1
|
Ghone D, Evans EL, Bandini M, Stephenson KG, Sherer NM, Suzuki A. HIV-1 Vif disrupts phosphatase feedback regulation at the kinetochore, leading to a pronounced pseudo-metaphase arrest. eLife 2025; 13:RP101136. [PMID: 40080415 PMCID: PMC11906157 DOI: 10.7554/elife.101136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025] Open
Abstract
Virion Infectivity Factor (Vif) of the Human Immunodeficiency Virus type 1 (HIV-1) targets and degrades cellular APOBEC3 proteins, key regulators of intrinsic and innate antiretroviral immune responses, thereby facilitating HIV-1 infection. While Vif's role in degrading APOBEC3G is well-studied, Vif is also known to cause cell cycle arrest, but the detailed nature of Vif's effects on the cell cycle has yet to be delineated. In this study, we employed high-temporal resolution single-cell live imaging and super-resolution microscopy to monitor individual cells during Vif-induced cell cycle arrest. Our findings reveal that Vif does not affect the G2/M boundary as previously thought. Instead, Vif triggers a unique and robust pseudo-metaphase arrest, distinct from the mild prometaphase arrest induced by Vpr. During this arrest, chromosomes align properly and form the metaphase plate, but later lose alignment, resulting in polar chromosomes. Notably, Vif, unlike Vpr, significantly reduces the levels of both Protein Phosphatase 1 (PP1) and 2 A (PP2A) at kinetochores, which regulate chromosome-microtubule interactions. These results unveil a novel role for Vif in kinetochore regulation that governs the spatial organization of chromosomes during mitosis.
Collapse
Affiliation(s)
- Dhaval Ghone
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-MadisonMadisonUnited States
- Biophysics Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Edward L Evans
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-MadisonMadisonUnited States
- Cancer Biology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
- Institute for Molecular Virology, University of Wisconsin-MadisonMadisonUnited States
| | - Madison Bandini
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-MadisonMadisonUnited States
- Cancer Biology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
- Institute for Molecular Virology, University of Wisconsin-MadisonMadisonUnited States
| | - Kaelyn G Stephenson
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-MadisonMadisonUnited States
| | - Nathan M Sherer
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-MadisonMadisonUnited States
- Institute for Molecular Virology, University of Wisconsin-MadisonMadisonUnited States
- Carbone Comprehensive Cancer Center, University of Wisconsin-MadisonMadisonUnited States
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-MadisonMadisonUnited States
- Carbone Comprehensive Cancer Center, University of Wisconsin-MadisonMadisonUnited States
| |
Collapse
|
2
|
Sang Y, Du J, Zulikala D, Sang Z. Mechanistic analysis of Tanshinone IIA's regulation of the ATM/GADD45/ORC signaling pathway to reduce myocardial ischemia-reperfusion injury. Front Pharmacol 2024; 15:1510380. [PMID: 39776578 PMCID: PMC11703710 DOI: 10.3389/fphar.2024.1510380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Background By far, one of the best treatments for myocardial ischemia is reperfusion therapy. The primary liposoluble component of Danshen, a traditional Chinese herbal remedy, Tanshinone ⅡA, has been shown to have cardiac healing properties. The purpose of this work is to investigate the processes by which Tanshinone ⅡA influences myocardial ischemia-reperfusion injury (MIRI) in the H9C2 cardiac myoblast cell line, as well as the association between Tanshinone ⅡA and MIRI. Methods and results The cardiac cells were divided into a normal group, a model group and Tanshinone ⅡA treatment groups. After 4 h of culture with the deprivation of oxygen and glucose, the cells were incubated normally for 2 h. The success of the model and the capacity of Tanshinone ⅡA to heal cardiac damage were validated by the outcomes of cell viability, morphology, and proliferation. The efficacy of Tanshinone ⅡA in treating MIRI was further confirmed by the scratch assay and biomarker measurement. The differentially expressed genes were examined using transcriptome sequencing. The Ataxia-Telangiectasia Mutated (ATM)/Growth Arrest and DNA Damage (GADD45)/Origin Recognition Complex (ORC) signaling pathway was identified as being crucial to this process by KEGG pathway analysis and GO enrichment. Molecular docking and RT-qPCR were used to confirm our results. The crucial function of the ATM/GADD45/ORC pathway was further confirmed by the addition of an ATM inhibitor, which inhibited the expression of ATM. Conclusion Tanshinone ⅡA can relieve the myocardial ischemia-reperfusion injury in cardiac cells by activating the ATM/GADD45/ORC pathway.
Collapse
Affiliation(s)
- Yiwei Sang
- Nature Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast, United Kingdom
| | - Jiangnan Du
- Nature Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast, United Kingdom
| | - Dilimulati Zulikala
- Nature Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast, United Kingdom
| | - Zhongqiang Sang
- Dermatology Department, Shanghai Zhongye Hospital, Shanghai, China
| |
Collapse
|
3
|
Ghone D, Evans EL, Bandini M, Stephenson KG, Sherer NM, Suzuki A. HIV-1 Vif disrupts phosphatase feedback regulation at the kinetochore, leading to a pronounced pseudo-metaphase arrest. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605839. [PMID: 39131328 PMCID: PMC11312601 DOI: 10.1101/2024.07.30.605839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Virion Infectivity Factor (Vif) of the Human Immunodeficiency Virus type 1 (HIV-1) targets and degrades cellular APOBEC3 proteins, key regulators of intrinsic and innate antiretroviral immune responses, thereby facilitating HIV-1 infection. While Vif's role in degrading APOBEC3G is well-studied, Vif is also known to cause cell cycle arrest, but the detailed nature of Vif's effects on the cell cycle has yet to be delineated. In this study, we employed high-temporal single-cell live imaging and super-resolution microscopy to monitor individual cells during Vif-induced cell cycle arrest. Our findings reveal that Vif does not affect the G2/M boundary as previously thought. Instead, Vif triggers a unique and robust pseudo-metaphase arrest, distinct from the mild prometaphase arrest induced by Vpr. During this arrest, chromosomes align properly and form the metaphase plate, but later lose alignment, resulting in polar chromosomes. Notably, Vif, unlike Vpr, significantly reduces the levels of both Protein Phosphatase 1 (PP1) and 2A (PP2A) at kinetochores, which regulate chromosome-microtubule interactions. These results unveil a novel role for Vif in kinetochore regulation that governs the spatial organization of chromosomes during mitosis.
Collapse
Affiliation(s)
- Dhaval Ghone
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Biophysics Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- These authors contributed equally
| | - Edward L. Evans
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Cancer Biology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Institute for Molecular Virology, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- These authors contributed equally
- Present address: Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Madison Bandini
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Cancer Biology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Institute for Molecular Virology, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Kaelyn G. Stephenson
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Nathan M. Sherer
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Institute for Molecular Virology, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Lead contact
| |
Collapse
|
4
|
Djordjevic S, Itzykson R, Hague F, Lebon D, Legrand J, Ouled‐Haddou H, Jedraszak G, Harbonnier J, Collet L, Paubelle E, Marolleau J, Garçon L, Boyer T. STIM2 is involved in the regulation of apoptosis and the cell cycle in normal and malignant monocytic cells. Mol Oncol 2024; 18:1571-1592. [PMID: 38234211 PMCID: PMC11161727 DOI: 10.1002/1878-0261.13584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/28/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024] Open
Abstract
Calcium is a ubiquitous messenger that regulates a wide range of cellular functions, but its involvement in the pathophysiology of acute myeloid leukemia (AML) is not widely investigated. Here, we identified, from an analysis of The Cancer Genome Atlas and genotype-tissue expression databases, stromal interaction molecule 2 (STIM2) as being highly expressed in AML with monocytic differentiation and negatively correlated with overall survival. This was confirmed on a validation cohort of 407 AML patients. We then investigated the role of STIM2 in cell proliferation, differentiation, and survival in two leukemic cell lines with monocytic potential and in normal hematopoietic stem cells. STIM2 expression increased at the RNA and protein levels upon monocyte differentiation. Phenotypically, STIM2 knockdown drastically inhibited cell proliferation and induced genomic stress with DNA double-strand breaks, as shown by increased levels of phosphorylate histone H2AXγ (p-H2AXγ), followed by activation of the cellular tumor antigen p53 pathway, decreased expression of cell cycle regulators such as cyclin-dependent kinase 1 (CDK1)-cyclin B1 and M-phase inducer phosphatase 3 (CDC25c), and a decreased apoptosis threshold with a low antiapoptotic/proapoptotic protein ratio. Our study reports STIM2 as a new actor regulating genomic stability and p53 response in terms of cell cycle and apoptosis of human normal and malignant monocytic cells.
Collapse
Affiliation(s)
| | - Raphaël Itzykson
- Département Hématologie et ImmunologieHôpital Saint‐Louis, Assistance Publique‐Hôpitaux de ParisFrance
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRSUniversité Paris CitéFrance
| | - Frédéric Hague
- Laboratoire de Physiologie Cellulaire et Moléculaire UR4667Université Picardie Jules VerneAmiensFrance
| | - Delphine Lebon
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Clinique et de Thérapie CellulaireCHU Amiens‐PicardieFrance
| | - Julien Legrand
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
| | | | - Guillaume Jedraszak
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Laboratoire de Génétique ConstitutionnelleCHU Amiens‐PicardieFrance
| | | | - Louison Collet
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
| | - Etienne Paubelle
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Clinique et de Thérapie CellulaireCHU Amiens‐PicardieFrance
| | - Jean‐Pierre Marolleau
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Clinique et de Thérapie CellulaireCHU Amiens‐PicardieFrance
| | - Loïc Garçon
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie BiologiqueCHU Amiens‐PicardieFrance
| | - Thomas Boyer
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie BiologiqueCHU Amiens‐PicardieFrance
| |
Collapse
|
5
|
Zhai M, Miao J, Zhang R, Liu R, Li F, Shen Y, Wang T, Xu X, Gao G, Hu J, He A, Bai J. KIF22 promotes multiple myeloma progression by regulating the CDC25C/CDK1/cyclinB1 pathway. J Cancer Res Clin Oncol 2024; 150:239. [PMID: 38713252 PMCID: PMC11076398 DOI: 10.1007/s00432-024-05747-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024]
Abstract
PURPOSE Multiple myeloma (MM) is an incurable hematological malignancy characterized by clonal proliferation of malignant plasma B cells in bone marrow, and its pathogenesis remains unknown. The aim of this study was to determine the role of kinesin family member 22 (KIF22) in MM and elucidate its molecular mechanism. METHODS The expression of KIF22 was detected in MM patients based upon the public datasets and clinical samples. Then, in vitro assays were performed to investigate the biological function of KIF22 in MM cell lines, and subcutaneous xenograft models in nude mice were conducted in vivo. Chromatin immunoprecipitation (ChIP) and luciferase reporter assay were used to determine the mechanism of KIF22-mediated regulation. RESULTS The results demonstrated that the expression of KIF22 in MM patients was associated with several clinical features, including gender (P = 0.016), LDH (P < 0.001), β2-MG (P = 0.003), percentage of tumor cells (BM) (P = 0.002) and poor prognosis (P < 0.0001). Furthermore, changing the expression of KIF22 mainly influenced the cell proliferation in vitro and tumor growth in vivo, and caused G2/M phase cell cycle dysfunction. Mechanically, KIF22 directly transcriptionally regulated cell division cycle 25C (CDC25C) by binding its promoter and indirectly influenced CDC25C expression by regulating the ERK pathway. KIF22 also regulated CDC25C/CDK1/cyclinB1 pathway. CONCLUSION KIF22 could promote cell proliferation and cell cycle progression by transcriptionally regulating CDC25C and its downstream CDC25C/CDK1/cyclinB1 pathway to facilitate MM progression, which might be a potential therapeutic target in MM.
Collapse
Affiliation(s)
- Meng Zhai
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an Key Laboratory of Hematological Diseases, Xi'an, China
| | - Jiyu Miao
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an Key Laboratory of Hematological Diseases, Xi'an, China
| | - Ru Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an Key Laboratory of Hematological Diseases, Xi'an, China
| | - Rui Liu
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an Key Laboratory of Hematological Diseases, Xi'an, China
| | - Fangmei Li
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an Key Laboratory of Hematological Diseases, Xi'an, China
| | - Ying Shen
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an Key Laboratory of Hematological Diseases, Xi'an, China
- National Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ting Wang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an Key Laboratory of Hematological Diseases, Xi'an, China
| | - Xuezhu Xu
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an Key Laboratory of Hematological Diseases, Xi'an, China
| | - Gongzhizi Gao
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an Key Laboratory of Hematological Diseases, Xi'an, China
| | - Jinsong Hu
- Department of Cell Biology and Genetics, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'anShaanxi, 710061, China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Xi'an Key Laboratory of Hematological Diseases, Xi'an, China.
- National Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Department of Tumor and Immunology in Precision Medical Institute, Xi'an Jiaotong University, Xi'an, China.
| | - Ju Bai
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Xi'an Key Laboratory of Hematological Diseases, Xi'an, China.
| |
Collapse
|
6
|
K AR, Arumugam S, Muninathan N, Baskar K, S D, D DR. P53 Gene as a Promising Biomarker and Potential Target for the Early Diagnosis of Reproductive Cancers. Cureus 2024; 16:e60125. [PMID: 38864057 PMCID: PMC11165294 DOI: 10.7759/cureus.60125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/10/2024] [Indexed: 06/13/2024] Open
Abstract
One of the crucial aspects of cancer research is diagnosis with specificity and accuracy. Early cancer detection mostly helps make appropriate decisions regarding treatment and metastasis. The well-studied transcription factor tumor suppressor protein p53 is essential for maintaining genetic integrity. p53 is a key tumor suppressor that recognizes the carcinogenic biological pathways and eradicates them by apoptosis. A wide range of carcinomas, especially gynecological such as ovarian, cervical, and endometrial cancers, frequently undergo TP53 gene mutations. This study evaluates the potential of the p53 gene as a biological marker for the diagnosis of reproductive system neoplasms. Immunohistochemistry of p53 is rapid, easy to accomplish, cost-effective, and preferred by pathologists as a surrogate for the analysis of TP53 mutation. Thus, this review lays a groundwork for future efforts to develop techniques using p53 for the early diagnosis of cancer.
Collapse
Affiliation(s)
- Aswathi R K
- Medical Biochemistry, Meenakshi Academy of Higher Education and Research, Chennai, IND
| | - Suresh Arumugam
- Central Research Laboratory, Meenakshi Medical College Hospital and Research Institute, Kanchipuram, IND
| | - Natrajan Muninathan
- Central Research Laboratory, Meenakshi Medical College Hospital and Research Institute, Kanchipuram, IND
| | - Kuppusamy Baskar
- Central Research Laboratory, Meenakshi Medical College Hospital and Research Institute, Kanchipuram, IND
| | - Deepthi S
- Research and Development, Meenakshi Academy of Higher Education and Research, Chennai, IND
| | - Dinesh Roy D
- Centre for Advanced Genetic Studies, Genetika, Thiruvananthapuram, IND
| |
Collapse
|
7
|
Wang C, Wang B, Mou Y, Liu X, Chen Q, Pu W, Rao Q, Wang C, Song J, Huang Y, Yan L, Huang L, Li Y. Design, Synthesis, and Anti-Leukemic Evaluation of a Series of Dianilinopyrimidines by Regulating the Ras/Raf/MEK/ERK and STAT3/c-Myc Pathways. Molecules 2024; 29:1597. [PMID: 38611876 PMCID: PMC11013136 DOI: 10.3390/molecules29071597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
Although the long-term survival rate for leukemia has made significant progress over the years with the development of chemotherapeutics, patients still suffer from relapse, leading to an unsatisfactory outcome. To discover the new effective anti-leukemia compounds, we synthesized a series of dianilinopyrimidines and evaluated the anti-leukemia activities of those compounds by using leukemia cell lines (HEL, Jurkat, and K562). The results showed that the dianilinopyrimidine analog H-120 predominantly displayed the highest cytotoxic potential in HEL cells. It remarkably induced apoptosis of HEL cells by activating the apoptosis-related proteins (cleaved caspase-3, cleaved caspase-9 and cleaved poly ADP-ribose polymerase (PARP)), increasing apoptosis protein Bad expression, and decreasing the expression of anti-apoptotic proteins (Bcl-2 and Bcl-xL). Furthermore, it induced cell cycle arrest in G2/M; concomitantly, we observed the activation of p53 and a reduction in phosphorylated cell division cycle 25C (p-CDC25C) / Cyclin B1 levels in treated cells. Additionally, the mechanism study revealed that H-120 decreased these phosphorylated signal transducers and activators of transcription 3, rat sarcoma, phosphorylated cellular RAF proto-oncogene serine / threonine kinase, phosphorylated mitogen-activated protein kinase kinase, phosphorylated extracellular signal-regulated kinase, and cellular myelocytomatosis oncogene (p-STAT3, Ras, p-C-Raf, p-MEK, p-MRK, and c-Myc) protein levels in HEL cells. Using the cytoplasmic and nuclear proteins isolation assay, we found for the first time that H-120 can inhibit the activation of STAT3 and c-Myc and block STAT3 phosphorylation and dimerization. Moreover, H-120 treatment effectively inhibited the disease progression of erythroleukemia mice by promoting erythroid differentiation into the maturation of erythrocytes and activating the immune cells. Significantly, H-120 also improved liver function in erythroleukemia mice. Therefore, H-120 may be a potential chemotherapeutic drug for leukemia patients.
Collapse
Affiliation(s)
- Chaoyan Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (C.W.); (B.W.); (Y.M.); (X.L.); (Q.C.); (W.P.); (Q.R.); (C.W.); (J.S.); (Y.H.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
- College of Pharmacy, Guizhou Medical University, Guiyang 550004, China
| | - Bo Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (C.W.); (B.W.); (Y.M.); (X.L.); (Q.C.); (W.P.); (Q.R.); (C.W.); (J.S.); (Y.H.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
- College of Basic Medical, Guizhou Medical University, Guiyang 550004, China
| | - Yu Mou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (C.W.); (B.W.); (Y.M.); (X.L.); (Q.C.); (W.P.); (Q.R.); (C.W.); (J.S.); (Y.H.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
- College of Pharmacy, Guizhou Medical University, Guiyang 550004, China
| | - Xiang Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (C.W.); (B.W.); (Y.M.); (X.L.); (Q.C.); (W.P.); (Q.R.); (C.W.); (J.S.); (Y.H.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
- College of Basic Medical, Guizhou Medical University, Guiyang 550004, China
| | - Qiqing Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (C.W.); (B.W.); (Y.M.); (X.L.); (Q.C.); (W.P.); (Q.R.); (C.W.); (J.S.); (Y.H.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
- College of Pharmacy, Guizhou Medical University, Guiyang 550004, China
| | - Weidong Pu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (C.W.); (B.W.); (Y.M.); (X.L.); (Q.C.); (W.P.); (Q.R.); (C.W.); (J.S.); (Y.H.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
- College of Pharmacy, Guizhou Medical University, Guiyang 550004, China
| | - Qing Rao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (C.W.); (B.W.); (Y.M.); (X.L.); (Q.C.); (W.P.); (Q.R.); (C.W.); (J.S.); (Y.H.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Chunlin Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (C.W.); (B.W.); (Y.M.); (X.L.); (Q.C.); (W.P.); (Q.R.); (C.W.); (J.S.); (Y.H.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Jingrui Song
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (C.W.); (B.W.); (Y.M.); (X.L.); (Q.C.); (W.P.); (Q.R.); (C.W.); (J.S.); (Y.H.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Yubing Huang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (C.W.); (B.W.); (Y.M.); (X.L.); (Q.C.); (W.P.); (Q.R.); (C.W.); (J.S.); (Y.H.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Longjia Yan
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550000, China
| | - Lei Huang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (C.W.); (B.W.); (Y.M.); (X.L.); (Q.C.); (W.P.); (Q.R.); (C.W.); (J.S.); (Y.H.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Yanmei Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (C.W.); (B.W.); (Y.M.); (X.L.); (Q.C.); (W.P.); (Q.R.); (C.W.); (J.S.); (Y.H.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| |
Collapse
|
8
|
Hanna DH, Aziz MM, Shafee EE. Effective-by-method for the preparation of folic acid-coated TiO 2 nanoparticles with high targeting potential for apoptosis induction against bladder cancer cells (T24). Biotechnol Appl Biochem 2023; 70:1597-1615. [PMID: 36905187 DOI: 10.1002/bab.2456] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 02/25/2023] [Indexed: 03/12/2023]
Abstract
The research's goal is to create the surfaces of titanium dioxide nanoparticles (TiO2 NPs) in a layer of folic acid (FA) that can effectively target human bladder cancer cells (T24). An efficient method for creating FA-coated TiO2 NPs was used, and many tools have been used to analyze its physicochemical properties. The cytotoxic effects of FA-coated NPs on T24 cells and the mechanisms of apoptosis generation were examined employing a variety of methodologies. The prepared FA-coated TiO2 NPs suspensions with a hydrodynamic diameter around 37 nm and a negative surface charge of -30 mV reduced T24 cell proliferation with stronger IC50 value (21.8 ± 1.9 μg/ml) than TiO2 NPs (47.8 ± 2.5 μg/ml). This toxicity resulted in apoptosis induction (16.63%) that was caused through enhanced reactive oxygen species formation and stopping the cell cycle over G2/M phase. Moreover, FA-TiO2 NPs raised the expression levels of P53, P21, BCL2L4, and cleaved Caspase-3, while decreasing Bcl-2, Cyclin B, and CDK1 in treated cells. Overall, these findings revealed efficient targeting of the FA-TiO2 NPs resulted in increasing cellular internalization caused increased apoptosis in T24 cells. As a result, FA-TiO2 NPs might be a viable treatment for human bladder cancer.
Collapse
Affiliation(s)
- Demiana H Hanna
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | - Marina M Aziz
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | - E El Shafee
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
9
|
Liu H, Zhang W, Jin L, Liu S, Liang L, Wei Y. Plumbagin Exhibits Genotoxicity and Induces G2/M Cell Cycle Arrest via ROS-Mediated Oxidative Stress and Activation of ATM-p53 Signaling Pathway in Hepatocellular Cells. Int J Mol Sci 2023; 24:ijms24076279. [PMID: 37047251 PMCID: PMC10094147 DOI: 10.3390/ijms24076279] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Plumbagin (5-hydroxy-2-methyl-1,4-naphthoquinone, PLB), a naturally occurring naphthoquinone mainly isolated from the plant Plumbago zeylanica L., has been proven to possess anticancer activities towards multiple types of cancer. Although there has been an increasing amount of research regarding its anticancer effects, the association between oxidative stress, genotoxicity and the cell cycle arrest induced by PLB still remains unclear. Therefore, it is important to investigate their potential connections and the involvement of DNA damage and the ataxia telangiectasia mutated protein (ATM)-p53 signaling pathway in PLB’s anticancer mechanism. The present study showed that PLB exposure significantly reduced HCC cell viability and colony formation. In addition, PLB-induced G2/M cell cycle arrest, oxidative stress, and DNA damage was detected, which could be almost blocked by NAC pretreatment. PLB could trigger a DNA damage response by activating cell cycle checkpoints such as ATM, checkpoint kinase 1 (Chk1), checkpoint kinase 2 (Chk2) and p53. Meanwhile, the key modulator of the G2/M transition factor, Cell Division Cycle 25C (cdc25C), was significantly downregulated in an ROS-dependent manner. Furthermore, pretreatment with ATM and p53 inhibitors (KU55933 and Pifithrin-α) could reduce the occurrence of G2/M cell cycle arrest by inhibiting the activation of the ATM-p53 pathway. Taken together, these results indicate that ROS-mediated oxidative stress plays a key role in PLB-induced G2/M cell cycle arrest mediated by the ATM-p53 pathway.
Collapse
Affiliation(s)
- Huan Liu
- Laboratory of Medical Molecular Biology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530024, China; (H.L.)
- Guangxi Key Laboratory of Molecular Biology of Preventive Medicine of Traditional Chinese Medicine, Nanning 530024, China
| | - Wenchao Zhang
- Research Center for Non-Food Biorefinery, Guangxi Academy of Science, Nanning 530001, China
| | - Lijie Jin
- Guangxi Key Laboratory of Molecular Biology of Preventive Medicine of Traditional Chinese Medicine, Nanning 530024, China
- Department of Physiology, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Shasha Liu
- Guangxi Key Laboratory of Molecular Biology of Preventive Medicine of Traditional Chinese Medicine, Nanning 530024, China
- Department of Physiology, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Liying Liang
- Laboratory of Medical Molecular Biology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530024, China; (H.L.)
- Guangxi Key Laboratory of Molecular Biology of Preventive Medicine of Traditional Chinese Medicine, Nanning 530024, China
| | - Yanfei Wei
- Guangxi Key Laboratory of Molecular Biology of Preventive Medicine of Traditional Chinese Medicine, Nanning 530024, China
- Department of Physiology, Guangxi University of Chinese Medicine, Nanning 530200, China
- Correspondence:
| |
Collapse
|
10
|
Malik S, Mintoo MJ, Reddy CN, Kumar R, Kotwal P, Bharate SB, Nandi U, Mondhe DM, Shukla SK. In vitro and in vivo anticancer potential and molecular targets of the new colchicine analog IIIM-067. JOURNAL OF INTEGRATIVE MEDICINE 2023; 21:62-76. [PMID: 36253285 DOI: 10.1016/j.joim.2022.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/14/2022] [Indexed: 01/12/2023]
Abstract
OBJECTIVE The current study evaluated various new colchicine analogs for their anticancer activity and to study the primary mechanism of apoptosis and in vivo antitumor activity of the analogs with selective anticancer properties and minimal toxicity to normal cells. METHODS Sulforhodamine B (SRB) assay was used to screen various colchicine analogs for their in vitro cytotoxicity. The effect of N-[(7S)-1,2,3-trimethoxy-9-oxo-10-(pyrrolidine-1-yl)5,6,7,9-tetrahydrobenzo[a] heptalene-7-yl] acetamide (IIIM-067) on clonogenicity, apoptotic induction, and invasiveness of A549 cells was determined using a clonogenic assay, scratch assay, and staining with 4',6-diamidino-2-phenylindole (DAPI) and annexin V/propidium iodide. Mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) levels were observed using fluorescence microscopy. Western blot analysis was used to quantify expression of proteins involved in apoptosis, cell cycle, and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling. Pharmacokinetic and in vivo efficacy studies against Ehrlich ascites carcinoma (EAC) and Ehrlich solid tumor models were conducted using Swiss albino mice. RESULTS IIIM-067 showed potent cytotoxicity and better selectivity than all other colchicine analogs screened in this study. The selective activity of IIIM-067 toward A549 cells was higher among other cancer cell lines, with a selectivity index (SI) value of 2.28. IIIM-067 demonstrated concentration- and time-dependent cytotoxicity against A549 cells with half-maximal inhibitory concentration values of 0.207, 0.150 and 0.106 μmol/L at 24, 48 and 72 h, respectively. It also had reduced toxicity to normal cells (SI > 1) than the parent compound colchicine (SI = 1). IIIM-067 reduced the clonogenic ability of A549 cells in a dose-dependent manner. IIIM-067 enhanced ROS production from 24.6% at 0.05 μmol/L to 82.1% at 0.4 μmol/L and substantially decreased the MMP (100% in control to 5.6% at 0.4 μmol/L). The annexin V-FITC assay demonstrated 78% apoptosis at 0.4 μmol/L. IIIM-067 significantly (P < 0.5) induced the expression of various intrinsic apoptotic pathway proteins, and it differentially regulated the PI3K/AKT/mTOR signaling pathway. Furthermore, IIIM-067 exhibited remarkable in vivo anticancer activity against the murine EAC model, with tumor growth inhibition (TGI) of 67.0% at a dose of 6 mg/kg (i.p.) and a reduced mortality compared to colchicine. IIIM-067 also effectively inhibited the tumor growth in the murine solid tumor model with TGI rates of 48.10%, 55.68% and 44.00% at doses of 5 mg/kg (i.p.), 6 mg/kg (i.p.) and 7 mg/kg (p.o.), respectively. CONCLUSION IIIM-067 exhibited significant anticancer activity with reduced toxicity both in vitro and in vivo and is a promising anticancer candidate. However, further studies are required in clinical settings to fully understand its potential.
Collapse
Affiliation(s)
- Sumera Malik
- Pharmacology Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Integrative Medicine, Jammu 180001, Jammu & Kashmir, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Mubashir J Mintoo
- Pharmacology Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Integrative Medicine, Jammu 180001, Jammu & Kashmir, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Chilakala Nagarjuna Reddy
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India; Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, Jammu & Kashmir, India
| | - Rajesh Kumar
- Pharmacology Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Integrative Medicine, Jammu 180001, Jammu & Kashmir, India
| | - Pankul Kotwal
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India; Pharmacokinetics-Pharmacodynamics (PK-PD), Toxicology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, Jammu & Kashmir, India
| | - Sandip B Bharate
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India; Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, Jammu & Kashmir, India
| | - Utpal Nandi
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India; Pharmacokinetics-Pharmacodynamics (PK-PD), Toxicology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, Jammu & Kashmir, India
| | - Dilip M Mondhe
- Pharmacology Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Integrative Medicine, Jammu 180001, Jammu & Kashmir, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India.
| | - Sanket K Shukla
- Pharmacology Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Integrative Medicine, Jammu 180001, Jammu & Kashmir, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India.
| |
Collapse
|
11
|
Qin S, Geng H, Wang G, Chen L, Xia C, Yao J, Bai Z, Deng L. Suffruticosol C-Mediated Autophagy and Cell Cycle Arrest via Inhibition of mTORC1 Signaling. Nutrients 2022; 14:nu14235000. [PMID: 36501031 PMCID: PMC9736330 DOI: 10.3390/nu14235000] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
Paeonia species are well-known ornamental plants that are used in traditional Chinese medicines. The seeds of these species are rich in stilbenes, which have wide-ranging health-promoting effects. In particular, resveratrol, which is a common stilbene, is widely known for its anticancer properties. Suffruticosol C, which is a trimer of resveratrol, is the most dominant stilbene found in peony seeds. However, it is not clear whether suffruticosol C has cancer regulating properties. Therefore, in the present study, we aimed to determine the effect of suffruticosol C against various cancer cell lines. Our findings showed that suffruticosol C induces autophagy and cell cycle arrest instead of cell apoptosis and ferroptosis. Mechanistically, suffruticosol C regulates autophagy and cell cycle via inhibiting the mechanistic target of rapamycin complex 1 (mTORC1) signaling. Thus, our findings imply that suffruticosol C regulates cancer cell viability by inducing autophagy and cell cycle arrest via the inhibition of mTORC1 signaling.
Collapse
Affiliation(s)
- Senlin Qin
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712000, China
| | - Huijun Geng
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712000, China
| | - Guoyan Wang
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712000, China
| | - Lei Chen
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712000, China
| | - Chao Xia
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712000, China
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712000, China
| | - Zhangzhen Bai
- College of Landscape Architecture and Arts, Northwest A&F University, Xianyang 712000, China
- Correspondence: (Z.B.); (L.D.); Tel.: +86-18829783704 (Z.B.); +86-18818275171 (L.D.)
| | - Lu Deng
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712000, China
- Correspondence: (Z.B.); (L.D.); Tel.: +86-18829783704 (Z.B.); +86-18818275171 (L.D.)
| |
Collapse
|
12
|
Zhou X, Zhou M, Zheng M, Tian S, Yang X, Ning Y, Li Y, Zhang S. Polyploid giant cancer cells and cancer progression. Front Cell Dev Biol 2022; 10:1017588. [PMID: 36274852 PMCID: PMC9581214 DOI: 10.3389/fcell.2022.1017588] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/21/2022] [Indexed: 12/02/2022] Open
Abstract
Polyploid giant cancer cells (PGCCs) are an important feature of cellular atypia, the detailed mechanisms of their formation and function remain unclear. PGCCs were previously thought to be derived from repeated mitosis/cytokinesis failure, with no intrinsic ability to proliferate and divide. However, recently, PGCCs have been confirmed to have cancer stem cell (CSC)-like characteristics, and generate progeny cells through asymmetric division, which express epithelial-mesenchymal transition-related markers to promote invasion and migration. The formation of PGCCs can be attributed to multiple stimulating factors, including hypoxia, chemotherapeutic reagents, and radiation, can induce the formation of PGCCs, by regulating the cell cycle and cell fusion-related protein expression. The properties of CSCs suggest that PGCCs can be induced to differentiate into non-tumor cells, and produce erythrocytes composed of embryonic hemoglobin, which have a high affinity for oxygen, and thereby allow PGCCs survival from the severe hypoxia. The number of PGCCs is associated with metastasis, chemoradiotherapy resistance, and recurrence of malignant tumors. Targeting relevant proteins or signaling pathways related with the formation and transdifferentiation of adipose tissue and cartilage in PGCCs may provide new strategies for solid tumor therapy.
Collapse
Affiliation(s)
- Xinyue Zhou
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Mingming Zhou
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Shifeng Tian
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Xiaohui Yang
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yidi Ning
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yuwei Li
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| |
Collapse
|
13
|
Symbiosis with Dinoflagellates Alters Cnidarian Cell-Cycle Gene Expression. Cell Microbiol 2022. [DOI: 10.1155/2022/3330160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In the cnidarian-dinoflagellate symbiosis, hosts show altered expression of genes involved in growth and proliferation when in the symbiotic state, but little is known about the molecular mechanisms that underlie the host’s altered growth rate. Using tissue-specific transcriptomics, we determined how symbiosis affects expression of cell cycle-associated genes, in the model symbiotic cnidarian Exaiptasia diaphana (Aiptasia). The presence of symbionts within the gastrodermis elicited cell-cycle arrest in the G1 phase in a larger proportion of host cells compared with the aposymbiotic gastrodermis. The symbiotic gastrodermis also showed a reduction in the amount of cells synthesizing their DNA and progressing through mitosis when compared with the aposymbiotic gastrodermis. Host apoptotic inhibitors (Mdm2) were elevated, while host apoptotic sensitizers (c-Myc) were depressed, in the symbiotic gastrodermis when compared with the aposymbiotic gastrodermis and epidermis of symbiotic anemones, respectively. This indicates that the presence of symbionts negatively regulates host apoptosis, possibly contributing to their persistence within the host. Transcripts (ATM/ATR) associated with DNA damage were also downregulated in symbiotic gastrodermal tissues. In epidermal cells, a single gene (Mob1) required for mitotic completion was upregulated in symbiotic compared with aposymbiotic anemones, suggesting that the presence of symbionts in the gastrodermis stimulates host cell division in the epidermis. To further corroborate this hypothesis, we performed microscopic analysis using an S-phase indicator (EdU), allowing us to evaluate cell cycling in host cells. Our results confirmed that there were significantly more proliferating host cells in both the gastrodermis and epidermis in the symbiotic state compared with the aposymbiotic state. Furthermore, when comparing between tissue layers in the presence of symbionts, the epidermis had significantly more proliferating host cells than the symbiont-containing gastrodermis. These results contribute to our understanding of the influence of symbionts on the mechanisms of cnidarian cell proliferation and mechanisms associated with symbiont maintenance.
Collapse
|
14
|
Kung CP, Weber JD. It’s Getting Complicated—A Fresh Look at p53-MDM2-ARF Triangle in Tumorigenesis and Cancer Therapy. Front Cell Dev Biol 2022; 10:818744. [PMID: 35155432 PMCID: PMC8833255 DOI: 10.3389/fcell.2022.818744] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/07/2022] [Indexed: 01/31/2023] Open
Abstract
Anti-tumorigenic mechanisms mediated by the tumor suppressor p53, upon oncogenic stresses, are our bodies’ greatest weapons to battle against cancer onset and development. Consequently, factors that possess significant p53-regulating activities have been subjects of serious interest from the cancer research community. Among them, MDM2 and ARF are considered the most influential p53 regulators due to their abilities to inhibit and activate p53 functions, respectively. MDM2 inhibits p53 by promoting ubiquitination and proteasome-mediated degradation of p53, while ARF activates p53 by physically interacting with MDM2 to block its access to p53. This conventional understanding of p53-MDM2-ARF functional triangle have guided the direction of p53 research, as well as the development of p53-based therapeutic strategies for the last 30 years. Our increasing knowledge of this triangle during this time, especially through identification of p53-independent functions of MDM2 and ARF, have uncovered many under-appreciated molecular mechanisms connecting these three proteins. Through recognizing both antagonizing and synergizing relationships among them, our consideration for harnessing these relationships to develop effective cancer therapies needs an update accordingly. In this review, we will re-visit the conventional wisdom regarding p53-MDM2-ARF tumor-regulating mechanisms, highlight impactful studies contributing to the modern look of their relationships, and summarize ongoing efforts to target this pathway for effective cancer treatments. A refreshed appreciation of p53-MDM2-ARF network can bring innovative approaches to develop new generations of genetically-informed and clinically-effective cancer therapies.
Collapse
Affiliation(s)
- Che-Pei Kung
- ICCE Institute, St. Louis, MO, United States
- Division of Molecular Oncology, Department of Medicine, St. Louis, MO, United States
- *Correspondence: Che-Pei Kung, ; Jason D. Weber,
| | - Jason D. Weber
- ICCE Institute, St. Louis, MO, United States
- Division of Molecular Oncology, Department of Medicine, St. Louis, MO, United States
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, United States
- *Correspondence: Che-Pei Kung, ; Jason D. Weber,
| |
Collapse
|
15
|
Ou A, Zhao X, Lu Z. The potential roles of p53 signaling reactivation in pancreatic cancer therapy. Biochim Biophys Acta Rev Cancer 2022; 1877:188662. [PMID: 34861354 DOI: 10.1016/j.bbcan.2021.188662] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/25/2022]
Abstract
Globally, pancreatic cancer (PC) is a common and highly malignant gastrointestinal tumor that is characterized by an insidious onset and ready metastasis and recurrence. Over recent decades, the incidence of PC has been increasing on an annual basis; however, the pathogenesis of this condition remains enigmatic. PC is not sensitive to radio- or chemotherapy, and except for early surgical resection, there is no curative treatment regime; consequently, the prognosis for patients with PC is extremely poor. Transcription factor p53 is known to play key roles in many important biological processes in vertebrates, including normal cell growth, differentiation, cell cycle progression, senescence, apoptosis, metabolism, and DNA damage repair. However, there is a significant paucity of basic and clinical studies to describe how p53 gene mutations or protein dysfunction facilitate the occurrence, progression, invasion, and resistance to therapy, of malignancies, including PC. Herein, we describe the involvement of p53 signaling reactivation in PC treatment as well as its underlying molecular mechanisms, thereby providing useful insights for targeting p53-related signal pathways in PC therapy.
Collapse
Affiliation(s)
- Aixin Ou
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, LN, China
| | - Xiangxuan Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, LN, China
| | - Zaiming Lu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, LN, China.
| |
Collapse
|
16
|
Ou A, Zhao X, Lu Z. The potential roles of p53 signaling reactivation in pancreatic cancer therapy. Biochim Biophys Acta Rev Cancer 2022; 1877:188662. [DOI: doi10.1016/j.bbcan.2021.188662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
17
|
Li Q, Yuan Q, Wang T, Zhan Y, Yang L, Fan Y, Lei H, Su J. Fumonisin B 1 Inhibits Cell Proliferation and Decreases Barrier Function of Swine Umbilical Vein Endothelial Cells. Toxins (Basel) 2021; 13:toxins13120863. [PMID: 34941701 PMCID: PMC8704807 DOI: 10.3390/toxins13120863] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 11/27/2022] Open
Abstract
The fumonisins are a group of common mycotoxins found around the world that mainly contaminate maize. As environmental toxins, they pose a threat to human and animal health. Fumonisin B1 (FB1) is the most widely distributed and the most toxic. FB1 can cause pulmonary edema in pigs. However, the current toxicity mechanism of fumonisins is still in the exploratory stage, which may be related to sphingolipid metabolism. Our study is designed to investigate the effect of FB1 on the cell proliferation and barrier function of swine umbilical vein endothelial cells (SUVECs). We show that FB1 can inhibit the cell viability of SUVECs. FB1 prevents cells from entering the S phase from the G1 phase by regulating the expression of the cell cycle-related genes cyclin B1, cyclin D1, cyclin E1, Cdc25c, and the cyclin-dependent kinase-4 (CDK-4). This results in an inhibition of cell proliferation. In addition, FB1 can also change the cell morphology, increase paracellular permeability, destroy tight junctions and the cytoskeleton, and reduce the expression of tight junction-related genes claudin 1, occludin, and ZO-1. This indicates that FB1 can cause cell barrier dysfunction of SUVECs and promote the weakening or even destruction of the connections between endothelial cells. In turn, this leads to increased blood vessel permeability and promotes exudation. Our findings suggest that FB1 induces toxicity in SUVECs by affecting cell proliferation and disrupting the barrier function.
Collapse
|
18
|
Hanna DH, R. Saad G. Induction of mitochondria mediated apoptosis in human ovarian cancer cells by folic acid coated tin oxide nanoparticles. PLoS One 2021; 16:e0258115. [PMID: 34597348 PMCID: PMC8486119 DOI: 10.1371/journal.pone.0258115] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/17/2021] [Indexed: 12/15/2022] Open
Abstract
PURPOSE This study aims to prepare folic acid coated tin oxide nanoparticles (FA-SnO2 NPs) for specifically targeting human ovarian cancer cells with minimum side effects against normal cells. METHODS The prepared FA-SnO2 NPs were characterized by FT-IR, UV-vis spectroscopy, XRD, SEM and TEM. The inhibition effects of FA-SnO2 NPs against SKOV3 cancer cell were tested by MTT and LDH assay. Apoptosis induction in FA-SnO2 NPs treated SKOV3 cells were investigated using Annexin V/PI, AO/EB and Comet assays and the possible mechanisms of the cytotoxic action were studied by Flow cytometry, qRT-PCR, Immunohistochemistry, and Western blotting analyses. The effects of FA-SnO2 NPs on reactive oxygen species generation in SKOV3 cells were also examined. Additionally, the safety of utilization FA-SnO2 NPs were studied in vivo using Wister rats. RESULTS The obtained FA-SnO2 NPs displayed amorphous spherical morphology with an average diameter of 157 nm and a zeta potential value of -24 mV. Comparing to uncoated SnO2 NPs, FA-SnO2 NPs had a superior inhibition effect towards SKOV3 cell growth that was suggested to be mediated through higher reactive oxygen species generation. It was showed that FA-SnO2 NPs increased significantly the % of apoptotic cells in the sub- G1 and G2/M phases with a higher intensity comet nucleus in SKOV3 treated cells. Furthermore, FA-SnO2 NPs was significantly increased the expression levels of P53, Bax, and cleaved Caspase-3 and accompanied with a significant decrease of Bcl-2 in the treated SKOV3 cells. CONCLUSION Overall, the results suggested that an increase in cellular FA-SnO2 NPs internalization resulted in a significant induced cytotoxicity in SKOV3 cancer cells in dose-dependent mode through ROS-mediated cell apoptosis that may have occurred through mitochondrial pathway. Additionally, the results confirmed the safety of utilization FA-SnO2 NPs against living systems. So, FA-SnO2 NPs with a specific targeting moiety may be a promising therapeutic candidate for human ovarian cancer.
Collapse
Affiliation(s)
- Demiana H. Hanna
- Faculty of Science, Department of Chemistry, Cairo University, Giza, Egypt
| | - Gamal R. Saad
- Faculty of Science, Department of Chemistry, Cairo University, Giza, Egypt
| |
Collapse
|
19
|
Tao Y, Hao X, Jing L, Sun L, Cherukupalli S, Liu S, Wu G, Xu S, Zhang X, Shi X, Song Y, Liu X, Zhan P. Discovery of potent and selective Cdc25 phosphatase inhibitors via rapid assembly and in situ screening of Quinonoid-focused libraries. Bioorg Chem 2021; 115:105254. [PMID: 34426152 DOI: 10.1016/j.bioorg.2021.105254] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/19/2021] [Accepted: 07/25/2021] [Indexed: 12/31/2022]
Abstract
Cell division cycle 25 (Cdc25) phosphatase is an attractive target for drug discovery. The rapid assembly and in situ screening of focused combinatorial fragment libraries using efficient modular reactions is a highly robust strategy for discovering bioactive molecules. In this study, we have utilized miniaturized synthesis to generate several quinonoid-focused libraries, by standard CuAAC reaction and HBTU-based amide coupling chemistry. Then the enzyme inhibition screening afforded some potent and selective Cdc25s inhibitors. Compound M5N36 (Cdc25A: IC50 = 0.15 ± 0.05 μM; Cdc25B: IC50 = 0.19 ± 0.06 μM; Cdc25C: IC50 = 0.06 ± 0.04 μM) exhibited higher inhibitory activity than the initial lead NSC663284 (Cdc25A: IC50 = 0.27 ± 0.02 μM; Cdc25B: IC50 = 0.42 ± 0.01 μM; Cdc25C: IC50 = 0.23 ± 0.01 μM). Moreover, M5N36 displayed about three-fold more potent against Cdc25C than Cdc25A and B, indicating that M5N36 could act as a relatively selective Cdc25C inhibitor. Cell viability evaluation, western blotting and molecular simulations provided a mechanistic understanding of the activity of M5N36. It showed promising anti-growth activity against the MDA-MB-231 cell line and desirable predicted physicochemical properties. Overall, M5N36 was proven to be a promising novel Cdc25C inhibitor.
Collapse
Affiliation(s)
- Yucen Tao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Chelloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Xia Hao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Chelloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Lanlan Jing
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Chelloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Lin Sun
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Chelloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Srinivasulu Cherukupalli
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Chelloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Shugong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Chelloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Gaochan Wu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Chelloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Shujing Xu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Chelloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Xujie Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Chelloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Xiaoyu Shi
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Chelloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Yuning Song
- Department of Clinical Pharmacy, Qilu Hospital of Shandong University, 250012 Jinan, China.
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Chelloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Chelloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| |
Collapse
|
20
|
Serpeloni JM, Specian AFL, Ribeiro DL, Tuttis K, Heredia-Vieira SC, Vilegas W, Martínez-López W, Varanda EA, de Syllos Cólus IM. Selective anticancer effects of Serjania marginata Casar. extract in gastric cells are mediated by antioxidant response. ENVIRONMENTAL TOXICOLOGY 2021; 36:1544-1556. [PMID: 33885224 DOI: 10.1002/tox.23151] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/17/2021] [Accepted: 04/11/2021] [Indexed: 06/12/2023]
Abstract
Gastric cancer is the fifth most common malignancy worldwide. Serjania marginata Casar. (SM) displays anti-inflammatory properties and has been used to treat gastrointestinal disorders. In the current study, we examined whether the hydroethanolic extract of SM leaves exerted cytotoxic, mutagenic, and protective effects in non-tumor gastric epithelium cells (MNP01) and gastric adenocarcinoma cells (ACP02) in vitro and analyzed whether its action was selective. Initially, cell viability (MTT assay), cell cycle kinetics (flow cytometry), and cell proliferation (total protein content) were analyzed. In addition, genomic instability (cytokinesis-block micronucleus cytome assay), anti/pro-oxidant status (CM-H2 DCFDA probe), and transcriptional expression (RT-qPCR) of genes related to cell cycle, cell death, and antioxidant defense were also evaluated. The SM extract was cytotoxic toward MNP01 and ACP02 cells at concentrations greater than 300 and 100 μg·ml-1 , respectively, and decreased protein content only toward ACP02 cells at 200 μg ml-1 . In ACP02 cells, the SM extract at 100 μg·ml-1 associated with doxorubicin (DXR; 0.2 μg ml-1 ) clearly promoted cell cycle arrest at the G2/M phase. The extract alone was not mutagenic to either cell type and reversed DXR-induced DNA damage and H2 O2 -induced oxidative stress in MNP01 cells. The gene expression experiments showed that SM hydroethanolic extract exerts an antioxidant response via NFE2L2 activation in non-tumor gastric cells, and cell cycle arrest (G2/M) in ACP02 gastric cancer cells via the TP53 pathway. The selective action of SM indicates that it is a promising therapeutic agent to treat gastric diseases and merits further studies.
Collapse
Affiliation(s)
- Juliana Mara Serpeloni
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina, Brazil
| | - Ana Flavia Leal Specian
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina, Brazil
| | - Diego Luis Ribeiro
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina, Brazil
| | - Katiuska Tuttis
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina, Brazil
| | | | - Wagner Vilegas
- Experimental Campus of São Vicente, São Paulo State University (UNESP), São Vicente, Brazil
| | - Wilner Martínez-López
- Epigenetics and Genomic Instability Laboratory, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Eliana Aparecida Varanda
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Ilce Mara de Syllos Cólus
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina, Brazil
| |
Collapse
|
21
|
Aliotta F, Nasso R, Rullo R, Arcucci A, Avagliano A, Simonetti M, Sanità G, Masullo M, Lavecchia A, Ruocco MR, Vendittis ED. Inhibition mechanism of naphthylphenylamine derivatives acting on the CDC25B dual phosphatase and analysis of the molecular processes involved in the high cytotoxicity exerted by one selected derivative in melanoma cells. J Enzyme Inhib Med Chem 2021; 35:1866-1878. [PMID: 32990107 PMCID: PMC7580834 DOI: 10.1080/14756366.2020.1819257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The dual phosphatases CDC25 are involved in cell cycle regulation and overexpressed in many tumours, including melanoma. CDC25 is a promising target for discovering anticancer drugs, and several studies focussed on characterisation of quinonoid CDC25 inhibitors, frequently causing undesired side toxic effects. Previous work described an optimisation of the inhibition properties by naphthylphenylamine (NPA) derivatives of NSC28620, a nonquinonoid CDC25 inhibitor. Now, the CDC25B•inhibitor interaction was investigated through fluorescence studies, shedding light on the different inhibition mechanism exerted by NPA derivatives. Among the molecular processes, mediating the specific and high cytotoxicity of one NPA derivative in melanoma cells, we observed decrease of phosphoAkt, increase of p53, reduction of CDC25 forms, cytochrome c cytosolic translocation and increase of caspase activity, that lead to the activation of an apoptotic programme. A basic knowledge on CDC25 inhibitors is relevant for discovering potent bioactive molecules, to be used as anticancer agents against the highly aggressive melanoma.
Collapse
Affiliation(s)
- Federica Aliotta
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Rosarita Nasso
- Department of Movement Sciences and Wellness, University of Naples "Parthenope", Naples, Italy
| | - Rosario Rullo
- Institute for the Animal Production Systems in the Mediterranean Environment, CNR, Naples, Italy
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Angelica Avagliano
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Martina Simonetti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Gennaro Sanità
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Mariorosario Masullo
- Department of Movement Sciences and Wellness, University of Naples "Parthenope", Naples, Italy
| | - Antonio Lavecchia
- Department of Pharmacy, "Drug Discovery" Laboratory, University of Naples Federico II, Naples, Italy
| | - Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Emmanuele De Vendittis
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
22
|
Ho NTT, Rahane CS, Pramanik S, Kim PS, Kutzner A, Heese K. FAM72, Glioblastoma Multiforme (GBM) and Beyond. Cancers (Basel) 2021; 13:cancers13051025. [PMID: 33804473 PMCID: PMC7957592 DOI: 10.3390/cancers13051025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/18/2021] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Glioblastoma multiforme (GBM) is a serious and aggressive cancer disease that has not allowed scientists to rest for decades. In this review, we consider the new gene pair |-SRGAP2–FAM72-| and discuss its role in the cell cycle and the possibility of defining new therapeutic approaches for the treatment of GBM and other cancers via this gene pair |-SRGAP2–FAM72-|. Abstract Neural stem cells (NSCs) offer great potential for regenerative medicine due to their excellent ability to differentiate into various specialized cell types of the brain. In the central nervous system (CNS), NSC renewal and differentiation are under strict control by the regulation of the pivotal SLIT-ROBO Rho GTPase activating protein 2 (SRGAP2)—Family with sequence similarity 72 (FAM72) master gene (i.e., |-SRGAP2–FAM72-|) via a divergent gene transcription activation mechanism. If the gene transcription control unit (i.e., the intergenic region of the two sub-gene units, SRGAP2 and FAM72) gets out of control, NSCs may transform into cancer stem cells and generate brain tumor cells responsible for brain cancer such as glioblastoma multiforme (GBM). Here, we discuss the surveillance of this |-SRGAP2–FAM72-| master gene and its role in GBM, and also in light of FAM72 for diagnosing various types of cancers outside of the CNS.
Collapse
Affiliation(s)
- Nguyen Thi Thanh Ho
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133-791, Korea;
| | - Chinmay Satish Rahane
- Maharashtra Institute of Medical Education and Research, Talegaon Dabhade, Maharashtra 410507, India;
| | - Subrata Pramanik
- Institute of Biotechnology, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany;
| | - Pok-Son Kim
- Department of Mathematics, Kookmin University, 77 Jeongneung-ro, Seongbuk-gu, Seoul 136-702, Korea;
| | - Arne Kutzner
- Department of Information Systems, College of Computer Science, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133-791, Korea;
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133-791, Korea;
- Correspondence:
| |
Collapse
|
23
|
Tiwari B, Jones AE, Caillet CJ, Das S, Royer SK, Abrams JM. p53 directly represses human LINE1 transposons. Genes Dev 2020; 34:1439-1451. [PMID: 33060137 PMCID: PMC7608743 DOI: 10.1101/gad.343186.120] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022]
Abstract
p53 is a potent tumor suppressor and commonly mutated in human cancers. Recently, we demonstrated that p53 genes act to restrict retrotransposons in germline tissues of flies and fish but whether this activity is conserved in somatic human cells is not known. Here we show that p53 constitutively restrains human LINE1s by cooperatively engaging sites in the 5'UTR and stimulating local deposition of repressive histone marks at these transposons. Consistent with this, the elimination of p53 or the removal of corresponding binding sites in LINE1s, prompted these retroelements to become hyperactive. Concurrently, p53 loss instigated chromosomal rearrangements linked to LINE sequences and also provoked inflammatory programs that were dependent on reverse transcriptase produced from LINE1s. Taken together, our observations establish that p53 continuously operates at the LINE1 promoter to restrict autonomous copies of these mobile elements in human cells. Our results further suggest that constitutive restriction of these retroelements may help to explain tumor suppression encoded by p53, since erupting LINE1s produced acute oncogenic threats when p53 was absent.
Collapse
Affiliation(s)
- Bhavana Tiwari
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Amanda E Jones
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Candace J Caillet
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Simanti Das
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Stephanie K Royer
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - John M Abrams
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
24
|
p73: From the p53 shadow to a major pharmacological target in anticancer therapy. Pharmacol Res 2020; 162:105245. [PMID: 33069756 DOI: 10.1016/j.phrs.2020.105245] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023]
Abstract
p73, along with p53 and p63, belongs to the p53 family of transcription factors. Besides the p53-like tumor suppressive activities, p73 has unique roles, namely in neuronal development and differentiation. In addition, the TP73 gene is rarely mutated in tumors. This makes p73 a highly appealing therapeutic target, particularly towards cancers with a null or disrupted p53 pathway. Distinct isoforms are transcribed from the TP73 locus either with (TAp73) and without (ΔNp73) the N-terminal transactivation domain. Conversely to TA tumor suppressors, ΔN proteins exhibit oncogenic properties by inhibiting p53 and TA protein functions. As such, p73 isoforms compose a puzzled and challenging regulatory pathway. This state-of-the-art review affords an update overview on p73 structure, biological functions and pharmacological regulation. Importantly, it addresses the relevance of p73 isoforms in carcinogenesis, highlighting their potential as drug targets in anticancer therapy. A critical discussion of major pharmacological approaches to promote p73 tumor suppressive activities, with relevant survival outcomes for cancer patients, is also provided.
Collapse
|
25
|
Abstract
The evolutionarily conserved p53 protein and its cellular pathways mediate tumour suppression through an informed, regulated and integrated set of responses to environmental perturbations resulting in either cellular death or the maintenance of cellular homeostasis. The p53 and MDM2 proteins form a central hub in this pathway that receives stressful inputs via MDM2 and respond via p53 by informing and altering a great many other pathways and functions in the cell. The MDM2-p53 hub is one of the hubs most highly connected to other signalling pathways in the cell, and this may be why TP53 is the most commonly mutated gene in human cancers. Initial or truncal TP53 gene mutations (the first mutations in a stem cell) are selected for early in cancer development inectodermal and mesodermal-derived tissue-specific stem and progenitor cells and then, following additional mutations, produce tumours from those tissue types. In endodermal-derived tissue-specific stem or progenitor cells, TP53 mutations are functionally selected as late mutations transitioning the mutated cell into a malignant tumour. The order in which oncogenes or tumour suppressor genes are functionally selected for in a stem cell impacts the timing and development of a tumour.
Collapse
Affiliation(s)
- Arnold J Levine
- Simons Center for Systems Biology, Institute for Advanced Study, Princeton, NJ, USA.
| |
Collapse
|
26
|
Liu K, Zheng M, Lu R, Du J, Zhao Q, Li Z, Li Y, Zhang S. The role of CDC25C in cell cycle regulation and clinical cancer therapy: a systematic review. Cancer Cell Int 2020; 20:213. [PMID: 32518522 PMCID: PMC7268735 DOI: 10.1186/s12935-020-01304-w] [Citation(s) in RCA: 169] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 05/28/2020] [Indexed: 12/24/2022] Open
Abstract
One of the most prominent features of tumor cells is uncontrolled cell proliferation caused by an abnormal cell cycle, and the abnormal expression of cell cycle-related proteins gives tumor cells their invasive, metastatic, drug-resistance, and anti-apoptotic abilities. Recently, an increasing number of cell cycle-associated proteins have become the candidate biomarkers for early diagnosis of malignant tumors and potential targets for cancer therapies. As an important cell cycle regulatory protein, Cell Division Cycle 25C (CDC25C) participates in regulating G2/M progression and in mediating DNA damage repair. CDC25C is a cyclin of the specific phosphatase family that activates the cyclin B1/CDK1 complex in cells for entering mitosis and regulates G2/M progression and plays an important role in checkpoint protein regulation in case of DNA damage, which can ensure accurate DNA information transmission to the daughter cells. The regulation of CDC25C in the cell cycle is affected by multiple signaling pathways, such as cyclin B1/CDK1, PLK1/Aurora A, ATR/CHK1, ATM/CHK2, CHK2/ERK, Wee1/Myt1, p53/Pin1, and ASK1/JNK-/38. Recently, it has evident that changes in the expression of CDC25C are closely related to tumorigenesis and tumor development and can be used as a potential target for cancer treatment. This review summarizes the role of CDC25C phosphatase in regulating cell cycle. Based on the role of CDC25 family proteins in the development of tumors, it will become a hot target for a new generation of cancer treatments.
Collapse
Affiliation(s)
- Kai Liu
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Rui Lu
- Department of Pathology, Tianjin Nankai Hospital, Tianjin, People’s Republic of China
| | - Jiaxing Du
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Qi Zhao
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Zugui Li
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Yuwei Li
- Departments of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| |
Collapse
|
27
|
He P, Li Z, Xu F, Ru G, Huang Y, Lin E, Peng S. AMPK Activity Contributes to G2 Arrest and DNA Damage Decrease via p53/p21 Pathways in Oxidatively Damaged Mouse Zygotes. Front Cell Dev Biol 2020; 8:539485. [PMID: 33015052 PMCID: PMC7505953 DOI: 10.3389/fcell.2020.539485] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 08/19/2020] [Indexed: 02/05/2023] Open
Abstract
In zygotes, the capacity of G2/M checkpoint and DNA repair mechanisms to respond to DNA damage varies depending on different external stressors. In our previous studies, we found that mild oxidative stress induced a G2/M phase delay in mouse zygotes fertilized in vitro, due to the activation of the spindle assembly checkpoint. However, it is unclear whether the G2/M phase delay involves G2 arrest, triggered by activation of the G2/M checkpoint, and whether AMPK, a highly conserved cellular energy sensor, is involved in G2 arrest and DNA damage repair in mouse zygotes. Here, we found that mouse zygotes treated with 0.03 mM H2O2 at 7 h post-insemination (G1 phase), went into G2 arrest in the first cleavage. Furthermore, phosphorylated H2AX, a specific DNA damage and repair marker, can be detected since the early S phase. We also observed that oxidative stress induced phosphorylation and activation of AMPK. Oxidative stress-activated AMPK first localized in the cytoplasm of the mouse zygotes in the late G1 phase and then translocated to the nucleus from the early S phase. Overall, most of the activated AMPK accumulated in the nuclei of mouse zygotes arrested in the G2 phase. Inhibition of AMPK activity with Compound C and SBI-0206965 abolished oxidative stress-induced G2 arrest, increased the activity of CDK1, and decreased the induction of cell cycle regulatory proteins p53 and p21. Moreover, bypassing G2 arrest after AMPK inhibition aggravated oxidative stress-induced DNA damage at M phase, increased the apoptotic rate of blastocysts, and reduced the formation rate of 4-cell embryos and blastocysts. Our results suggest the G2/M checkpoint and DNA repair mechanisms are operative in coping with mild oxidative stress-induced DNA damage. Further, AMPK activation plays a vital role in the regulation of the oxidative stress-induced G2 arrest through the inhibition of CDK1 activity via p53/p21 pathways, thereby facilitating the repair of DNA damage and the development and survival of oxidative stress-damaged embryos. Our study provides insights into the molecular mechanisms underlying oxidative-stress induced embryonic developmental arrest, which is crucial for the development of novel strategies to ensure viable embryo generation.
Collapse
Affiliation(s)
- Pei He
- Department of Reproductive Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Guangdong Key Laboratory of Medical Molecular Imaging, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zhiling Li
- Department of Reproductive Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Guangdong Key Laboratory of Medical Molecular Imaging, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- *Correspondence: Zhiling Li,
| | - Feng Xu
- Department of Respiratory Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Gaizhen Ru
- Department of Reproductive Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yue Huang
- Department of Reproductive Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - En Lin
- Department of Reproductive Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Sanfeng Peng
- Department of Reproductive Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
28
|
Flem-Karlsen K, McFadden E, Omar N, Haugen MH, Øy GF, Ryder T, Gullestad HP, Hermann R, Mælandsmo GM, Flørenes VA. Targeting AXL and the DNA Damage Response Pathway as a Novel Therapeutic Strategy in Melanoma. Mol Cancer Ther 2019; 19:895-905. [PMID: 31871265 DOI: 10.1158/1535-7163.mct-19-0290] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 08/28/2019] [Accepted: 12/10/2019] [Indexed: 11/16/2022]
Abstract
Receptor tyrosine kinase AXL is found upregulated in various types of cancer, including melanoma, and correlates with an aggressive cancer phenotype, inducing cell proliferation and epithelial-to-mesenchymal transition. In addition, AXL has recently been linked to chemotherapy resistance, and inhibition of AXL is found to increase DNA damage and reduce expression of DNA repair proteins. In light of this, we aimed to investigate whether targeting AXL together with DNA damage response proteins would be therapeutically beneficial. Using melanoma cell lines, we observed that combined reduction of AXL and CHK1/CHK2 signaling decreased proliferation, deregulated cell-cycle progression, increased apoptosis, and reduced expression of DNA damage response proteins. Enhanced therapeutic effect of combined treatment, as compared with mono-treatment, was further observed in a patient-derived xenograft model and, of particular interest, when applying a three-dimensional ex vivo spheroid drug sensitivity assay on tumor cells harvested directly from 27 patients with melanoma lymph node metastases. Together, these results indicate that targeting AXL together with the DNA damage response pathway could be a promising treatment strategy in melanoma, and that further investigations in patient groups lacking treatment alternatives should be pursued.
Collapse
Affiliation(s)
- Karine Flem-Karlsen
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway. .,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Erin McFadden
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Nasrin Omar
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Mads H Haugen
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Geir Frode Øy
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Truls Ryder
- Department of Plastic and Reconstructive Surgery, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Hans Petter Gullestad
- Department of Plastic and Reconstructive Surgery, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Robert Hermann
- Department of Plastic and Reconstructive Surgery, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Gunhild Mari Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Institute of Medical Biology, Faculty of Health Sciences, UiT-Arctic University of Norway, Tromsø, Norway
| | - Vivi Ann Flørenes
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
29
|
Abstract
The transition between proliferating and quiescent states must be carefully regulated to ensure that cells divide to create the cells an organism needs only at the appropriate time and place. Cyclin-dependent kinases (CDKs) are critical for both transitioning cells from one cell cycle state to the next, and for regulating whether cells are proliferating or quiescent. CDKs are regulated by association with cognate cyclins, activating and inhibitory phosphorylation events, and proteins that bind to them and inhibit their activity. The substrates of these kinases, including the retinoblastoma protein, enforce the changes in cell cycle status. Single cell analysis has clarified that competition among factors that activate and inhibit CDK activity leads to the cell's decision to enter the cell cycle, a decision the cell makes before S phase. Signaling pathways that control the activity of CDKs regulate the transition between quiescence and proliferation in stem cells, including stem cells that generate muscle and neurons. © 2020 American Physiological Society. Compr Physiol 10:317-344, 2020.
Collapse
Affiliation(s)
- Hilary A Coller
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, California, USA.,Department of Biological Chemistry, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California, USA.,Molecular Biology Institute, University of California, Los Angeles, California, USA
| |
Collapse
|
30
|
Chen YT, Chen JJ, Wang HT. Targeting RNA Polymerase I with Hernandonine Inhibits Ribosomal RNA Synthesis and Tumor Cell Growth. Mol Cancer Res 2019; 17:2294-2305. [PMID: 31409627 DOI: 10.1158/1541-7786.mcr-19-0402] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/08/2019] [Accepted: 08/08/2019] [Indexed: 11/16/2022]
Abstract
RNA polymerase I (RNA Pol. I) activity is consistently expanded in multiplying cells to continue the expanded interest for ribosome generation and protein synthesis, which are fundamental for cell development and division. Thus, selective inhibitors of RNA Pol. I may offer a general helpful intends to block cancer cell multiplication. Hernandonine, isolated from the root wood of Hernandia nymphaeifolia, causes rearrangement of nucleolar proteins consistent with segregation of the nucleolus, a hallmark of RNA Pol. I transcription stress. Furthermore, the compound destabilizes RPA194, the large catalytic protein of RNA Pol. I, in a proteasome-dependent manner and inhibits nascent rRNA synthesis and expression of the 45S rRNA precursor. Finally, hernandonine induces cellular apoptosis through a p53-dependent or p53-independent process in solid tumor cell lines. These outcomes feature the prevailing effect of RNA Pol. I transcription stress on apoptosis pathway initiation and present a synthetically novel and significant molecule that represses RNA Pol. I, making it a potential objective for malignancy treatment. IMPLICATIONS: Our findings position hernandonine as a potential, particular, and orally administered cancer treatment agent appropriate for use in investigational clinical trials.
Collapse
Affiliation(s)
- Yen-Ting Chen
- Department of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Jih-Jung Chen
- Faculty of Pharmacy, School of Pharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Hsiang-Tsui Wang
- Department of Pharmacology, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
31
|
Aru B, Güzelmeric E, Akgül A, Demirel GY, Kırmızıbekmez H. Antiproliferative Activity of Chemically Characterized Propolis from Turkey and Its Mechanisms of Action. Chem Biodivers 2019; 16:e1900189. [DOI: 10.1002/cbdv.201900189] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 05/20/2019] [Indexed: 12/26/2022]
Affiliation(s)
- Başak Aru
- Department of ImmunologyFaculty of MedicineYeditepe University TR-34755 Kayışdağı İstanbul Turkey
| | - Etil Güzelmeric
- Department of PharmacognosyFaculty of PharmacyYeditepe University TR-34755 Kayışdağı İstanbul Turkey
| | - Aslı Akgül
- Faculty of PharmacyYeditepe University TR-34755 Kayışdağı İstanbul Turkey
| | | | - Hasan Kırmızıbekmez
- Department of PharmacognosyFaculty of PharmacyYeditepe University TR-34755 Kayışdağı İstanbul Turkey
| |
Collapse
|
32
|
Anvarnia A, Mohaddes‐Gharamaleki F, Asadi M, Akbari M, Yousefi B, Shanehbandi D. Dysregulated microRNAs in colorectal carcinogenesis: New insight to cell survival and apoptosis regulation. J Cell Physiol 2019; 234:21683-21693. [DOI: 10.1002/jcp.28872] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Alireza Anvarnia
- Department of Biochemistry and Clinical Laboratories Faculty of Medicine, Tabriz University of Medical Sciences Tabriz Iran
- Student Research Committee, Tabriz University of Medical Sciences Tabriz Iran
| | - Farzad Mohaddes‐Gharamaleki
- Department of Biochemistry and Clinical Laboratories Faculty of Medicine, Tabriz University of Medical Sciences Tabriz Iran
- Student Research Committee, Tabriz University of Medical Sciences Tabriz Iran
| | - Milad Asadi
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | - Bahman Yousefi
- Department of Biochemistry and Clinical Laboratories Faculty of Medicine, Tabriz University of Medical Sciences Tabriz Iran
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
33
|
Prognostic significance of CDC25C in lung adenocarcinoma: An analysis of TCGA data. Cancer Genet 2019; 233-234:67-74. [PMID: 31109596 DOI: 10.1016/j.cancergen.2019.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/16/2019] [Accepted: 04/03/2019] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Cell division cycle 25C (CDC25C) is involved in the regulation of the G2/M phase transition and is associated with various cancers, including non-small cell lung cancer. We evaluated its prognostic value in lung adenocarcinoma (LUAD) based on data from The Cancer Genome Atlas (TCGA). METHODS Kruskal-Wallis test, Wilcoxon signed-rank test, and logistic regression were used to evaluate relationships between clinical-pathologic features and CDC25C expression. Cox regression analyses and the Kaplan-Meier method were used to evaluate factors contributing to prognosis. Gene set enrichment analysis (GSEA) was performed. RESULTS High CDC25C expression in LUAD was associated with a high tumor extent (odds ratio (OR) = 2.23 (1.52-3.29), P < 0.001), regional lymph node invasion (OR = 2.18 (1.48-3.22), P < 0.001), OR = advanced stage (OR = 2.47 (1.72-3.59), P < 0.001), and poor status (OR = 1.87 (1.19-2.96), P = 0.007). A univariate analysis showed that high CDC25C expression is associated with a short overall survival (OS) (HR: 1.873; 95% CI: 1.385-2.535; P < 0.001) and poor progression-free survival (HR: 1.503; 95% CI: 1.173-1.926; P = 0.0012). In a multivariate analysis, high CDC25C expression was associated with poor OS (HR = 2.193; CI: 1.394-3.452, P = 0.001). GSEA showed the enrichment of cell cycle, apoptosis, p53-dependent G1 DNA damage response, S-phase, mitotic M-M G1 phases, and FA-mediated cell death in the CDC25C high-expression phenotype. CONCLUSIONS CDC25C predicts poor prognosis in LUAD and may function in cell cycle regulation and FAS-mediated apoptosis.
Collapse
|
34
|
Hu Y, Jin R, Gao M, Xu H, Zou S, Li X, Xing C, Wang Q, Wang H, Feng J, Hu M, Song L. Transcriptional repression of IKKβ by p53 in arsenite-induced GADD45α accumulation and apoptosis. Oncogene 2019; 38:731-746. [PMID: 30177839 PMCID: PMC6355650 DOI: 10.1038/s41388-018-0478-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 08/03/2018] [Accepted: 08/06/2018] [Indexed: 02/01/2023]
Abstract
Our previous studies revealed that GADD45α is a liable protein, which undergoes MDM2-dependent constitutive ubiquitination and degradation in resting HepG2 hepatoma cells. Arsenite exposure induces ribosomal stress responses mediated by the ribosomal protein S7, which can block MDM2 activity and result in GADD45α accumulation and cell apoptosis. In the present study, we found that one of the catalytic subunits of IκB kinase (IKK), IKKβ, exerted a novel IKKα- and NF-κB-independent function in stabilizing MDM2 and therefore contributed to ubiquitination-dependent degradation of GADD45α in resting HepG2 cells. Arsenite stimulation induced transactivation of p53, which formed a complex with its downstream target, Ets-1, and then synergistically repressed IKKβ transcription, reduced MDM2 stability, and ultimately removed the inhibitory effect of MDM2 on GADD45α induction. In addition, DAPK1 functioned as an upstream protein kinase triggering p53/Ets-1-dependent IKKβ and MDM2 reduction and GADD45α accumulation, thus promoting apoptosis in HepG2 cells. Subsequent studies further revealed that the activation of the DAPK1/p53/Ets-1/IKKβ/MDM2/GADD45α cascade was a common signaling event in mediating apoptosis of diverse cancer cells induced by arsenite and other tumor therapeutic agents. Therefore, we conclude that data in the current study have revealed a novel role for IKKβ in negatively regulating GADD45α protein stability and the contribution of p53-dependent IKKβ reduction to mediating cancer cell apoptosis.
Collapse
Affiliation(s)
- Yongliang Hu
- Department of Neuroimmunology, Beijing Institute of Brain Sciences, 27 Taiping Road, Beijing, 100850, China
- Department of Dermatology, The 309 Hospital of PLA, 17 Heishanhu Street, Beijing, 100091, P. R. China
| | - Rui Jin
- Department of Tumor Biology, Beijing Institute of Biotechnology, 27 Taiping Road, Beijing, 100850, China
| | - Ming Gao
- Department of Neuroimmunology, Beijing Institute of Brain Sciences, 27 Taiping Road, Beijing, 100850, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Beijing, 100085, P. R. China
| | - Huan Xu
- Department of Neuroimmunology, Beijing Institute of Brain Sciences, 27 Taiping Road, Beijing, 100850, China
| | - Shuxian Zou
- Department of Neuroimmunology, Beijing Institute of Brain Sciences, 27 Taiping Road, Beijing, 100850, China
- Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Xiaoguang Li
- Department of Neuroimmunology, Beijing Institute of Brain Sciences, 27 Taiping Road, Beijing, 100850, China
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, 270 Dong'an Road, Shanghai, 200032, P. R. China
| | - Chen Xing
- Department of Neuroimmunology, Beijing Institute of Brain Sciences, 27 Taiping Road, Beijing, 100850, China
| | - Qiyu Wang
- Department of Neuroimmunology, Beijing Institute of Brain Sciences, 27 Taiping Road, Beijing, 100850, China
| | - Hongli Wang
- Department of Neuroimmunology, Beijing Institute of Brain Sciences, 27 Taiping Road, Beijing, 100850, China
| | - Jiannan Feng
- Department of Neuroimmunology, Beijing Institute of Brain Sciences, 27 Taiping Road, Beijing, 100850, China
| | - Meiru Hu
- Department of Neuroimmunology, Beijing Institute of Brain Sciences, 27 Taiping Road, Beijing, 100850, China
| | - Lun Song
- Department of Neuroimmunology, Beijing Institute of Brain Sciences, 27 Taiping Road, Beijing, 100850, China.
- Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China.
- Anhui Medical University, 81 Meishan Road, Hefei, 230032, China.
| |
Collapse
|
35
|
Wang J, Shen T, Zhu W, Dou L, Gu H, Zhang L, Yang Z, Chen H, Zhou Q, Sánchez ER, Field LJ, Mayo LD, Xie Z, Xiao D, Lin X, Shou W, Yong W. Protein phosphatase 5 and the tumor suppressor p53 down-regulate each other's activities in mice. J Biol Chem 2018; 293:18218-18229. [PMID: 30262665 DOI: 10.1074/jbc.ra118.004256] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/17/2018] [Indexed: 12/20/2022] Open
Abstract
Protein phosphatase 5 (PP5), a serine/threonine phosphatase, has a wide range of biological functions and exhibits elevated expression in tumor cells. We previously reported that pp5-deficient mice have altered ataxia-telangiectasia mutated (ATM)-mediated signaling and function. However, this regulation was likely indirect, as ATM is not a known PP5 substrate. In the current study, we found that pp5-deficient mice are hypersensitive to genotoxic stress. This hypersensitivity was associated with the marked up-regulation of the tumor suppressor tumor protein p53 and its downstream targets cyclin-dependent kinase inhibitor 1A (p21), MDM2 proto-oncogene (MDM2), and phosphatase and tensin homolog (PTEN) in pp5-deficient tissues and cells. These observations suggested that PP5 plays a role in regulating p53 stability and function. Experiments conducted with p53 +/- pp5 +/- or p53 +/- pp5 -/- mice revealed that complete loss of PP5 reduces tumorigenesis in the p53 +/- mice. Biochemical analyses further revealed that PP5 directly interacts with and dephosphorylates p53 at multiple serine/threonine residues, resulting in inhibition of p53-mediated transcriptional activity. Interestingly, PP5 expression was significantly up-regulated in p53-deficient cells, and further analysis of pp5 promoter activity revealed that p53 strongly represses PP5 transcription. Our results suggest a reciprocal regulatory interplay between PP5 and p53, providing an important feedback mechanism for the cellular response to genotoxic stress.
Collapse
Affiliation(s)
- Jun Wang
- From the Comparative Medical Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing 100021, China,; School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Tao Shen
- DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030
| | - Wuqiang Zhu
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Longyu Dou
- From the Comparative Medical Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Hao Gu
- From the Comparative Medical Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Lingling Zhang
- From the Comparative Medical Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Zhenyun Yang
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Hanying Chen
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Qi Zhou
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Edwin R Sánchez
- Department of Physiology and Pharmacology, College of Medicine, University of Toledo, Toledo, Ohio 43614, and
| | - Loren J Field
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Lindsey D Mayo
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Zhongwen Xie
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Deyong Xiao
- Fountain Valley Institute of Life Sciences and Fountain Valley Biomedical Technology Company, Dalian Hi-Tech Industrial Zone, Dalian 116023, China
| | - Xia Lin
- DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030
| | - Weinian Shou
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana 46202,.
| | - Weidong Yong
- From the Comparative Medical Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing 100021, China,; Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana 46202,.
| |
Collapse
|
36
|
Ogita N, Okushima Y, Tokizawa M, Yamamoto YY, Tanaka M, Seki M, Makita Y, Matsui M, Okamoto-Yoshiyama K, Sakamoto T, Kurata T, Hiruma K, Saijo Y, Takahashi N, Umeda M. Identifying the target genes of SUPPRESSOR OF GAMMA RESPONSE 1, a master transcription factor controlling DNA damage response in Arabidopsis. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2018; 94:439-453. [PMID: 29430765 DOI: 10.1111/tpj.13866] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 05/17/2023]
Abstract
In mammalian cells, the transcription factor p53 plays a crucial role in transmitting DNA damage signals to maintain genome integrity. However, in plants, orthologous genes for p53 and checkpoint proteins are absent. Instead, the plant-specific transcription factor SUPPRESSOR OF GAMMA RESPONSE 1 (SOG1) controls most of the genes induced by gamma irradiation and promotes DNA repair, cell cycle arrest, and stem cell death. To date, the genes directly controlled by SOG1 remain largely unknown, limiting the understanding of DNA damage signaling in plants. Here, we conducted a microarray analysis and chromatin immunoprecipitation (ChIP)-sequencing, and identified 146 Arabidopsis genes as direct targets of SOG1. By using ChIP-sequencing data, we extracted the palindromic motif [CTT(N)7 AAG] as a consensus SOG1-binding sequence, which mediates target gene induction in response to DNA damage. Furthermore, DNA damage-triggered phosphorylation of SOG1 is required for efficient binding to the SOG1-binding sequence. Comparison between SOG1 and p53 target genes showed that both transcription factors control genes responsible for cell cycle regulation, such as CDK inhibitors, and DNA repair, whereas SOG1 preferentially targets genes involved in homologous recombination. We also found that defense-related genes were enriched in the SOG1 target genes. Consistent with this finding, SOG1 is required for resistance against the hemi-biotrophic fungus Colletotrichum higginsianum, suggesting that SOG1 has a unique function in controlling the immune response.
Collapse
Affiliation(s)
- Nobuo Ogita
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Yoko Okushima
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Mutsutomo Tokizawa
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Gifu, 501-1193, Japan
| | - Yoshiharu Y Yamamoto
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Gifu, 501-1193, Japan
| | - Maho Tanaka
- RIKEN Center for Sustainable Resource Science, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Motoaki Seki
- RIKEN Center for Sustainable Resource Science, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- JST, CREST, Kawaguchi, Saitama, 332-0012, Japan
| | - Yuko Makita
- RIKEN Center for Sustainable Resource Science, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Minami Matsui
- RIKEN Center for Sustainable Resource Science, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Kaoru Okamoto-Yoshiyama
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Tomoaki Sakamoto
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Tetsuya Kurata
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Kei Hiruma
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Yusuke Saijo
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Naoki Takahashi
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Masaaki Umeda
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
- JST, CREST, Ikoma, Nara, 630-0192, Japan
| |
Collapse
|
37
|
Benatti P, Basile V, Dolfini D, Belluti S, Tomei M, Imbriano C. NF-Y loss triggers p53 stabilization and apoptosis in HPV18-positive cells by affecting E6 transcription. Oncotarget 2018; 7:45901-45915. [PMID: 27323853 PMCID: PMC5216769 DOI: 10.18632/oncotarget.9974] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 06/01/2016] [Indexed: 12/28/2022] Open
Abstract
The expression of the high risk HPV18 E6 and E7 oncogenic proteins induces the transformation of epithelial cells, through the disruption of p53 and Rb function. The binding of cellular transcription factors to cis-regulatory elements in the viral Upstream Regulatory Region (URR) stimulates E6/E7 transcription. Here, we demonstrate that the CCAAT-transcription factor NF-Y binds to a non-canonical motif within the URR and activates viral gene expression. In addition, NF-Y indirectly up-regulates HPV18 transcription through the transactivation of multiple cellular transcription factors. NF-YA depletion inhibits the expression of E6 and E7 genes and re-establishes functional p53. The activation of p53 target genes in turn leads to apoptotic cell death. Finally, we show that NF-YA loss sensitizes HPV18-positive cells toward the DNA damaging agent Doxorubicin, via p53-mediated transcriptional response.
Collapse
Affiliation(s)
- Paolo Benatti
- Dipartimento di Scienze della Vita, Università di Modena e Reggio Emilia, 41125 Modena, Italy
| | - Valentina Basile
- Dipartimento di Scienze della Vita, Università di Modena e Reggio Emilia, 41125 Modena, Italy
| | - Diletta Dolfini
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20133 Milano, Italy
| | - Silvia Belluti
- Dipartimento di Scienze della Vita, Università di Modena e Reggio Emilia, 41125 Modena, Italy
| | - Margherita Tomei
- Dipartimento di Scienze della Vita, Università di Modena e Reggio Emilia, 41125 Modena, Italy
| | - Carol Imbriano
- Dipartimento di Scienze della Vita, Università di Modena e Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
38
|
Giono LE, Resnick-Silverman L, Carvajal LA, St Clair S, Manfredi JJ. Mdm2 promotes Cdc25C protein degradation and delays cell cycle progression through the G2/M phase. Oncogene 2017; 36:6762-6773. [PMID: 28806397 PMCID: PMC6002854 DOI: 10.1038/onc.2017.254] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 06/15/2017] [Accepted: 06/27/2017] [Indexed: 12/16/2022]
Abstract
Upon different types of stress, the gene encoding the mitosis-promoting phosphatase Cdc25C is transcriptionally repressed by p53, contributing to p53's enforcement of a G2 cell cycle arrest. In addition, Cdc25C protein stability is also decreased following DNA damage. Mdm2, another p53 target gene, encodes a ubiquitin ligase that negatively regulates p53 levels by ubiquitination. Ablation of Mdm2 by siRNA led to an increase in p53 protein and repression of Cdc25C gene expression. However, Cdc25C protein levels were actually increased following Mdm2 depletion. Mdm2 is shown to negatively regulate Cdc25C protein levels by reducing its half-life independently of the presence of p53. Further, Mdm2 physically interacts with Cdc25C and promotes its degradation through the proteasome in a ubiquitin-independent manner. Either Mdm2 overexpression or Cdc25C downregulation delays cell cycle progression through the G2/M phase. Thus, the repression of the Cdc25C promoter by p53, together with p53-dependent induction of Mdm2 and subsequent degradation of Cdc25C, could provide a dual mechanism by which p53 can enforce and maintain a G2/M cell cycle arrest.
Collapse
Affiliation(s)
- L E Giono
- Department of Oncological Sciences and Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - L Resnick-Silverman
- Department of Oncological Sciences and Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - L A Carvajal
- Department of Oncological Sciences and Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - S St Clair
- Department of Oncological Sciences and Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J J Manfredi
- Department of Oncological Sciences and Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
39
|
Fischer M. Census and evaluation of p53 target genes. Oncogene 2017; 36:3943-3956. [PMID: 28288132 PMCID: PMC5511239 DOI: 10.1038/onc.2016.502] [Citation(s) in RCA: 668] [Impact Index Per Article: 83.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/23/2016] [Accepted: 11/29/2016] [Indexed: 12/17/2022]
Abstract
The tumor suppressor p53 functions primarily as a transcription factor. Mutation of the TP53 gene alters its response pathway, and is central to the development of many cancers. The discovery of a large number of p53 target genes, which confer p53's tumor suppressor function, has led to increasingly complex models of p53 function. Recent meta-analysis approaches, however, are simplifying our understanding of how p53 functions as a transcription factor. In the survey presented here, a total set of 3661 direct p53 target genes is identified that comprise 3509 potential targets from 13 high-throughput studies, and 346 target genes from individual gene analyses. Comparison of the p53 target genes reported in individual studies with those identified in 13 high-throughput studies reveals limited consistency. Here, p53 target genes have been evaluated based on the meta-analysis data, and the results show that high-confidence p53 target genes are involved in multiple cellular responses, including cell cycle arrest, DNA repair, apoptosis, metabolism, autophagy, mRNA translation and feedback mechanisms. However, many p53 target genes are identified only in a small number of studies and have a higher likelihood of being false positives. While numerous mechanisms have been proposed for mediating gene regulation in response to p53, recent advances in our understanding of p53 function show that p53 itself is solely an activator of transcription, and gene downregulation by p53 is indirect and requires p21. Taking into account the function of p53 as an activator of transcription, recent results point to an unsophisticated means of regulation.
Collapse
Affiliation(s)
- M Fischer
- Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
40
|
Li X, Jiang Z, Feng J, Zhang X, Wu J, Chen W. 2-Acetylamino-3-[4-(2-acetylamino-2-carboxyethylsulfanylcarbonylamino) phenyl carbamoylsulfanyl] propionic acid, a glutathione reductase inhibitor, induces G 2/M cell cycle arrest through generation of thiol oxidative stress in human esophageal cancer cells. Oncotarget 2017; 8:61846-61860. [PMID: 28977909 PMCID: PMC5617469 DOI: 10.18632/oncotarget.18705] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 05/22/2017] [Indexed: 02/07/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a highly malignant cancer with poor response to both of chemotherapy and radiotherapy. 2-Acetylamino-3-[4-(2-acetylamino-2-carboxyethylsulfanylcarbonylamino) phenyl carbamoylsulfanyl] propionic acid (2-AAPA), an irreversible inhibitor of glutathione reductase (GR), is able to induce intracellular oxidative stress, and has shown anticancer activity in many cancer cell lines. In this study, we investigated the effects of 2-AAPA on the cell proliferation, cell cycle and apoptosis and aimed to explore its mechanism of action in human esophageal cancer TE-13 cells. It was found that 2-AAPA inhibited growth of ESCC cells in a dose-dependent manner and it did not deplete reduced glutathione (GSH), but significantly increased the oxidized form glutathione (GSSG), resulting in decreased GSH/GSSG ratio. In consequence, significant reactive oxygen species (ROS) production was observed. The flow cytometric analysis revealed that 2-AAPA inhibited growth of esophageal cancer cells through arresting cell cycle in G2/M phase, but apoptosis-independent mechanism. The G2/M arrest was partially contributed by down-regulation of protein expression of Cdc-25c and up-regulation of phosphorylated Cdc-2 (Tyr15), Cyclin B1 (Ser147) and p53. Meanwhile, 2-AAPA-induced thiol oxidative stress led to increased protein S-glutathionylation, which resulted in α-tubulin S-glutathionylation-dependent depolymerization of microtubule in the TE-13 cells. In conclusion, we identified that 2-AAPA as an effective thiol oxidative stress inducer and proliferation of TE-13 cells were suppressed by G2/M phase cell cycle arrest, mainly, through α-tubulin S-glutathionylation-mediated microtubule depolymerization. Our results may introduce new target and approach for esophageal cancer therapy through generation of GR-mediated thiol oxidative stress.
Collapse
Affiliation(s)
- Xia Li
- Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, Zhejiang Cancer Center, Hangzhou, Zhejiang 310022, China.,Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus), Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Zhiming Jiang
- Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, Zhejiang Cancer Center, Hangzhou, Zhejiang 310022, China.,Zhejiang Key Laboratory of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Jianguo Feng
- Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, Zhejiang Cancer Center, Hangzhou, Zhejiang 310022, China.,Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus), Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | | | - Junzhou Wu
- Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, Zhejiang Cancer Center, Hangzhou, Zhejiang 310022, China.,Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus), Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Wei Chen
- Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, Zhejiang Cancer Center, Hangzhou, Zhejiang 310022, China.,Zhejiang Key Laboratory of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
41
|
Liu X, Wang Y, Ji H, Aihara K, Chen L. Personalized characterization of diseases using sample-specific networks. Nucleic Acids Res 2016; 44:e164. [PMID: 27596597 PMCID: PMC5159538 DOI: 10.1093/nar/gkw772] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 08/17/2016] [Accepted: 08/23/2016] [Indexed: 01/20/2023] Open
Abstract
A complex disease generally results not from malfunction of individual molecules but from dysfunction of the relevant system or network, which dynamically changes with time and conditions. Thus, estimating a condition-specific network from a single sample is crucial to elucidating the molecular mechanisms of complex diseases at the system level. However, there is currently no effective way to construct such an individual-specific network by expression profiling of a single sample because of the requirement of multiple samples for computing correlations. We developed here with a statistical method, i.e. a sample-specific network (SSN) method, which allows us to construct individual-specific networks based on molecular expressions of a single sample. Using this method, we can characterize various human diseases at a network level. In particular, such SSNs can lead to the identification of individual-specific disease modules as well as driver genes, even without gene sequencing information. Extensive analysis by using the Cancer Genome Atlas data not only demonstrated the effectiveness of the method, but also found new individual-specific driver genes and network patterns for various types of cancer. Biological experiments on drug resistance further validated one important advantage of our method over the traditional methods, i.e. we can even identify such drug resistance genes that actually have no clear differential expression between samples with and without the resistance, due to the additional network information.
Collapse
Affiliation(s)
- Xiaoping Liu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- Institute of Industrial Science, University of Tokyo, Tokyo 153-8505, Japan
| | - Yuetong Wang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongbin Ji
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Kazuyuki Aihara
- Institute of Industrial Science, University of Tokyo, Tokyo 153-8505, Japan
| | - Luonan Chen
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- Institute of Industrial Science, University of Tokyo, Tokyo 153-8505, Japan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| |
Collapse
|
42
|
Ligand-based chemoinformatic discovery of a novel small molecule inhibitor targeting CDC25 dual specificity phosphatases and displaying in vitro efficacy against melanoma cells. Oncotarget 2016; 6:40202-22. [PMID: 26474275 PMCID: PMC4741889 DOI: 10.18632/oncotarget.5473] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Accepted: 10/02/2015] [Indexed: 12/20/2022] Open
Abstract
CDC25 phosphatases are important regulators of the cell cycle and represent promising targets for anticancer drug discovery. We recently identified NSC 119915 as a new quinonoid CDC25 inhibitor with potent anticancer activity. In order to discover more active analogs of NSC 119915, we performed a range of ligand-based chemoinformatic methods against the full ZINC drug-like subset and the NCI lead-like set. Nine compounds (3, 5-9, 21, 24, and 25) were identified with Ki values for CDC25A, -B and -C ranging from 0.01 to 4.4 μM. One of these analogs, 7, showed a high antiproliferative effect on human melanoma cell lines, A2058 and SAN. Compound 7 arrested melanoma cells in G2/M, causing a reduction of the protein levels of CDC25A and, more consistently, of CDC25C. Furthermore, an intrinsic apoptotic pathway was induced, which was mediated by ROS, because it was reverted in the presence of antioxidant N-acetyl-cysteine (NAC). Finally, 7 decreased the protein levels of phosphorylated Akt and increased those of p53, thus contributing to the regulation of chemosensitivity through the control of downstream Akt pathways in melanoma cells. Taken together, our data emphasize that CDC25 could be considered as a possible oncotarget in melanoma cells and that compound 7 is a small molecule CDC25 inhibitor that merits to be further evaluated as a chemotherapeutic agent for melanoma, likely in combination with other therapeutic compounds.
Collapse
|
43
|
Kozakai Y, Kamada R, Furuta J, Kiyota Y, Chuman Y, Sakaguchi K. PPM1D controls nucleolar formation by up-regulating phosphorylation of nucleophosmin. Sci Rep 2016; 6:33272. [PMID: 27619510 PMCID: PMC5020408 DOI: 10.1038/srep33272] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 08/24/2016] [Indexed: 01/01/2023] Open
Abstract
An increase of nucleolar number and size has made nucleoli essential markers for cytology and tumour development. However, the underlying basis for their structural integrity and abundance remains unclear. Protein phosphatase PPM1D was found to be up-regulated in different carcinomas including breast cancers. Here, we demonstrate for the first time that PPM1D regulates nucleolar formation via inducing an increased phosphorylation of the nucleolar protein NPM. We show that PPM1D overexpression induces an increase in the nucleolar number regardless of p53 status. We also demonstrated that specific sequential phosphorylation of NPM is important for nucleolar formation and that PPM1D is a novel upstream regulator of this phosphorylation pathway. These results enhance our understanding of the molecular mechanisms that govern nucleoli formation by demonstrating that PPM1D regulates nucleolar formation by regulating NPM phosphorylation status through a novel signalling pathway, PPM1D-CDC25C-CDK1-PLK1.
Collapse
Affiliation(s)
- Yuuki Kozakai
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Rui Kamada
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Junya Furuta
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Yuhei Kiyota
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Yoshiro Chuman
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Kazuyasu Sakaguchi
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Japan
| |
Collapse
|
44
|
Chen J. The Cell-Cycle Arrest and Apoptotic Functions of p53 in Tumor Initiation and Progression. Cold Spring Harb Perspect Med 2016; 6:a026104. [PMID: 26931810 DOI: 10.1101/cshperspect.a026104] [Citation(s) in RCA: 792] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
P53 is a transcription factor highly inducible by many stress signals such as DNA damage, oncogene activation, and nutrient deprivation. Cell-cycle arrest and apoptosis are the most prominent outcomes of p53 activation. Many studies showed that p53 cell-cycle and apoptosis functions are important for preventing tumor development. p53 also regulates many cellular processes including metabolism, antioxidant response, and DNA repair. Emerging evidence suggests that these noncanonical p53 activities may also have potent antitumor effects within certain context. This review focuses on the cell-cycle arrest and apoptosis functions of p53, their roles in tumor suppression, and the regulation of cell fate decision after p53 activation.
Collapse
Affiliation(s)
- Jiandong Chen
- Molecular Oncology Department, Moffitt Cancer Center, Tampa, Florida 33612
| |
Collapse
|
45
|
Gabrielli B, Burgess A. Cdc25 Family Phosphatases in Cancer. PROTEIN TYROSINE PHOSPHATASES IN CANCER 2016:283-306. [DOI: 10.1007/978-1-4939-3649-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
46
|
Fischer M, Quaas M, Nickel A, Engeland K. Indirect p53-dependent transcriptional repression of Survivin, CDC25C, and PLK1 genes requires the cyclin-dependent kinase inhibitor p21/CDKN1A and CDE/CHR promoter sites binding the DREAM complex. Oncotarget 2015; 6:41402-17. [PMID: 26595675 PMCID: PMC4747163 DOI: 10.18632/oncotarget.6356] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 11/11/2015] [Indexed: 12/15/2022] Open
Abstract
The transcription factor p53 is central to cell cycle control by downregulation of cell cycle-promoting genes upon cell stress such as DNA damage. Survivin (BIRC5), CDC25C, and PLK1 encode important cell cycle regulators that are repressed following p53 activation. Here, we provide evidence that p53-dependent repression of these genes requires activation of p21 (CDKN1A, WAF1, CIP1). Chromatin immunoprecipitation (ChIP) data indicate that promoter binding of B-MYB switches to binding of E2F4 and p130 resulting in a replacement of the MMB (Myb-MuvB) by the DREAM complex. We demonstrate that this replacement depends on p21. Furthermore, transcriptional repression by p53 requires intact DREAM binding sites in the target promoters. The CDE and CHR cell cycle promoter elements are the sites for DREAM binding. These elements as well as the p53 response of Survivin, CDC25C, and PLK1 are evolutionarily conserved. No binding of p53 to these genes is detected by ChIP and mutation of proposed p53 binding sites does not alter the p53 response. Thus, a mechanism for direct p53-dependent transcriptional repression is not supported by the data. In contrast, repression by DREAM is consistent with most previous findings and unifies models based on p21-, E2F4-, p130-, and CDE/CHR-dependent repression by p53. In conclusion, the presented data suggest that the p53-p21-DREAM-CDE/CHR pathway regulates p53-dependent repression of Survivin, CDC25C, and PLK1.
Collapse
Affiliation(s)
- Martin Fischer
- Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
- Department of Medical Oncology, Dana–Farber Cancer Institute, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marianne Quaas
- Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
| | - Annina Nickel
- Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
| | - Kurt Engeland
- Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
| |
Collapse
|
47
|
Long Noncoding RNA MEG3 Interacts with p53 Protein and Regulates Partial p53 Target Genes in Hepatoma Cells. PLoS One 2015; 10:e0139790. [PMID: 26444285 PMCID: PMC4596861 DOI: 10.1371/journal.pone.0139790] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 09/17/2015] [Indexed: 12/13/2022] Open
Abstract
Maternally Expressed Gene 3 (MEG3) encodes a lncRNA which is suggested to function as a tumor suppressor. Previous studies suggested that MEG3 functioned through activation of p53, however, the functional properties of MEG3 remain obscure and their relevance to human diseases is under continuous investigation. Here, we try to illuminate the relationship of MEG3 and p53, and the consequence in hepatoma cells. We find that transfection of expression construct of MEG3 enhances stability and transcriptional activity of p53. Deletion analysis of MEG3 confirms that full length and intact structure of MEG3 are critical for it to activate p53-mediated transactivation. Interestingly, our results demonstrate for the first time that MEG3 can interact with p53 DNA binding domain and various p53 target genes are deregulated after overexpression of MEG3 in hepatoma cells. Furthermore, results of qRT-PCR have shown that MEG3 RNA is lost or reduced in the majority of HCC samples compared with adjacent non-tumorous samples. Ectopic expression of MEG3 in hepatoma cells significantly inhibits proliferation and induces apoptosis. In conclusion, our data demonstrates that MEG3 functions as a tumor suppressor in hepatoma cells through interacting with p53 protein to activate p53-mediated transcriptional activity and influence the expression of partial p53 target genes.
Collapse
|
48
|
Müllers E, Silva Cascales H, Jaiswal H, Saurin AT, Lindqvist A. Nuclear translocation of Cyclin B1 marks the restriction point for terminal cell cycle exit in G2 phase. Cell Cycle 2015; 13:2733-43. [PMID: 25486360 PMCID: PMC4615111 DOI: 10.4161/15384101.2015.945831] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Upon DNA damage, cell cycle progression is temporally blocked to avoid propagation of mutations. While transformed cells largely maintain the competence to recover from a cell cycle arrest, untransformed cells past the G1/S transition lose mitotic inducers, and thus the ability to resume cell division. This permanent cell cycle exit depends on p21, p53, and APC/CCdh1. However, when and how permanent cell cycle exit occurs remains unclear. Here, we have investigated the cell cycle response to DNA damage in single cells that express Cyclin B1 fused to eYFP at the endogenous locus. We find that upon DNA damage Cyclin B1-eYFP continues to accumulate up to a threshold level, which is reached only in G2 phase. Above this threshold, a p21 and p53-dependent nuclear translocation required for APC/CCdh1-mediated Cyclin B1-eYFP degradation is initiated. Thus, cell cycle exit is decoupled from activation of the DNA damage response in a manner that correlates to Cyclin B1 levels, suggesting that G2 activities directly feed into the decision for cell cycle exit. Once Cyclin B1-eYFP nuclear translocation occurs, checkpoint inhibition can no longer promote mitotic entry or re-expression of mitotic inducers, suggesting that nuclear translocation of Cyclin B1 marks the restriction point for permanent cell cycle exit in G2 phase.
Collapse
Key Words
- APC/C, anaphase-promoting complex/cyclosome
- ATM, Ataxia telangiectasia mutated kinase
- ATR, Ataxia telangiectasia and Rad3 related kinase
- AU, arbitrary units
- Cdk, cyclin-dependent kinase
- Chk1/2, checkpoint kinase 1/2
- Cyclin B1
- DDR, DNA damage response
- DNA damage response
- DNA-PK, DNA-dependent protein kinase
- G2 phase
- H2AX, phosphorylated on serine 139
- LMB, Leptomycin B
- MK2, MAPKAP kinase 2
- Mdm2, mouse double minute 2 homolog
- NCS, Neocarzinostatin
- Plk1, polo-like kinase 1
- cell cycle
- checkpoint recovery
- nuclear translocation recovery competence
- senescence
- γH2AX, histone variant
Collapse
Affiliation(s)
- Erik Müllers
- a Department of Cell and Molecular Biology; Karolinska Institutet ; Stockholm , Sweden
| | | | | | | | | |
Collapse
|
49
|
Tsuchiya Y, Murai S, Yamashita S. Dual inhibition of Cdc2 protein kinase activation during apoptosis inXenopusegg extracts. FEBS J 2015; 282:1256-70. [DOI: 10.1111/febs.13217] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 01/12/2015] [Accepted: 01/26/2015] [Indexed: 11/27/2022]
Affiliation(s)
- Yuichi Tsuchiya
- Department of Biochemistry; Toho University School of Medicine; Ota-ku Tokyo Japan
| | - Shin Murai
- Department of Biochemistry; Toho University School of Medicine; Ota-ku Tokyo Japan
| | - Shigeru Yamashita
- Department of Biochemistry; Toho University School of Medicine; Ota-ku Tokyo Japan
| |
Collapse
|
50
|
Expression of p53 target genes in the early phase of long-term potentiation in the rat hippocampal CA1 area. Neural Plast 2015; 2015:242158. [PMID: 25767724 PMCID: PMC4341845 DOI: 10.1155/2015/242158] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/27/2015] [Indexed: 01/09/2023] Open
Abstract
Gene expression plays an important role in the mechanisms of long-term potentiation (LTP), which is a widely accepted experimental model of synaptic plasticity. We have studied the expression of at least 50 genes that are transcriptionally regulated by p53, as well as other genes that are related to p53-dependent processes, in the early phase of LTP. Within 30 min after Schaffer collaterals (SC) tetanization, increases in the mRNA and protein levels of Bax, which are upregulated by p53, and a decrease in the mRNA and protein levels of Bcl2, which are downregulated by p53, were observed. The inhibition of Mdm2 by nutlin-3 increased the basal p53 protein level and rescued its tetanization-induced depletion, which suggested the involvement of Mdm2 in the control over p53 during LTP. Furthermore, nutlin-3 caused an increase in the basal expression of Bax and a decrease in the basal expression of Bcl2, whereas tetanization-induced changes in their expression were occluded. These results support the hypothesis that p53 may be involved in transcriptional regulation during the early phase of LTP. We hope that the presented data may aid in the understanding of the contribution of p53 and related genes in the processes that are associated with synaptic plasticity.
Collapse
|