1
|
Gibbs E, Miao Q, Ferrolino M, Bajpai R, Hassan A, Phillips AH, Pitre A, Kümmerle R, Miller S, Nagy G, Leite W, Heller W, Stanley C, Perrone B, Kriwacki R. p14 ARF forms meso-scale assemblies upon phase separation with NPM1. Nat Commun 2024; 15:9531. [PMID: 39528457 PMCID: PMC11555371 DOI: 10.1038/s41467-024-53904-z] [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] [Received: 11/26/2023] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
NPM1 is an abundant nucleolar chaperone that, in addition to facilitating ribosome biogenesis, contributes to nucleolar stress responses and tumor suppression through its regulation of the p14 Alternative Reading Frame tumor suppressor protein (p14ARF). Oncogenic stress induces p14ARF to inhibit MDM2, stabilize p53 and arrest the cell cycle. Under non-stress conditions, NPM1 stabilizes p14ARF in nucleoli, preventing its degradation and blocking p53 activation. However, the mechanisms underlying the regulation of p14ARF by NPM1 are unclear because the structural features of the p14ARF-NPM1 complex were elusive. Here we show that p14ARF assembles into a gel-like meso-scale network upon phase separation with NPM1. This assembly is mediated by intermolecular contacts formed by hydrophobic residues in an α-helix and β-strands within a partially folded N-terminal portion of p14ARF. These hydrophobic interactions promote phase separation with NPM1, enhance p14ARF nucleolar partitioning, restrict NPM1 diffusion within condensates and nucleoli, and reduce cellular proliferation. Our structural analysis provides insights into the multifaceted chaperone function of NPM1 in nucleoli by mechanistically linking the nucleolar localization of p14ARF to its partial folding and meso-scale assembly upon phase separation with NPM1.
Collapse
Affiliation(s)
- Eric Gibbs
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Qi Miao
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Mylene Ferrolino
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Richa Bajpai
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Aila Hassan
- Bruker Switzerland AG, Fällanden, Switzerland
| | - Aaron H Phillips
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Aaron Pitre
- Cell and Tissue Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Shondra Miller
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Gergely Nagy
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Wellington Leite
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - William Heller
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Chris Stanley
- Computational Sciences and Engineering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | | | - Richard Kriwacki
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Sciences Center, Memphis, Tennessee, USA.
| |
Collapse
|
2
|
Zhang P, Sun C, Yin T, Guo J, Chong D, Tang Y, Liu Y, Li Y, Gu Y, Lu L. ESF1 positively regulates MDM2 and promotes tumorigenesis. Int J Biol Macromol 2024; 276:133652. [PMID: 38971273 DOI: 10.1016/j.ijbiomac.2024.133652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 06/05/2024] [Accepted: 06/20/2024] [Indexed: 07/08/2024]
Abstract
Eighteen S rRNA factor 1 (ESF1) is a predominantly nucleolar protein essential for embryogenesis. Our previous studies have suggested that Esf1 is a negative regulator of the tumor suppressor protein p53. However, it remains unclear whether ESF1 contributes to tumorigenesis. In this current research, we find that increased ESF1 expression correlates with poor survival in multiple tumors including pancreatic cancer. ESF1 is able to regulate cell proliferation, migration, DNA damage-induced apoptosis, and tumorigenesis. Mechanistically, ESF1 physically interacts with MDM2 and is essential for maintaining the stability of MDM2 protein by inhibiting its ubiquitination. Additionally, ESF1 also prevented stress-induced stabilization of p53 in multiple cancer cells. Hence, our findings suggest that ESF1 is a potent regulator of the MDM2-p53 pathway and promotes tumor progression.
Collapse
Affiliation(s)
- Pei Zhang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao, China
| | - Changning Sun
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao, China; College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Tiantian Yin
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao, China
| | - Jiang Guo
- Department of Interventional Oncology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Daochen Chong
- Pathology Department, Navy 971 Hospital of PLA, Qingdao, China
| | - Yanfei Tang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao, China
| | - Yunzhang Liu
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao, China
| | - Yun Li
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao, China
| | - Yuchao Gu
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Ling Lu
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao, China.
| |
Collapse
|
3
|
Jansen J, Bohnsack KE, Böhlken-Fascher S, Bohnsack MT, Dobbelstein M. The ribosomal protein L22 binds the MDM4 pre-mRNA and promotes exon skipping to activate p53 upon nucleolar stress. Cell Rep 2024; 43:114610. [PMID: 39116201 DOI: 10.1016/j.celrep.2024.114610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/09/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
The tumor suppressor p53 and its antagonists MDM2 and MDM4 integrate stress signaling. For instance, dysbalanced assembly of ribosomes in nucleoli induces p53. Here, we show that the ribosomal protein L22 (RPL22; eL22), under conditions of ribosomal and nucleolar stress, promotes the skipping of MDM4 exon 6. Upon L22 depletion, more full-length MDM4 is maintained, leading to diminished p53 activity and enhanced cellular proliferation. L22 binds to specific RNA elements within intron 6 of MDM4 that correspond to a stem-loop consensus, leading to exon 6 skipping. Targeted deletion of these intronic elements largely abolishes L22-mediated exon skipping and re-enables cell proliferation, despite nucleolar stress. L22 also governs alternative splicing of the L22L1 (RPL22L1) and UBAP2L mRNAs. Thus, L22 serves as a signaling intermediate that integrates different layers of gene expression. Defects in ribosome synthesis lead to specific alternative splicing, ultimately triggering p53-mediated transcription and arresting cell proliferation.
Collapse
Affiliation(s)
- Jennifer Jansen
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Katherine E Bohnsack
- Department of Molecular Biology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Susanne Böhlken-Fascher
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Markus T Bohnsack
- Department of Molecular Biology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Matthias Dobbelstein
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany.
| |
Collapse
|
4
|
Filippopoulou C, Thomé CC, Perdikari S, Ntini E, Simos G, Bohnsack KE, Chachami G. Hypoxia-driven deSUMOylation of EXOSC10 promotes adaptive changes in the transcriptome profile. Cell Mol Life Sci 2024; 81:58. [PMID: 38279024 PMCID: PMC10817850 DOI: 10.1007/s00018-023-05035-9] [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: 07/10/2023] [Revised: 10/12/2023] [Accepted: 11/06/2023] [Indexed: 01/28/2024]
Abstract
Reduced oxygen availability (hypoxia) triggers adaptive cellular responses via hypoxia-inducible factor (HIF)-dependent transcriptional activation. Adaptation to hypoxia also involves transcription-independent processes like post-translational modifications; however, these mechanisms are poorly characterized. Investigating the involvement of protein SUMOylation in response to hypoxia, we discovered that hypoxia strongly decreases the SUMOylation of Exosome subunit 10 (EXOSC10), the catalytic subunit of the RNA exosome, in an HIF-independent manner. EXOSC10 is a multifunctional exoribonuclease enriched in the nucleolus that mediates the processing and degradation of various RNA species. We demonstrate that the ubiquitin-specific protease 36 (USP36) SUMOylates EXOSC10 and we reveal SUMO1/sentrin-specific peptidase 3 (SENP3) as the enzyme-mediating deSUMOylation of EXOSC10. Under hypoxia, EXOSC10 dissociates from USP36 and translocates from the nucleolus to the nucleoplasm concomitant with its deSUMOylation. Loss of EXOSC10 SUMOylation does not detectably affect rRNA maturation but affects the mRNA transcriptome by modulating the expression levels of hypoxia-related genes. Our data suggest that dynamic modulation of EXOSC10 SUMOylation and localization under hypoxia regulates the RNA degradation machinery to facilitate cellular adaptation to low oxygen conditions.
Collapse
Affiliation(s)
- Chrysa Filippopoulou
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - Chairini C Thomé
- Department of Molecular Biology, University Medical Center Göttingen, 37073, Göttingen, Germany
| | - Sofia Perdikari
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (FORTH), 70013, Heraklion, Greece
| | - Evgenia Ntini
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (FORTH), 70013, Heraklion, Greece
| | - George Simos
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, Biopolis, 41500, Larissa, Greece
- Gerald Bronfman Department of Oncology, Faculty of Medicine, McGill University, Montreal, Canada
| | - Katherine E Bohnsack
- Department of Molecular Biology, University Medical Center Göttingen, 37073, Göttingen, Germany
| | - Georgia Chachami
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, Biopolis, 41500, Larissa, Greece.
| |
Collapse
|
5
|
Zhou Y, Nakajima R, Shirasawa M, Fikriyanti M, Zhao L, Iwanaga R, Bradford AP, Kurayoshi K, Araki K, Ohtani K. Expanding Roles of the E2F-RB-p53 Pathway in Tumor Suppression. BIOLOGY 2023; 12:1511. [PMID: 38132337 PMCID: PMC10740672 DOI: 10.3390/biology12121511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/03/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
The transcription factor E2F links the RB pathway to the p53 pathway upon loss of function of pRB, thereby playing a pivotal role in the suppression of tumorigenesis. E2F fulfills a major role in cell proliferation by controlling a variety of growth-associated genes. The activity of E2F is controlled by the tumor suppressor pRB, which binds to E2F and actively suppresses target gene expression, thereby restraining cell proliferation. Signaling pathways originating from growth stimulative and growth suppressive signals converge on pRB (the RB pathway) to regulate E2F activity. In most cancers, the function of pRB is compromised by oncogenic mutations, and E2F activity is enhanced, thereby facilitating cell proliferation to promote tumorigenesis. Upon such events, E2F activates the Arf tumor suppressor gene, leading to activation of the tumor suppressor p53 to protect cells from tumorigenesis. ARF inactivates MDM2, which facilitates degradation of p53 through proteasome by ubiquitination (the p53 pathway). P53 suppresses tumorigenesis by inducing cellular senescence or apoptosis. Hence, in almost all cancers, the p53 pathway is also disabled. Here we will introduce the canonical functions of the RB-E2F-p53 pathway first and then the non-classical functions of each component, which may be relevant to cancer biology.
Collapse
Affiliation(s)
- Yaxuan Zhou
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| | - Rinka Nakajima
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| | - Mashiro Shirasawa
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| | - Mariana Fikriyanti
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| | - Lin Zhao
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| | - Ritsuko Iwanaga
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA; (R.I.); (A.P.B.)
| | - Andrew P. Bradford
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA; (R.I.); (A.P.B.)
| | - Kenta Kurayoshi
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
| | - Keigo Araki
- Department of Morphological Biology, Ohu University School of Dentistry, 31-1 Misumido Tomitamachi, Koriyama, Fukushima 963-8611, Japan;
| | - Kiyoshi Ohtani
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| |
Collapse
|
6
|
Gibbs E, Miao Q, Ferrolino M, Bajpai R, Hassan A, Phillips AH, Pitre A, Kümmerle R, Miller S, Heller W, Stanley C, Perrone B, Kriwacki R. p14 ARF forms meso-scale assemblies upon phase separation with NPM1. RESEARCH SQUARE 2023:rs.3.rs-3592059. [PMID: 38106181 PMCID: PMC10723529 DOI: 10.21203/rs.3.rs-3592059/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
NPM1 is an abundant nucleolar chaperone that, in addition to facilitating ribosome biogenesis, contributes to nucleolar stress responses and tumor suppression through its regulation of the p14 Alternative Reading Frame tumor suppressor protein (p14ARF). Oncogenic stress induces p14ARF to inhibit MDM2, stabilize p53 and arrest the cell cycle. Under non-stress conditions, NPM1 stabilizes p14ARF in nucleoli, preventing its degradation and blocking p53 activation. However, the mechanisms underlying the regulation of p14ARF by NPM1 are unclear because the structural features of the p14ARF-NPM1 complex remain elusive. Here we show that NPM1 sequesters p14ARF within phase-separated condensates, facilitating the assembly of p14ARF into a gel-like meso-scale network. This assembly is mediated by intermolecular contacts formed by hydrophobic residues in an α-helix and β-strands within a partially folded N-terminal domain of p14ARF. Those hydrophobic interactions promote phase separation with NPM1, enhance nucleolar partitioning of p14ARF, restrict p14ARF and NPM1 diffusion within condensates and in nucleoli, and reduce cell viability. Our structural model provides novel insights into the multifaceted chaperone function of NPM1 in nucleoli by mechanistically linking the nucleolar localization of p14ARF to its partial folding and meso-scale assembly upon phase separation with NPM1.
Collapse
Affiliation(s)
- Eric Gibbs
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Qi Miao
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Mylene Ferrolino
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Richa Bajpai
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Aila Hassan
- Bruker Switzerland AG, Fällanden, Switzerland
| | - Aaron H. Phillips
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Aaron Pitre
- Cell and Tissue Imaging Shared Resource, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | | | - Shondra Miller
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - William Heller
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Chris Stanley
- Computational Sciences and Engineering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | | | - Richard Kriwacki
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Sciences Center, Memphis, Tennessee, USA
| |
Collapse
|
7
|
Zhan Q, Zhang H, Wu B, Zhang N, Zhang L. E3 ubiquitin ligases in the acute leukemic signaling pathways. Front Physiol 2022; 13:1004330. [PMID: 36439256 PMCID: PMC9691902 DOI: 10.3389/fphys.2022.1004330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/28/2022] [Indexed: 11/13/2022] Open
Abstract
Acute leukemia is a common hematologic tumor with highly genetic heterogeneity, and many factors are involved in the pathogenesis and drug-resistance mechanism. Emerging evidence proves that E3 ubiquitin ligases participate in the acute leukemic signaling pathways via regulating substrates. This review summarized the E3 ligases which can affect the leukemic signal. It is worth noting that the abnormal signal is often caused by a deficiency or a mutation of the E3 ligases. In view of this phenomenon, we envisioned perspectives associated with targeted agonists of E3 ligases and proteolysis-targeting chimera technology. Moreover, we emphasized the significance of research into the upstream factors regulating the expression of E3 ubiquitin ligases. It is expected that the understanding of the mechanism of leukemic signaling pathways with which that E3 ligases are involved will be beneficial to accelerating the process of therapeutic strategy improvement for acute leukemia.
Collapse
Affiliation(s)
- Qianru Zhan
- Department of Hematology, The First Hospital of China Medical University, Shenyang, China
| | - Heyang Zhang
- Department of Hematology, The First Hospital of China Medical University, Shenyang, China
| | - Boquan Wu
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Naijin Zhang
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
- *Correspondence: Lijun Zhang, ; Naijin Zhang,
| | - Lijun Zhang
- Department of Hematology, The First Hospital of China Medical University, Shenyang, China
- *Correspondence: Lijun Zhang, ; Naijin Zhang,
| |
Collapse
|
8
|
Damasio MPS, Nascimento CS, Andrade LM, de Oliveira VL, Calzavara-Silva CE. The role of T-cells in head and neck squamous cell carcinoma: From immunity to immunotherapy. Front Oncol 2022; 12:1021609. [PMID: 36338731 PMCID: PMC9632296 DOI: 10.3389/fonc.2022.1021609] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/10/2022] [Indexed: 12/24/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) encompass a group of complex entities of tumours affecting the aerodigestive upper tract. The main risk factors are strongly related to tobacco and alcohol consumption, but also HPV infection is often associated. Surgery, radiotherapy and/or chemotherapy are the standard treatments, though the 5-year overall survival is less than 50%. The advances in genomics, molecular medicine, immunology, and nanotechnology have shed a light on tumour biology which helps clinical researchers to obtain more efficacious and less toxic therapies. Head and neck tumours possess different immune escape mechanisms including diminishing the immune response through modulating immune checkpoints, in addition to the recruitment and differentiation of suppressive immune cells. The insights into the HNSCC biology and its strong interaction with the tumour microenvironment highlights the role of immunomodulating agents. Recently, the knowledge of the immunological features of these tumours has paved the way for the discovery of effective biomarkers that allow a better selection of patients with odds of improving overall survival through immunotherapy. Specially biomarkers regarding immune checkpoint inhibitors antibodies, such as anti-PD-1/PD-L1 and anti-CTLA-4 in combination with standard therapy or as monotherapy. New immunotherapies to treat head and neck cancer carcinomas, such as CAR T cells and nanoparticles have been the center of attention and in this review, we discuss the necessity of finding targets for the T cell in the cancer cells to generate CAR T cells, but also the relevance of evaluating specificity and safety of those therapies.
Collapse
Affiliation(s)
- Marcos Paulo S. Damasio
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Camila Sales Nascimento
- Grupo de pesquisa em Imunologia Celular e Molecular, Fundação Oswaldo Cruz, Instituto Rene Rachou, Belo Horizonte, MG, Brazil
| | - Lidia M. Andrade
- Departamento de Genética, Ecologia e Evolução, Departamento de Física, Nanobiomedical Research Group, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vivian L. de Oliveira
- Universidade Federal do ABC, Centro de Ciências Naturais e Humanas, São Paulo, Brazil
- Laboratório de Imunologia, LIM19, Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
| | - Carlos Eduardo Calzavara-Silva
- Grupo de pesquisa em Imunologia Celular e Molecular, Fundação Oswaldo Cruz, Instituto Rene Rachou, Belo Horizonte, MG, Brazil
| |
Collapse
|
9
|
Nucleolus and Nucleolar Stress: From Cell Fate Decision to Disease Development. Cells 2022; 11:cells11193017. [PMID: 36230979 PMCID: PMC9563748 DOI: 10.3390/cells11193017] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/19/2022] [Accepted: 09/22/2022] [Indexed: 11/30/2022] Open
Abstract
Besides the canonical function in ribosome biogenesis, there have been significant recent advances towards the fascinating roles of the nucleolus in stress response, cell destiny decision and disease progression. Nucleolar stress, an emerging concept describing aberrant nucleolar structure and function as a result of impaired rRNA synthesis and ribosome biogenesis under stress conditions, has been linked to a variety of signaling transductions, including but not limited to Mdm2-p53, NF-κB and HIF-1α pathways. Studies have uncovered that nucleolus is a stress sensor and signaling hub when cells encounter various stress conditions, such as nutrient deprivation, DNA damage and oxidative and thermal stress. Consequently, nucleolar stress plays a pivotal role in the determination of cell fate, such as apoptosis, senescence, autophagy and differentiation, in response to stress-induced damage. Nucleolar homeostasis has been involved in the pathogenesis of various chronic diseases, particularly tumorigenesis, neurodegenerative diseases and metabolic disorders. Mechanistic insights have revealed the indispensable role of nucleolus-initiated signaling in the progression of these diseases. Accordingly, the intervention of nucleolar stress may pave the path for developing novel therapies against these diseases. In this review, we systemically summarize recent findings linking the nucleolus to stress responses, signaling transduction and cell-fate decision, set the spotlight on the mechanisms by which nucleolar stress drives disease progression, and highlight the merit of the intervening nucleolus in disease treatment.
Collapse
|
10
|
Obaidi I, Blanco Fernández A, McMorrow T. Curcumin Sensitises Cancerous Kidney Cells to TRAIL Induced Apoptosis via Let-7C Mediated Deregulation of Cell Cycle Proteins and Cellular Metabolism. Int J Mol Sci 2022; 23:ijms23179569. [PMID: 36076967 PMCID: PMC9455736 DOI: 10.3390/ijms23179569] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 12/31/2022] Open
Abstract
Targeted therapies are the most attractive options in the treatment of different tumours, including kidney cancers. Such therapies have entered a golden era due to advancements in research, breakthroughs in scientific knowledge, and a better understanding of cancer therapy mechanisms, which significantly improve the survival rates and life expectancy of patients. The use of tumour necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL) as an anticancer therapy has attracted the attention of the scientific community and created great excitement due to its selectivity in targeting cancerous cells with no toxic impacts on normal tissues. However, clinical studies disappointingly showed the emergence of resistance against TRAIL. This study aimed to employ curcumin to sensitise TRAIL-resistant kidney cancerous ACHN cells, as well as to gain insight into the molecular mechanisms of TRAIL sensitization. Curcumin deregulated the expression of apoptosis-regulating micro Ribonucleic Acid (miRNAs), most notably, let-7C. Transfecting ACHN cells with a let-7C antagomir significantly increased the expression of several cell cycle protein, namely beta (β)-catenin, cyclin dependent kinase (CDK)1/2/4/6 and cyclin B/D. Further, it overexpressed the expression of the two key glycolysis regulating proteins including hypoxia-inducible factor 1-alpha (HIF-1α) and pyruvate dehydrogenase kinase 1 (PDK1). Curcumin also suppressed the expression of the overexpressed proteins when added to the antagomir transfected cells. Overall, curcumin targeted ACHN cell cycle and cellular metabolism by promoting the differential expression of let-7C. To the best of our knowledge, this is the first study to mechanistically report the cancer chemosensitisation potential of curcumin in kidney cancer cells via induction of let-7C.
Collapse
Affiliation(s)
- Ismael Obaidi
- NatPro Centre for Natural Product Research, School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, D02 W272 Dublin, Ireland
- College of Pharmacy, University of Babylon, Babylon 51002, Iraq
- Correspondence: (I.O.); (T.M.); Tel.: +353-8-6064-2626 (I.O.); +353-1-716-2317 (ext. 6819) (T.M.)
| | - Alfonso Blanco Fernández
- Flow Cytometry Core Technology, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Tara McMorrow
- Centre for Toxicology, School of Biomedical and Biomolecular Sciences, Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
- Correspondence: (I.O.); (T.M.); Tel.: +353-8-6064-2626 (I.O.); +353-1-716-2317 (ext. 6819) (T.M.)
| |
Collapse
|
11
|
Kowalczyk D, Nakasone MA, Smith BO, Huang DT. Bivalent binding of p14ARF to MDM2 RING and acidic domains inhibits E3 ligase function. Life Sci Alliance 2022; 5:e202201472. [PMID: 35944929 PMCID: PMC9366199 DOI: 10.26508/lsa.202201472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 12/29/2022] Open
Abstract
ARF tumor suppressor protein is a key regulator of the MDM2-p53 signaling axis. ARF interferes with MDM2-mediated ubiquitination and degradation of p53 by sequestering MDM2 in the nucleolus and preventing MDM2-p53 interaction and nuclear export of p53. Moreover, ARF also directly inhibits MDM2 ubiquitin ligase (E3) activity, but the mechanism remains elusive. Here, we apply nuclear magnetic resonance and biochemical analyses to uncover the mechanism of ARF-mediated inhibition of MDM2 E3 activity. We show that MDM2 acidic and zinc finger domains (AD-ZnF) form a weak intramolecular interaction with the RING domain, where the binding site overlaps with the E2∼ubiquitin binding surface and thereby partially reduces MDM2 E3 activity. Binding of human N-terminal 32 residues of p14ARF to the acidic domain of MDM2 strengthens the AD-ZnF-RING domain interaction. Furthermore, the N-terminal RxFxV motifs of p14ARF participate directly in the MDM2 RING domain interaction. This bivalent binding mode of p14ARF to MDM2 acidic and RING domains restricts E2∼ubiquitin recruitment and massively hinders MDM2 E3 activity. These findings elucidate the mechanism by which ARF inhibits MDM2 E3 activity.
Collapse
Affiliation(s)
| | | | - Brian O Smith
- Institute of Molecular Cell and System Biology, University of Glasgow, Glasgow, UK
| | - Danny T Huang
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
12
|
Huang X, Wang B, Shen H, Huang D, Shi G. Farnesoid X receptor functions in cervical cancer via the p14 ARF-mouse double minute 2-p53 pathway. Mol Biol Rep 2022; 49:3617-3625. [PMID: 35347542 PMCID: PMC9174312 DOI: 10.1007/s11033-022-07201-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/25/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Cervical cancer is the second most common cancer among women living in developing countries. Farnesoid X receptor (FXR) is a member of the nuclear receptor family, which regulates the development and proliferation of cancer. However, the role of and molecular mechanism by which FXR acts in cervical cancer are still unknown. METHODS AND RESULTS The relationship between FXR and the proliferation of cervical cancer cell lines was detected by MTT and colony formation assays. Immunohistochemistry was used to detect the expression of FXR in cervical cancer tissue slides. Western blotting was used to detect the expression of p14ARF, mouse double minute 2 (MDM2) and p53 when FXR was overexpressed or siRNA was applied. Western blotting was used to detect the expression of MDM2 and p53 when pifithrin-α (PFT-α) was applied. FXR activation inhibited the proliferation of cervical cancer cell lines. FXR was significantly decreased in cervical squamous cell carcinoma, which was correlated with TNM stage, but not with metastasis. Overexpression of FXR activated the p14ARF-MDM2-p53 pathway. As a p53 inhibitor, PFT-α increased MDM2 in Lenti-vector cells, but had no effect on MDM2 in Lenti-FXR cells. CONCLUSIONS FXR inhibits cervical cancer by upregulating the p14ARF-MDM2-p53 pathway. Activation of FXR may be a potential strategy for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Xiaohua Huang
- Department of Pediatrics, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Bin Wang
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Huimin Shen
- Department of Neurology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Danmei Huang
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Ganggang Shi
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| |
Collapse
|
13
|
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
|
14
|
Rathi A, Kumar D, Hasan GM, Haque MM, Hassan MI. Therapeutic targeting of PIM KINASE signaling in cancer therapy: Structural and clinical prospects. Biochim Biophys Acta Gen Subj 2021; 1865:129995. [PMID: 34455019 DOI: 10.1016/j.bbagen.2021.129995] [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: 05/25/2021] [Revised: 07/28/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND PIM kinases are well-studied drug targets for cancer, belonging to Serine/Threonine kinases family. They are the downstream target of various signaling pathways, and their up/down-regulation affects various physiological processes. PIM family comprises three isoforms, namely, PIM-1, PIM-2, and PIM-3, on alternative initiation of translation and they have different levels of expression in different types of cancers. Its structure shows a unique ATP-binding site in the hinge region which makes it unique among other kinases. SCOPE OF REVIEW PIM kinases are widely reported in hematological malignancies along with prostate and breast cancers. Currently, many drugs are used as inhibitors of PIM kinases. In this review, we highlighted the physiological significance of PIM kinases in the context of disease progression and therapeutic targeting. We comprehensively reviewed the PIM kinases in terms of their expression and regulation of different physiological roles. We further predicted functional partners of PIM kinases to elucidate their role in the cellular physiology of different cancer and mapped their interaction network. MAJOR CONCLUSIONS A deeper mechanistic insight into the PIM signaling involved in regulating different cellular processes, including transcription, apoptosis, cell cycle regulation, cell proliferation, cell migration and senescence, is provided. Furthermore, structural features of PIM have been dissected to understand the mechanism of inhibition and subsequent implication of designed inhibitors towards therapeutic management of prostate, breast and other cancers. GENERAL SIGNIFICANCE Being a potential drug target for cancer therapy, available drugs and PIM inhibitors at different stages of clinical trials are discussed in detail.
Collapse
Affiliation(s)
- Aanchal Rathi
- Department of Biotechnology, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Dhiraj Kumar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | | | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
15
|
Xue M, Liu X, Cheng B, Rui X, Wu M, Lv J. Epigallocatechin Gallate Enhances Inhibition Effect of DDP on the Proliferation of Gastric Cancer BGC-823 Cells by Regulating p19Arf-p53-p21Cip1 Signaling Pathway. Asian Pac J Cancer Prev 2021; 22:1263-1270. [PMID: 33906321 PMCID: PMC8325134 DOI: 10.31557/apjcp.2021.22.4.1263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/17/2021] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE To indicate the effect of Epigallocatechin gallate (EGCG) and Cisplatin (DDP) on proliferation of gastric cancer BGC-823 cells and the relative underlying mechanism. METHODS Cultured BGC-823 cells were treated by 5 μg/mL DDP, 25 μg/mL EGCG and combined 5 μg/mL DDP with 25 μg/mL EGCG, a blank group was used as control. Cell morphology was observed by 4',6-diamidino-2-phenylindole (DAPI) staining. The ability of cell proliferation was detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)assay. The cell cloning rate was determined by colony formation assay. The ability of cell migration was detected by cell scratch test. The cell cycle distributions and apoptosis were analyzed by flow cytometry, The expression of p19Arf, p53, p21Cip1 mRNA was determined by RT-qPCR. The protein levels of p19Arf, p53, p21Cip1 were measured by Western blot. RESULTS Compared with DDP or EGCG treatment alone, EGCG combined with DDP treatment significantly caused nuclear shrinkage, reduced the proliferation rate, the ability of cell clone and migration. EGCG combined with DDP treatment caused cell cycle arrest in G1 phase in BGC-823 cells, increase of apoptosis (21.3%) vs EGCG (7.25%) and DDP (3.86%) single-use group (p <0.01), up-regulated gene and protein expressions of p19Arf, p53, p21Cip1 (p <0.01). CONCLUSION EGCG can enhance the effect of DDP on inhibiting BGC-823 cell proliferation and inducing apoptosis via activating the p19Arf-p53-p21Cip1 signaling pathway.
Collapse
Affiliation(s)
- Mengya Xue
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China.
| | - XiaoLv Liu
- Wuhu Sub-center of Anhui International Travel Health Care Center, Wuhu, China.
| | - Bing Cheng
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China.
| | - XingKai Rui
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China.
| | - MingCai Wu
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China.
| | - Jun Lv
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China.
| |
Collapse
|
16
|
Sok V, Jacinto AZ, Peng N, Eldemerdash M, Le L, Tran PD, Feng LF, Patel JR, Gi M, Ammon JC, So CH. G protein coupled receptor kinase 5 modifies the nucleolar stress response activated by actinomycin D. Biochem Cell Biol 2021; 99:508-518. [PMID: 33507833 DOI: 10.1139/bcb-2020-0480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
G protein coupled receptor kinase 5 (GRK5) is localized within the nucleus and moderates functions such as DNA transcription, in addition to its localization at the plasma membrane. In this report, we show that GRK5 modifies the nucleolar stress response activated by the DNA polymerase inhibitor, actinomycin D (ActD). We show an increased sensitivity to the apoptotic effects of ActD on cervical HeLa cells and the breast cancer cell line MDA MB 231 with reduced protein expression of GRK5. We also tested two types of breast cancer cells (MDA MB 231 and MCF7 cells) and found that the rate of response to ActD varied between them because they have innate differences in the protein expression of GRK5. We also found that GRK5 phosphorylates nucleophosmin (NPM1) at T199 before and during the early stages of ActD treatment. Phosphorylation at T199 increases the ability of NPM1 to interact with p14ARF in vitro, which may affect the protein expression levels of p14ARF. We found that the expression levels of p14ARF were lower in the cells transfected with the control shRNA, but higher in cells transfected with GRK5 shRNA. Collectively, this suggests that GRK5 modifies the nucleolar stress response associated with ActD.
Collapse
Affiliation(s)
- Vanessa Sok
- Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA.,Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA
| | - Alec Z Jacinto
- Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA.,Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA
| | - Natalie Peng
- Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA.,Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA
| | - Mohamed Eldemerdash
- Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA.,Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA
| | - Lysa Le
- Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA.,Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA
| | - Philip D Tran
- Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA.,Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA
| | - Li Feng Feng
- Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA.,Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA
| | - Jigisha R Patel
- Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA.,Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA
| | - Michael Gi
- Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA.,Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA
| | - Jane C Ammon
- Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA.,Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA
| | - Christopher H So
- Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA.,Roseman University of Health Sciences, School of Pharmacy, Henderson, NV 89014, USA
| |
Collapse
|
17
|
Dutta Gupta S, Pan CH. Recent update on discovery and development of Hsp90 inhibitors as senolytic agents. Int J Biol Macromol 2020; 161:1086-1098. [DOI: 10.1016/j.ijbiomac.2020.06.115] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/22/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023]
|
18
|
Piazzi M, Bavelloni A, Gallo A, Faenza I, Blalock WL. Signal Transduction in Ribosome Biogenesis: A Recipe to Avoid Disaster. Int J Mol Sci 2019; 20:ijms20112718. [PMID: 31163577 PMCID: PMC6600399 DOI: 10.3390/ijms20112718] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 12/27/2022] Open
Abstract
Energetically speaking, ribosome biogenesis is by far the most costly process of the cell and, therefore, must be highly regulated in order to avoid unnecessary energy expenditure. Not only must ribosomal RNA (rRNA) synthesis, ribosomal protein (RP) transcription, translation, and nuclear import, as well as ribosome assembly, be tightly controlled, these events must be coordinated with other cellular events, such as cell division and differentiation. In addition, ribosome biogenesis must respond rapidly to environmental cues mediated by internal and cell surface receptors, or stress (oxidative stress, DNA damage, amino acid depletion, etc.). This review examines some of the well-studied pathways known to control ribosome biogenesis (PI3K-AKT-mTOR, RB-p53, MYC) and how they may interact with some of the less well studied pathways (eIF2α kinase and RNA editing/splicing) in higher eukaryotes to regulate ribosome biogenesis, assembly, and protein translation in a dynamic manner.
Collapse
Affiliation(s)
- Manuela Piazzi
- Istituto di Genetica Molecolare-Luigi Luca Cavalli Sforza, UOS Bologna, Consiglio Nazionale delle Ricerche (IGM-CNR), 40136 Bologna, Italy.
- IRCCS, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | | | - Angela Gallo
- RNA Editing Laboratory, Dipartimento di Oncoematologia, IRCCS, Ospedale Pediatrica Bambino Gesù, 00146 Rome, Italy.
| | - Irene Faenza
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, 40126 Bologna, Italy.
| | - William L Blalock
- Istituto di Genetica Molecolare-Luigi Luca Cavalli Sforza, UOS Bologna, Consiglio Nazionale delle Ricerche (IGM-CNR), 40136 Bologna, Italy.
- IRCCS, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| |
Collapse
|
19
|
Fontana R, Ranieri M, La Mantia G, Vivo M. Dual Role of the Alternative Reading Frame ARF Protein in Cancer. Biomolecules 2019; 9:E87. [PMID: 30836703 PMCID: PMC6468759 DOI: 10.3390/biom9030087] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 02/07/2023] Open
Abstract
The CDKN2a/ARF locus expresses two partially overlapping transcripts that encode two distinct proteins, namely p14ARF (p19Arf in mouse) and p16INK4a, which present no sequence identity. Initial data obtained in mice showed that both proteins are potent tumor suppressors. In line with a tumor-suppressive role, ARF-deficient mice develop lymphomas, sarcomas, and adenocarcinomas, with a median survival rate of one year of age. In humans, the importance of ARF inactivation in cancer is less clear whereas a more obvious role has been documented for p16INK4a. Indeed, many alterations in human tumors result in the elimination of the entire locus, while the majority of point mutations affect p16INK4a. Nevertheless, specific mutations of p14ARF have been described in different types of human cancers such as colorectal and gastric carcinomas, melanoma and glioblastoma. The activity of the tumor suppressor ARF has been shown to rely on both p53-dependent and independent functions. However, novel data collected in the last years has challenged the traditional and established role of this protein as a tumor suppressor. In particular, tumors retaining ARF expression evolve to metastatic and invasive phenotypes and in humans are associated with a poor prognosis. In this review, the recent evidence and the molecular mechanisms of a novel role played by ARF will be presented and discussed, both in pathological and physiological contexts.
Collapse
Affiliation(s)
- Rosa Fontana
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Michela Ranieri
- Division of Hematology and Medical Oncology, Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY 10016, USA.
| | - Girolama La Mantia
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy.
| | - Maria Vivo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy.
| |
Collapse
|
20
|
McWilliams RR, Wieben ED, Chaffee KG, Antwi SO, Raskin L, Olopade OI, Li D, Highsmith WE, Colon-Otero G, Khanna LG, Permuth JB, Olson JE, Frucht H, Genkinger J, Zheng W, Blot WJ, Wu L, Almada LL, Fernandez-Zapico ME, Sicotte H, Pedersen KS, Petersen GM. CDKN2A Germline Rare Coding Variants and Risk of Pancreatic Cancer in Minority Populations. Cancer Epidemiol Biomarkers Prev 2018; 27:1364-1370. [PMID: 30038052 PMCID: PMC6214745 DOI: 10.1158/1055-9965.epi-17-1065] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 02/13/2018] [Accepted: 07/11/2018] [Indexed: 12/20/2022] Open
Abstract
Background: Pathogenic germline mutations in the CDKN2A tumor suppressor gene are rare and associated with highly penetrant familial melanoma and pancreatic cancer in non-Hispanic whites (NHW). To date, the prevalence and impact of CDKN2A rare coding variants (RCV) in racial minority groups remain poorly characterized. We examined the role of CDKN2A RCVs on the risk of pancreatic cancer among minority subjects.Methods: We sequenced CDKN2A in 220 African American (AA) pancreatic cancer cases, 900 noncancer AA controls, and 183 Nigerian controls. RCV frequencies were determined for each group and compared with that of 1,537 NHW patients with pancreatic cancer. Odds ratios (OR) and 95% confidence intervals (CI) were calculated for both a case-case comparison of RCV frequencies in AAs versus NHWs, and case-control comparison between AA cases versus noncancer AA controls plus Nigerian controls. Smaller sets of Hispanic and Native American cases and controls also were sequenced.Results: One novel missense RCV and one novel frameshift RCV were found among AA patients: 400G>A and 258_278del. RCV carrier status was associated with increased risk of pancreatic cancer among AA cases (11/220; OR, 3.3; 95% CI, 1.5-7.1; P = 0.004) compared with AA and Nigerian controls (17/1,083). Further, AA cases had higher frequency of RCVs: 5.0% (OR, 13.4; 95% CI, 4.9-36.7; P < 0.001) compared with NHW cases (0.4%).Conclusions: CDKN2A RCVs are more common in AA than in NHW patients with pancreatic cancer and associated with moderately increased pancreatic cancer risk among AAs.Impact: RCVs in CDKN2A are frequent in AAs and are associated with risk for pancreatic cancer. Cancer Epidemiol Biomarkers Prev; 27(11); 1364-70. ©2018 AACR.
Collapse
Affiliation(s)
| | - Eric D Wieben
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Kari G Chaffee
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Samuel O Antwi
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida
| | - Leon Raskin
- Division of Epidemiology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Olufunmilayo I Olopade
- Departments of Medicine and Human Genetics, University of Chicago Medical Center, Chicago, Illinois
| | - Donghui Li
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - W Edward Highsmith
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Gerardo Colon-Otero
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Jacksonville, Florida
| | - Lauren G Khanna
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Jennifer B Permuth
- Departments of Cancer Epidemiology and Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Janet E Olson
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Harold Frucht
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Jeanine Genkinger
- Department of Epidemiology, Columbia University Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, New York, New York
| | - Wei Zheng
- Division of Epidemiology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - William J Blot
- Division of Epidemiology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Lang Wu
- Division of Epidemiology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Luciana L Almada
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, Minnesota
| | - Martin E Fernandez-Zapico
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, Minnesota
| | - Hugues Sicotte
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | - Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
21
|
Fontana R, Vivo M. Dynamics of p14ARF and Focal Adhesion Kinase-Mediated Autophagy in Cancer. Cancers (Basel) 2018; 10:cancers10070221. [PMID: 29966311 PMCID: PMC6071150 DOI: 10.3390/cancers10070221] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 06/22/2018] [Accepted: 06/26/2018] [Indexed: 12/23/2022] Open
Abstract
It has been widely shown that the focal adhesion kinase (FAK) is involved in nearly every aspect of cancer, from invasion to metastasis to epithelial–mesenchymal transition and maintenance of cancer stem cells. FAK has been shown to interact with p14ARF (alternative reading frame)—a well-established tumor suppressor—and functions in the negative regulation of cancer through both p53-dependent and -independent pathways. Interestingly, both FAK and ARF (human and mouse counterpart) proteins, as well as p53, are involved in autophagy—a process of “self-digestion”—whose main function is the recycling of cellular components and quality control of proteins and organelles. In the last years, an unexpected role of p14ARF in the survival of cancer cells has been underlined in different cellular contexts, suggesting a novel pro-oncogenic function of this protein. In this review, the mechanisms whereby ARF and FAK control autophagy are presented, as well as the role of autophagy in cell migration and spreading. Integrated investigation of these cell functions is extremely important to understand the mechanism of the basis of cell transformation and migration and thus cancer development.
Collapse
Affiliation(s)
- Rosa Fontana
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy.
| | - Maria Vivo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy.
| |
Collapse
|
22
|
Moulder DE, Hatoum D, Tay E, Lin Y, McGowan EM. The Roles of p53 in Mitochondrial Dynamics and Cancer Metabolism: The Pendulum between Survival and Death in Breast Cancer? Cancers (Basel) 2018; 10:cancers10060189. [PMID: 29890631 PMCID: PMC6024909 DOI: 10.3390/cancers10060189] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/01/2018] [Accepted: 06/05/2018] [Indexed: 12/29/2022] Open
Abstract
Cancer research has been heavily geared towards genomic events in the development and progression of cancer. In contrast, metabolic regulation, such as aberrant metabolism in cancer, is poorly understood. Alteration in cellular metabolism was once regarded simply as a consequence of cancer rather than as playing a primary role in cancer promotion and maintenance. Resurgence of cancer metabolism research has identified critical metabolic reprogramming events within biosynthetic and bioenergetic pathways needed to fulfill the requirements of cancer cell growth and maintenance. The tumor suppressor protein p53 is emerging as a key regulator of metabolic processes and metabolic reprogramming in cancer cells—balancing the pendulum between cell death and survival. This review provides an overview of the classical and emerging non-classical tumor suppressor roles of p53 in regulating mitochondrial dynamics: mitochondrial engagement in cell death processes in the prevention of cancer. On the other hand, we discuss p53 as a key metabolic switch in cellular function and survival. The focus is then on the conceivable roles of p53 in breast cancer metabolism. Understanding the metabolic functions of p53 within breast cancer metabolism will, in due course, reveal critical metabolic hotspots that cancers advantageously re-engineer for sustenance. Illustration of these events will pave the way for finding novel therapeutics that target cancer metabolism and serve to overcome the breast cancer burden.
Collapse
Affiliation(s)
- David E Moulder
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo NSW 2007, Australia.
| | - Diana Hatoum
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo NSW 2007, Australia.
| | - Enoch Tay
- Viral Hepatitis Pathogenesis Group, The Westmead Institute for Medical Research, University of Sydney, 176 Hawkesbury Road, Westmead NSW 2145, Australia.
| | - Yiguang Lin
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo NSW 2007, Australia.
| | - Eileen M McGowan
- Central Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China.
| |
Collapse
|
23
|
Abstract
The nucleolus is a prominent subnuclear compartment, where ribosome biosynthesis takes place. Recently, the nucleolus has gained attention for its novel role in the regulation of cellular stress. Nucleolar stress is emerging as a new concept, which is characterized by diverse cellular insult-induced abnormalities in nucleolar structure and function, ultimately leading to activation of p53 or other stress signaling pathways and alterations in cell behavior. Despite a number of comprehensive reviews on this concept, straightforward and clear-cut way criteria for a nucleolar stress state, regarding the factors that elicit this state, the morphological and functional alterations as well as the rationale for p53 activation are still missing. Based on literature of the past two decades, we herein summarize the evolution of the concept and provide hallmarks of nucleolar stress. Along with updated information and thorough discussion of existing confusions in the field, we pay particular attention to the current understanding of the sensing mechanisms, i.e., how stress is integrated by p53. In addition, we propose our own emphasis regarding the role of nucleolar protein NPM1 in the hallmarks of nucleolar stress and sensing mechanisms. Finally, the links of nucleolar stress to human diseases are briefly and selectively introduced.
Collapse
Affiliation(s)
- Kai Yang
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.,Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Jie Yang
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Jing Yi
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| |
Collapse
|
24
|
PKC Dependent p14ARF Phosphorylation on Threonine 8 Drives Cell Proliferation. Sci Rep 2018; 8:7056. [PMID: 29728595 PMCID: PMC5935756 DOI: 10.1038/s41598-018-25496-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 04/24/2018] [Indexed: 01/11/2023] Open
Abstract
ARF role as tumor suppressor has been challenged in the last years by several findings of different groups ultimately showing that its functions can be strictly context dependent. We previously showed that ARF loss in HeLa cells induces spreading defects, evident as rounded morphology of depleted cells, accompanied by a decrease of phosphorylated Focal Adhesion Kinase (FAK) protein levels and anoikis. These data, together with previous finding that a PKC dependent signalling pathway can lead to ARF stabilization, led us to the hypothesis that ARF functions in cell proliferation might be regulated by phosphorylation. In line with this, we show here that upon spreading ARF is induced through PKC activation. A constitutive-phosphorylated ARF mutant on the conserved threonine 8 (T8D) is able to mediate both cell spreading and FAK activation. Finally, ARF-T8D expression confers growth advantage to cells thus leading to the intriguing hypothesis that ARF phosphorylation could be a mechanism through which pro-proliferative or anti proliferative signals could be transduced inside the cells in both physiological and pathological conditions.
Collapse
|
25
|
[Shaoyangzhugu Formula regulates p19 Arf-p53-p21 Cip1 signaling pathway to ameliorate cartilage degeneration in aged cynomolgus monkeys with knee osteoarthritis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38. [PMID: 29643043 PMCID: PMC6744164 DOI: 10.3969/j.issn.1673-4254.2018.03.17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To study the effect of Shaoyangzhugu (SYZG) Formula (a formula consisting of 9 traditional Chinese drugs) in delaying the degeneration of articular cartilage and the role p19Arf-p53-p21Cip1 signaling pathway in mediating this effect. METHOD Thirteen aged cynomolgus monkeys with degenerative knee joints were selected based on X-ray findings, and one of them was randomly selected for pathological observation. The other monkeys were randomized equally into SYZG Formula group (treated with SYZG decoction), ammonia moxime group and saline group. All the monkeys were sacrificed after 8 weeks of treatment with intragastric administration of the drugs or saline. The pathology in the knee joint articular cartilage was observed and the mRNA and protein expressions of p19Arf, p53, and p21Cip1 in the articular cartilage were detected using RT-qPCR and Western blotting. RESULTS The pathological findings of the articular cartilage in old cynomolgus monkeys were consistent with the characteristics of knee osteoarthritis (KOA). Mankin scores of the cynomolgus monkeys were 7.38∓0.52 in SYZG Formula group, 7.88∓0.83 in ammonia moxime group, and 8.38∓0.74 in saline group, showing a significant difference between SYZG Formula group and saline group (P<0.05). The expressions of p19Arf, p53, and p21Cip1 were the lowest in SYZG Formula group and the highest in saline group with significant differences among the 3 groups (P<0.05). CONCLUSION SYZG Formula can delay chondrocyte senescence by regulating p19Arf-p53-p21Cip1 signaling pathway to delay articular cartilage degeneration in aged cynomolgus monkeys.
Collapse
|
26
|
p14ARF interacts with the focal adhesion kinase and protects cells from anoikis. Oncogene 2017; 36:4913-4928. [PMID: 28436949 PMCID: PMC5582215 DOI: 10.1038/onc.2017.104] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 03/01/2017] [Accepted: 03/06/2017] [Indexed: 12/13/2022]
Abstract
The ARF protein functions as an important sensor of hyper-proliferative stimuli restricting cell proliferation through both p53-dependent and -independent pathways. Although to date the majority of studies on ARF have focused on its anti-proliferative role, few studies have addressed whether ARF may also have pro-survival functions. Here we show for the first time that during the process of adhesion and spreading ARF re-localizes to sites of active actin polymerization and to focal adhesion points where it interacts with the phosphorylated focal adhesion kinase. In line with its recruitment to focal adhesions, we observe that hampering ARF function in cancer cells leads to gross defects in cytoskeleton organization resulting in apoptosis through a mechanism dependent on the Death-Associated Protein Kinase. Our data uncover a novel function for p14ARF in protecting cells from anoikis that may reflect its role in anchorage independence, a hallmark of malignant tumor cells.
Collapse
|
27
|
Zhang J, Sun Z, Han Y, Yao R, Yue L, Xu Y, Zhang J. Rnf2 knockdown reduces cell viability and promotes cell cycle arrest in gastric cancer cells. Oncol Lett 2017; 13:3817-3822. [PMID: 28529595 DOI: 10.3892/ol.2017.5868] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/26/2017] [Indexed: 01/08/2023] Open
Abstract
Rnf2 is a fundamental component of the polycomb repressive complex 1and acts as the really interesting new gene finger E3 ligase, which is responsible for histone 2A modification. Previous studies have shown that the ring finger protein 2 (Rnf2) is overexpressed in various types of tumor and has a close association with tumor development. However, few studies have been carried out into the expression and biological function of Rnf2 in gastric cancer cells. The present study measured the expression of Rnf2 in gastric cancer cells and normal epithelial gastric cells. The results demonstrate that Rnf2 is upregulated in gastric cancer cells. In addition, the knockdown of Rnf2 inhibited the cell viability and induced increased G1 phase followed by a substantial reduction of the G2/M phase. The expression levels of p21 and p27 were also significantly elevated by the knockdown of Rnf2. These results provide evidence of the oncogenic function of Rnf2 in gastric cancer, possibly through an inhibition of cellular proliferation and a delay of the G2/M phase. Therefore, Rnf2 may be a novel target for the prognosis and therapy of gastric cancer.
Collapse
Affiliation(s)
- Jingfang Zhang
- Department of Pathology, Taishan Medical University, Taian, Shandong 271000, P.R. China
| | - Zhenni Sun
- Department of Oncology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong 266011, P.R. China
| | - Yafei Han
- The Center of Medical Research, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Ruyong Yao
- The Center of Medical Research, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Lu Yue
- Department of Oncology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong 266011, P.R. China
| | - Yan Xu
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Jisheng Zhang
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
28
|
Yang K, Wang M, Zhao Y, Sun X, Yang Y, Li X, Zhou A, Chu H, Zhou H, Xu J, Wu M, Yang J, Yi J. A redox mechanism underlying nucleolar stress sensing by nucleophosmin. Nat Commun 2016; 7:13599. [PMID: 27886181 PMCID: PMC5133708 DOI: 10.1038/ncomms13599] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 10/18/2016] [Indexed: 12/24/2022] Open
Abstract
The nucleolus has been recently described as a stress sensor. The nucleoplasmic translocation of nucleolar protein nucleophosmin (NPM1) is a hallmark of nucleolar stress; however, the causes of this translocation and its connection to p53 activation are unclear. Using single live-cell imaging and the redox biosensors, we demonstrate that nucleolar oxidation is a general response to various cellular stresses. During nucleolar oxidation, NPM1 undergoes S-glutathionylation on cysteine 275, which triggers the dissociation of NPM1 from nucleolar nucleic acids. The C275S mutant NPM1, unable to be glutathionylated, remains in the nucleolus under nucleolar stress. Compared with wild-type NPM1 that can disrupt the p53–HDM2 interaction, the C275S mutant greatly compromises the activation of p53, highlighting that nucleoplasmic translocation of NPM1 is a prerequisite for stress-induced activation of p53. This study elucidates a redox mechanism for the nucleolar stress sensing and may help the development of therapeutic strategies. Nucleoplasmic translocation of NPM1 is integral to nucleolar stress sensing. Here, the authors show that nucleolar oxidation is a general cellular stress response, and that oxidation-related glutathionylation of NPM1 triggers its translocation and facilitates p53 activation.
Collapse
Affiliation(s)
- Kai Yang
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Ming Wang
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Yuzheng Zhao
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Xuxu Sun
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Yi Yang
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Xie Li
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Aiwu Zhou
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Huilin Chu
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Hu Zhou
- Shanghai Institute of Materia Medica, 555 Zu Chong Zhi Road, Zhang Jiang Hi-Tech Park, Shanghai 201203, China
| | - Jianrong Xu
- Department of Pharmacology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Mian Wu
- School of Life Sciences, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230022, China
| | - Jie Yang
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Jing Yi
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| |
Collapse
|
29
|
Wasylishen AR, Lozano G. Attenuating the p53 Pathway in Human Cancers: Many Means to the Same End. Cold Spring Harb Perspect Med 2016; 6:cshperspect.a026211. [PMID: 27329033 DOI: 10.1101/cshperspect.a026211] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The p53 pathway is perturbed in the majority of human cancers. Although this most frequently occurs through the direct mutation or deletion of p53 itself, there are a number of other alterations that can attenuate the pathway and contribute to tumorigenesis. For example, amplification of important negative regulators, MDM2 and MDM4, occurs in a number of cancers. In this work, we will review both the normal regulation of the p53 pathway and the different mechanisms of pathway inhibition in cancer, discuss these alterations in the context of the global genomic analyses that have been conducted across tumor types, and highlight the translational implications for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Amanda R Wasylishen
- Department of Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Guillermina Lozano
- Department of Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
30
|
Oh AY, Jung YS, Kim J, Lee JH, Cho JH, Chun HY, Park S, Park H, Lim S, Ha NC, Park JS, Park CS, Song GY, Park BJ. Inhibiting DX2-p14/ARF Interaction Exerts Antitumor Effects in Lung Cancer and Delays Tumor Progression. Cancer Res 2016; 76:4791-804. [PMID: 27302160 DOI: 10.1158/0008-5472.can-15-1025] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/03/2016] [Indexed: 11/16/2022]
Abstract
The aminoacyl tRNA synthetase complex-interacting multifunctional protein 2 (AIMP2) splice variant designated DX2 is induced by cigarette smoke carcinogens and is often detected in human lung cancer specimens. However, the function of DX2 in lung carcinogenesis is obscure. In this study, we found that DX2 expression was induced by oncogenes in human lung cancer tissues and cells. DX2 prevented oncogene-induced apoptosis and senescence and promoted drug resistance by directly binding to and inhibiting p14/ARF. Through chemical screening, we identified SLCB050, a novel compound that blocks the interaction between DX2 and p14/ARF in vitro and in vivo SLCB050 reduced the viability of human lung cancer cells, especially small cell lung cancer cells, in a p14/ARF-dependent manner. Moreover, in a mouse model of K-Ras-driven lung tumorigenesis, ectopic expression of DX2 induced small cell and non-small cell lung cancers, both of which could be suppressed by SLCB050 treatment. Taken together, our findings show how DX2 promotes lung cancer progression and how its activity may be thwarted as a strategy to treat patients with lung cancers exhibiting elevated DX2 levels. Cancer Res; 76(16); 4791-804. ©2016 AACR.
Collapse
Affiliation(s)
- Ah-Young Oh
- Department of Molecular Biology, Pusan National University, Busan, Republic of Korea (South)
| | - Youn Sang Jung
- Department of Molecular Biology, Pusan National University, Busan, Republic of Korea (South)
| | - Jiseon Kim
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea (South)
| | - Jee-Hyun Lee
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea (South)
| | - Jung-Hyun Cho
- Department of Molecular Biology, Pusan National University, Busan, Republic of Korea (South)
| | - Ho-Young Chun
- Department of Molecular Biology, Pusan National University, Busan, Republic of Korea (South)
| | - Soyoung Park
- Department of Molecular Biology, Pusan National University, Busan, Republic of Korea (South)
| | - Hyunchul Park
- Forensic DNA Division, National Forensic Service, Wonju, Republic of Korea (South)
| | - Sikeun Lim
- Forensic DNA Division, National Forensic Service, Wonju, Republic of Korea (South)
| | - Nam-Chul Ha
- Program in Food Science and Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea (South)
| | - Jong Sook Park
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Gyeonggi Do, Republic of Korea (South)
| | - Choon-Sik Park
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Gyeonggi Do, Republic of Korea (South)
| | - Gyu-Yong Song
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea (South).
| | - Bum-Joon Park
- Department of Molecular Biology, Pusan National University, Busan, Republic of Korea (South).
| |
Collapse
|
31
|
HEY1 functions are regulated by its phosphorylation at Ser-68. Biosci Rep 2016; 36:BSR20160123. [PMID: 27129302 PMCID: PMC5293587 DOI: 10.1042/bsr20160123] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 04/27/2016] [Indexed: 01/25/2023] Open
Abstract
HEY1-dependent activation of the p53 tumour suppressor pathway can be inhibited through direct phosphorylation of HEY1 at Ser-68 located in the bHLH domain. STK38 and STK38L serine/threonine kinases can phosphorylate HEY1 Ser-68 and could modulate its biological function. HEY1 (hairy/enhancer-of-split related with YRPW motif 1) is a member of the basic helix–loop–helix-orange (bHLH-O) family of transcription repressors that mediate Notch signalling. HEY1 acts as a positive regulator of the tumour suppressor p53 via still unknown mechanisms. A MALDI-TOF/TOF MS analysis has uncovered a novel HEY1 regulatory phosphorylation event at Ser-68. Strikingly, this single phosphorylation event controls HEY1 stability and function: simulation of HEY1 Ser-68 phosphorylation increases HEY1 protein stability but inhibits its ability to enhance p53 transcriptional activity. Unlike wild-type HEY1, expression of the phosphomimetic mutant HEY1-S68D failed to induce p53-dependent cell cycle arrest and it did not sensitize U2OS cells to p53-activating chemotherapeutic drugs. We have identified two related kinases, STK38 (serine/threonine kinase 38) and STK38L (serine/threonine kinase 38 like), which interact with and phosphorylate HEY1 at Ser-68. HEY1 is phosphorylated at Ser-68 during mitosis and it accumulates in the centrosomes of mitotic cells, suggesting a possible integration of HEY1-dependent signalling in centrosome function. Moreover, HEY1 interacts with a subset of p53-activating ribosomal proteins. Ribosomal stress causes HEY1 relocalization from the nucleoplasm to perinucleolar structures termed nucleolar caps. HEY1 interacts physically with at least one of the ribosomal proteins, RPL11, and both proteins cooperate in the inhibition of MDM2-mediated p53 degradation resulting in a synergistic positive effect on p53 transcriptional activity. HEY1 itself also interacts directly with MDM2 and it is subjected to MDM2-mediated degradation. Simulation of HEY1 Ser-68 phosphorylation prevents its interaction with p53, RPL11 and MDM2 and abolishes HEY1 migration to nucleolar caps upon ribosomal stress. Our findings uncover a novel mechanism for cross-talk between Notch signalling and nucleolar stress.
Collapse
|
32
|
Stępiński D. Nucleolus-derived mediators in oncogenic stress response and activation of p53-dependent pathways. Histochem Cell Biol 2016; 146:119-39. [PMID: 27142852 DOI: 10.1007/s00418-016-1443-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2016] [Indexed: 12/12/2022]
Abstract
Rapid growth and division of cells, including tumor ones, is correlated with intensive protein biosynthesis. The output of nucleoli, organelles where translational machineries are formed, depends on a rate of particular stages of ribosome production and on accessibility of elements crucial for their effective functioning, including substrates, enzymes as well as energy resources. Different factors that induce cellular stress also often lead to nucleolar dysfunction which results in ribosome biogenesis impairment. Such nucleolar disorders, called nucleolar or ribosomal stress, usually affect cellular functioning which in fact is a result of p53-dependent pathway activation, elicited as a response to stress. These pathways direct cells to new destinations such as cell cycle arrest, damage repair, differentiation, autophagy, programmed cell death or aging. In the case of impaired nucleolar functioning, nucleolar and ribosomal proteins mediate activation of the p53 pathways. They are also triggered as a response to oncogenic factor overexpression to protect tissues and organs against extensive proliferation of abnormal cells. Intentional impairment of any step of ribosome biosynthesis which would direct the cells to these destinations could be a strategy used in anticancer therapy. This review presents current knowledge on a nucleolus, mainly in relation to cancer biology, which is an important and extremely sensitive element of the mechanism participating in cellular stress reaction mediating activation of the p53 pathways in order to counteract stress effects, especially cancer development.
Collapse
Affiliation(s)
- Dariusz Stępiński
- Department of Cytophysiology, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, 90-236, Łódź, Poland.
| |
Collapse
|
33
|
Abstract
A veritable explosion of primary research papers within the past 10 years focuses on nucleolar and ribosomal stress, and for good reason: with ribosome biosynthesis consuming ~80% of a cell’s energy, nearly all metabolic and signaling pathways lead ultimately to or from the nucleolus. We begin by describing p53 activation upon nucleolar stress resulting in cell cycle arrest or apoptosis. The significance of this mechanism cannot be understated, as oncologists are now inducing nucleolar stress strategically in cancer cells as a potential anti-cancer therapy. We also summarize the human ribosomopathies, syndromes in which ribosome biogenesis or function are impaired leading to birth defects or bone narrow failures; the perplexing problem in the ribosomopathies is why only certain cells are affected despite the fact that the causative mutation is systemic. We then describe p53-independent nucleolar stress, first in yeast which lacks p53, and then in other model metazoans that lack MDM2, the critical E3 ubiquitin ligase that normally inactivates p53. Do these presumably ancient p53-independent nucleolar stress pathways remain latent in human cells? If they still exist, can we use them to target >50% of known human cancers that lack functional p53?
Collapse
Affiliation(s)
- Allison James
- a Department of Biological Sciences; Louisiana State University; Baton Rouge, LA USA
| | | | | | | | | |
Collapse
|
34
|
Vivo M, Matarese M, Sepe M, Di Martino R, Festa L, Calabrò V, Mantia GL, Pollice A. MDM2-mediated degradation of p14ARF: a novel mechanism to control ARF levels in cancer cells. PLoS One 2015; 10:e0117252. [PMID: 25723571 PMCID: PMC4344200 DOI: 10.1371/journal.pone.0117252] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 12/19/2014] [Indexed: 01/22/2023] Open
Abstract
We here show a new relationship between the human p14ARF oncosuppressor and the MDM2 oncoprotein. MDM2 overexpression in various cancer cell lines causes p14ARF reduction inducing its degradation through the proteasome. The effect does not require the ubiquitin ligase activity of MDM2 and preferentially occurs in the cytoplasm. Interestingly, treatment with inhibitors of the PKC (Protein Kinase C) pathway and use of p14ARF phosphorylation mutants indicate that ARF phosphorylation could play a role in MDM2 mediated ARF degradation reinforcing our previous observations that ARF phosphorylation influences its stability and biological activity. Our study uncovers a new potentially important mechanism through which ARF and MDM2 can counterbalance each other during the tumorigenic process.
Collapse
Affiliation(s)
- Maria Vivo
- Dipartimento di Biologia, Università di Napoli Federico II, Naples, Italy
| | - Maria Matarese
- Dipartimento di Biologia, Università di Napoli Federico II, Naples, Italy
- Istituto di Genetica e Biofisica (IGB)—Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Maria Sepe
- Dipartimento di Biologia, Università di Napoli Federico II, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche- Università di Napoli Federico II, Naples, Italy
| | - Rosaria Di Martino
- Dipartimento di Biologia, Università di Napoli Federico II, Naples, Italy
- Istituto di Biochimica delle Proteine (IBP)—Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Luisa Festa
- Dipartimento di Biologia, Università di Napoli Federico II, Naples, Italy
- Diagnostica e Farmaceutica Molecolare- Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Viola Calabrò
- Dipartimento di Biologia, Università di Napoli Federico II, Naples, Italy
| | - Girolama La Mantia
- Dipartimento di Biologia, Università di Napoli Federico II, Naples, Italy
| | - Alessandra Pollice
- Dipartimento di Biologia, Università di Napoli Federico II, Naples, Italy
- * E-mail:
| |
Collapse
|
35
|
Mondello P, Cuzzocrea S, Mian M. Pim kinases in hematological malignancies: where are we now and where are we going? J Hematol Oncol 2014; 7:95. [PMID: 25491234 PMCID: PMC4266197 DOI: 10.1186/s13045-014-0095-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/04/2014] [Indexed: 12/21/2022] Open
Abstract
The proviral insertion in murine (PIM) lymphoma proteins are a serine/threonine kinase family composed of three isoformes: Pim-1, Pim-2 and Pim-3. They play a critical role in the control of cell proliferation, survival, homing and migration. Recently, overexpression of Pim kinases has been reported in human tumors, mainly in hematologic malignancies. In vitro and in vivo studies have confirmed their oncogenic potential. Indeed, PIM kinases have shown to be involved in tumorgenesis, to enhance tumor growth and to induce chemo-resistance, which is why they have become an attractive therapeutic target for cancer therapy. Novel molecules inhibiting Pim kinases have been evaluated in preclinical studies, demonstrating to be effective and with a favorable toxicity profile. Given the promising results, some of these compounds are currently under investigation in clinical trials. Herein, we provide an overview of the biological activity of PIM-kinases, their role in hematologic malignancies and future therapeutic opportunities.
Collapse
Affiliation(s)
- Patrizia Mondello
- Department of Human Pathology, University of Messina, Via Consolare Valeria, 98125, Messina, Italy. .,Department of Biological and Environmental Sciences, University of Messina, Messina, Italy.
| | - Salvatore Cuzzocrea
- Department of Biological and Environmental Sciences, University of Messina, Messina, Italy.
| | - Michael Mian
- Department of Hematology, Hospital S. Maurizio, Bolzano/Bozen, Italy. .,Department of Internal Medicine V, Hematology & Oncology, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
36
|
Reed SM, Hagen J, Muniz VP, Rosean TR, Borcherding N, Sciegienka S, Goeken JA, Naumann PW, Zhang W, Tompkins VS, Janz S, Meyerholz DK, Quelle DE. NIAM-deficient mice are predisposed to the development of proliferative lesions including B-cell lymphomas. PLoS One 2014; 9:e112126. [PMID: 25393878 PMCID: PMC4231569 DOI: 10.1371/journal.pone.0112126] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 10/12/2014] [Indexed: 01/10/2023] Open
Abstract
Nuclear Interactor of ARF and Mdm2 (NIAM, gene designation Tbrg1) is a largely unstudied inhibitor of cell proliferation that helps maintain chromosomal stability. It is a novel activator of the ARF-Mdm2-Tip60-p53 tumor suppressor pathway as well as other undefined pathways important for genome maintenance. To examine its predicted role as a tumor suppressor, we generated NIAM mutant (NIAMm/m) mice homozygous for a β-galactosidase expressing gene-trap cassette in the endogenous gene. The mutant mice expressed significantly lower levels of NIAM protein in tissues compared to wild-type animals. Fifty percent of aged NIAM deficient mice (14 to 21 months) developed proliferative lesions, including a uterine hemangioma, pulmonary papillary adenoma, and a Harderian gland adenoma. No age-matched wild-type or NIAM+/m heterozygous animals developed lesions. In the spleen, NIAMm/m mice had prominent white pulp expansion which correlated with enhanced increased reactive lymphoid hyperplasia and evidence of systemic inflammation. Notably, 17% of NIAM mutant mice had splenic white pulp features indicating early B-cell lymphoma. This correlated with selective expansion of marginal zone B cells in the spleens of younger, tumor-free NIAM-deficient mice. Unexpectedly, basal p53 expression and activity was largely unaffected by NIAM loss in isolated splenic B cells. In sum, NIAM down-regulation in vivo results in a significant predisposition to developing benign tumors or early stage cancers. These mice represent an outstanding platform for dissecting NIAM's role in tumorigenesis and various anti-cancer pathways, including p53 signaling.
Collapse
Affiliation(s)
- Sara M. Reed
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, United States of America
- Medical Scientist Training Program, University of Iowa, Iowa City, Iowa, United States of America
| | - Jussara Hagen
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, United States of America
| | - Viviane P. Muniz
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, United States of America
- Molecular and Cellular Biology Program, University of Iowa, Iowa City, Iowa, United States of America
| | - Timothy R. Rosean
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Nick Borcherding
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Sebastian Sciegienka
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, United States of America
| | - J. Adam Goeken
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Paul W. Naumann
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Weizhou Zhang
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Van S. Tompkins
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Siegfried Janz
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - David K. Meyerholz
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Dawn E. Quelle
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, United States of America
- Medical Scientist Training Program, University of Iowa, Iowa City, Iowa, United States of America
- Molecular and Cellular Biology Program, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
37
|
Hamilton G, Abraham AG, Morton J, Sampson O, Pefani DE, Khoronenkova S, Grawenda A, Papaspyropoulos A, Jamieson N, McKay C, Sansom O, Dianov GL, O'Neill E. AKT regulates NPM dependent ARF localization and p53mut stability in tumors. Oncotarget 2014; 5:6142-67. [PMID: 25071014 PMCID: PMC4171619 DOI: 10.18632/oncotarget.2178] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 07/07/2014] [Indexed: 12/31/2022] Open
Abstract
Nucleophosmin (NPM) is known to regulate ARF subcellular localization and MDM2 activity in response to oncogenic stress, though the precise mechanism has remained elusive. Here we describe how NPM and ARF associate in the nucleoplasm to form a MDM2 inhibitory complex. We find that oligomerization of NPM drives nucleolar accumulation of ARF. Moreover, the formation of NPM and ARF oligomers antagonizes MDM2 association with the inhibitory complex, leading to activation of MDM2 E3-ligase activity and targeting of p53. We find that AKT phosphorylation of NPM-Ser48 prevents oligomerization that results in nucleoplasmic localization of ARF, constitutive MDM2 inhibition and stabilization of p53. We also show that ARF promotes p53 mutant stability in tumors and suppresses p73 mediated p21 expression and senescence. We demonstrate that AKT and PI3K inhibitors may be effective in treatment of therapeutically resistant tumors with elevated AKT and carrying gain of function mutations in p53. Our results show that the clinical candidate AKT inhibitor MK-2206 promotes ARF nucleolar localization, reduced p53(mut) stability and increased sensitivity to ionizing radiation in a xenograft model of pancreatic cancer. Analysis of human tumors indicates that phospho-S48-NPM may be a useful biomarker for monitoring AKT activity and in vivo efficacy of AKT inhibitor treatment. Critically, we propose that combination therapy involving PI3K-AKT inhibitors would benefit from a patient stratification rationale based on ARF and p53(mut) status.
Collapse
Affiliation(s)
- Garth Hamilton
- Cancer Research UK/MRC Oxford Institute, Department of Oncology, University of Oxford, Old Road Campus, Roosevelt Drive, UK; These authors contributed equally to this work
| | - Aswin G Abraham
- Cancer Research UK/MRC Oxford Institute, Department of Oncology, University of Oxford, Old Road Campus, Roosevelt Drive, UK; These authors contributed equally to this work
| | - Jennifer Morton
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow, UK
| | - Oliver Sampson
- Cancer Research UK/MRC Oxford Institute, Department of Oncology, University of Oxford, Old Road Campus, Roosevelt Drive, UK
| | - Dafni E Pefani
- Cancer Research UK/MRC Oxford Institute, Department of Oncology, University of Oxford, Old Road Campus, Roosevelt Drive, UK
| | - Svetlana Khoronenkova
- Cancer Research UK/MRC Oxford Institute, Department of Oncology, University of Oxford, Old Road Campus, Roosevelt Drive, UK
| | - Anna Grawenda
- Cancer Research UK/MRC Oxford Institute, Department of Oncology, University of Oxford, Old Road Campus, Roosevelt Drive, UK
| | - Angelos Papaspyropoulos
- Cancer Research UK/MRC Oxford Institute, Department of Oncology, University of Oxford, Old Road Campus, Roosevelt Drive, UK
| | - Nigel Jamieson
- West of Scotland Pancreatic Unit and University Department of Surgery, Glasgow Royal Infirmary, Alexandra Parade. Glasgow
| | - Colin McKay
- West of Scotland Pancreatic Unit and University Department of Surgery, Glasgow Royal Infirmary, Alexandra Parade. Glasgow
| | - Owen Sansom
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow, UK
| | - Grigory L Dianov
- Cancer Research UK/MRC Oxford Institute, Department of Oncology, University of Oxford, Old Road Campus, Roosevelt Drive, UK
| | - Eric O'Neill
- Cancer Research UK/MRC Oxford Institute, Department of Oncology, University of Oxford, Old Road Campus, Roosevelt Drive, UK
| |
Collapse
|
38
|
Ghosh M, Ryan RO. Curcumin homing to the nucleolus: mechanism for initiation of an apoptotic program. J Nutr Biochem 2014; 25:1117-1123. [PMID: 25172633 DOI: 10.1016/j.jnutbio.2014.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/07/2014] [Accepted: 06/12/2014] [Indexed: 11/15/2022]
Abstract
Curcumin is a plant-derived polyphenol that displays antitumor properties. Incubation of cultured SF-767 glioma cells with curcumin gave rise to intense intranuclear foci of curcumin fluorescence. In vitro studies revealed that nuclear homing by curcumin is not a result of DNA/chromatin binding. On the other hand, curcumin fluorescence colocalized with nucleophosmin, a nucleolus marker protein. To determine the temporal relationship between curcumin-induced apoptosis and nucleolar homing, confocal live cell imaging was performed. The data show that curcumin localization to the nucleolus occurs prior to cell surface exposure of phosphatidylserine. In studies of the mechanism of curcumin-induced apoptosis in SF-767 cells, its effect on the subcellular location of p14(ARF) was determined. Whereas p14(ARF) was confined to the nucleolus in untreated cells, 2 h following incubation with curcumin, it displayed a diffuse nuclear distribution. Given the role of nuclear p14(ARF) in binding the E3 ubiquitin ligase, mouse double minute 2 homolog (MDM2), the effect of curcumin treatment on cellular levels of the tumor suppressor protein, p53, was examined. Between 2 and 4 h following curcumin treatment, p53 levels increased with maximum levels reached by 8 h. Thus, curcumin homing to the nucleolus induces redistribution of p14(ARF) to the nucleoplasm where interaction with MDM2 leads to stabilization of p53, with subsequent initiation of apoptosis.
Collapse
Affiliation(s)
- Mistuni Ghosh
- Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr. Way, Oakland CA 94609, USA
| | - Robert O Ryan
- Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr. Way, Oakland CA 94609, USA.
| |
Collapse
|
39
|
Sun XX, Dai MS. Deubiquitinating enzyme regulation of the p53 pathway: A lesson from Otub1. World J Biol Chem 2014; 5:75-84. [PMID: 24920999 PMCID: PMC4050119 DOI: 10.4331/wjbc.v5.i2.75] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 01/11/2014] [Accepted: 03/14/2014] [Indexed: 02/05/2023] Open
Abstract
Deubiquitination has emerged as an important mechanism of p53 regulation. A number of deubiquitinating enzymes (DUBs) from the ubiquitin-specific protease family have been shown to regulate the p53-MDM2-MDMX networks. We recently reported that Otub1, a DUB from the OTU-domain containing protease family, is a novel p53 regulator. Interestingly, Otub1 abrogates p53 ubiquitination and stabilizes and activates p53 in cells independently of its deubiquitinating enzyme activity. Instead, it does so by inhibiting the MDM2 cognate ubiquitin-conjugating enzyme (E2) UbcH5. Otub1 also regulates other biological signaling through this non-canonical mechanism, suppression of E2, including the inhibition of DNA-damage-induced chromatin ubiquitination. Thus, Otub1 evolves as a unique DUB that mainly suppresses E2 to regulate substrates. Here we review the current progress made towards the understanding of the complex regulation of the p53 tumor suppressor pathway by DUBs, the biological function of Otub1 including its positive regulation of p53, and the mechanistic insights into how Otub1 suppresses E2.
Collapse
|
40
|
Eischen CM, Lozano G. The Mdm network and its regulation of p53 activities: a rheostat of cancer risk. Hum Mutat 2014; 35:728-37. [PMID: 24488925 DOI: 10.1002/humu.22524] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 01/31/2014] [Indexed: 11/07/2022]
Abstract
The potent transcriptional activity of p53 (Trp53, TP53) must be kept in check for normal cell growth and survival. Tumors, which drastically deviate from these parameters, have evolved multiple mechanisms to inactivate TP53, the most prevalent of which is the emergence of TP53 missense mutations, some of which have gain-of-function activities. Another important mechanism by which tumors bypass TP53 functions is via increased levels of two TP53 inhibitors, MDM2, and MDM4. Studies in humans and in mice reveal the complexity of TP53 regulation and the exquisite sensitivity of this pathway to small changes in regulation. Here, we summarize the factors that impinge on TP53 activity and thus cell death/arrest or tumor development.
Collapse
Affiliation(s)
- Christine M Eischen
- Vanderbilt University Medical Center, Department of Pathology, Microbiology and Immunology, Nashville, Tennessee
| | | |
Collapse
|
41
|
Vlatković N, Boyd MT, Rubbi CP. Nucleolar control of p53: a cellular Achilles' heel and a target for cancer therapy. Cell Mol Life Sci 2014; 71:771-91. [PMID: 23685903 PMCID: PMC11113510 DOI: 10.1007/s00018-013-1361-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 04/07/2013] [Accepted: 04/30/2013] [Indexed: 02/07/2023]
Abstract
Nucleoli perform a crucial cell function, ribosome biogenesis, and of critical relevance to the subject of this review, they are also extremely sensitive to cellular stresses, which can cause loss of function and/or associated structural disruption. In recent years, we have learned that cells take advantage of this stress sensitivity of nucleoli, using them as stress sensors. One major protein regulated by this role of nucleoli is the tumor suppressor p53, which is activated in response to diverse cellular injuries in order to exert its onco-protective effects. Here we discuss a model of nucleolar regulation of p53, which proposes that key steps in the promotion of p53 degradation by the ubiquitin ligase MDM2 occur in nucleoli, thus providing an explanation for the observed link between nucleolar disruption and p53 stability. We review current evidence for this compartmentalization in p53 homeostasis and highlight current limitations of the model. Interestingly, a number of current chemotherapeutic agents capable of inducing a p53 response are likely to do so by targeting nucleolar functions and these compounds may serve to inform further improved therapeutic targeting of nucleoli.
Collapse
Affiliation(s)
- Nikolina Vlatković
- Cancer Research Centre, University of Liverpool, 200 London Rd, Liverpool, L3 9TA UK
| | - Mark T. Boyd
- Cancer Research Centre, University of Liverpool, 200 London Rd, Liverpool, L3 9TA UK
| | - Carlos P. Rubbi
- Cancer Research Centre, University of Liverpool, 200 London Rd, Liverpool, L3 9TA UK
| |
Collapse
|
42
|
Reed SM, Hagen J, Tompkins VS, Thies K, Quelle FW, Quelle DE. Nuclear interactor of ARF and Mdm2 regulates multiple pathways to activate p53. Cell Cycle 2014; 13:1288-98. [PMID: 24621507 DOI: 10.4161/cc.28202] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The p53 tumor suppressor is controlled by an interactive network of factors that stimulate or inhibit its transcriptional activity. Within that network, Mdm2 functions as the major antagonist of p53 by promoting its ubiquitylation and degradation. Conversely, Tip60 activates p53 through direct association on target promoters as well as acetylation of p53 at lysine 120 (K120). This study examines the functional relationship between Mdm2 and Tip60 with a novel p53 regulator, NIAM (nuclear interactor of ARF and Mdm2). Previous work showed NIAM can suppress proliferation and activate p53 independently of ARF, indicating that other factors mediate those activities. Here, we demonstrate that NIAM is a chromatin-associated protein that binds Tip60. NIAM can promote p53 K120 acetylation, although that modification is not required for NIAM to inhibit proliferation or induce p53 transactivation of the p21 promoter. Notably, Tip60 silencing showed it contributes to but is not sufficient for NIAM-mediated p53 activation, suggesting other mechanisms are involved. Indeed, growth-inhibitory forms of NIAM also bind to Mdm2, and increased NIAM expression levels disrupt p53-Mdm2 association, inhibit p53 polyubiquitylation, and prevent Mdm2-mediated inhibition of p53 transcriptional activity. Importantly, loss of NIAM significantly impairs p53 activation. Together, these results show that NIAM activates p53 through multiple mechanisms involving Tip60 association and Mdm2 inhibition. Thus, NIAM regulates 2 critical pathways that control p53 function and are altered in human cancers, implying an important role for NIAM in tumorigenesis.
Collapse
Affiliation(s)
- Sara M Reed
- Department of Pharmacology; University of Iowa College of Medicine; Iowa City, IA USA; Medical Scientist Training Program; University of Iowa College of Medicine; Iowa City, IA USA
| | - Jussara Hagen
- Department of Pharmacology; University of Iowa College of Medicine; Iowa City, IA USA
| | - Van S Tompkins
- Department of Pathology; University of Iowa College of Medicine; Iowa City, IA USA
| | - Katie Thies
- Department of Pharmacology; University of Iowa College of Medicine; Iowa City, IA USA
| | - Frederick W Quelle
- Department of Pharmacology; University of Iowa College of Medicine; Iowa City, IA USA
| | - Dawn E Quelle
- Department of Pharmacology; University of Iowa College of Medicine; Iowa City, IA USA; Medical Scientist Training Program; University of Iowa College of Medicine; Iowa City, IA USA; Department of Pathology; University of Iowa College of Medicine; Iowa City, IA USA
| |
Collapse
|
43
|
Heyne K, Förster J, Schüle R, Roemer K. Transcriptional repressor NIR interacts with the p53-inhibiting ubiquitin ligase MDM2. Nucleic Acids Res 2014; 42:3565-79. [PMID: 24413661 PMCID: PMC3973334 DOI: 10.1093/nar/gkt1371] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
NIR (novel INHAT repressor) can bind to p53 at promoters and inhibit p53-mediated gene transactivation by blocking histone acetylation carried out by p300/CBP. Like NIR, the E3 ubiquitin ligase MDM2 can also bind and inhibit p53 at promoters. Here, we present data indicating that NIR, which shuttles between the nucleolus and nucleoplasm, not only binds to p53 but also directly to MDM2, in part via the central acidic and zinc finger domain of MDM2 that is also contacted by several other nucleolus-based MDM2/p53-regulating proteins. Like some of these, NIR was able to inhibit the ubiquitination of MDM2 and stabilize MDM2; however, unlike these nucleolus-based MDM2 regulators, NIR did not inhibit MDM2 to activate p53. Rather, NIR cooperated with MDM2 to repress p53-induced transactivation. This cooperative repression may at least in part involve p300/CBP. We show that NIR can block the acetylation of p53 and MDM2. Non-acetylated p53 has been documented previously to more readily associate with inhibitory MDM2. NIR may thus help to sustain the inhibitory p53:MDM2 complex, and we present evidence suggesting that all three proteins can indeed form a ternary complex. In sum, our findings suggest that NIR can support MDM2 to suppress p53 as a transcriptional activator.
Collapse
Affiliation(s)
- Kristina Heyne
- José Carreras Research Center and Internal Medicine I, University of Saarland Medical Center, 66421 Homburg/Saar, Germany and Department of Urology, Center for Clinical Research, University of Freiburg, 79106 Freiburg, Germany
| | | | | | | |
Collapse
|
44
|
Levav-Cohen Y, Goldberg Z, Tan KH, Alsheich-Bartok O, Zuckerman V, Haupt S, Haupt Y. The p53-Mdm2 loop: a critical juncture of stress response. Subcell Biochem 2014; 85:161-86. [PMID: 25201194 DOI: 10.1007/978-94-017-9211-0_9] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The presence of a functional p53 protein is a key factor for the proper suppression of cancer development. A loss of p53 activity, by mutations or inhibition, is often associated with human malignancies. The p53 protein integrates various stress signals into a growth restrictive cellular response. In this way, p53 eliminates cells with a potential to become cancerous. Being a powerful decision maker, it is imperative that p53 will be activated properly, efficiently and temporarily in response to stress. Equally important is that p53 activation will be extinguished upon recovery from stress, and that improper activation of p53 will be avoided. Failure to achieve these aims is likely to have catastrophic consequences for the organism. The machinery that governs this tight regulation is largely based on the major inhibitor of p53, Mdm2, which both blocks p53 activities and promotes its destabilization. The interplay between p53 and Mdm2 involves a complex network of positive and negative feedback loops. Relief from Mdm2 suppression is required for p53 to be stabilized and activated in response to stress. Protection from Mdm2 entails a concerted action of modifying enzymes and partner proteins. The association of p53 with the PML-nuclear bodies may provide an infrastructure in which this complex regulatory network can be orchestrated. In this chapter we use examples to illustrate the regulatory machinery that drives this network.
Collapse
Affiliation(s)
- Yaara Levav-Cohen
- Lautenberg Center, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | | | | | | | | | | | |
Collapse
|
45
|
Zhang X, Hagen J, Muniz VP, Smith T, Coombs GS, Eischen CM, Mackie DI, Roman DL, Van Rheeden R, Darbro B, Tompkins VS, Quelle DE. RABL6A, a novel RAB-like protein, controls centrosome amplification and chromosome instability in primary fibroblasts. PLoS One 2013; 8:e80228. [PMID: 24282525 PMCID: PMC3839920 DOI: 10.1371/journal.pone.0080228] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 10/01/2013] [Indexed: 12/18/2022] Open
Abstract
RABL6A (RAB-like 6 isoform A) is a novel protein that was originally identified based on its association with the Alternative Reading Frame (ARF) tumor suppressor. ARF acts through multiple p53-dependent and p53-independent pathways to prevent cancer. How RABL6A functions, to what extent it depends on ARF and p53 activity, and its importance in normal cell biology are entirely unknown. We examined the biological consequences of RABL6A silencing in primary mouse embryo fibroblasts (MEFs) that express or lack ARF, p53 or both proteins. We found that RABL6A depletion caused centrosome amplification, aneuploidy and multinucleation in MEFs regardless of ARF and p53 status. The centrosome amplification in RABL6A depleted p53−/− MEFs resulted from centrosome reduplication via Cdk2-mediated hyperphosphorylation of nucleophosmin (NPM) at threonine-199. Thus, RABL6A prevents centrosome amplification through an ARF/p53-independent mechanism that restricts NPM-T199 phosphorylation. These findings demonstrate an essential role for RABL6A in centrosome regulation and maintenance of chromosome stability in non-transformed cells, key processes that ensure genomic integrity and prevent tumorigenesis.
Collapse
Affiliation(s)
- Xuefeng Zhang
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, United States of America
| | - Jussara Hagen
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, United States of America
| | - Viviane P. Muniz
- The Molecular and Cellular Biology Graduate Program, University of Iowa, Iowa City, Iowa, United States of America
| | - Tarik Smith
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, United States of America
| | - Gary S. Coombs
- Department of Biology, Waldorf College, Forest City, Iowa, United States of America
| | - Christine M. Eischen
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Duncan I. Mackie
- Division of Medicinal and Natural Products Chemistry, University of Iowa, Iowa City, Iowa, United States of America
| | - David L. Roman
- Division of Medicinal and Natural Products Chemistry, University of Iowa, Iowa City, Iowa, United States of America
| | - Richard Van Rheeden
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, United States of America
| | - Benjamin Darbro
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, United States of America
| | - Van S. Tompkins
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Dawn E. Quelle
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, United States of America
- The Molecular and Cellular Biology Graduate Program, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
46
|
Devine T, Dai MS. Targeting the ubiquitin-mediated proteasome degradation of p53 for cancer therapy. Curr Pharm Des 2013; 19:3248-62. [PMID: 23151129 DOI: 10.2174/1381612811319180009] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 11/01/2012] [Indexed: 02/08/2023]
Abstract
Within the past decade, there has been a revolution in the types of drugs developed to treat cancer. Therapies that selectively target cancer-specific aberrations, such as kinase inhibitors, have made a dramatic impact on a subset of patients. In spite of these successes, there is still a dearth of treatment options for the vast majority of patients. Therefore, there is a need to design therapies with broader efficacy. The p53 tumor suppressor pathway is one of the most frequently altered in human cancers. However, about half of all cancers retain wild-type p53, yet through various mechanisms, the p53 pathway is otherwise inactivated. Targeting this pathway for reactivation truly represents the "holy grail" in cancer treatment. Most commonly, destabilization of p53 by various components of ubiquitin- proteasome system, notably the ubiquitin ligase MDM2 and its partner MDMX as well as various deubiquitinating enzymes (DUBs), render p53 inert and unresponsive to stress signals. Reinstating its function in cancer has been a long sought-after goal. Towards this end, a great deal of work has been devoted to the development of compounds that either interfere with the p53-MDM2 and p53- MDMX interactions, inhibit MDM2 E3 activity, or target individual DUBs. Here we review the current progress that has been made in the field, with a special emphasis on both MDM2 and DUB inhibitors. Developing inhibitors targeting the upstream of the p53 ubiquitination pathway will likely also be a valuable option.
Collapse
Affiliation(s)
- Tiffany Devine
- Department of Molecular & Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | | |
Collapse
|
47
|
Bursac S, Brdovcak MC, Donati G, Volarevic S. Activation of the tumor suppressor p53 upon impairment of ribosome biogenesis. Biochim Biophys Acta Mol Basis Dis 2013; 1842:817-30. [PMID: 24514102 DOI: 10.1016/j.bbadis.2013.08.014] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 08/27/2013] [Indexed: 12/31/2022]
Abstract
Errors in ribosome biogenesis can result in quantitative or qualitative defects in protein synthesis and consequently lead to improper execution of the genetic program and the development of specific diseases. Evidence has accumulated over the last decade suggesting that perturbation of ribosome biogenesis triggers a p53-activating checkpoint signaling pathway, often referred to as the ribosome biogenesis stress checkpoint pathway. Although it was originally suggested that p53 has a prominent role in preventing diseases by monitoring the fidelity of ribosome biogenesis, recent work has demonstrated that p53 activation upon impairment of ribosome biogenesis also mediates pathological manifestations in humans. Perturbations of ribosome biogenesis can trigger a p53-dependent checkpoint signaling pathway independent of DNA damage and the tumor suppressor ARF through inhibitory interactions of specific ribosomal components with the p53 negative regulator, Mdm2. Here we review the recent advances made toward understanding of this newly-recognized checkpoint signaling pathway, its role in health and disease, and discuss possible future directions in this exciting research field. This article is part of a Special Issue entitled: Role of the Nucleolus in Human Disease.
Collapse
Affiliation(s)
- Sladana Bursac
- Department of Molecular Medicine and Biotechnology, School of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Maja Cokaric Brdovcak
- Department of Molecular Medicine and Biotechnology, School of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Giulio Donati
- Catalan Institute of Oncology, Bellvitge Biomedical Research Institute, Institut d'Investigacio' Biome'dica de Bellvitge (IDIBELL), 08908 Hospitalet de Llobregat, Barcelona, Spain
| | - Sinisa Volarevic
- Department of Molecular Medicine and Biotechnology, School of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia.
| |
Collapse
|
48
|
Haga A, Ogawara Y, Kubota D, Kitabayashi I, Murakami Y, Kondo T. Interactomic approach for evaluating nucleophosmin-binding proteins as biomarkers for Ewing's sarcoma. Electrophoresis 2013; 34:1670-8. [PMID: 23483572 DOI: 10.1002/elps.201200661] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/21/2013] [Accepted: 01/29/2013] [Indexed: 02/02/2023]
Abstract
Nucleophosmin (NPM) is a novel prognostic biomarker for Ewing's sarcoma. To evaluate the prognostic utility of NPM, we conducted an interactomic approach to characterize the NPM protein complex in Ewing's sarcoma cells. A gene suppression assay revealed that NPM promoted cell proliferation and the invasive properties of Ewing's sarcoma cells. FLAG-tag-based affinity purification coupled with liquid chromatography-tandem mass spectrometry identified 106 proteins in the NPM protein complex. The functional classification suggested that the NPM complex participates in critical biological events, including ribosome biogenesis, regulation of transcription and translation, and protein folding, that are mediated by these proteins. In addition to JAK1, a candidate prognostic biomarker for Ewing's sarcoma, the NPM complex, includes 11 proteins known as prognostic biomarkers for other malignancies. Meta-analysis of gene expression profiles of 32 patients with Ewing's sarcoma revealed that 6 of 106 were significantly and independently associated with survival period. These observations suggest a functional role as well as prognostic value of these NPM complex proteins in Ewing's sarcoma. Further, our study suggests the potential applications of interactomics in conjunction with meta-analysis for biomarker discovery.
Collapse
Affiliation(s)
- Ayako Haga
- Division of Pharmaco-proteomics, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
49
|
Sloan K, Bohnsack M, Watkins N. The 5S RNP couples p53 homeostasis to ribosome biogenesis and nucleolar stress. Cell Rep 2013; 5:237-47. [PMID: 24120868 PMCID: PMC3808153 DOI: 10.1016/j.celrep.2013.08.049] [Citation(s) in RCA: 225] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 08/16/2013] [Accepted: 08/30/2013] [Indexed: 01/25/2023] Open
Abstract
Several proto-oncogenes and tumor suppressors regulate the production of ribosomes. Ribosome biogenesis is a major consumer of cellular energy, and defects result in p53 activation via repression of mouse double minute 2 (MDM2) homolog by the ribosomal proteins RPL5 and RPL11. Here, we report that RPL5 and RPL11 regulate p53 from the context of a ribosomal subcomplex, the 5S ribonucleoprotein particle (RNP). We provide evidence that the third component of this complex, the 5S rRNA, is critical for p53 regulation. In addition, we show that the 5S RNP is essential for the activation of p53 by p14ARF, a protein that is activated by oncogene overexpression. Our data show that the abundance of the 5S RNP, and therefore p53 levels, is determined by factors regulating 5S complex formation and ribosome integration, including the tumor suppressor PICT1. The 5S RNP therefore emerges as the critical coordinator of signaling pathways that couple cell proliferation with ribosome production. The 5S RNP (5S rRNA/RPL5/RPL11) activates p53 when ribosome production is blocked The 5S RNP is required for p53 activation by the tumor suppressor p14ARF PICT1 controls p53 levels by regulating 5S RNP integration into the ribosome Ribosome biogenesis is directly coupled to cellular proliferation via the 5S RNP
Collapse
Affiliation(s)
- Katherine E. Sloan
- ICaMB, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Centre for Biochemistry and Molecular Cell Biology, Institute for Molecular Biology, Medical Faculty, Georg-August University, 37073 Goettingen, Germany
| | - Markus T. Bohnsack
- Centre for Biochemistry and Molecular Cell Biology, Institute for Molecular Biology, Medical Faculty, Georg-August University, 37073 Goettingen, Germany
| | - Nicholas J. Watkins
- ICaMB, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Corresponding author
| |
Collapse
|
50
|
Chaar I, Amara S, Elamine OE, Khiari M, Ounissi D, Khalfallah T, Ben Hmida A, Mzabi S, Bouraoui S. Biological significance of promoter hypermethylation of p14/ARF gene: relationships to p53 mutational status in Tunisian population with colorectal carcinoma. Tumour Biol 2013; 35:1439-49. [PMID: 24065196 PMCID: PMC3932170 DOI: 10.1007/s13277-013-1198-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 09/11/2013] [Indexed: 11/26/2022] Open
Abstract
One of the most important pathways which are frequently affected in colorectal cancer is p53/ (MDM2)/p14ARF pathway. We aim to determine the methylation pattern of p14/ARF in relation to mutation of p53. This correlation was studied to investigate whether their alterations could be considered as a predictor factor of prognosis in colorectal cancer and whether it can be useful in early-stage diagnosis. Statistical analyses show that p14/ARF hypermethylation was correlated with rectum location (p = 0.004), primary TNM stage (p = 0.016), and advanced Astler–Coller stage (p = 0.024). The RT-PCR that revel 31 % of patients did not express p14/ARF mRNA or at very low level. A high concordance between CpG hypermethylation and the low levels (p < 0.005) was shown. In addition, our analyses demonstrate that patients with mutation in the p53 gene have a lack of the protein expression (p < 0.005). This category with negative expression of p53 had a shorter survival rate (p < 0.005). On the one hand, MSP pattern of p14/ARF were correlated with a lack of p53 expression (p = 0.007). We found that p53/p14ARF pathway was frequently deregulated among our patients. In our study, we demonstrate that hypermethylation of p14/ARF occurs early during CRC tumorogenesis. However, we did not find correlation between p14/ARF and survival. These results suggest that p14/ARF methylation pattern may constitute a predictor factor of CRC in early stage but it could not be considered as a prognostic factor. On the other hand and because of the reversibility of the methylation mechanism, it may be appropriate to target the demethylation of p14/ARF to develop new drogues for CRC.
Collapse
Affiliation(s)
- Ines Chaar
- Laboratory of Colorectal Cancer Research UR03ES04, Science University Tunis, Tunis, Tunisia,
| | | | | | | | | | | | | | | | | |
Collapse
|