1
|
Tosios KI, Kalogirou EM, Koutlas IG. Association of MDM2 Overexpression in Ameloblastomas with MDM2 Amplification and BRAF V600E Expression. Int J Mol Sci 2024; 25:2238. [PMID: 38396916 PMCID: PMC10889355 DOI: 10.3390/ijms25042238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/01/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Ameloblastoma is a rare tumor but represents the most common odontogenic neoplasm. It is localized in the jaws and, although it is a benign, slow-growing tumor, it has an aggressive local behavior and high recurrence rate. Therefore, alternative treatment options or complementary to surgery have been evaluated, with the most promising one among them being a targeted therapy with the v-Raf murine sarcoma viral oncogene homologue B (BRAF), as in ameloblastoma the activating mutation V600E in BRAF is common. Studies in other tumors have shown that the synchronous inhibition of BRAF and human murine double minute 2 homologue (MDM2 or HDM2) protein is more effective than BRAF monotherapy, particularly in the presence of wild type p53 (WTp53). To investigate the MDM2 protein expression and gene amplification in ameloblastoma, in association with BRAFV600E and p53 expression. Forty-four cases of ameloblastoma fixed in 10% buffered formalin and embedded in paraffin were examined for MDM2 overexpression and BRAFV600E and p53 expression by immunohistochemistry, and for MDM2 ploidy with fluorescence in situ hybridization. Sixteen of forty-four (36.36%) cases of ameloblastoma showed MDM2 overexpression. Seven of sixteen MDM2-positive ameloblastomas (43.75%) were BRAFV600E positive and fifteen of sixteen MDM2-positive ameloblastomas (93.75%) were p53 negative. All MDM2 overexpressing tumors did not show copy number alterations for MDM2. Overexpression of MDM2 in ameloblastomas is not associated with MDM2 amplification, but most probably with MAPK activation and WTp53 expression. Further verification of those findings could form the basis for the use of MDM2 expression as a marker of MAPK activation in ameloblastomas and the trial of dual BRAF/MDM2 inhibition in the management of MDM2-overexpressing/BRAFV600E-positive/WTp53 ameloblastomas.
Collapse
Affiliation(s)
- Konstantinos I. Tosios
- Department of Oral Pathology & Medicine and Hospital Dentistry, School of Dentistry, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Eleni-Marina Kalogirou
- Faculty of Health and Rehabilitation Sciences, Metropolitan College, 15125 Athens, Greece;
| | - Ioannis G. Koutlas
- Division of Oral Pathology, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
2
|
Lin W, Yan Y, Huang Q, Zheng D. MDMX in Cancer: A Partner of p53 and a p53-Independent Effector. Biologics 2024; 18:61-78. [PMID: 38318098 PMCID: PMC10839028 DOI: 10.2147/btt.s436629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/08/2023] [Indexed: 02/07/2024]
Abstract
The p53 tumor suppressor protein plays an important role in physiological and pathological processes. MDM2 and its homolog MDMX are the most important negative regulators of p53. Many studies have shown that MDMX promotes the growth of cancer cells by influencing the regulation of the downstream target gene of tumor suppressor p53. Studies have found that inhibiting the MDMX-p53 interaction can effectively restore the tumor suppressor activity of p53. MDMX has growth-promoting activities without p53 or in the presence of mutant p53. Therefore, it is extremely important to study the function of MDMX in tumorigenesis, progression and prognosis. This article mainly reviews the current research progress and mechanism on MDMX function, summarizes known MDMX inhibitors and provides new ideas for the development of more specific and effective MDMX inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Wu Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Yuxiang Yan
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Qingling Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Dali Zheng
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People’s Republic of China
| |
Collapse
|
3
|
López-Iniesta MJ, Parkar SN, Ramalho AC, Lacerda R, Costa IF, Zhao J, Romão L, Candeias MM. Conserved Double Translation Initiation Site for Δ160p53 Protein Hints at Isoform's Key Role in Mammalian Physiology. Int J Mol Sci 2022; 23:ijms232415844. [PMID: 36555484 PMCID: PMC9779343 DOI: 10.3390/ijms232415844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
p53 is the most commonly mutated gene in human cancers. Two fundamental reasons for this are its long protein isoforms protect from cancer, while its shorter C-terminal isoforms can support cancer and metastasis. Previously, we have shown that the Δ160p53 protein isoform enhances survival and the invasive character of cancer cells. Here, we identified a translation initiation site nine codons downstream of codon 160-the known initiation codon for the translation of Δ160p53-that is recognized by the translation machinery. When translation failed to initiate from AUG160 due to mutation, it initiated from AUG169 instead, producing similar levels of a similar protein, Δ169p53, which promoted cell survival as efficiently as Δ160p53 following DNA damage. Interestingly, almost all mammalian species with an orthologue to human AUG160 also possess one for AUG169, while none of the non-mammalian species lacking AUG160 have AUG169, even if that region of the p53 gene is well conserved. In view of our findings, we do not believe that Δ169p53 acts as a different p53 protein isoform; instead, we propose that the double translation initiation site strengthens the translation of these products with a critical role in cell homeostasis. Future studies will help verify if this is a more general mechanism for the expression of essential proteins in mammals.
Collapse
Affiliation(s)
- Maria José López-Iniesta
- MaRCU—Molecular and RNA Cancer Unit, Kyoto 606-8501, Japan
- Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Shrutee N. Parkar
- MaRCU—Molecular and RNA Cancer Unit, Kyoto 606-8501, Japan
- Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Ana Catarina Ramalho
- MaRCU—Molecular and RNA Cancer Unit, Kyoto 606-8501, Japan
- Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
- Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge, 1649-016 Lisbon, Portugal
- BioISI–Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
- Laboratory of Cancer cell Biology, Graduate School of Biostudies, Kyoto University, 606-8501 Kyoto, Japan
| | - Rafaela Lacerda
- MaRCU—Molecular and RNA Cancer Unit, Kyoto 606-8501, Japan
- Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge, 1649-016 Lisbon, Portugal
- BioISI–Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Inês F. Costa
- MaRCU—Molecular and RNA Cancer Unit, Kyoto 606-8501, Japan
- Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge, 1649-016 Lisbon, Portugal
- BioISI–Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Jingyuan Zhao
- MaRCU—Molecular and RNA Cancer Unit, Kyoto 606-8501, Japan
- Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Luísa Romão
- Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge, 1649-016 Lisbon, Portugal
- BioISI–Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Marco M. Candeias
- MaRCU—Molecular and RNA Cancer Unit, Kyoto 606-8501, Japan
- Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
- Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge, 1649-016 Lisbon, Portugal
- BioISI–Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
- Correspondence: ; Tel.: +81-(0)75-753-9297
| |
Collapse
|
4
|
Liu Y, Cao B, Hu L, Ye J, Tian W, He X. The Dual Roles of MAGE-C2 in p53 Ubiquitination and Cell Proliferation Through E3 Ligases MDM2 and TRIM28. Front Cell Dev Biol 2022; 10:922675. [PMID: 35927984 PMCID: PMC9344466 DOI: 10.3389/fcell.2022.922675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/17/2022] [Indexed: 01/10/2023] Open
Abstract
The tumor suppressor p53 is critical for the maintenance of genome stability and protection against tumor malignant transformation, and its homeostasis is usually regulated by ubiquitination. MDM2 is a major E3 ligase of p53 ubiquitination, and its activity is enhanced by TRIM28. TRIM28 also independently ubiquitinates p53 as an E3 ligase activated by MAGE-C2. Moreover, MAGE-C2 is highly expressed in various cancers, but the detailed mechanisms of MAGE-C2 involved in MDM2/TRIM28-mediated p53 ubiquitination remain unknown. Here, we found that MAGE-C2 directly interacts with MDM2 through its conserved MHD domain to inhibit the activity of MDM2 on p53 ubiquitination. Furthermore, TRIM28 acts as an MAGE-C2 binding partner and directly competes with MAGE-C2 for MDM2 interaction, thus releasing the inhibitory role of MAGE-C2 and promoting p53 ubiquitination. MAGE-C2 suppresses cell proliferation in TRIM28-deficient cells, but the overexpression of TRIM28 antagonizes the inhibitory role of MAGE-C2 and accumulates p53 ubiquitination to promote cell proliferation. This study clarified the molecular link of MAGE-C2 in two major E3 systems MDM2 and TRIM28 on p53 ubiquitination. Our results revealed the molecular function of how MAGE-C2 and TRIM28 contribute to p53 ubiquitination and cell proliferation, in which MAGE-C2 acts as a potential inhibitor of MDM2 and TRIM28 is a vital regulator for MAGE-C2 function in p53 protein level and cell proliferation. This work would be helpful to understand the regulation mechanism of tumor suppressor p53.
Collapse
|
5
|
Huang X, Ma C, Huang W, Dai M, Yang J, Xu X, Deng Y, Yang Y, Zhang H. Environmental BPDE induced human trophoblast cell apoptosis by up-regulating lnc-HZ01/p53 positive feedback loop. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 237:113564. [PMID: 35483139 DOI: 10.1016/j.ecoenv.2022.113564] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/08/2022] [Accepted: 04/21/2022] [Indexed: 06/14/2023]
Abstract
Human trophoblast cell apoptosis may induce miscarriage. Trophoblast cells are sensitive to environmental BaP-7,8-dihydrodiol-9,10-epoxide (BPDE). However, how BPDE induces human trophoblast cell apoptosis is still largely elusive. In this work, we used BPDE-treated human trophoblast cells and villous tissues collected from recurrent miscarriage and health control groups to explore the underlying mechanism of BPDE-induced human trophoblast cell apoptosis. Continued with our recent work, we found that lncRNA HZ01 (lnc-HZ01) could induce human trophoblast cell apoptosis. In mechanism, lnc-HZ01 up-regulated p53 expression level by suppressing its MDM2-mediated proteasomal degradation. Meanwhile, we found that p53 acted as lnc-HZ01 transcription factor and promoted lnc-HZ01 transcription. Thus, lnc-HZ01 and p53 composed a positive feedback loop in human trophoblast cells. In normal trophoblast cells, relatively low levels of lnc-HZ01 and p53 suppressed p53/caspase-3 apoptosis pathway, giving normal pregnancy. Upon BPDE exposure, BPDE up-regulated the expression levels of lnc-HZ01 and p53, triggered this positive feedback loop, activated the p53/caspase-3 apoptosis pathway, and then induced miscarriage. Collectively, we discovered new mechanism by which lnc-HZ01 regulated BPDE-induced human trophoblast cell apoptosis, providing scientific basis for the diagnosis and treatment of unexplained recurrent miscarriage.
Collapse
MESH Headings
- 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/metabolism
- 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/toxicity
- Abortion, Habitual/chemically induced
- Abortion, Habitual/metabolism
- Apoptosis
- Caspase 3/metabolism
- Feedback
- Female
- Humans
- Pregnancy
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Trophoblasts/metabolism
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Xinying Huang
- Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Chenglong Ma
- Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Wenxin Huang
- Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Mengyuan Dai
- Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Jingjing Yang
- Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Xiaole Xu
- Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Yuanlv Deng
- Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Yang Yang
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Huidong Zhang
- Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China.
| |
Collapse
|
6
|
Hu H, Ye L, Liu Z. GINS2 regulates the proliferation and apoptosis of colon cancer cells through PTP4A1. Mol Med Rep 2022; 25:117. [PMID: 35137928 PMCID: PMC8855163 DOI: 10.3892/mmr.2022.12633] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/24/2021] [Indexed: 12/24/2022] Open
Abstract
Colon cancer is associated with high death rates worldwide and poses a serious threat to public health. GINS complex subunit 2 (GINS2) serves a carcinogenic role in many cancers, including gastric adenocarcinoma, ovarian cancer and pancreatic cancer. However, the specific function of GINS2 in the development of colon cancer has not been described in detail. The present study aimed to clarify the role of GINS2 in colon cancer. A Cell Counting Kit‑8 assay, EdU staining, TUNEL and flow cytometry analyses were performed to determine the levels of cell viability, proliferation and apoptosis and to evaluate the cell cycle. Through the analysis of BioGrid, a Protein‑Protein Interaction database, it was hypothesized that protein tyrosine phosphatase 4A1 (PTP4A1) is a protein that might interact with GINS2, which was then validated using a co‑immunoprecipitation assay. mRNA and protein levels were measured using reverse transcription‑quantitative PCR and western blotting, respectively. The results of the present study demonstrated that GINS2 expression levels were increased in colon cancer cells. Furthermore, GINS2 knockdown inhibited the proliferation of colon cancer cells, while the levels of cell cycle arrest and apoptosis were increased. By interacting with PTP4A1, GINS2 promoted the expression of PTP4A1, a novel p53 target. GINS2 knockdown was increased, while PTP4A1 overexpression decreased the protein level of p53. Notably, PTP4A1 overexpression partly reversed the effects of GINS2 downregulation on colon cancer cells. Therefore, the present study demonstrated that GINS2 regulated the proliferation and apoptosis of colon cancer cells through PTP4A1/p53 pathway, highlighting that GINS2 may serve as a novel molecular marker for colon cancer prevention and therapy.
Collapse
Affiliation(s)
- Hao Hu
- Department of Endoscopy, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Lina Ye
- Department of Endoscopy, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Zhe Liu
- Department of Gastroenterology, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, P.R. China
| |
Collapse
|
7
|
Li H, Fang H, Chang L, Qiu S, Ren X, Cao L, Bian J, Wang Z, Guo Y, Lv J, Sun Z, Wang T, Li B. TC2N: A Novel Vital Oncogene or Tumor Suppressor Gene In Cancers. Front Immunol 2021; 12:764749. [PMID: 34925334 PMCID: PMC8674203 DOI: 10.3389/fimmu.2021.764749] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022] Open
Abstract
Several C2 domain-containing proteins play key roles in tumorigenesis, signal transduction, and mediating protein–protein interactions. Tandem C2 domains nuclear protein (TC2N) is a tandem C2 domain-containing protein that is differentially expressed in several types of cancers and is closely associated with tumorigenesis and tumor progression. Notably, TC2N has been identified as an oncogene in lung and gastric cancer but as a tumor suppressor gene in breast cancer. Recently, a large number of tumor-associated antigens (TAAs), such as heat shock proteins, alpha-fetoprotein, and carcinoembryonic antigen, have been identified in a variety of malignant tumors. Differences in the expression levels of TAAs between cancer cells and normal cells have led to these antigens being investigated as diagnostic and prognostic biomarkers and as novel targets in cancer treatment. In this review, we summarize the clinical characteristics of TC2N-positive cancers and potential mechanisms of action of TC2N in the occurrence and development of specific cancers. This article provides an exploration of TC2N as a potential target for the diagnosis and treatment of different types of cancers.
Collapse
Affiliation(s)
- Hanyang Li
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
- Department of Thyroid Surgery, The Second Hospital of Jilin University, Changchun, China
| | - He Fang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Li Chang
- Department of Pathology, The Second Hospital of Jilin University, Changchun, China
| | - Shuang Qiu
- Department of Biobank, The China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiaojun Ren
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Lidong Cao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jinda Bian
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Zhenxiao Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Yi Guo
- Department of Breast Surgery, The Affiliated Hospital Changchun University of Chinese Medicine, Changchun, China
| | - Jiayin Lv
- Department of Orthopedics, The China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhihui Sun
- Department of Pharmacy, The Second Hospital of Jilin University, Changchun, China
| | - Tiejun Wang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Tiejun Wang, ; Bingjin Li,
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Tiejun Wang, ; Bingjin Li,
| |
Collapse
|
8
|
Erden Y. Sour black mulberry (Morus nigra L.) causes cell death by decreasing mutant p53 expression in HT-29 human colon cancer cells. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2021.101113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
9
|
Hou Y, Li S, Du W, Li H, Wen R. The Tumor Suppressor Role of the Ras Association Domain Family 10. Anticancer Agents Med Chem 2021; 20:2207-2215. [PMID: 32664845 DOI: 10.2174/1871520620666200714141906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/30/2020] [Accepted: 05/17/2020] [Indexed: 11/22/2022]
Abstract
The Ras association domain family 10(RASSF10), a tumor suppressor gene, is located on human chromosome 11p15.2, which is one of the members homologous to other N-terminal RASSF families obtained through structural prediction. RASSF10 plays an important role in inhibiting proliferation, invasion, and migration, inducing apoptosis, making cancer cells sensitive to docetaxel, and capturing G2/M phase. Some studies have found that RASSF10 may inhibit the occurrence and development of tumors by regulating Wnt/β-catenin, P53, and MMP2. Methylation of tumor suppressor gene promoter is a key factor in the development and progression of many tumors. Various methylation detection methods confirmed that the methylation and downregulation of RASSF10 often occur in various tumors, such as gastric cancer, lung cancer, colon cancer, breast cancer, and leukemia. The status of RASSF10 methylation is positively correlated with tumor size, tumor type, and TNM stage. RASSF10 methylation can be used as a prognostic factor for overall survival and disease-free survival, and is also a sign of tumor diagnosis and sensitivity to docetaxel chemotherapy. In this review, we mainly elucidate the acknowledged structure and progress in the verified functions of RASSF10 and the probably relevant signaling pathways.
Collapse
Affiliation(s)
- Yulong Hou
- Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Shuofeng Li
- Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Wei Du
- Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Hailong Li
- Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Rumin Wen
- Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| |
Collapse
|
10
|
Yu DH, Xu ZY, Mo S, Yuan L, Cheng XD, Qin JJ. Targeting MDMX for Cancer Therapy: Rationale, Strategies, and Challenges. Front Oncol 2020; 10:1389. [PMID: 32850448 PMCID: PMC7419686 DOI: 10.3389/fonc.2020.01389] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022] Open
Abstract
The oncogene MDMX, also known as MDM4 is a critical negative regulator of the tumor suppressor p53 and has been implicated in the initiation and progression of human cancers. Increasing evidence indicates that MDMX is often amplified and highly expressed in human cancers, promotes cancer cell growth, and inhibits apoptosis by dampening p53-mediated transcription of its target genes. Inhibiting MDMX-p53 interaction has been found to be effective for restoring the tumor suppressor activity of p53. Therefore, MDMX is becoming one of the most promising molecular targets for developing anticancer therapeutics. In the present review, we mainly focus on the current MDMX-targeting strategies and known MDMX inhibitors, as well as their mechanisms of action and in vitro and in vivo anticancer activities. We also propose other potential targeting strategies for developing more specific and effective MDMX inhibitors for cancer therapy.
Collapse
Affiliation(s)
- De-Hua Yu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhi-Yuan Xu
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shaowei Mo
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yuan
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiang-Dong Cheng
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jiang-Jiang Qin
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
11
|
SOX2 and p53 Expression Control Converges in PI3K/AKT Signaling with Versatile Implications for Stemness and Cancer. Int J Mol Sci 2020; 21:ijms21144902. [PMID: 32664542 PMCID: PMC7402325 DOI: 10.3390/ijms21144902] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022] Open
Abstract
Stemness and reprogramming involve transcriptional master regulators that suppress cell differentiation while promoting self-renewal. A distinguished example thereof is SOX2, a high mobility group (HMG)-box transcription factor (TF), whose subcellular localization and turnover regulation in embryonic, induced-pluripotent, and cancer stem cells (ESCs, iPSCs, and CSCs, respectively) is mediated by the PI3K/AKT/SOX2 axis, a stem cell-specific branch of the PI3K/AKT signaling pathway. Further effector functions associated with PI3K/AKT induction include cell cycle progression, cellular (mass) growth, and the suppression of apoptosis. Apoptosis, however, is a central element of DNA damage response (DDR), where it provides a default mechanism for cell clearance when DNA integrity cannot be maintained. A key player in DDR is tumor suppressor p53, which accumulates upon DNA-damage and is counter-balanced by PI3K/AKT enforced turnover. Accordingly, stemness sustaining SOX2 expression and p53-dependent DDR mechanisms show molecular–functional overlap in PI3K/AKT signaling. This constellation proves challenging for stem cells whose genomic integrity is a functional imperative for normative ontogenesis. Unresolved mutations in stem and early progenitor cells may in fact provoke transformation and cancer development. Such mechanisms are also particularly relevant for iPSCs, where genetic changes imposed through somatic cell reprogramming may promote DNA damage. The current review aims to summarize the latest advances in the understanding of PI3K/AKT/SOX2-driven stemness and its intertwined relations to p53-signaling in DDR under conditions of pluripotency, reprogramming, and transformation.
Collapse
|
12
|
Wang Y, Jiang X, Feng F, Liu W, Sun H. Degradation of proteins by PROTACs and other strategies. Acta Pharm Sin B 2020; 10:207-238. [PMID: 32082969 PMCID: PMC7016280 DOI: 10.1016/j.apsb.2019.08.001] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/19/2019] [Accepted: 07/30/2019] [Indexed: 12/13/2022] Open
Abstract
Blocking the biological functions of scaffold proteins and aggregated proteins is a challenging goal. PROTAC proteolysis-targeting chimaera (PROTAC) technology may be the solution, considering its ability to selectively degrade target proteins. Recent progress in the PROTAC strategy include identification of the structure of the first ternary eutectic complex, extra-terminal domain-4-PROTAC-Von-Hippel-Lindau (BRD4-PROTAC-VHL), and PROTAC ARV-110 has entered clinical trials for the treatment of prostate cancer in 2019. These discoveries strongly proved the value of the PROTAC strategy. In this perspective, we summarized recent meaningful research of PROTAC, including the types of degradation proteins, preliminary biological data in vitro and in vivo, and new E3 ubiquitin ligases. Importantly, the molecular design, optimization strategy and clinical application of candidate molecules are highlighted in detail. Future perspectives for development of advanced PROTAC in medical fields have also been discussed systematically.
Collapse
Affiliation(s)
- Yang Wang
- Department of Pharmaceutical Analysis, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Xueyang Jiang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Feng Feng
- Jiangsu Food and Pharmaceutical Science College, Huaian 223003, China
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Haopeng Sun
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
13
|
Abdel-Rashid RS, Omar SM, Teiama MS, Khairy A, Magdy M, Anis B. Fabrication Of Gold Nanoparticles In Absence Of Surfactant As In Vitro Carrier Of Plasmid DNA. Int J Nanomedicine 2019; 14:8399-8408. [PMID: 31695373 PMCID: PMC6815217 DOI: 10.2147/ijn.s226498] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/09/2019] [Indexed: 01/17/2023] Open
Abstract
PURPOSE This work aimed to synthesize surfactant-free AuNPs for targeted delivery of plasmid DNA encoded p53 gene and to avoid conventional production method of Gold nanoparticles (AuNPs) which may adversely affect the final shape, diversity, and size due to accumulation of the formulated surfactant - gold complex to the surface. METHODS The AuNPs were fabricated using seeded-growth method with L-Cystine methyl ester hydrochloride as capping agent, then loaded with plasmid DNA encoded p53 gene. The resultant AuNPs and AuNPs-p53 complex were evaluated for physical characteristics and morphology. Confirmation of complex formation was performed using gel electrophoresis. Furthermore, the efficient delivery and cytotoxicity behavior of the encoded gene were examined on both healthy lung cells (WI38) and cancerous lung cells (A549). RESULTS L-cysteine methyl ester hydrochloride AuNPs showed acceptable physical characteristics (30 nm, +36.9 mv, and spherical morphology). P53 attachment to AuNPs was confirmed by gel electrophoresis. The RT-PCR proved the overexpression of p53 by the fabricated AuNPs-p53 complex. The high percentage of cell viability in normal lung cell line (WI 38) proved the safety of L-cysteine methyl ester functionalized AuNPs. Additionally, the apoptotic effect due to expression of p53 gene loaded on AuNPs was only prominent in lung cancer cell line (A549), revealing selectivity and targeting efficiency of anticancer AuNPs-p53 complex. CONCLUSION AuNPs can be considered as a potential delivery system for effective transfection of plasmid DNA which can be used for successful treatment of cancer.
Collapse
Affiliation(s)
- Rania S Abdel-Rashid
- Department of Pharmaceutics, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Samia M Omar
- Department of Pharmaceutics, Faculty of Pharmacy, Helwan University, Cairo, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Mohammed S Teiama
- Department of Pharmaceutics, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Ahmed Khairy
- Department of Pharmaceutics, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt
| | - Mohamed Magdy
- Ministry of Interior, Administration of Criminal Evidence, Cairo, Egypt
| | - Badawi Anis
- Spectroscopy Department, Physics Division, National Research Centre, Giza, Egypt
| |
Collapse
|
14
|
Evaluating dose-limiting toxicities of MDM2 inhibitors in patients with solid organ and hematologic malignancies: A systematic review of the literature. Leuk Res 2019; 86:106222. [PMID: 31522038 DOI: 10.1016/j.leukres.2019.106222] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/30/2019] [Accepted: 09/05/2019] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Mouse double minute 2 protein (MDM2), a negative regulator of the p53 tumour suppressor gene, is frequently amplified in malignancies. MDM2 antagonists have shown efficacy in treating malignancies with MDM2 overexpression and can overcome chemoresistance in acute myeloid leukemia. We systematically evaluated the safety profile of MDM2 inhibitors in the treatment of solid organ and hematologic malignancies. MATERIALS AND METHODS We searched Medline and EMBASE from January 1947 to November 2018 for prospective clinical studies, in English or French, investigating any MDM2 inhibitor in pediatric or adult cancers, and reporting dose and toxicity outcomes. Primary outcome was dose-limiting toxicity (DLT) and secondary outcome was death. RESULTS The search yielded 493 non-duplicate citations. Eighteen studies of 10 inhibitors met inclusion criteria (total N = 1005 patients). Two-thirds of included studies did not define DLTs and the reporting of toxicities was highly variable. The most commonly reported DLTs were cytopenias, gastrointestinal toxicity, metabolic disturbances, fatigue and cardiovascular toxicity; there was one death attributed to treatment toxicity. CONCLUSION MDM2 antagonists have been studied in a variety of malignancies with toxicities similar to other commonly used chemotherapy agents and may represent a safe adjuvant treatment for further study in in acute leukemia.
Collapse
|
15
|
Wang Y, Ding Q, Lu YC, Cao SY, Liu QX, Zhang L. Interferon-stimulated gene 15 enters posttranslational modifications of p53. J Cell Physiol 2018; 234:5507-5518. [PMID: 30317575 DOI: 10.1002/jcp.27347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/17/2018] [Indexed: 12/27/2022]
Abstract
The tumor suppressor protein p53 is a central governor of various cellular signals. It is well accepted that ubiquitination as well as ubiquitin-like (UBL) modifications of p53 protein is critical in the control of its activity. Interferon-stimulated gene 15 (ISG15) is a well-known UBL protein with pleiotropic functions, serving both as a free intracellular molecule and as a modifier by conjugating to target proteins. Initially, attentions have historically focused on the antiviral effects of ISG15 pathway. Remarkably, a significant role in the processes of autophagy, DNA repair, and protein translation provided considerable insight into the new functions of ISG15 pathway. Despite the deterministic revelation of the relation between ISG15 and p53, the functional consequence of p53 ISGylation appears somewhat confused. More important, more recent studies have hinted p53 ubiquitination or other UBL modifications that might interconnect with its ISGylation. Here, we aim to summarize the current knowledge of p53 ISGylation and the differences in other significant modifications, which would be beneficial for the development of p53-based cancer therapy.
Collapse
Affiliation(s)
- Yang Wang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Disease, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Qi Ding
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Disease, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Yu-Chen Lu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Disease, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Shi-Yang Cao
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Disease, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Qing-Xue Liu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Disease, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Lei Zhang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Disease, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| |
Collapse
|
16
|
Olotu FA, Soliman MES. Dynamic perspectives into the mechanisms of mutation-induced p53-DNA binding loss and inactivation using active perturbation theory: Structural and molecular insights toward the design of potent reactivators in cancer therapy. J Cell Biochem 2018; 120:951-966. [PMID: 30160791 DOI: 10.1002/jcb.27458] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/19/2018] [Indexed: 01/08/2023]
Abstract
The DNA-binding ability of p53 represents the crux of its tumor suppressive activities, which involves transcriptional activation of target genes responsible for apoptosis and cell-cycle arrest. Mutational occurrences within or in close proximity to the DNA-binding surface of p53 have accounted for the loss of direct DNA-binding ability and inactivation implicated in many cases of cancer. Moreover, the design of therapeutic compounds that can restore DNA-binding ability in p53 mutants has been identified as a way forward in curtailing their oncogenic activities. However, there is still the need for more insights into evaluate the perturbations that occur at the DNA-binding interface of mp53 relative to DNA-binding loss, inactivation, and design of potent reactivators, hence the purpose of this study. Therefore, we evaluated p53-structural (R175H) and contact (R273C) mutational effects using tunnel perturbation analysis and other computational tools. We identified significant perturbations in the active tunnels of p53, which resulted in altered geometry and loss, unlike in the wild-type p53. This corroborated with structural, DNA-binding, and interaction network analysis, which showed that loss of flexibility, repulsion of DNA-interactive residues, and instability occurred at the binding interface of both mutants. Also, these mutations altered bonding interactions and network topology at the DNA-binding interface, resulting in the reduction of p53-DNA binding proximity and affinity. Therefore, these findings would aid the structure-based design of novel chemical entities capable of restoring p53-DNA binding and activation.
Collapse
Affiliation(s)
- Fisayo A Olotu
- Molecular Bio-Computation and Drug Design Laboratory, Department of Pharmaceutical Chemistry, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - Mahmoud E S Soliman
- Molecular Bio-Computation and Drug Design Laboratory, Department of Pharmaceutical Chemistry, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| |
Collapse
|
17
|
Alaee M, Padda A, Mehrabani V, Churchill L, Pasdar M. The physical interaction of p53 and plakoglobin is necessary for their synergistic inhibition of migration and invasion. Oncotarget 2018; 7:26898-915. [PMID: 27058623 PMCID: PMC5042024 DOI: 10.18632/oncotarget.8616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/14/2016] [Indexed: 01/15/2023] Open
Abstract
Plakoglobin (PG) is a paralog of β-catenin with similar adhesive, but contrasting signalling functions. Although β-catenin has well-known oncogenic function, PG generally acts as a tumor/metastasis suppressor by mechanisms that are just beginning to be deciphered. Previously, we showed that PG interacted with wild type (WT) and a number of mutant p53s, and that its tumor/metastasis suppressor activity may be mediated, at least partially, by this interaction. Here, carcinoma cell lines deficient in both p53 and PG (H1299), or expressing mutant p53 in the absence of PG (SCC9), were transfected with expression constructs encoding WT and different fragments and deletions of p53 and PG, individually or in pairs. Transfectants were characterized for their in vitro growth, migratory and invasive properties and for mapping the interacting domain of p53 and PG. We showed that when coexpressed, p53-WT and PG-WT cooperated to decrease growth, and acted synergistically to significantly reduce cell migration and invasion. The DNA-binding domain of p53 and C-terminal domain of PG mediated p53/PG interaction, and furthermore, the C-terminus of PG played a central role in the inhibition of invasion in association with p53.
Collapse
Affiliation(s)
- Mahsa Alaee
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| | - Amarjot Padda
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| | - Vahedah Mehrabani
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| | - Lucas Churchill
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| | - Manijeh Pasdar
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| |
Collapse
|
18
|
Chen S, Du Z, Wu B, Shen H, Liu C, Qiu X, Zhang Y, Xu L, Li E, Zhong Z. STAT1, IGF1, RAC1, and MDM2 Are Associated with Recurrence of Giant Cell Tumor of Bone. J Immunol Res 2018; 2018:4564328. [PMID: 29651441 PMCID: PMC5831922 DOI: 10.1155/2018/4564328] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/08/2017] [Accepted: 11/28/2017] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND In our previous study, mouse double minute 2 homolog (MDM2), insulin-like growth factor 1 (IGF1), signal transducer and activator of transcription 1 (STAT1), and Rac family small GTPase 1 (RAC1) were correlated with the recurrence of giant cell tumor of bone (GCT). The aim of this study is to use a large cohort study to confirm the involvement of these four genes in GCT recurrence. METHODS The expression of these four genes was detected and compared between GCT patients with or without recurrence. The correlation between the expression of these four genes and clinical characteristics was evaluated. Protein-protein interaction (PPI) network was constructed for functional enrichment analysis. RESULTS It showed that the expression levels of MDM2, IGF1, STAT1, and RAC1 in GCT patients with recurrence were significantly higher than those in GCT patients without recurrence (P < 0.05). Multivariate logistic regression analysis suggested that several clinical characteristics may influence prognosis. A PPI network was constructed using the four genes as hub genes. Functional enrichment analysis showed that this network involves many important biological progress mediated by these four genes, including immune response. CONCLUSION MDM2, IGF1, STAT1, and RAC1 are associated with GCT recurrence, which might serve as biomarkers for GCT recurrence.
Collapse
Affiliation(s)
- Shuxin Chen
- Department of Orthopedic Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, China
| | - Zepeng Du
- Department of Pathology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, China
| | - Bingli Wu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Huiyang Shen
- Department of Orthopedic Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, China
| | - Chunpeng Liu
- Department of Orthopedic Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, China
| | - Xueli Qiu
- Department of Orthopedic Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, China
| | - Yufeng Zhang
- Department of Orthopedic Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, China
| | - Liyan Xu
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Enmin Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Zhigang Zhong
- Department of Orthopedic Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, China
| |
Collapse
|
19
|
Parrales A, Thoenen E, Iwakuma T. The interplay between mutant p53 and the mevalonate pathway. Cell Death Differ 2017; 25:460-470. [PMID: 29238070 DOI: 10.1038/s41418-017-0026-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 10/02/2017] [Accepted: 10/25/2017] [Indexed: 02/08/2023] Open
Abstract
Missense mutations in the TP53 gene lead to accumulation of dysfunctional TP53 proteins in tumors, showing oncogenic gain-of-function (GOF) activities. Stabilization of mutant TP53 (mutp53) is required for the GOF; however, the mechanisms by which mutp53 promotes cancer progression and how mutp53 stability is regulated are not completely understood. Recent work from our laboratory has identified statins, inhibitors of the mevalonate pathway, as degraders of conformational mutp53. Specific reduction of mevalonate-5-phosphate (MVP), a metabolic intermediate in the mevalonate pathway, by statins or mevalonate kinase (MVK) knockdown triggers CHIP ubiquitin ligase-mediated degradation of conformational mutp53 by inhibiting interaction between mutp53 and DNAJA1, a Hsp40 family member. Thus, the mevalonate pathway contributes to mutp53 stabilization. Given that mutp53 is shown to promote cancer progression by upregulating mRNA expression of mevalonate pathway enzymes by binding to the sterol regulatory element-binding protein 2 (SREBP2) and subsequently increasing activities of mevalonate pathway-associated oncogenic proteins (e.g., Ras, Rho, YAP/TAZ), there is a positive-feedback loop between mutp53 and the mevalonate pathway. Here, we summarize recent evidence linking the mevalonate pathway-mutp53 axis with cancer progression and further discuss the clinical relevance of this axis.
Collapse
Affiliation(s)
- Alejandro Parrales
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Elizabeth Thoenen
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Tomoo Iwakuma
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
20
|
Meitinger F, Anzola JV, Kaulich M, Richardson A, Stender JD, Benner C, Glass CK, Dowdy SF, Desai A, Shiau AK, Oegema K. 53BP1 and USP28 mediate p53 activation and G1 arrest after centrosome loss or extended mitotic duration. J Cell Biol 2017; 214:155-66. [PMID: 27432897 PMCID: PMC4949453 DOI: 10.1083/jcb.201604081] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/24/2016] [Indexed: 12/14/2022] Open
Abstract
In normal human cells, centrosome loss induced by centrinone-a specific centrosome duplication inhibitor-leads to irreversible, p53-dependent G1 arrest by an unknown mechanism. A genome-wide CRISPR/Cas9 screen for centrinone resistance identified genes encoding the p53-binding protein 53BP1, the deubiquitinase USP28, and the ubiquitin ligase TRIM37. Deletion of TP53BP1, USP28, or TRIM37 prevented p53 elevation in response to centrosome loss but did not affect cytokinesis failure-induced arrest or p53 elevation after doxorubicin-induced DNA damage. Deletion of TP53BP1 and USP28, but not TRIM37, prevented growth arrest in response to prolonged mitotic duration. TRIM37 knockout cells formed ectopic centrosomal-component foci that suppressed mitotic defects associated with centrosome loss. TP53BP1 and USP28 knockouts exhibited compromised proliferation after centrosome removal, suggesting that centrosome-independent proliferation is not conferred solely by the inability to sense centrosome loss. Thus, analysis of centrinone resistance identified a 53BP1-USP28 module as critical for communicating mitotic challenges to the p53 circuit and TRIM37 as an enforcer of the singularity of centrosome assembly.
Collapse
Affiliation(s)
- Franz Meitinger
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093 Ludwig Institute for Cancer Research, La Jolla, CA 92093
| | - John V Anzola
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, La Jolla, CA 92093
| | - Manuel Kaulich
- Institute of Biochemistry II, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Amelia Richardson
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093 Ludwig Institute for Cancer Research, La Jolla, CA 92093
| | - Joshua D Stender
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Christopher Benner
- Department of Medicine, University of California, San Diego, La Jolla, California 92093
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093 Department of Medicine, University of California, San Diego, La Jolla, California 92093
| | - Steven F Dowdy
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Arshad Desai
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093 Ludwig Institute for Cancer Research, La Jolla, CA 92093
| | - Andrew K Shiau
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, La Jolla, CA 92093
| | - Karen Oegema
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093 Ludwig Institute for Cancer Research, La Jolla, CA 92093
| |
Collapse
|
21
|
Kendig RD, Kai F, Fry EA, Inoue K. Stabilization of the p53-DNA Complex by the Nuclear Protein Dmp1α. Cancer Invest 2017; 35:301-312. [PMID: 28406729 PMCID: PMC6262109 DOI: 10.1080/07357907.2017.1303505] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/14/2016] [Accepted: 03/03/2017] [Indexed: 01/19/2023]
Abstract
We recently reported the existence of a physical interaction between the Myb-like transcription factor Dmp1 (Dmtf1) and p53 in which Dmp1 antagonized polyubiquitination of p53 by Mdm2 and promoted its nuclear localization. Dmp1 significantly stabilized p53-DNA complexes on promoters that contained p53-consensus sequences, which were either supershifted or disrupted with antibodies to Dmp1. Lysates from mice injected with doxorubicin showed that Dmp1 bound to p21Cip1, Bbc3, and Thbs1 gene regulatory regions in a p53-dependent fashion. Our data suggest that acceleration of DNA-binding of p53 by Dmp1 is a critical process for Dmp1 to increase the p53 function in Arf-deficient cells.
Collapse
Affiliation(s)
- Robert D Kendig
- a Department of Pathology , Wake Forest University School of Medicine , Winston-Salem , North Carolina , USA
| | - Fumitake Kai
- a Department of Pathology , Wake Forest University School of Medicine , Winston-Salem , North Carolina , USA
| | - Elizabeth A Fry
- a Department of Pathology , Wake Forest University School of Medicine , Winston-Salem , North Carolina , USA
| | - Kazushi Inoue
- a Department of Pathology , Wake Forest University School of Medicine , Winston-Salem , North Carolina , USA
| |
Collapse
|
22
|
Fan C, Wang X. Mdm2 Splice isoforms regulate the p53/Mdm2/Mdm4 regulatory circuit via RING domain-mediated ubiquitination of p53 and Mdm4. Cell Cycle 2017; 16:660-664. [PMID: 28166445 DOI: 10.1080/15384101.2017.1288327] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
p53 is regulated by heterodimer E3 ligase Mdm2-Mdm4 via RING domain interaction. Mdm2 transcripts undergo alternative splicing, and Mdm2 splice isoforms are increased in cancer and induced by DNA damage. Although 2 major Mdm2 splice isoforms that do not bind to p53 were reported to impact the p53 pathway, the underlying biochemical mechanisms were not understood. Here, we show that these Mdm2 splice isoforms ubiquitinate Mdm2 and Mdm4 in vivo and regulate the activity of Mdm2-Mdm4 E3 complex in cells. The Mdm2 isoforms are capable of promoting p53 ubiquitination in the absence of Mdm2 or Mdm4. The 2 isoforms stimulate Mdm2 or Mdm4 activity for p53 ubiquitination in vivo and promote degradation of p53 and Mdm4 in cells. However, the Mdm2 isoforms have opposing effects on the steady-state p53 levels depending on the stoichiometric ratios of Mdm2, Mdm4 and the isoforms, causing either decreased or increased p53 levels in cells. Our data indicate that the Mdm2 splice isoforms can act as independent E3 ligases for p53 when Mdm2 and Mdm4 are absent, form potent heterodimer E3 ligases with either Mdm2 or Mdm4 for targeting p53 degradation, or act as inhibitory regulators of Mdm2-Mdm4 E3 ligase activity by downregulating Mdm4. These findings suggest that Mdm2 splice isoforms may play critical roles in the regulatory loop of p53/Mdm2-Mdm4 via a RING domain-mediated biochemical mechanism.
Collapse
Affiliation(s)
- Chuandong Fan
- a Department of Pharmacology and Therapeutics , Roswell Park Cancer Institute , Buffalo , NY , USA
| | - Xinjiang Wang
- a Department of Pharmacology and Therapeutics , Roswell Park Cancer Institute , Buffalo , NY , USA
| |
Collapse
|
23
|
Bonham-Carter O, Thapa I, From S, Bastola D. A study of bias and increasing organismal complexity from their post-translational modifications and reaction site interplays. Brief Bioinform 2017; 18:69-84. [PMID: 26764274 PMCID: PMC5221421 DOI: 10.1093/bib/bbv111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 12/08/2015] [Indexed: 01/21/2023] Open
Abstract
Post-translational modifications (PTMs) are important steps in the biosynthesis of proteins. Aside from their integral contributions to protein development, i.e. perform specialized proteolytic cleavage of regulatory subunits, the covalent addition of functional groups of proteins or the degradation of entire proteins, PTMs are also involved in enabling proteins to withstand and recover from temporary environmental stresses (heat shock, microgravity and many others). The literature supports evidence of thousands of recently discovered PTMs, many of which may likely contribute similarly (perhaps, even, interchangeably) to protein stress response. Although there are many PTM actors on the biological stage, our study determines that these PTMs are generally cast into organism-specific, preferential roles. In this work, we study the PTM compositions across the mitochondrial (Mt) and non-Mt proteomes of 11 diverse organisms to illustrate that each organism appears to have a unique list of PTMs, and an equally unique list of PTM-associated residue reaction sites (RSs), where PTMs interact with protein. Despite the present limitation of available PTM data across different species, we apply existing and current protein data to illustrate particular organismal biases. We explore the relative frequencies of observed PTMs, the RSs and general amino-acid compositions of Mt and non-Mt proteomes. We apply these data to create networks and heatmaps to illustrate the evidence of bias. We show that the number of PTMs and RSs appears to grow along with organismal complexity, which may imply that environmental stress could play a role in this bias.
Collapse
|
24
|
Parrales A, Ranjan A, Iyer SV, Padhye S, Weir SJ, Roy A, Iwakuma T. DNAJA1 controls the fate of misfolded mutant p53 through the mevalonate pathway. Nat Cell Biol 2016; 18:1233-1243. [PMID: 27775703 DOI: 10.1038/ncb3427] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 09/21/2016] [Indexed: 12/20/2022]
Abstract
Stabilization of mutant p53 (mutp53) in tumours greatly contributes to malignant progression. However, little is known about the underlying mechanisms and therapeutic approaches to destabilize mutp53. Here, through high-throughput screening we identify statins, cholesterol-lowering drugs, as degradation inducers for conformational or misfolded p53 mutants with minimal effects on wild-type p53 (wtp53) and DNA contact mutants. Statins preferentially suppress mutp53-expressing cancer cell growth. Specific reduction of mevalonate-5-phosphate by statins or mevalonate kinase knockdown induces CHIP ubiquitin ligase-mediated nuclear export, ubiquitylation, and degradation of mutp53 by impairing interaction of mutp53 with DNAJA1, a Hsp40 family member. Knockdown of DNAJA1 also induces CHIP-mediated mutp53 degradation, while its overexpression antagonizes statin-induced mutp53 degradation. Our study reveals that DNAJA1 controls the fate of misfolded mutp53, provides insights into potential strategies to deplete mutp53 through the mevalonate pathway-DNAJA1 axis, and highlights the significance of p53 status in impacting statins' efficacy on cancer therapy.
Collapse
Affiliation(s)
- Alejandro Parrales
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Atul Ranjan
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Swathi V Iyer
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Subhash Padhye
- Department of Chemistry, Abeda Inamdar Senior College, Pune, Maharashtra 411001, India
| | - Scott J Weir
- Department of Pharmacology, Toxicology and Therapeutics, Institute for Advancing Medical Innovation, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Anuradha Roy
- High Throughput Screening Laboratory, University of Kansas, Lawrence, Kansas 66047, USA
| | - Tomoo Iwakuma
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| |
Collapse
|
25
|
NORE1A Regulates MDM2 Via β-TrCP. Cancers (Basel) 2016; 8:cancers8040039. [PMID: 27023610 PMCID: PMC4846848 DOI: 10.3390/cancers8040039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/09/2016] [Accepted: 03/14/2016] [Indexed: 12/11/2022] Open
Abstract
Mouse Double Minute 2 Homolog (MDM2) is a key negative regulator of the master tumor suppressor p53. MDM2 regulates p53 on multiple levels, including acting as an ubiquitin ligase for the protein, thereby promoting its degradation by the proteasome. MDM2 is oncogenic and is frequently found to be over-expressed in human tumors, suggesting its dysregulation plays an important role in human cancers. We have recently found that the Ras effector and RASSF (Ras Association Domain Family) family member RASSF5/NORE1A enhances the levels of nuclear p53. We have also found that NORE1A (Novel Ras Effector 1A) binds the substrate recognition component of the SCF-ubiquitin ligase complex β-TrCP. Here, we now show that NORE1A regulates MDM2 protein levels by targeting it for ubiquitination by SCF-β-TrCP. We also show the suppression of NORE1A protein levels enhances MDM2 protein expression. Finally, we show that MDM2 can suppress the potent senescence phenotype induced by NORE1A over-expression. Thus, we identify a mechanism by which Ras/NORE1A can modulate p53 protein levels. As MDM2 has several important targets in addition to p53, this finding has broad implications for cancer biology in tumor cells that have lost expression of NORE1A due to promoter methylation.
Collapse
|
26
|
Chaffee BR, Hoang TV, Leonard MR, Bruney DG, Wagner BD, Dowd JR, Leone G, Ostrowski MC, Robinson ML. FGFR and PTEN signaling interact during lens development to regulate cell survival. Dev Biol 2016; 410:150-163. [PMID: 26764128 DOI: 10.1016/j.ydbio.2015.12.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 12/31/2015] [Accepted: 12/31/2015] [Indexed: 12/13/2022]
Abstract
Lens epithelial cells express many receptor tyrosine kinases (RTKs) that stimulate PI3K-AKT and RAS-RAF-MEK-ERK intracellular signaling pathways. These pathways ultimately activate the phosphorylation of key cellular transcription factors and other proteins that control proliferation, survival, metabolism, and differentiation in virtually all cells. Among RTKs in the lens, only stimulation of fibroblast growth factor receptors (FGFRs) elicits a lens epithelial cell to fiber cell differentiation response in mammals. Moreover, although the lens expresses three different Fgfr genes, the isolated removal of Fgfr2 at the lens placode stage inhibits both lens cell survival and fiber cell differentiation. Phosphatase and tensin homolog (PTEN), commonly known as a tumor suppressor, inhibits ERK and AKT activation and initiates both apoptotic pathways, and cell cycle arrest. Here, we show that the combined deletion of Fgfr2 and Pten rescues the cell death phenotype associated with Fgfr2 loss alone. Additionally, Pten removal increased AKT and ERK activation, above the levels of controls, in the presence or absence of Fgfr2. However, isolated deletion of Pten failed to stimulate ectopic fiber cell differentiation, and the combined deletion of Pten and Fgfr2 failed to restore differentiation-specific Aquaporin0 and DnaseIIβ expression in the lens fiber cells.
Collapse
Affiliation(s)
- Blake R Chaffee
- Department of Biology, Cell Molecular and Structural Biology Graduate Program, Miami University, Oxford, OH, USA
| | - Thanh V Hoang
- Department of Biology, Cell Molecular and Structural Biology Graduate Program, Miami University, Oxford, OH, USA
| | - Melissa R Leonard
- Department of Biology, Cell Molecular and Structural Biology Graduate Program, Miami University, Oxford, OH, USA
| | - Devin G Bruney
- Department of Biology, Cell Molecular and Structural Biology Graduate Program, Miami University, Oxford, OH, USA
| | - Brad D Wagner
- Department of Biology, Cell Molecular and Structural Biology Graduate Program, Miami University, Oxford, OH, USA
| | - Joseph Richard Dowd
- Department of Biology, Cell Molecular and Structural Biology Graduate Program, Miami University, Oxford, OH, USA
| | - Gustavo Leone
- Department of Molecular Virology, Immunology and Medical Genetics, Department of Molecular Genetics, The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Michael C Ostrowski
- Department of Molecular Virology, Immunology and Medical Genetics, Department of Molecular Genetics, The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Michael L Robinson
- Department of Biology, Cell Molecular and Structural Biology Graduate Program, Miami University, Oxford, OH, USA.
| |
Collapse
|
27
|
Abstract
The role of p73, the homologue of the tumor suppressor p53, in regulating angiogenesis has recently been extensively investigated, resulting in the publication of five articles. Of these, two studies suggested a suppressive role, while the others implied a stimulatory role for the p73 isoforms in regulating angiogenesis. A negative role for TAp73, the full-length form that is often associated with tumor suppression, in blood vessel formation, is consistent with its general attributes and was proposed to be effected indirectly through the degradation of hypoxia-inducible factor 1α (HIF1-α), the master angiogenic regulator. In contrast, a positive role for TAp73 coincides with its recently understood role in supporting cellular survival and thus tumorigenesis, consistent with TAp73 being not-mutated but rather often overexpressed in clinical contexts. In the latter case, TAp73 expression was induced by hypoxia via HIF1-α, and it appears to directly promote angiogenic target gene activation and blood vessel formation independent of HIF1-α. This mini review will provide an overview of these seemingly opposite recent findings as well as earlier data, which collectively establish the definite possibility that TAp73 is indeed capable of both promoting and inhibiting angiogenesis, depending on the cellular context.
Collapse
|
28
|
Parrales A, Iwakuma T. Targeting Oncogenic Mutant p53 for Cancer Therapy. Front Oncol 2015; 5:288. [PMID: 26732534 PMCID: PMC4685147 DOI: 10.3389/fonc.2015.00288] [Citation(s) in RCA: 231] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/07/2015] [Indexed: 12/14/2022] Open
Abstract
Among genetic alterations in human cancers, mutations in the tumor suppressor p53 gene are the most common, occurring in over 50% of human cancers. The majority of p53 mutations are missense mutations and result in the accumulation of dysfunctional p53 protein in tumors. These mutants frequently have oncogenic gain-of-function activities and exacerbate malignant properties of cancer cells, such as metastasis and drug resistance. Increasing evidence reveals that stabilization of mutant p53 in tumors is crucial for its oncogenic activities, while depletion of mutant p53 attenuates malignant properties of cancer cells. Thus, mutant p53 is an attractive druggable target for cancer therapy. Different approaches have been taken to develop small-molecule compounds that specifically target mutant p53. These include compounds that restore wild-type conformation and transcriptional activity of mutant p53, induce depletion of mutant p53, inhibit downstream pathways of oncogenic mutant p53, and induce synthetic lethality to mutant p53. In this review article, we comprehensively discuss the current strategies targeting oncogenic mutant p53 in cancers, with special focus on compounds that restore wild-type p53 transcriptional activity of mutant p53 and those reducing mutant p53 levels.
Collapse
Affiliation(s)
- Alejandro Parrales
- Department of Cancer Biology, University of Kansas Medical Center , Kansas City, KS , USA
| | - Tomoo Iwakuma
- Department of Cancer Biology, University of Kansas Medical Center , Kansas City, KS , USA
| |
Collapse
|
29
|
Ho YH, Gasch AP. Exploiting the yeast stress-activated signaling network to inform on stress biology and disease signaling. Curr Genet 2015; 61:503-11. [PMID: 25957506 DOI: 10.1007/s00294-015-0491-0] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 04/23/2015] [Accepted: 04/24/2015] [Indexed: 12/24/2022]
Abstract
Healthy cells utilize intricate systems to monitor their environment and mount robust responses in the event of cellular stress. Whether stress arises from external insults or defects due to mutation and disease, cells must be able to respond precisely to mount the appropriate defenses. Multi-faceted stress responses are generally coupled with arrest of growth and cell-cycle progression, which both limits the transmission of damaged materials and serves to reallocate limited cellular resources toward defense. Therefore, stress defense versus rapid growth represent competing interests in the cell. How eukaryotic cells set the balance between defense versus proliferation, and in particular knowledge of the regulatory networks that control this decision, are poorly understood. In this perspective, we expand upon our recent work inferring the stress-activated signaling network in budding yeast, which captures pathways controlling stress defense and regulators of growth and cell-cycle progression. We highlight similarities between the yeast and mammalian stress responses and explore how stress-activated signaling networks in yeast can inform on signaling defects in human cancers.
Collapse
Affiliation(s)
- Yi-Hsuan Ho
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Audrey P Gasch
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| |
Collapse
|
30
|
Park HJ, Kim MM. Flavonoids in Ginkgo biloba fallen leaves induce apoptosis through modulation of p53 activation in melanoma cells. Oncol Rep 2014; 33:433-8. [PMID: 25394497 DOI: 10.3892/or.2014.3602] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 10/21/2014] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to examine the apoptotic effect of flavonoids in methanol extracts of Ginkgo biloba fallen leaves (MEGFL) on melanoma cells. Ginkgo biloba is a deciduous castle chaplain and its leaves include various types of flavonoids such as flavonol-O-glycosides. Ginkgo biloba is known to have therapeutic properties against a number of diseases such as cerebrovascular diseases, blood circulation disease and hypertension. In the present study MEGFL exhibited a higher cytotoxic effect on melanoma cells than Ginkgo biloba leaves (MEGL). It was also found that MEGFL induced apoptotic cell death which was characterized by DNA fragmentation. During the cell death process following treatment with MEGFL, the expression of a variety of death-associated proteins including p53, caspase-3, caspase-9, cytochrome c and Bax were analyzed in the cytosol of melanoma cells. MEGFL significantly increased the expression levels of caspase-3, caspase-9 and p53 in a dose-dependent manner. Our results indicate that MEGFL induced apoptotic cell death by increasing the expression of cell death-associated proteins in melanoma cells.
Collapse
Affiliation(s)
- Hye-Jung Park
- Department of Chemistry, Dong-Eui University, Busan 614-714, Republic of Korea
| | - Moon-Moo Kim
- Department of Chemistry, Dong-Eui University, Busan 614-714, Republic of Korea
| |
Collapse
|