of genotoxic stresses, frequently via post-translational modification. Using a knock in mouse model with a Ser312 to Ala mutation, we show here that phosphorylation of p53 on Ser312 helps to prevent tumour induction by the alkylating agent MNU, which predominantly caused T cell lymphomas. This is consistent with our previous observation that p53312A/A mice are more susceptible to X-ray induced tumourigenesis. Phosphorylation on Ser312 aids p53's interaction with E2F1, and enhances p53-mediated apoptosis. Loss of E2F1 alone does not affect tumour susceptibility to MNU, but its absence partially rescues tumour formation in p53312A/A mice, thus reflecting the oncogenic properties of E2F1. Our data confirms the participation of Ser312 phosphorylation in tumour suppression by p53.
Collapse
|
42
|
Another fork in the road--life or death decisions by the tumour suppressor p53. EMBO Rep 2013; 14:414-21. [PMID: 23588418 DOI: 10.1038/embor.2013.25] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 02/13/2013] [Indexed: 12/18/2022] Open
Abstract
In response to cellular stress signals, the tumour suppressor p53 accumulates and triggers a host of antineoplastic responses. For instance, DNA damage activates two main p53-dependent responses: cell cycle arrest and attendant DNA repair or apoptosis (cell death). It is broadly accepted that, in response to DNA damage, the function of p53 as a sequence-specific transcription factor is crucial for tumour suppression. The molecular determinants, however, that favour the initiation of either a p53-dependent cell cycle arrest (life) or apoptotic (death) transcriptional programme remain elusive. Gaining a clear understanding of the mechanisms controlling cell fate determination by p53 could lead to the identification of molecular targets for therapy, which could selectively sensitize cancer cells to apoptosis. This review summarizes the literature addressing this important question in the field. Special emphasis is given to the role of the p53 response element, post-translational modifications and protein-protein interactions on cell fate decisions made by p53 in response to DNA damage.
Collapse
|
43
|
Stracker TH, Roig I, Knobel PA, Marjanović M. The ATM signaling network in development and disease. Front Genet 2013; 4:37. [PMID: 23532176 PMCID: PMC3607076 DOI: 10.3389/fgene.2013.00037] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 03/04/2013] [Indexed: 12/15/2022] Open
Abstract
The DNA damage response (DDR) rapidly recognizes DNA lesions and initiates the appropriate cellular programs to maintain genome integrity. This includes the coordination of cell cycle checkpoints, transcription, translation, DNA repair, metabolism, and cell fate decisions, such as apoptosis or senescence (Jackson and Bartek, 2009). DNA double-strand breaks (DSBs) represent one of the most cytotoxic DNA lesions and defects in their metabolism underlie many human hereditary diseases characterized by genomic instability (Stracker and Petrini, 2011; McKinnon, 2012). Patients with hereditary defects in the DDR display defects in development, particularly affecting the central nervous system, the immune system and the germline, as well as aberrant metabolic regulation and cancer predisposition. Central to the DDR to DSBs is the ataxia-telangiectasia mutated (ATM) kinase, a master controller of signal transduction. Understanding how ATM signaling regulates various aspects of the DDR and its roles in vivo is critical for our understanding of human disease, its diagnosis and its treatment. This review will describe the general roles of ATM signaling and highlight some recent advances that have shed light on the diverse roles of ATM and related proteins in human disease.
Collapse
Affiliation(s)
- Travis H. Stracker
- Oncology Programme, Institute for Research in Biomedicine (IRB Barcelona)Barcelona, Spain
| | - Ignasi Roig
- Departament de Biologia Cellular, Fisiologia i Immunologia, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de BarcelonBarcelona, Spain
| | - Philip A. Knobel
- Oncology Programme, Institute for Research in Biomedicine (IRB Barcelona)Barcelona, Spain
| | - Marko Marjanović
- Oncology Programme, Institute for Research in Biomedicine (IRB Barcelona)Barcelona, Spain
| |
Collapse
|
44
|
Abstract
BACKGROUND The p53 gene is the most frequently mutated gene in cancer and accordingly has been the subject of intensive investigation for almost 30 years. Loss of p53 function due to mutations has been unequivocally demonstrated to promote cancer in both humans and in model systems. As a consequence, there exists an enormous body of information regarding the function of normal p53 in biology and the pathobiological consequences of p53 mutation. It has long been recognised that analysis of p53 has considerable potential as a tool for use in both diagnostic and, to a greater extent, prognostic settings and some significant progress has been made in both of these arenas. OBJECTIVE To provide an overview of the biology of p53, particularly in the context of uses of p53 as a diagnostic tool. METHODS A literature review focused upon the methods and uses of p53 analysis in the diagnosis of sporadic cancers, rare genetic disorders and in detection of residual disease. CONCLUSION p53 is currently an essential diagnostic for the rare inherited cancer prone syndrome (Li-Fraumeni) and is an important diagnostic in only a limited number of settings in sporadic disease. Research in specific cancers indicates that the uses of increasingly well informed p53 mutational analysis are likely to expand to other cancers.
Collapse
Affiliation(s)
- Mark T Boyd
- Reader in Molecular Oncology and Director of Laboratories University of Liverpool, p53/MDM2 Research Team, Division of Surgery and Oncology, School of Cancer Studies, 5th Floor, UCD Building, Daulby Street, Liverpool L69 3GA, UK +44 151 706 4185 ; +44 151 706 5826 ;
| | | |
Collapse
|
45
|
Reimand J, Bader GD. Systematic analysis of somatic mutations in phosphorylation signaling predicts novel cancer drivers. Mol Syst Biol 2013; 9:637. [PMID: 23340843 PMCID: PMC3564258 DOI: 10.1038/msb.2012.68] [Citation(s) in RCA: 201] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 12/06/2012] [Indexed: 12/20/2022] Open
Abstract
Large-scale cancer genome sequencing has uncovered thousands of gene mutations, but distinguishing tumor driver genes from functionally neutral passenger mutations is a major challenge. We analyzed 800 cancer genomes of eight types to find single-nucleotide variants (SNVs) that precisely target phosphorylation machinery, important in cancer development and drug targeting. Assuming that cancer-related biological systems involve unexpectedly frequent mutations, we used novel algorithms to identify genes with significant phosphorylation-associated SNVs (pSNVs), phospho-mutated pathways, kinase networks, drug targets, and clinically correlated signaling modules. We highlight increased survival of patients with TP53 pSNVs, hierarchically organized cancer kinase modules, a novel pSNV in EGFR, and an immune-related network of pSNVs that correlates with prolonged survival in ovarian cancer. Our findings include multiple actionable cancer gene candidates (FLNB, GRM1, POU2F1), protein complexes (HCF1, ASF1), and kinases (PRKCZ). This study demonstrates new ways of interpreting cancer genomes and presents new leads for cancer research.
Collapse
Affiliation(s)
- Jüri Reimand
- The Donnelly Centre, University of Toronto, Toronto, Canada
| | - Gary D Bader
- The Donnelly Centre, University of Toronto, Toronto, Canada
| |
Collapse
|
46
|
Chen J. The Roles of MDM2 and MDMX Phosphorylation in Stress Signaling to p53. Genes Cancer 2012; 3:274-82. [PMID: 23150760 DOI: 10.1177/1947601912454733] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The p53 tumor suppressor is highly responsive to different physiological stresses such as abnormal cell proliferation, nutrient deprivation, and DNA damage. Distinct signaling mechanisms have evolved to activate p53, which in turn modulate numerous pathways to enhance fitness and survival of the organism. Elucidating the molecular mechanisms of these signaling events is critical for understanding tumor suppression by p53 and development of novel therapeutics. Studies in the past decade have established that MDM2 and MDMX are important targets of signaling input from different pathways. Here, we focus our discussion on MDM2 and MDMX phosphorylation, which is important for p53 activation by DNA damage. Investigations in this area have generated new insight into the inner workings of MDM2 and MDMX and underscore the importance of allosteric communication between different domains in achieving an efficient response to phosphorylation. It is likely that MDM2 and MDMX regulation by phosphorylation will share mechanistic similarities to other signaling hub molecules. Phosphorylation-independent p53 activators such as ARF and ribosomal proteins ultimately achieve the same outcome as phosphorylation, suggesting that they may induce similar changes in the structure and function of MDM2 and MDMX through protein-protein interactions.
Collapse
Affiliation(s)
- Jiandong Chen
- Molecular Oncology Department, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
47
|
Gannon HS, Jones SN. Using Mouse Models to Explore MDM-p53 Signaling in Development, Cell Growth, and Tumorigenesis. Genes Cancer 2012; 3:209-18. [PMID: 23150754 DOI: 10.1177/1947601912455324] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The p53 transcription factor regulates the expression of numerous genes whose products affect cell proliferation, senescence, cellular metabolism, apoptosis, and DNA repair. These p53-mediated effects can inhibit the growth of stressed or mutated cells and suppress tumorigenesis in the organism. However, the various growth-inhibitory properties of p53 must be kept in check in nondamaged cells in order to facilitate proper embryogenesis or the homeostatic maintenance of adult tissues. This requisite inhibition of p53 is performed primarily by the MDM oncoproteins, Mdm2 and MdmX. These p53-binding proteins limit p53 activity both in normal cells and in stressed cells seeking to promote resolution of their p53-stress response. Many mouse models bearing genetic alterations in Mdm2 or MdmX have been generated to explore the function and regulation of MDM-p53 signaling in development, in tissue homeostasis, in aging, and in cancer. These models not only have demonstrated a critical need for Mdm2 and MdmX in normal cell growth and in development but more recently have identified the MDM-p53 signaling axis as a key regulator of the cellular response to a wide variety of genetic or metabolic stresses. In this review, we discuss what has been learned from various studies of these Mdm2 and MdmX mouse models and highlight a few of the many important remaining questions.
Collapse
Affiliation(s)
- Hugh S Gannon
- University of Massachusetts Medical School, Worcester, MA, USA
| | | |
Collapse
|
48
|
Abstract
Aberrations in the p53 tumor suppressor pathway are associated with hematologic malignancies. p53-dependent cell cycle control, senescence, and apoptosis functions are actively involved in maintaining hematopoietic homeostasis under normal and stress conditions. Whereas loss of p53 function promotes leukemia and lymphoma development in humans and mice, increased p53 activity inhibits hematopoietic stem cell function and results in myelodysplasia. Thus, exquisite regulation of p53 activity is critical for homeostasis. Most of our understanding of p53 function in hematopoiesis is derived from genetically engineered mice. Here we summarize some of these models, the various mechanisms that disrupt the regulation of p53 activity, and their relevance to human disease.
Collapse
|
49
|
Lake BB, Fink J, Klemetsaune L, Fu X, Jeffers JR, Zambetti GP, Xu Y. Context-dependent enhancement of induced pluripotent stem cell reprogramming by silencing Puma. Stem Cells 2012; 30:888-97. [PMID: 22311782 DOI: 10.1002/stem.1054] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Reprogramming of the somatic state to pluripotency can be induced by a defined set of transcription factors including Oct3/4, Sox2, Klf4, and c-Myc [Cell 2006;126:663-676]. These induced pluripotent stem cells (iPSCs) hold great promise in human therapy and disease modeling. However, tumor suppressive activities of p53, which are necessary to prevent persistence of DNA damage in mammalian cells, have proven a serious impediment to formation of iPSCs [Nat Methods 2011;8:409-412]. We examined the requirement for downstream p53 activities in suppressing efficiency of reprogramming as well as preventing persistence of DNA damage into the early iPSCs. We discovered that the majority of the p53 activation occurred through early reprogramming-induced DNA damage with the activated expression of the apoptotic inducer Puma and the cell cycle inhibitor p21. While Puma deficiency increases reprogramming efficiency only in the absence of c-Myc, double deficiency of Puma and p21 has achieved a level of efficiency that exceeded that of p53 deficiency alone. We further demonstrated that, in both the presence and absence of p21, Puma deficiency was able to prevent any increase in persistent DNA damage in early iPSCs. This may be due to a compensatory cellular senescent response to reprogramming-induced DNA damage in pre-iPSCs. Therefore, our findings provide a potentially safe approach to enhance iPSC derivation by transiently silencing Puma and p21 without compromising genomic integrity.
Collapse
Affiliation(s)
- Blue B Lake
- Division of Biological Sciences, University of California, 9500 Gilman Drive, La Jolla, California, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Gannon HS, Woda BA, Jones SN. ATM phosphorylation of Mdm2 Ser394 regulates the amplitude and duration of the DNA damage response in mice. Cancer Cell 2012; 21:668-679. [PMID: 22624716 PMCID: PMC3360886 DOI: 10.1016/j.ccr.2012.04.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 12/13/2011] [Accepted: 04/03/2012] [Indexed: 11/25/2022]
Abstract
DNA damage induced by ionizing radiation activates the ATM kinase, which subsequently stabilizes and activates the p53 tumor suppressor protein. Although phosphorylation of p53 by ATM was found previously to modulate p53 levels and transcriptional activities in vivo, it does not appear to be a major regulator of p53 stability. We have utilized mice bearing altered Mdm2 alleles to demonstrate that ATM phosphorylation of Mdm2 serine 394 is required for robust p53 stabilization and activation after DNA damage. In addition, we demonstrate that dephosphorylation of Mdm2 Ser394 regulates attenuation of the p53-mediated response to DNA damage. Therefore, the phosphorylation status of Mdm2 Ser394 governs p53 protein levels and functions in cells undergoing DNA damage.
Collapse
Affiliation(s)
- Hugh S Gannon
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Bruce A Woda
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Stephen N Jones
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA; Department of Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| |
Collapse
|