1
|
Holoubek A, Strachotová D, Wolfová K, Otevřelova P, Belejová S, Röselová P, Benda A, Brodská B, Herman P. Correlation of p53 oligomeric status and its subcellular localization in the presence of the AML-associated NPM mutant. PLoS One 2025; 20:e0322096. [PMID: 40334261 PMCID: PMC12058200 DOI: 10.1371/journal.pone.0322096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/17/2025] [Indexed: 05/09/2025] Open
Abstract
Tumor suppressor p53 is a key player in the cell response to DNA damage that suffers by frequent inactivating aberrations. Some of them disturb p53 oligomerization and influence cell decision between proliferation, growth arrest and apoptosis. Active p53 resides mostly in the nucleus, degradation occurs in the cytoplasm. Acute myeloid leukemia (AML)-related mutation of NPM (NPMmut) induces massive mislocalization of p53 to the cytoplasm, which might be related to leukemia initiation. Since both proteins interact and execute their function as oligomers, we investigated the role of perturbed p53 oligomerization in the p53 mislocalization process in live cells by FLIM (fluorescence lifetime imaging microscopy), fluorescence anisotropy imaging (FAIM), fluorescence cross-correlation spectroscopy (FCCS) and immunochemical methods. On a set of fluorescently labeled p53 variants, monomeric R337G and L344P, dimeric L344A, and multimeric D352G and A353S, we correlated their cellular localization, oligomerization and interaction with NPMmut. Interplay between nuclear export signal (NES) and nuclear localization signal (NLS) of p53 was investigated as well. While NLS was found critical for the nuclear p53 localization, NES plays less significant role. We observed cytoplasmic translocation only for multimeric A353S variant with sufficient stability and strong interaction with NPMmut. Less stable multimer D352G and L344A dimer were not translocated, monomeric p53 variants always resided in the nucleus independently of the presence of NPMmut and NES intactness. Oligomeric state of NPMmut is not required for p53 translocation, which happens also in the presence of the nonoligomerizing NPMmut variant. The prominent structural and functional role of the R337 residue is shown.
Collapse
Affiliation(s)
- Aleš Holoubek
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Dita Strachotová
- Faculty of Mathematics and Physics, Institute of Physics, Charles University, Prague, Czech Republic
| | - Kateřina Wolfová
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Petra Otevřelova
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Sára Belejová
- Faculty of Mathematics and Physics, Institute of Physics, Charles University, Prague, Czech Republic
| | - Pavla Röselová
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Aleš Benda
- Imaging Methods Core Facility at BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Barbora Brodská
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Petr Herman
- Faculty of Mathematics and Physics, Institute of Physics, Charles University, Prague, Czech Republic
| |
Collapse
|
2
|
Kumari A, Vertii A. Perspective: "Current understanding of NADs dynamics and mechanisms of Disease". Gene 2024; 894:147960. [PMID: 37923094 DOI: 10.1016/j.gene.2023.147960] [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: 07/27/2023] [Revised: 10/09/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Chromatin architecture is essential for gene regulation, and multiple levels of the 3D chromatin organization exhibit dynamic changes during organismal development and cell differentiation. Heterochromatin, termed compartment B in Hi-C datasets, is a phase-separating gene-silencing form of chromatin, preferentially located at the two nuclear sites, nuclear (lamina-associate chromatin domains, LADs) and nucleoli (nucleoli-associated chromatin domains, NADs) peripheries. LADs and NADs contain both interchangeable and location-specific chromatin domains. Recent studies suggest striking dynamics in LADs and NADs during the differentiation of embryonic stem cells into neural progenitors and neurons. Here we discuss recent advances in understanding NADs changes during neuronal differentiation and future questions on how NADs integrity can contribute to healthy neurodevelopment and neurodevelopment diseases.
Collapse
Affiliation(s)
- Amrita Kumari
- Department of Molecular, Cellular and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 1605, US
| | - Anastassiia Vertii
- Department of Molecular, Cellular and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 1605, US.
| |
Collapse
|
3
|
Escarcega RD, Patil AA, Moruno-Manchon JF, Urayama A, Marrelli SP, Kim N, Monchaud D, McCullough LD, Tsvetkov AS. Pirh2-dependent DNA damage in neurons induced by the G-quadruplex ligand pyridostatin. J Biol Chem 2023; 299:105157. [PMID: 37579947 PMCID: PMC10534229 DOI: 10.1016/j.jbc.2023.105157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/28/2023] [Accepted: 08/07/2023] [Indexed: 08/16/2023] Open
Abstract
Noncanonical base pairing between four guanines (G) within single-stranded G-rich sequences leads to formation of а G-quartet. Self-stacking of G-quartets results in a columnar four-stranded DNA structure known as the G-quadruplex (G4 or G4-DNA). In cancer cells, G4-DNA regulates multiple DNA-dependent processes, including transcription, replication, and telomere function. How G4s function in neurons is poorly understood. Here, we performed a genome-wide gene expression analysis (RNA-Seq) to identify genes modulated by a G4-DNA ligand, pyridostatin (PDS), in primary cultured neurons. PDS promotes stabilization of G4 structures, thus allowing us to define genes directly or indirectly responsive to G4 regulation. We found that 901 genes were differentially expressed in neurons treated with PDS out of a total of 18,745 genes with measured expression. Of these, 505 genes were downregulated and 396 genes were upregulated and included gene networks regulating p53 signaling, the immune response, learning and memory, and cellular senescence. Within the p53 network, the E3 ubiquitin ligase Pirh2 (Rchy1), a modulator of DNA damage responses, was upregulated by PDS. Ectopically overexpressing Pirh2 promoted the formation of DNA double-strand breaks, suggesting a new DNA damage mechanism in neurons that is regulated by G4 stabilization. Pirh2 downregulated DDX21, an RNA helicase that unfolds G4-RNA and R-loops. Finally, we demonstrated that Pirh2 increased G4-DNA levels in the neuronal nucleolus. Our data reveal the genes that are responsive to PDS treatment and suggest similar transcriptional regulation by endogenous G4-DNA ligands. They also connect G4-dependent regulation of transcription and DNA damage mechanisms in neuronal cells.
Collapse
Affiliation(s)
- Rocio Diaz Escarcega
- Department of Neurology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Abhijeet A Patil
- Department of Neurology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Jose F Moruno-Manchon
- Department of Neurology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Akihiko Urayama
- Department of Neurology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Sean P Marrelli
- Department of Neurology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Nayun Kim
- Department of Microbiology and Molecular Genetics, The University of Texas McGovern Medical School at Houston, Houston, Texas, USA
| | - David Monchaud
- Institut de Chimie Moléculaire (ICMUB), UBFC Dijon, CNRS UMR6302, Dijon, France
| | - Louise D McCullough
- Department of Neurology, The University of Texas McGovern Medical School, Houston, Texas, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Andrey S Tsvetkov
- Department of Neurology, The University of Texas McGovern Medical School, Houston, Texas, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA; UTHealth Consortium on Aging, The University of Texas McGovern Medical School, Houston, Texas, USA.
| |
Collapse
|
4
|
Sazonova EN, Gusev IA, Filatova TS. Effects of Non-Opiate Analogue of Leu-Enkephalin on the Ion Currents, Number of Nucleoli, and p53 Expression in Isolated Cardiomyocytes of Albino Rats. Bull Exp Biol Med 2023; 175:544-548. [PMID: 37768450 DOI: 10.1007/s10517-023-05902-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Indexed: 09/29/2023]
Abstract
Acute exposure of isolated ventricular cardiomyocytes to non-opiate analogue of leu-enkephalin (NALE peptide: Phe-D-Ala-Gly-Phe-Leu-Arg) in a concentration of 100 μg/liter and 6-h incubation in NALE solution did not significantly change ATP-dependent K+ current, L-type Ca2+ current, p53 protein expression, and number of nucleoli in the cardiomyocyte nuclei. Incubation of cardiomyocytes with NALE (100 μg/liter) in combination with NOP receptor blocker J-113397 (1 mg/liter) was followed by an increase in Ca2+ L-type current and the number of p53+ cells. The exposure of cardiomyocytes to NALE in a concentration 1000 μg/liter induced similar changes in the studied parameters (increase in Ca2+ L-type current and number of p53+ cardiomyocytes); an increase in the mean number of nucleoli was also observed. Our findings suggest that NALE peptide has direct effect on cardiomyocytes and NOP receptors are involved in this effect.
Collapse
Affiliation(s)
- E N Sazonova
- Far Eastern State Medical University, Ministry of Health of the Russian Federation, Khabarovsk, Russia
- Khabarovsk Branch of Far-Eastern Research Center of Physiology and Pathology of Breath, Research Institute of Maternity and Childhood Protection, Khabarovsk, Russia
| | - I A Gusev
- Far Eastern State Medical University, Ministry of Health of the Russian Federation, Khabarovsk, Russia.
| | - T S Filatova
- Department of Human and Animal Physiology, Faculty of Biology, M. V. Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
5
|
Trifault B, Mamontova V, Burger K. In vivo Proximity Labeling of Nuclear and Nucleolar Proteins by a Stably Expressed, DNA Damage-Responsive NONO-APEX2 Fusion Protein. Front Mol Biosci 2022; 9:914873. [PMID: 35733943 PMCID: PMC9207311 DOI: 10.3389/fmolb.2022.914873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular stress can induce DNA lesions that threaten the stability of genes. The DNA damage response (DDR) recognises and repairs broken DNA to maintain genome stability. Intriguingly, components of nuclear paraspeckles like the non-POU domain containing octamer-binding protein (NONO) participate in the repair of DNA double-strand breaks (DSBs). NONO is a multifunctional RNA-binding protein (RBP) that facilitates the retention and editing of messenger (m)RNA as well as pre-mRNA processing. However, the role of NONO in the DDR is poorly understood. Here, we establish a novel human U2OS cell line that expresses NONO fused to the engineered ascorbate peroxidase 2 (U2OS:NONO-APEX2-HA). We show that NONO-APEX2-HA accumulates in the nucleolus in response to DNA damage. Combining viability assays, subcellular localisation studies, coimmunoprecipitation experiments and in vivo proximity labeling, we demonstrate that NONO-APEX2-HA is a stably expressed fusion protein that mimics endogenous NONO in terms of expression, localisation and bona fide interactors. We propose that in vivo proximity labeling in U2OS:NONO-APEX2-HA cells is capable for the assessment of NONO interactomes by downstream assays. U2OS:NONO-APEX2-HA cells will likely be a valuable resource for the investigation of NONO interactome dynamics in response to DNA damage and other stimuli.
Collapse
|
6
|
Nucleolus localization of SpyCas9 affects its stability and interferes with host protein translation in mammalian cells. Genes Dis 2022; 9:731-740. [PMID: 35782966 PMCID: PMC9243344 DOI: 10.1016/j.gendis.2020.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/28/2022] Open
Abstract
The CRISPR/Cas9 system, originally derived from the prokaryotic adaptive immune system, has been developed as efficient genome editing tools. It enables precise gene manipulation on chromosomal DNA through the specific binding of programmable sgRNA to target DNA, and the Cas9 protein, which has endonuclease activity, will cut a double strand break at specific locus. However, Cas9 is a foreign protein in mammalian cells, and the potential risks associated with its introduction into mammalian cells are not fully understood. In this study, we performed pull-down and mass spectrometry (MS) analysis of Streptococcus pyogenes Cas9 (SpyCas9) interacting proteins in HEK293T cells and showed that the majority of Cas9-associated proteins identified by MS were localized in the nucleolus. Interestingly, we further discovered that the Cas9 protein contains a sequence encoding a nucleolus detention signal (NoDS). Compared with wild-type (WT) Cas9, NoDS-mutated variants of Cas9 (mCas9) are less stable, although their gene editing activity is minimally affected. Overexpression of WT Cas9, but not mCas9, causes general effects on transcription and protein translation in the host cell. Overall, identification of NoDS in Cas9 will improve the understanding of Cas9's biological function in vivo, and the removal of NoDS in Cas9 may enhance its safety for future clinical use.
Collapse
|
7
|
Roqanian S, Ahmadian S, Nabavi SM, Pakdaman H, Shafiezadeh M, Goudarzi G, Shahpasand K. Tau nuclear translocation is a leading step in tau pathology process through P53 stabilization and nucleolar dispersion. J Neurosci Res 2022; 100:1084-1104. [PMID: 35170061 DOI: 10.1002/jnr.25024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022]
Abstract
Tau protein abnormalities are associated with various neurodegenerative disorders, including Alzheimer's disease (AD) and traumatic brain injury (TBI). In tau-overexpressing SHSY5Y cells and iPSC-derived neuron models of frontotemporal dementia (FTD), axonal tau translocates into the nuclear compartment, resulting in neuronal dysfunction. Despite extensive research, the mechanisms by which tau translocation results in neurodegeneration remain elusive thus far. We studied the nuclear displacement of different P-tau species [Cis phosphorylated Thr231-tau (cis P-tau), phosphorylated Ser202/Thr205-tau (AT8 P-tau), and phosphorylated Thr212/Ser214-tau (AT100 P-tau)] at various time points using starvation in primary cortical neurons and single severe TBI (ssTBI) in male mouse cerebral cortices as tauopathy models. While all P-tau species translocated into the somatodendritic compartment in response to stress, cis P-tau did so more rapidly than the other species. Notably, nuclear localization of P-tau was associated with p53 apoptotic stabilization and nucleolar stress, both of which resulted in neurodegeneration. In summary, our findings indicate that P-tau nuclear translocation results in p53-dependent apoptosis and nucleolar dispersion, which is consistent with neurodegeneration.
Collapse
Affiliation(s)
- Shaqayeq Roqanian
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.,Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shahin Ahmadian
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Seyed Masood Nabavi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Pakdaman
- Brain Mapping Research Center, Department of Neurology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahshid Shafiezadeh
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ghazaleh Goudarzi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
8
|
Usluer S, Spreitzer E, Bourgeois B, Madl T. p53 Transactivation Domain Mediates Binding and Phase Separation with Poly-PR/GR. Int J Mol Sci 2021; 22:11431. [PMID: 34768862 PMCID: PMC8583712 DOI: 10.3390/ijms222111431] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/06/2021] [Accepted: 10/18/2021] [Indexed: 12/23/2022] Open
Abstract
The most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is the presence of poly-PR/GR dipeptide repeats, which are encoded by the chromosome 9 open reading frame 72 (C9orf72) gene. Recently, it was shown that poly-PR/GR alters chromatin accessibility, which results in the stabilization and enhancement of transcriptional activity of the tumor suppressor p53 in several neurodegenerative disease models. A reduction in p53 protein levels protects against poly-PR and partially against poly-GR neurotoxicity in cells. Moreover, in model organisms, a reduction of p53 protein levels protects against neurotoxicity of poly-PR. Here, we aimed to study the detailed molecular mechanisms of how p53 contributes to poly-PR/GR-mediated neurodegeneration. Using a combination of biophysical techniques such as nuclear magnetic resonance (NMR) spectroscopy, fluorescence polarization, turbidity assays, and differential interference contrast (DIC) microscopy, we found that p53 physically interacts with poly-PR/GR and triggers liquid-liquid phase separation of p53. We identified the p53 transactivation domain 2 (TAD2) as the main binding site for PR25/GR25 and showed that binding of poly-PR/GR to p53 is mediated by a network of electrostatic and/or hydrophobic interactions. Our findings might help to understand the mechanistic role of p53 in poly-PR/GR-associated neurodegeneration.
Collapse
Affiliation(s)
- Sinem Usluer
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Department of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (S.U.); (E.S.); (B.B.)
| | - Emil Spreitzer
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Department of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (S.U.); (E.S.); (B.B.)
| | - Benjamin Bourgeois
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Department of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (S.U.); (E.S.); (B.B.)
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Department of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (S.U.); (E.S.); (B.B.)
- BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|
9
|
Iarovaia OV, Ioudinkova ES, Velichko AK, Razin SV. Manipulation of Cellular Processes via Nucleolus Hijaking in the Course of Viral Infection in Mammals. Cells 2021; 10:cells10071597. [PMID: 34202380 PMCID: PMC8303250 DOI: 10.3390/cells10071597] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/16/2022] Open
Abstract
Due to their exceptional simplicity of organization, viruses rely on the resources, molecular mechanisms, macromolecular complexes, regulatory pathways, and functional compartments of the host cell for an effective infection process. The nucleolus plays an important role in the process of interaction between the virus and the infected cell. The interactions of viral proteins and nucleic acids with the nucleolus during the infection process are universal phenomena and have been described for almost all taxonomic groups. During infection, proteins of the nucleolus in association with viral components can be directly used for the processes of replication and transcription of viral nucleic acids and the assembly and transport of viral particles. In the course of a viral infection, the usurpation of the nucleolus functions occurs and the usurpation is accompanied by profound changes in ribosome biogenesis. Recent studies have demonstrated that the nucleolus is a multifunctional and dynamic compartment. In addition to the biogenesis of ribosomes, it is involved in regulating the cell cycle and apoptosis, responding to cellular stress, repairing DNA, and transcribing RNA polymerase II-dependent genes. A viral infection can be accompanied by targeted transport of viral proteins to the nucleolus, massive release of resident proteins of the nucleolus into the nucleoplasm and cytoplasm, the movement of non-nucleolar proteins into the nucleolar compartment, and the temporary localization of viral nucleic acids in the nucleolus. The interaction of viral and nucleolar proteins interferes with canonical and non-canonical functions of the nucleolus and results in a change in the physiology of the host cell: cell cycle arrest, intensification or arrest of ribosome biogenesis, induction or inhibition of apoptosis, and the modification of signaling cascades involved in the stress response. The nucleolus is, therefore, an important target during viral infection. In this review, we discuss the functional impact of viral proteins and nucleic acid interaction with the nucleolus during infection.
Collapse
|
10
|
Cui X, Pan G, Chen Y, Guo X, Liu T, Zhang J, Yang X, Cheng M, Gao H, Jiang F. The p53 pathway in vasculature revisited: A therapeutic target for pathological vascular remodeling? Pharmacol Res 2021; 169:105683. [PMID: 34019981 DOI: 10.1016/j.phrs.2021.105683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/26/2021] [Accepted: 05/14/2021] [Indexed: 02/08/2023]
Abstract
Pathological vascular remodeling contributes to the development of restenosis following intraluminal interventions, transplant vasculopathy, and pulmonary arterial hypertension. Activation of the tumor suppressor p53 may counteract vascular remodeling by inhibiting aberrant proliferation of vascular smooth muscle cells and repressing vascular inflammation. In particular, the development of different lines of small-molecule p53 activators ignites the hope of treating remodeling-associated vascular diseases by targeting p53 pharmacologically. In this review, we discuss the relationships between p53 and pathological vascular remodeling, and summarize current experimental data suggesting that drugging the p53 pathway may represent a novel strategy to prevent the development of vascular remodeling.
Collapse
Affiliation(s)
- Xiaopei Cui
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Guopin Pan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China; Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Ye Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Xiaosun Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Tengfei Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Jing Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Xiaofan Yang
- Department of Pediatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Mei Cheng
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Haiqing Gao
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Fan Jiang
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
11
|
Solomon DA, Smikle R, Reid MJ, Mizielinska S. Altered Phase Separation and Cellular Impact in C9orf72-Linked ALS/FTD. Front Cell Neurosci 2021; 15:664151. [PMID: 33967699 PMCID: PMC8096919 DOI: 10.3389/fncel.2021.664151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/19/2021] [Indexed: 12/21/2022] Open
Abstract
Since the discovery of the C9orf72 repeat expansion mutation as causative for chromosome 9-linked amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) in 2011, a multitude of cellular pathways have been implicated. However, evidence has also been accumulating for a key mechanism of cellular compartmentalization—phase separation. Liquid-liquid phase separation (LLPS) is fundamental for the formation of membraneless organelles including stress granules, the nucleolus, Cajal bodies, nuclear speckles and the central channel of the nuclear pore. Evidence has now accumulated showing that the formation and function of these membraneless organelles is impaired by both the toxic arginine rich dipeptide repeat proteins (DPRs), translated from the C9orf72 repeat RNA transcript, and the repeat RNA itself. Both the arginine rich DPRs and repeat RNA themselves undergo phase separation and disrupt the physiological phase separation of proteins involved in the formation of these liquid-like organelles. Hence abnormal phase separation may explain a number of pathological cellular phenomena associated with C9orf72-ALS/FTD. In this review article, we will discuss the principles of phase separation, phase separation of the DPRs and repeat RNA themselves and how they perturb LLPS associated with membraneless organelles and the functional consequences of this. We will then discuss how phase separation may impact the major pathological feature of C9orf72-ALS/FTD, TDP-43 proteinopathy, and how LLPS may be targeted therapeutically in disease.
Collapse
Affiliation(s)
- Daniel A Solomon
- UK Dementia Research Institute at King's College London, London, United Kingdom.,Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Rebekah Smikle
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Matthew J Reid
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Sarah Mizielinska
- UK Dementia Research Institute at King's College London, London, United Kingdom.,Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| |
Collapse
|
12
|
Arva A, Kasu YAT, Duncan J, Alkhatatbeh MA, Brower CS. The Ligand of Ate1 is intrinsically disordered and participates in nucleolar phase separation regulated by Jumonji Domain Containing 6. Proc Natl Acad Sci U S A 2021; 118:e2015887118. [PMID: 33443146 PMCID: PMC7817205 DOI: 10.1073/pnas.2015887118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The Ligand of Ate1 (Liat1) is a protein of unknown function that was originally discovered through its interaction with arginyl-tRNA protein transferase 1 (Ate1), a component of the Arg/N-degron pathway of protein degradation. Here, we characterized the functional domains of mouse Liat1 and found that its N-terminal half comprises an intrinsically disordered region (IDR) that facilitates its liquid-liquid phase separation (LLPS) in the nucleolus. Using bimolecular fluorescence complementation and immunocytochemistry, we found that Liat1 is targeted to the nucleolus by a low-complexity poly-K region within its IDR. We also found that the lysyl-hydroxylase activity of Jumonji Domain Containing 6 (Jmjd6) modifies Liat1, in a manner that requires the Liat1 poly-K region, and inhibits its nucleolar targeting and potential functions. In sum, this study reveals that Liat1 participates in nucleolar LLPS regulated by Jmjd6.
Collapse
Affiliation(s)
- Akshaya Arva
- Department of Biology, Texas Woman's University, Denton, TX 76204
| | | | - Jennifer Duncan
- Department of Biology, Texas Woman's University, Denton, TX 76204
| | | | | |
Collapse
|
13
|
Botti V, Urbanelli L, Sagini K, Tarpani L, Cesaretti A, Fortuna CG, Elisei F. Quaternized styryl-azinium fluorophores as cellular RNA-binders. Photochem Photobiol Sci 2020; 19:362-370. [PMID: 32147676 DOI: 10.1039/c9pp00465c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The capability of three quaternized styryl-azinium iodides to bind cellular RNA has been tested by means of Fluorescence Confocal Microscopy imaging of stained MCF-7 cells treated with RNase. Their association constants have been estimated through spectrophotometric and fluorimetric titrations with tRNA and compared to their affinity toward DNA. Transient absorption spectroscopy with femtosecond resolution confirmed the binding of the investigated compounds with tRNA and shed new light on the excited state dynamics of their complexes, by revealing a significant lengthening of the lifetime of S1 upon complexation, which parallels the fluorescence quantum yield enhancement.
Collapse
Affiliation(s)
- Valentina Botti
- Department of Chemistry, Biology and Biotechnology and Center of Excellence on Innovative Nanostructured Materials (CEMIN), University of Perugia, via Elce di Sotto 8, 06123, Perugia, Italy.
| | - Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, via del Giochetto, 06126, Perugia, Italy
| | - Krizia Sagini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, via del Giochetto, 06126, Perugia, Italy
| | - Luigi Tarpani
- Department of Chemistry, Biology and Biotechnology and Center of Excellence on Innovative Nanostructured Materials (CEMIN), University of Perugia, via Elce di Sotto 8, 06123, Perugia, Italy
| | - Alessio Cesaretti
- Department of Chemistry, Biology and Biotechnology and Center of Excellence on Innovative Nanostructured Materials (CEMIN), University of Perugia, via Elce di Sotto 8, 06123, Perugia, Italy
| | - Cosimo G Fortuna
- Department of Chemical Sciences, University of Catania, viale Andrea Doria 6, 95125, Catania, Italy
| | - Fausto Elisei
- Department of Chemistry, Biology and Biotechnology and Center of Excellence on Innovative Nanostructured Materials (CEMIN), University of Perugia, via Elce di Sotto 8, 06123, Perugia, Italy
| |
Collapse
|
14
|
Pirogov SA, Gvozdev VA, Klenov MS. Long Noncoding RNAs and Stress Response in the Nucleolus. Cells 2019; 8:cells8070668. [PMID: 31269716 PMCID: PMC6678565 DOI: 10.3390/cells8070668] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 12/15/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) perform diverse functions in the regulation of cellular processes. Here we consider a variety of lncRNAs found in the ribosome production center, the nucleolus, and focus on their role in the response to environmental stressors. Nucleolar lncRNAs ensure stress adaptation by cessation of resource-intensive ribosomal RNA (rRNA) synthesis and by inducing the massive sequestration of proteins within the nucleolus. Different cell states like quiescence and cancer are also controlled by specific lncRNAs in the nucleolus. Taken together, recent findings allow us to consider lncRNAs as multifunctional regulators of nucleolar activities, which are responsive to various physiological conditions.
Collapse
Affiliation(s)
- Sergei A Pirogov
- Institute of Molecular Genetics, Russian Academy of Sciences, 2 Kurchatov Sq., 123182 Moscow, Russia
| | - Vladimir A Gvozdev
- Institute of Molecular Genetics, Russian Academy of Sciences, 2 Kurchatov Sq., 123182 Moscow, Russia.
| | - Mikhail S Klenov
- Institute of Molecular Genetics, Russian Academy of Sciences, 2 Kurchatov Sq., 123182 Moscow, Russia.
| |
Collapse
|
15
|
Wang Q, Huang WR, Chih WY, Chuang KP, Chang CD, Wu Y, Huang Y, Liu HJ. Cdc20 and molecular chaperone CCT2 and CCT5 are required for the Muscovy duck reovirus p10.8-induced cell cycle arrest and apoptosis. Vet Microbiol 2019; 235:151-163. [PMID: 31282373 DOI: 10.1016/j.vetmic.2019.06.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/19/2019] [Accepted: 06/22/2019] [Indexed: 01/22/2023]
Abstract
This study demonstrates that the Muscovy duck reovirus (MDRV) p10.8 protein is one of many viral non-structural proteins that induces both cell cycle arrest and apoptosis. The p10.8 but not σC is a nuclear targeting protein that shuttles between the nucleus and the cytoplasm. Our results reveal that p10.8-induced apoptosis in cultured cells occurs by the nucleoporin Tpr/p53-dependent and Fas/caspase 8-mediated pathways. Furthermore, a compelling finding from this study is that the p10.8 and σC proteins of MDRV facilitate CDK2 and CDK4 degradation via the ubiquitin-proteasome pathway. We found that depletion of Cdc20 reversed the p10.8- and σC- mediated CDK4 degradation and p10.8-induced apoptosis, suggesting that Cdc20 plays a critical role in modulating p10.8-mediated cell cycle and apoptosis. Furthermore, we found that depletion of chaperonin-containing tailless complex polypeptide 1 (CCT) 2 and CCT5 reduced the level of Cdc20 and reversed the p10.8- and σC-mediated CDK4 degradation and p10.8-induced apoptosis, indicating that molecular chaperone CCT2 and CCT5 are required for stabilization of Ccd20 for mediating both cell cycle arrest and apoptosis. This study provides mechanistic insights into how p10.8 induces both cell cycle arrest and apoptosis.
Collapse
Affiliation(s)
- Quanxi Wang
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Wei-Ru Huang
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan
| | - Wan-Yi Chih
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan
| | - Kuo-Pin Chuang
- Graduate Institute of Animal Vaccine Technology, National Pingtung University of Science and Technology, Pingtung, 912, Taiwan
| | - Ching-Dong Chang
- Department of Veterinary medicine, National Pingtung University of Science and Technology, Pingtung, 912, Taiwan
| | - Yijian Wu
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Yifan Huang
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Hung-Jen Liu
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan; The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; Ph. D Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
16
|
Acetylation of Werner protein at K1127 and K1117 is important for nuclear trafficking and DNA repair. DNA Repair (Amst) 2019; 79:22-31. [PMID: 31085421 DOI: 10.1016/j.dnarep.2019.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 03/28/2019] [Accepted: 04/24/2019] [Indexed: 11/20/2022]
Abstract
Werner syndrome is a rare autosomal recessive disorder where Werner (WRN) gene is mutated. Being a nucleolar protein, during DNA damage, WRN translocates at the damage site where its catalytic function is required in DNA repair. Several studies have indicated that WRN acetylation may modulate WRN trafficking and catalytic function (Blander et al., 2002; Lozada et al., 2014). Among the six acetylation sites in WRN protein identified by mass-spectrometry analysis (Li et al., 2010) we here explore the role of acetylation sites in C-terminal of WRN (K1127, K1117, K1389, K1413) because the C- terminal domain is the hub for protein- protein interaction and DNA binding activity (Brosh et al. [4]; Muftuoglu et al., 2008; Huang et al., 2006). To explore their functional activity, we created mutations in these sites by changing the acetylation residue lysine (K) to a non-acetylation residue arginine (R) and expressed them in WRN mutant cell lines. We observed that K1127R and K1117R mutants are sensitive to the DNA damaging agents etoposide and mitomycin C and display deficient DNA repair. Importantly, deacetylation of WRN by SIRT1 (Mammalian Sir2) is necessary for restoration of WRN localization at nucleoli after completion of DNA repair. Among all putative acetylation sites, K1127R, K1117R and the double mutant K1127R/K1117R showed significantly delayed re-entry to the nucleolus after damage recovery, even when SIRT1 is overexpressed. These mutants showed partial interaction with SIRT1 compared to WT WRN. Thus, our results suggest that K1127 and K1117 are the major sites of acetylation, necessary for DNA repair. These results elucidate the mechanism by which SIRT1 regulates WRN trafficking via these acetylation sites during DNA damage.
Collapse
|
17
|
Bi X, Ye Q, Li D, Peng Q, Wang Z, Wu X, Zhang Y, Zhang Q, Jiang F. Inhibition of nucleolar stress response by Sirt1: A potential mechanism of acetylation-independent regulation of p53 accumulation. Aging Cell 2019; 18:e12900. [PMID: 30623565 PMCID: PMC6413664 DOI: 10.1111/acel.12900] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 11/28/2018] [Accepted: 12/08/2018] [Indexed: 02/04/2023] Open
Abstract
The mammalian Sirt1 deacetylase is generally thought to be a nuclear protein, but some pilot studies have suggested that Sirt1 may also be involved in orchestrating nucleolar functions. Here, we show that nucleolar stress response is a ubiquitous cellular reaction that can be induced by different types of stress conditions, and Sirt1 is an endogenous suppressor of nucleolar stress response. Using stable isotope labeling by amino acids in cell culture approach, we have identified a physical interaction of between Sirt1 and the nucleolar protein nucleophosmin, and this protein-protein interaction appears to be necessary for Sirt1 inhibition on nucleolar stress, whereas the deacetylase activity of Sirt1 is not strictly required. Based on the reported prerequisite role of nucleolar stress response in stress-induced p53 protein accumulation, we have also provided evidence suggesting that Sirt1-mediated inhibition on nucleolar stress response may represent a novel mechanism by which Sirt1 can modulate intracellular p53 accumulation independent of lysine deacetylation. This process may represent an alternative mechanism by which Sirt1 regulates functions of the p53 pathway.
Collapse
Affiliation(s)
- Xiaolei Bi
- School of Basic MedicineShandong UniversityJinanShandong ProvinceChina
- Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical SciencesJinanChina
- The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
- Present address:
Department of CardiologyQingdao Municipal HospitalQingdaoShandong ProvinceChina
| | - Qing Ye
- School of Basic MedicineShandong UniversityJinanShandong ProvinceChina
- Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical SciencesJinanChina
- The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Daoyuan Li
- National Glycoengineering Research CenterShandong UniversityJinanChina
| | - Qisheng Peng
- Key Laboratory of Zoonosis ResearchJilin UniversityChangchunJilin ProvinceChina
| | - Zhe Wang
- Division of Endocrinology and MetabolismShandong Provincial Hospital affiliated to Shandong UniversityJinanChina
| | - Xiao Wu
- Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical SciencesJinanChina
- The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Yun Zhang
- Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical SciencesJinanChina
- The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Qunye Zhang
- Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical SciencesJinanChina
- The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Fan Jiang
- School of Basic MedicineShandong UniversityJinanShandong ProvinceChina
- Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical SciencesJinanChina
- The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| |
Collapse
|
18
|
|
19
|
Sen Gupta A, Joshi G, Pawar S, Sengupta K. Nucleolin modulates compartmentalization and dynamics of histone 2B-ECFP in the nucleolus. Nucleus 2018; 9:350-367. [PMID: 29943658 PMCID: PMC6165600 DOI: 10.1080/19491034.2018.1471936] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Eukaryotic cells have 2 to 3 discrete nucleoli required for ribosome synthesis. Nucleoli are phase separated nuclear sub-organelles. Here we examined the role of nuclear Lamins and nucleolar factors in modulating the compartmentalization and dynamics of histone 2B (H2B-ECFP) in the nucleolus. Live imaging and Fluorescence Recovery After Photobleaching (FRAP) of labelled H2B, showed that the depletion of Lamin B1, Fibrillarin (FBL) or Nucleostemin (GNL3), enhances H2B-ECFP mobility in the nucleolus. Furthermore, Nucleolin knockdown significantly decreases H2B-ECFP compartmentalization in the nucleolus, while H2B-ECFP residence and mobility in the nucleolus was prolonged upon Nucleolin overexpression. Co-expression of N-terminal and RNA binding domain (RBD) deletion mutants of Nucleolin or inhibiting 45S rRNA synthesis reduces the sequestration of H2B-ECFP in the nucleolus. Taken together, these studies reveal a crucial role of Nucleolin-rRNA complex in modulating the compartmentalization, stability and dynamics of H2B within the nucleolus.
Collapse
Affiliation(s)
- Ayantika Sen Gupta
- Biology, Indian Institute of Science Education and Research (IISER), Pune, India
| | - Gaurav Joshi
- Biology, Indian Institute of Science Education and Research (IISER), Pune, India
| | - Sumit Pawar
- Biology, Indian Institute of Science Education and Research (IISER), Pune, India
| | - Kundan Sengupta
- Biology, Indian Institute of Science Education and Research (IISER), Pune, India
| |
Collapse
|
20
|
Brun S, Abella N, Berciano MT, Tapia O, Jaumot M, Freire R, Lafarga M, Agell N. SUMO regulates p21Cip1 intracellular distribution and with p21Cip1 facilitates multiprotein complex formation in the nucleolus upon DNA damage. PLoS One 2017; 12:e0178925. [PMID: 28582471 PMCID: PMC5459497 DOI: 10.1371/journal.pone.0178925] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/22/2017] [Indexed: 01/06/2023] Open
Abstract
We previously showed that p21Cip1 transits through the nucleolus on its way from the nucleus to the cytoplasm and that DNA damage inhibits this transit and induces the formation of p21Cip1-containing intranucleolar bodies (INoBs). Here, we demonstrate that these INoBs also contain SUMO-1 and UBC9, the E2 SUMO-conjugating enzyme. Furthermore, whereas wild type SUMO-1 localized in INoBs, a SUMO-1 mutant, which is unable to conjugate with proteins, does not, suggesting the presence of SUMOylated proteins at INoBs. Moreover, depletion of the SUMO-conjugating enzyme UBC9 or the sumo hydrolase SENP2 changed p21Cip1 intracellular distribution. In addition to SUMO-1 and p21Cip1, cell cycle regulators and DNA damage checkpoint proteins, including Cdk2, Cyclin E, PCNA, p53 and Mdm2, and PML were also detected in INoBs. Importantly, depletion of UBC9 or p21Cip1 impacted INoB biogenesis and the nucleolar accumulation of the cell cycle regulators and DNA damage checkpoint proteins following DNA damage. The impact of p21Cip1 and SUMO-1 on the accumulation of proteins in INoBs extends also to CRM1, a nuclear exportin that is also important for protein translocation from the cytoplasm to the nucleolus. Thus, SUMO and p21Cip1 regulate the transit of proteins through the nucleolus, and that disruption of nucleolar export by DNA damage induces SUMO and p21Cip1 to act as hub proteins to form a multiprotein complex in the nucleolus.
Collapse
Affiliation(s)
- Sonia Brun
- Departament Biomedicina, Universitat de Barcelona, IDIBAPS, Barcelona, Spain
| | - Neus Abella
- Departament Biomedicina, Universitat de Barcelona, IDIBAPS, Barcelona, Spain
| | - Maria T. Berciano
- Departamento de Anatomía y Biología Celular, Universidad de Cantabria-IDIVAL, Santander, Spain
| | - Olga Tapia
- Departamento de Anatomía y Biología Celular, Universidad de Cantabria-IDIVAL, Santander, Spain
| | - Montserrat Jaumot
- Departament Biomedicina, Universitat de Barcelona, IDIBAPS, Barcelona, Spain
| | - Raimundo Freire
- Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Tecnologías Biomédicas, Tenerife, Spain
| | - Miguel Lafarga
- Departamento de Anatomía y Biología Celular, Universidad de Cantabria-IDIVAL, Santander, Spain
| | - Neus Agell
- Departament Biomedicina, Universitat de Barcelona, IDIBAPS, Barcelona, Spain
| |
Collapse
|
21
|
Slomnicki LP, Hallgren J, Vashishta A, Smith SC, Ellis SR, Hetman M. Proapoptotic Requirement of Ribosomal Protein L11 in Ribosomal Stress-Challenged Cortical Neurons. Mol Neurobiol 2016; 55:538-553. [PMID: 27975169 DOI: 10.1007/s12035-016-0336-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 11/30/2016] [Indexed: 01/05/2023]
Abstract
While impaired ribosomal biogenesis is observed in neurodegenerative diseases, its pathogenic contributions are not clear. For instance, it is well established that in rodent neurons, genetic inhibition of RNA-polymerase 1 that transcribes rRNA results in structural disruption of the nucleolus, neuronal apoptosis, and neurodegeneration. However, in most neurodegenerative diseases, nucleolar morphology is unaffected. It is reported here that in primary cortical neurons from newborn rats, inhibition of ribosomal biogenesis by shRNA-mediated knockdowns of several ribosomal proteins including S6, S14, or L4 resulted in p53-mediated apoptosis despite absence of structural disruption of the nucleolus. Conversely, knockdown of the RP L11, which in nonneuronal systems mediates p53 activation downstream of ribosomal stress, protected neurons against inhibition of ribosomal biogenesis but not staurosporine. Moreover, overexpression of L11 enhanced p53-driven transcription and increased neuronal apoptosis. In addition, inhibition of p53, or L11 knockdown, blocked apoptosis in response to the RNA analog 5-fluorouridine which perturbed nucleolar structure, inhibited ribosomal synthesis, and activated p53. Although the DNA double-strand break (DSB) inducer etoposide activated p53, nucleolar structure appeared intact. However, by activating the DNA damage response kinase ATM, etoposide increased 47S pre-rRNA levels, and enhanced nucleolar accumulation of nascent RNA, suggesting slower rRNA processing and/or increased Pol1 activity. In addition, shL11 reduced etoposide-induced apoptosis. Therefore, seemingly normal morphology of the neuronal nucleolus does not exclude presence of ribosomal stress. Conversely, targeting the ribosomal stress-specific signaling mediators including L11 offers a novel approach to uncover neurodegenerative contributions of deregulated ribosomal synthesis as exemplified in DSB-challenged neurons.
Collapse
Affiliation(s)
- Lukasz P Slomnicki
- KY Spinal Cord Injury Research Center and the Department of Neurological Surgery, University of Louisville, 511 S. Floyd St., MDR616, Louisville, KY, 40292, USA
| | - Justin Hallgren
- KY Spinal Cord Injury Research Center and the Department of Neurological Surgery, University of Louisville, 511 S. Floyd St., MDR616, Louisville, KY, 40292, USA.,Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40292, USA
| | - Aruna Vashishta
- KY Spinal Cord Injury Research Center and the Department of Neurological Surgery, University of Louisville, 511 S. Floyd St., MDR616, Louisville, KY, 40292, USA
| | - Scott C Smith
- KY Spinal Cord Injury Research Center and the Department of Neurological Surgery, University of Louisville, 511 S. Floyd St., MDR616, Louisville, KY, 40292, USA
| | - Steven R Ellis
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, KY, 40292, USA
| | - Michal Hetman
- KY Spinal Cord Injury Research Center and the Department of Neurological Surgery, University of Louisville, 511 S. Floyd St., MDR616, Louisville, KY, 40292, USA. .,Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40292, USA.
| |
Collapse
|
22
|
Chang TH, Wang SS, Chen LW, Shih YJ, Chang LK, Liu ST, Chang PJ. Regulation of the Abundance of Kaposi's Sarcoma-Associated Herpesvirus ORF50 Protein by Oncoprotein MDM2. PLoS Pathog 2016; 12:e1005918. [PMID: 27698494 PMCID: PMC5047794 DOI: 10.1371/journal.ppat.1005918] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/06/2016] [Indexed: 01/10/2023] Open
Abstract
The switch between latency and the lytic cycle of Kaposi's sarcoma-associated herpesvirus (KSHV) is controlled by the expression of virally encoded ORF50 protein. Thus far, the regulatory mechanism underlying the protein stability of ORF50 is unknown. Our earlier studies have demonstrated that a protein abundance regulatory signal (PARS) at the ORF50 C-terminal region modulates its protein abundance. The PARS region consists of PARS-I (aa 490-535) and PARS-II (aa 590-650), and mutations in either component result in abundant expression of ORF50. Here, we show that ORF50 protein is polyubiquitinated and its abundance is controlled through the proteasomal degradation pathway. The PARS-I motif mainly functions as a nuclear localization signal in the control of ORF50 abundance, whereas the PARS-II motif is required for the binding of ubiquitin enzymes in the nucleus. We find that human oncoprotein MDM2, an ubiquitin E3 ligase, is capable of interacting with ORF50 and promoting ORF50 degradation in cells. The interaction domains between both proteins are mapped to the PARS region of ORF50 and the N-terminal 220-aa region of MDM2. Additionally, we identify lysine residues at positions 152 and 154 in the N-terminal domain of ORF50 critically involved in MDM2-mediated downregulation of ORF50 levels. Within KSHV-infected cells, the levels of MDM2 were greatly reduced during viral lytic cycle and genetic knockdown of MDM2 in these cells favored the enhancement of ORF50 expression, supporting that MDM2 is a negative regulator of ORF50 expression. Collectively, the study elucidates the regulatory mechanism of ORF50 stability and implicates that MDM2 may have a significant role in the maintenance of viral latency by lowering basal level of ORF50.
Collapse
Affiliation(s)
- Tzu-Hsuan Chang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | - Shie-Shan Wang
- Department of Pediatric Surgery, Chang-Gung Memorial Hospital, Chiayi, Taiwan
| | - Lee-Wen Chen
- Department of Respiratory Care, Chang-Gung University of Science and Technology, Chiayi, Taiwan
| | - Ying-Ju Shih
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | - Li-Kwan Chang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Shih-Tung Liu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang-Gung University, Taoyuan, Taiwan
- Department of Microbiology and Immunology, College of Medicine, Chang-Gung University, Taoyuan, Taiwan
- * E-mail: (STL); (PJC)
| | - Pey-Jium Chang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang-Gung University, Taoyuan, Taiwan
- Department of Nephrology, Chang-Gung Memorial Hospital, Chiayi, Taiwan
- * E-mail: (STL); (PJC)
| |
Collapse
|
23
|
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
|
24
|
Slomnicki LP, Malinowska A, Kistowski M, Palusinski A, Zheng JJ, Sepp M, Timmusk T, Dadlez M, Hetman M. Nucleolar Enrichment of Brain Proteins with Critical Roles in Human Neurodevelopment. Mol Cell Proteomics 2016; 15:2055-75. [PMID: 27053602 DOI: 10.1074/mcp.m115.051920] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Indexed: 11/06/2022] Open
Abstract
To study nucleolar involvement in brain development, the nuclear and nucleolar proteomes from the rat cerebral cortex at postnatal day 7 were analyzed using LC-MS/iTRAQ methodology. Data of the analysis are available via ProteomeXchange with identifier PXD002188. Among 504 candidate nucleolar proteins, the overrepresented gene ontology terms included such cellular compartmentcategories as "nucleolus", "ribosome" and "chromatin". Consistent with such classification, the most overrepresented functional gene ontology terms were related to RNA metabolism/ribosomal biogenesis, translation, and chromatin organization. Sixteen putative nucleolar proteins were associated with neurodevelopmental phenotypes in humans. Microcephaly and/or cognitive impairment were the most common phenotypic manifestations. Although several such proteins have links to ribosomal biogenesis and/or genomic stability/chromatin structure (e.g. EMG1, RPL10, DKC1, EIF4A3, FLNA, SMC1, ATRX, MCM4, NSD1, LMNA, or CUL4B), others including ADAR, LARP7, GTF2I, or TCF4 have no such connections known. Although neither the Alazami syndrome-associated LARP7nor the Pitt-Hopkins syndrome-associated TCF4 were reported in nucleoli of non-neural cells, in neurons, their nucleolar localization was confirmed by immunostaining. In cultured rat hippocampal neurons, knockdown of LARP7 reduced both perikaryal ribosome content and general protein synthesis. Similar anti-ribosomal/anti-translation effects were observed after knockdown of the ribosomal biogenesis factor EMG1 whose deficiency underlies Bowen-Conradi syndrome. Finally, moderate reduction of ribosome content and general protein synthesis followed overexpression of two Pitt-Hopkins syndrome mutant variants of TCF4. Therefore, dysregulation of ribosomal biogenesis and/or other functions of the nucleolus may disrupt neurodevelopment resulting in such phenotypes as microcephaly and/or cognitive impairment.
Collapse
Affiliation(s)
- Lukasz P Slomnicki
- From the ‡Kentucky Spinal Cord Injury Research Center and the Departments of Neurological Surgery and
| | - Agata Malinowska
- ¶Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Michal Kistowski
- ¶Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Antoni Palusinski
- ‖Department of Systems Biology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Jing-Juan Zheng
- From the ‡Kentucky Spinal Cord Injury Research Center and the Departments of Neurological Surgery and
| | - Mari Sepp
- **Department of Gene Technology, Tallinn University of Technology, Tallinn, Estonia
| | - Tonis Timmusk
- **Department of Gene Technology, Tallinn University of Technology, Tallinn, Estonia
| | - Michal Dadlez
- ¶Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Michal Hetman
- From the ‡Kentucky Spinal Cord Injury Research Center and the Departments of Neurological Surgery and §Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky;
| |
Collapse
|
25
|
Cabrera CM, Fernández-Grande E, Urra JM. Serological profile and clinical features of nucleolar antinuclear pattern in patients with systemic lupus erythematosus from southwestern Spain. Lupus 2016; 25:980-7. [DOI: 10.1177/0961203316629557] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/07/2016] [Indexed: 12/29/2022]
Abstract
Nucleolar staining of antinuclear antibodies (ANAs) is not exclusive to patients suffering systemic sclerosis (SSc) since it can occur in other autoimmune diseases, such as systemic lupus erythematosus (SLE). The nucleolar ANA pattern presents a low incidence in patients with SLE, with less than 9% reported in some studies. The significance of nucleolar staining and antinucleolar antibodies (ANoA) in SLE is still unknown, as is its association with clinical manifestations. To address these issues, a case-control study was carried out. Twenty-eight cases of SLE with nucleolar staining were enrolled, as well as 73 controls with no nucleolar staining and different ANA patterns (homogeneous, speckled, and combined homogeneous and speckled). The homogeneous nucleolar pattern was the most frequent (27 out of 28), and in 75% was combined with other ANA patterns. The anti-double stranded DNA antibodies showed no differences between the two groups of patients, nor the auto-antibodies detected by line immunoassay (LIA). However, we have found an increased frequency of anti-PM-Scl antibodies with respect to the controls ( p = 0.02), in addition to the association between Raynaud's phenomenon (RP) and anti-PM-Scl antibodies (OR = 20.72, 95% CI 1.33–323.19, p = 0.03). Moreover, the cases of SLE showed a 7.78-fold increase in the risk of developing cancer (95%, CI 1.85–32.75, p = 0.005) with respect to the control group. Taken together these findings suggest that nucleolar staining represents a comorbidity factor in patients with SLE, although its significance must still be determined.
Collapse
Affiliation(s)
- C M Cabrera
- Immunology Section, Servicio de Hematología, Hospital de Jerez de la Frontera, Cádiz, Spain
| | - E Fernández-Grande
- Immunology Section, Servicio de Análisis Clínicos, Hospital General Universitario de Ciudad Real, Spain
| | - J M Urra
- Immunology Section, Servicio de Análisis Clínicos, Hospital General Universitario de Ciudad Real, Spain
| |
Collapse
|
26
|
Han KJ, Foster D, Harhaj EW, Dzieciatkowska M, Hansen K, Liu CW. Monoubiquitination of survival motor neuron regulates its cellular localization and Cajal body integrity. Hum Mol Genet 2016; 25:1392-405. [PMID: 26908624 DOI: 10.1093/hmg/ddw021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 01/25/2016] [Indexed: 12/13/2022] Open
Abstract
Low levels of the survival motor neuron (SMN) protein cause spinal muscular atrophy, the leading genetic disorder for infant mortality. SMN is ubiquitously expressed in various cell types and localizes in both the cytoplasm and the nucleus, where it concentrates in two subnuclear structures termed Cajal body (CB) and gems. In addition, SMN can also be detected in the nucleolus of neurons. Mechanisms that control SMN sorting in the cell remain largely unknown. Here, we report that the ubiquitin (Ub) ligase Itch directly interacts with and monoubiquitinates SMN. Monoubiquitination of SMN has a mild effect on promoting proteasomal degradation of SMN. We generated two SMN mutants, SMN(K0), in which all lysines are mutated to arginines and thereby abolishing SMN ubiquitination, and Ub-SMN(K0), in which a single Ub moiety is fused at the N-terminus of SMN(K0) and thereby mimicking SMN monoubiquitination. Immunostaining assays showed that SMN(K0) mainly localizes in the nucleus, whereas Ub-SMN(K0) localizes in both the cytoplasm and the nucleolus in neuronal SH-SY5Y cells. Interestingly, canonical CB foci and coilin/small nuclear ribonucleoprotein (snRNP) co-localization are significantly impaired in SH-SY5Y cells stably expressing SMN(K0) or Ub-SMN(K0). Thus, our studies discover that Itch monoubiquitinates SMN and monoubiquitination of SMN plays an important role in regulating its cellular localization. Moreover, mislocalization of SMN disrupts CB integrity and likely impairs snRNP maturation.
Collapse
Affiliation(s)
- Ke-Jun Han
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80015, USA and
| | - Daniel Foster
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80015, USA and
| | - Edward W Harhaj
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80015, USA and
| | - Kirk Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80015, USA and
| | - Chang-Wei Liu
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80015, USA and
| |
Collapse
|
27
|
RRP12 is a crucial nucleolar protein that regulates p53 activity in osteosarcoma cells. Tumour Biol 2015; 37:4351-8. [PMID: 26499779 DOI: 10.1007/s13277-015-4062-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/04/2015] [Indexed: 10/22/2022] Open
Abstract
RRP12 (ribosomal RNA processing 12 homolog), a nucleolar protein, plays important roles in cell cycle progression and the response to deoxyribonucleic acid (DNA) damage in yeast cells. However, its role has not been investigated in mammalian cells that possess p53, which has close functional association to nucleolus. We explored the role of RRP12 in nucleolar stress condition using an osteosarcoma cell line, U2OS. To induce DNA damage and nucleolar disruption, two cytotoxic drugs, doxorubicin and actinomycin D were used. Cytotoxic stress resulted nucleolar disruption induced cell cycle arrest and apoptosis in U2OS cells. However, RRP12 overexpression promoted resistance to cytotoxic stress. In contrast, RRP12 silencing enhanced susceptibility to cytotoxic stress. During drug treatment, p53 activity and cell death were suppressed by RRP12 overexpression but promoted by RRP12 silencing. This study demonstrated that RRP12 was crucial for cell survival during cytotoxic stress via the repression of p53 stability. Thus, targeting RRP12 may enhance chemotherapeutic effect in cancers.
Collapse
|
28
|
Wang C, Feng W, Zhang C. The Expression and Function of NUMB in Endometrial Cancer and the Interaction with HDM2 and P53. J Cancer 2015; 6:1030-40. [PMID: 26366217 PMCID: PMC4565853 DOI: 10.7150/jca.11970] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/12/2015] [Indexed: 02/06/2023] Open
Abstract
Background: Since more and more evidences support that NUMB orchestrates many cell physiological and pathological processes of diseases including cancer, based on our previous work, we studied deeply the function of NUMB in endometrial cancer (EC) and tried to understand the mechanism of NUMB's nucleus translocation which might be relative to the occurrence of EC and will contribute to find a new targeting therapeutic strategy for EC. Methods: Immunohistochemistry was employed to test NUMB and HDM2 expression in endometrial cancer tissue from clinical patients. CCK-8 assay, cell cycle tested by Flow cytometer and PCNA determined by RT-PCR were employed to test the effects of NUMB on cell proliferation and apoptosis. In order to investigate the mechanism how NUMB, HDM2 and p53 interact in EC cell, western blot, Co-IP and immunofluorescent were used to observe the combination and location of NUMB, HDM2 and p53 as well as the interaction among them. Results: Both NUMB and HDM2 expressed greater in endometrial cancer tissues than in normal endometrial tissues. Overexpression of NUMB induced apoptosis in Ishikawa cell while inhibition of NUMB increased cell proliferation. NUMB could combine HDM2 and p53, moreover the PTB domain of NUMB is the main site combining with p53. The effects of NUMB in cell was closely associated with p53. Not only NUMB regulated P53 expression level but also NUMB acts depending on P53, in turn p53 impacted the NUMB level as a feedback. Overexpression of NUMB could not bring itself into nuclear. Both siHDM2 and siP53 didn't bring NUMB into nucleus, However overexpression of HDM2 and p53 increased the NUMB level in nucleus, and the NUMB nuclear location induced by overexpression of HDM2 was stronger than that of p53 overexpression. Conclusions: Based on present data, we think NUMB acts as an anti-oncogene role and could regulate p53 level and function in endometrial cancer like in other cancers, meanwhile, the function of NUMB depend on P53. On the other hand, the location of NUMB could be regulated mainly by HDM2. So far we are not able to explain why endometrial cancer patients had high NUMB expression level since NUMB was regarded as a tumor suppressor, which is worthy studying further to explore underlying mechanism.
Collapse
Affiliation(s)
- Chao Wang
- Department of Obstetrics & Gynecology, Obstetrics & Gynecology hospital of Fu Dan University, Shang Hai, China
| | - Weiwei Feng
- Department of Obstetrics & Gynecology, Obstetrics & Gynecology hospital of Fu Dan University, Shang Hai, China
| | - Chuyao Zhang
- Department of Obstetrics & Gynecology, Obstetrics & Gynecology hospital of Fu Dan University, Shang Hai, China
| |
Collapse
|
29
|
Goudarzi KM, Nistér M, Lindström MS. mTOR inhibitors blunt the p53 response to nucleolar stress by regulating RPL11 and MDM2 levels. Cancer Biol Ther 2015; 15:1499-514. [PMID: 25482947 DOI: 10.4161/15384047.2014.955743] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a master regulator of cell growth through its ability to stimulate ribosome biogenesis and mRNA translation. In contrast, the p53 tumor suppressor negatively controls cell growth and is activated by a wide range of insults to the cell. The mTOR and p53 signaling pathways are connected by a number of different mechanisms. Chemotherapeutics that inhibit ribosome biogenesis often induce nucleolar stress and activation of p53. Here we have investigated how the p53 response to nucleolar stress is affected by simultaneous mTOR inhibition in osteosarcoma and glioma cell lines. We found that inhibitors of the mTOR pathway including rapamycin, wortmannin, and caffeine blunted the p53 response to nucleolar stress induced by actinomycin D. Synthetic inhibitors of mTOR (temsirolimus, LY294.002 and PP242) also impaired actinomycin D triggered p53 stabilization and induction of p21. Ribosomal protein (RPL11) is known to be required for p53 protein stabilization following nucleolar stress. Treatment of cells with mTOR inhibitors may lead to reduced synthesis of RPL11 and thereby destabilize p53. We found that rapamycin mimicked the effect of RPL11 depletion in terms of blunting the p53 response to nucleolar stress. However, the extent to which the levels of p53 and RPL11 were reduced by rapamycin varied between cell lines. Additional mechanisms whereby rapamycin blunts the p53 response to nucleolar stress are likely to be involved. Indeed, rapamycin increased the levels of endogenous MDM2 despite inhibition of its phosphorylation at Ser-166. Our findings may have implications for the design of combinatorial cancer treatments with mTOR pathway inhibitors.
Collapse
Key Words
- 5-FU, 5-fluorouracil
- Act D, actinomycin D
- BrdU, bromodeoxyuridine
- CHX, cycloheximide
- DMSO, dimethylsulphoxide
- DOX, doxorubicin
- EGCG, epigallocatechin-3-gallate
- FACS, fluorescence-activated cell sorting
- MPA, mycophenolic acid
- MTT, (3-[4, 5-dimethylthiazol-2-yl]-2, 5 diphenyl tetrazolium bromide)
- PI, propidium iodide
- actinomycin D
- caffeine
- glioma
- mTOR
- mTOR, mechanistic target of rapamycin
- nutlin-3
- p21
- p53
- rapamycin
- ribosomal protein L11
- ribosome biogenesis
Collapse
Affiliation(s)
- Kaveh M Goudarzi
- a Department of Oncology-Pathology; Karolinska Institutet; Cancer Center Karolinska ; Karolinska University Hospital ; Stockholm , Sweden
| | | | | |
Collapse
|
30
|
Aldrich JC, Maggert KA. Transgenerational inheritance of diet-induced genome rearrangements in Drosophila. PLoS Genet 2015; 11:e1005148. [PMID: 25885886 PMCID: PMC4401788 DOI: 10.1371/journal.pgen.1005148] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 03/17/2015] [Indexed: 12/17/2022] Open
Abstract
Ribosomal RNA gene (rDNA) copy number variation modulates heterochromatin formation and influences the expression of a large fraction of the Drosophila genome. This discovery, along with the link between rDNA, aging, and disease, high-lights the importance of understanding how natural rDNA copy number variation arises. Pursuing the relationship between rDNA expression and stability, we have discovered that increased dietary yeast concentration, emulating periods of dietary excess during life, results in somatic rDNA instability and copy number reduction. Modulation of Insulin/TOR signaling produces similar results, indicating a role for known nutrient sensing signaling pathways in this process. Furthermore, adults fed elevated dietary yeast concentrations produce offspring with fewer rDNA copies demonstrating that these effects also occur in the germline, and are transgenerationally heritable. This finding explains one source of natural rDNA copy number variation revealing a clear long-term consequence of diet.
Collapse
Affiliation(s)
- John C. Aldrich
- Department of Biology, College of Science, Texas A&M University, College Station, Texas, United States of America
| | - Keith A. Maggert
- Department of Biology, College of Science, Texas A&M University, College Station, Texas, United States of America
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, Arizona, United States of America
- * E-mail:
| |
Collapse
|
31
|
Pérez-García C, Rouxel J, Akcha F. Development of a comet-FISH assay for the detection of DNA damage in hemocytes of Crassostrea gigas. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 161:189-195. [PMID: 25710447 DOI: 10.1016/j.aquatox.2015.01.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 01/14/2015] [Accepted: 01/25/2015] [Indexed: 06/04/2023]
Abstract
In this work, the DNA-damaging effect of hydrogen peroxide on the structural integrity of nucleolar organizer regions (NORs) was studied for the first time by comet-FISH in the Pacific oyster Crassostrea gigas. Global DNA damage was assessed in hemocytes using an alkaline version of the comet assay. Next, NOR sensitivity was analyzed by mapping major rDNA repeat unit by fluorescence in situ hybridization (FISH) on the same comet slides. Exposure of hemocytes to 100 μM of hydrogen peroxide induced a significant increase in both DNA damage and number of FISH-signals of major ribosomal genes versus the control. Moreover, a significant positive correlation was shown between DNA damage as measured by the comet assay (percentage of DNA in comet tail) and the number of signals present in comet tails. This study demonstrates the potential value of the comet-FISH assay for the study of DNA damage induced by genotoxicant exposure of target genes. It offers a perspective for better understanding the impact of genotoxicity on animal physiology and fitness.
Collapse
Affiliation(s)
- C Pérez-García
- IFREMER, Department of Biogeochemistry and Ecotoxicology, Laboratory of Ecotoxicology, Rue de l'Ile d'Yeu, BP 21105, 44311 Nantes Cedex 03, France.
| | - J Rouxel
- IFREMER, Department of Biogeochemistry and Ecotoxicology, Laboratory of Ecotoxicology, Rue de l'Ile d'Yeu, BP 21105, 44311 Nantes Cedex 03, France
| | - F Akcha
- IFREMER, Department of Biogeochemistry and Ecotoxicology, Laboratory of Ecotoxicology, Rue de l'Ile d'Yeu, BP 21105, 44311 Nantes Cedex 03, France.
| |
Collapse
|
32
|
Ehm P, Nalaskowski MM, Wundenberg T, Jücker M. The tumor suppressor SHIP1 colocalizes in nucleolar cavities with p53 and components of PML nuclear bodies. Nucleus 2015; 6:154-64. [PMID: 25723258 DOI: 10.1080/19491034.2015.1022701] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The inositol 5-phosphatase SHIP1 is a negative regulator of signaling processes in haematopoietic cells. By converting PI(3,4,5)P3 to PtdIns(3,4)P2 at the plasma membrane, SHIP1 modifies PI3-kinase mediated signaling. We have recently demonstrated that SHIP1 is a nucleo-cytoplasmic shuttling protein and SHIP1 nuclear puncta partially colocalize with FLASH, a component of nuclear bodies. In this study, we demonstrate that endogenous SHIP1 localizes to intranucleolar regions of both normal and leukemic haematopoietic cells. In addition, we report that ectopically expressed SHIP1 accumulates in nucleolar cavities and colocalizes with the tumor suppressor protein p53 and components of PML nuclear bodies (e.g. SP100, SUMO-1 and CK2). Moreover, SHIP1 also colocalizes in nucleolar cavities with components of the ubiquitin-proteasome pathway. By using confocal microscopy data, we generated 3D-models revealing the enormous extent of the SHIP1 aggresomes in the nucleolus. Furthermore, treatment of cells with the proteasome inhibitor MG132 causes an enlargement of nucleolar SHIP1 containing structures. Unexpectedly, this accumulation can be partially prevented by treatment with the inhibitor of nuclear protein export Leptomycin B. In recent years, several proteins aggregating in nucleolar cavities were shown to be key factors of neurodegenerative diseases and cancerogenesis. Our findings support current relevance of nuclear localized SHIP1.
Collapse
Key Words
- DFC, dense fibrillar component
- DIC, Differential interference contrast
- EGFP, enhanced green fluorescent protein
- FC, fibrillar center
- GC, granular component
- LMB, leptomycin B
- MG132
- NES, nuclear export signal
- PBMC, Peripheral Blood Mononuclear Cell
- PML bodies
- PML, Promyelocytic Leukemia
- PtdIns(3, 4)P2, phosphatidylinositol-(3, 4)-bisphosphate
- PtdIns(3, 4, 5)P3, phosphatidylinositol-(3, 4, 5)-trisphosphate
- RNA pol, RNA polymerase
- SHIP1
- SHIP1, src homology 2 domain-containing inositol phosphatase 1
- UPP, ubiquitin-proteasome pathway.
- aggresome
- cancer
- leptomycin B
- nucleolar cavities
- nucleus
- p53
- ubiquitin proteasome pathway
Collapse
Affiliation(s)
- Patrick Ehm
- a Institute of Biochemistry and Signal Transduction ; University Medical Center Hamburg-Eppendorf ; Hamburg , Germany
| | | | | | | |
Collapse
|
33
|
Barranger A, Benabdelmouna A, Dégremont L, Burgeot T, Akcha F. Parental exposure to environmental concentrations of diuron leads to aneuploidy in embryos of the Pacific oyster, as evidenced by fluorescent in situ hybridization. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 159:36-43. [PMID: 25498420 DOI: 10.1016/j.aquatox.2014.11.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 10/29/2014] [Accepted: 11/13/2014] [Indexed: 06/04/2023]
Abstract
Changes in normal chromosome numbers (i.e. aneuploidy) due to abnormal chromosome segregation may arise either spontaneously or as a result of chemical/radiation exposure, particularly during cell division. Coastal ecosystems are continuously subjected to various contaminants originating from urban, industrial and agricultural activities. Genotoxicity is common to several families of major environmental pollutants, including pesticides, which therefore represent a potential important environmental hazard for marine organisms. A previous study demonstrated the vertical transmission of DNA damage by subjecting oyster genitors to short-term exposure to the herbicide diuron at environmental concentrations during gametogenesis. In this paper, Fluorescent in situ hybridization (FISH) was used to further characterize diuron-induced DNA damage at the chromosomal level. rDNA genes (5S and 18-5.8-28S), previously mapped onto Crassostrea gigas chromosomes 4, 5 and 10, were used as probes on the interphase nuclei of embryo preparations. Our results conclusively show higher aneuploidy (hypo- or hyperdiploidy) level in embryos from diuron-exposed genitors, with damage to the three studied chromosomal regions. This study suggests that sexually developing oysters are vulnerable to diuron exposure, incurring a negative impact on reproductive success and oyster recruitment.
Collapse
Affiliation(s)
- Audrey Barranger
- Ifremer, SG2M, Laboratory of Genetics and Pathology of Marine Molluscs, Avenue de Mus du Loup, 17390 La Tremblade, France; Ifremer, Department of Biogeochemistry and Ecotoxicology, Laboratory of Ecotoxicology, Rue de l'Ile d'Yeu, BP 21105, 44311 Nantes Cedex 03, France.
| | - Abdellah Benabdelmouna
- Ifremer, SG2M, Laboratory of Genetics and Pathology of Marine Molluscs, Avenue de Mus du Loup, 17390 La Tremblade, France.
| | - Lionel Dégremont
- Ifremer, SG2M, Laboratory of Genetics and Pathology of Marine Molluscs, Avenue de Mus du Loup, 17390 La Tremblade, France
| | - Thierry Burgeot
- Ifremer, Department of Biogeochemistry and Ecotoxicology, Laboratory of Ecotoxicology, Rue de l'Ile d'Yeu, BP 21105, 44311 Nantes Cedex 03, France
| | - Farida Akcha
- Ifremer, Department of Biogeochemistry and Ecotoxicology, Laboratory of Ecotoxicology, Rue de l'Ile d'Yeu, BP 21105, 44311 Nantes Cedex 03, France
| |
Collapse
|
34
|
Batnasan E, Wang R, Wen J, Ke Y, Li X, Bohio AA, Zeng X, Huo H, Han L, Boldogh I, Ba X. 17-beta estradiol inhibits oxidative stress-induced accumulation of AIF into nucleolus and PARP1-dependent cell death via estrogen receptor alpha. Toxicol Lett 2015; 232:1-9. [DOI: 10.1016/j.toxlet.2014.09.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/03/2014] [Accepted: 09/28/2014] [Indexed: 12/13/2022]
|
35
|
Wang W, Nag S, Zhang X, Wang MH, Wang H, Zhou J, Zhang R. Ribosomal proteins and human diseases: pathogenesis, molecular mechanisms, and therapeutic implications. Med Res Rev 2014; 35:225-85. [PMID: 25164622 DOI: 10.1002/med.21327] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ribosomes are essential components of the protein synthesis machinery. The process of ribosome biogenesis is well organized and tightly regulated. Recent studies have shown that ribosomal proteins (RPs) have extraribosomal functions that are involved in cell proliferation, differentiation, apoptosis, DNA repair, and other cellular processes. The dysfunction of RPs has been linked to the development and progression of hematological, metabolic, and cardiovascular diseases and cancer. Perturbation of ribosome biogenesis results in ribosomal stress, which triggers activation of the p53 signaling pathway through RPs-MDM2 interactions, resulting in p53-dependent cell cycle arrest and apoptosis. RPs also regulate cellular functions through p53-independent mechanisms. We herein review the recent advances in several forefronts of RP research, including the understanding of their biological features and roles in regulating cellular functions, maintaining cell homeostasis, and their involvement in the pathogenesis of human diseases. We also highlight the translational potential of this research for the identification of molecular biomarkers, and in the discovery and development of novel treatments for human diseases.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, 79106; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, 79106
| | | | | | | | | | | | | |
Collapse
|
36
|
Rodríguez JA. Interplay between nuclear transport and ubiquitin/SUMO modifications in the regulation of cancer-related proteins. Semin Cancer Biol 2014; 27:11-9. [DOI: 10.1016/j.semcancer.2014.03.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 03/22/2014] [Accepted: 03/25/2014] [Indexed: 11/25/2022]
|
37
|
Yoshimura M, Ishizawa J, Ruvolo V, Dilip A, Quintás-Cardama A, McDonnell TJ, Neelapu SS, Kwak LW, Shacham S, Kauffman M, Tabe Y, Yokoo M, Kimura S, Andreeff M, Kojima K. Induction of p53-mediated transcription and apoptosis by exportin-1 (XPO1) inhibition in mantle cell lymphoma. Cancer Sci 2014; 105:795-801. [PMID: 24766216 PMCID: PMC4106990 DOI: 10.1111/cas.12430] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 04/21/2014] [Accepted: 04/22/2014] [Indexed: 12/17/2022] Open
Abstract
The nuclear transporter exportin-1 (XPO1) is highly expressed in mantle cell lymphoma (MCL) cells, and is believed to be associated with the pathogenesis of this disease. XPO1-selective inhibitors of nuclear export (SINE) compounds have been shown to induce apoptosis in MCL cells. Given that p53 is a cargo protein of XPO1, we sought to determine the significance of p53 activation through XPO1 inhibition in SINE-induced apoptosis of MCL cells. We investigated the prognostic impact of XPO1 expression in MCL cells using Oncomine analysis. The significance of p53 mutational/functional status on sensitivity to XPO1 inhibition in cell models and primary MCL samples, and the functional role of p53-mediated apoptosis signaling, were also examined. Increased XPO1 expression was associated with poor prognosis in MCL patients. The XPO1 inhibitor KPT-185 induced apoptosis in MCL cells through p53-dependent and -independent mechanisms, and p53 status was a critical determinant of its apoptosis induction. The KPT-185-induced, p53-mediated apoptosis in the MCL cells occurred in a transcription-dependent manner. Exportin-1 appears to influence patient survival in MCL, and the SINE XPO1 antagonist KPT-185 effectively activates p53-mediated transcription and apoptosis, which would provide a novel strategy for the therapy of MCL.
Collapse
MESH Headings
- Acrylates/pharmacology
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Gene Expression Regulation, Neoplastic
- Genes, p53
- Humans
- Karyopherins/genetics
- Karyopherins/metabolism
- Lymphoma, Mantle-Cell/genetics
- Lymphoma, Mantle-Cell/mortality
- Lymphoma, Mantle-Cell/pathology
- Mice
- Mice, Transgenic
- Mutation
- Prognosis
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Transcription, Genetic
- Triazoles/pharmacology
- Tumor Cells, Cultured
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Exportin 1 Protein
Collapse
Affiliation(s)
- Mariko Yoshimura
- Hematology, Respiratory Medicine and Oncology, Department of Medicine, Saga UniversitySaga, Japan
| | - Jo Ishizawa
- Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer CenterHouston, Texas
| | - Vivian Ruvolo
- Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer CenterHouston, Texas
| | - Archana Dilip
- Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer CenterHouston, Texas
| | | | - Timothy J McDonnell
- Department of Hematopathology, The University of Texas MD Anderson Cancer CenterHouston, Texas
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer CenterHouston, Texas
| | - Larry W Kwak
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer CenterHouston, Texas
| | | | | | - Yoko Tabe
- Department of Clinical Laboratory Medicine, Juntendo University School of MedicineTokyo, Japan
| | - Masako Yokoo
- Hematology, Respiratory Medicine and Oncology, Department of Medicine, Saga UniversitySaga, Japan
| | - Shinya Kimura
- Hematology, Respiratory Medicine and Oncology, Department of Medicine, Saga UniversitySaga, Japan
| | - Michael Andreeff
- Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer CenterHouston, Texas
| | - Kensuke Kojima
- Hematology, Respiratory Medicine and Oncology, Department of Medicine, Saga UniversitySaga, Japan
- Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer CenterHouston, Texas
| |
Collapse
|
38
|
Abstract
This review summarizes the current understanding of the role of nuclear bodies in regulating gene expression. The compartmentalization of cellular processes, such as ribosome biogenesis, RNA processing, cellular response to stress, transcription, modification and assembly of spliceosomal snRNPs, histone gene synthesis and nuclear RNA retention, has significant implications for gene regulation. These functional nuclear domains include the nucleolus, nuclear speckle, nuclear stress body, transcription factory, Cajal body, Gemini of Cajal body, histone locus body and paraspeckle. We herein review the roles of nuclear bodies in regulating gene expression and their relation to human health and disease.
Collapse
Affiliation(s)
| | - Cornelius F. Boerkoel
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-604-875-2157; Fax: +1-604-875-2376
| |
Collapse
|
39
|
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: 43] [Impact Index Per Article: 3.9] [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
|
40
|
Antoniali G, Lirussi L, Poletto M, Tell G. Emerging roles of the nucleolus in regulating the DNA damage response: the noncanonical DNA repair enzyme APE1/Ref-1 as a paradigmatical example. Antioxid Redox Signal 2014; 20:621-39. [PMID: 23879289 PMCID: PMC3901381 DOI: 10.1089/ars.2013.5491] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 07/22/2013] [Indexed: 12/25/2022]
Abstract
SIGNIFICANCE An emerging concept in DNA repair mechanisms is the evidence that some key enzymes, besides their role in the maintenance of genome stability, display also unexpected noncanonical functions associated with RNA metabolism in specific subcellular districts (e.g., nucleoli). During the evolution of these key enzymes, the acquisition of unfolded domains significantly amplified the possibility to interact with different partners and substrates, possibly explaining their phylogenetic gain of functions. RECENT ADVANCES After nucleolar stress or DNA damage, many DNA repair proteins can freely relocalize from nucleoli to the nucleoplasm. This process may represent a surveillance mechanism to monitor the synthesis and correct assembly of ribosomal units affecting cell cycle progression or inducing p53-mediated apoptosis or senescence. CRITICAL ISSUES A paradigm for this kind of regulation is represented by some enzymes of the DNA base excision repair (BER) pathway, such as apurinic/apyrimidinic endonuclease 1 (APE1). In this review, the role of the nucleolus and the noncanonical functions of the APE1 protein are discussed in light of their possible implications in human pathologies. FUTURE DIRECTIONS A productive cross-talk between DNA repair enzymes and proteins involved in RNA metabolism seems reasonable as the nucleolus is emerging as a dynamic functional hub that coordinates cell growth arrest and DNA repair mechanisms. These findings will drive further analyses on other BER proteins and might imply that nucleic acid processing enzymes are more versatile than originally thought having evolved DNA-targeted functions after a previous life in the early RNA world.
Collapse
Affiliation(s)
- Giulia Antoniali
- Department of Medical and Biological Sciences, University of Udine , Udine, Italy
| | | | | | | |
Collapse
|
41
|
Quin JE, Devlin JR, Cameron D, Hannan KM, Pearson RB, Hannan RD. Targeting the nucleolus for cancer intervention. Biochim Biophys Acta Mol Basis Dis 2014; 1842:802-16. [PMID: 24389329 DOI: 10.1016/j.bbadis.2013.12.009] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 12/17/2013] [Indexed: 12/17/2022]
Abstract
The contribution of the nucleolus to cancer is well established with respect to its traditional role in facilitating ribosome biogenesis and proliferative capacity. More contemporary studies however, infer that nucleoli contribute a much broader role in malignant transformation. Specifically, extra-ribosomal functions of the nucleolus position it as a central integrator of cellular proliferation and stress signaling, and are emerging as important mechanisms for modulating how oncogenes and tumor suppressors operate in normal and malignant cells. The dependence of certain tumor cells to co-opt nucleolar processes to maintain their cancer phenotypes has now clearly been demonstrated by the application of small molecule inhibitors of RNA Polymerase I to block ribosomal DNA transcription and disrupt nucleolar function (Bywater et al., 2012 [1]). These drugs, which selectively kill tumor cells in vivo while sparing normal cells, have now progressed to clinical trials. It is likely that we have only just begun to scratch the surface of the potential of the nucleolus as a new target for cancer therapy, with "suppression of nucleolar stress" representing an emerging "hallmark" of cancer. This article is part of a Special Issue entitled: Role of the Nucleolus in Human Disease.
Collapse
Affiliation(s)
- Jaclyn E Quin
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jennifer R Devlin
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Donald Cameron
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Kate M Hannan
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Richard B Pearson
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Ross D Hannan
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia; Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia; School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia.
| |
Collapse
|
42
|
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
|
43
|
Burger K, Mühl B, Kellner M, Rohrmoser M, Gruber-Eber A, Windhager L, Friedel CC, Dölken L, Eick D. 4-thiouridine inhibits rRNA synthesis and causes a nucleolar stress response. RNA Biol 2013; 10:1623-30. [PMID: 24025460 DOI: 10.4161/rna.26214] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
High concentrations (> 100 µM) of the ribonucleoside analog 4-thiouridine (4sU) is widely used in methods for RNA analysis like photoactivatable-ribonucleoside-enhanced crosslinking and immunoprecipitation (PAR-CLIP) and nascent messenger (m)RNA labeling (4sU-tagging). Here, we show that 4sU-tagging at low concentrations ≤ 10 µM can be used to measure production and processing of ribosomal (r)RNA. However, elevated concentrations of 4sU (> 50 µM), which are usually used for mRNA labeling experiments, inhibit production and processing of 47S rRNA. The inhibition of rRNA synthesis is accompanied by nucleoplasmic translocation of nucleolar nucleophosmin (NPM1), induction of the tumor suppressor p53, and inhibition of proliferation. We conclude that metabolic labeling of RNA by 4sU triggers a nucleolar stress response, which might influence the interpretation of results. Therefore, functional ribosome biogenesis, nucleolar integrity, and cell cycle should be addressed in 4sU labeling experiments.
Collapse
Affiliation(s)
- Kaspar Burger
- Department of Molecular Epigenetics; Helmholtz Center Munich; Center for Integrated Protein Science Munich (CIPSM); Munich, Germany
| | - Bastian Mühl
- Department of Molecular Epigenetics; Helmholtz Center Munich; Center for Integrated Protein Science Munich (CIPSM); Munich, Germany
| | - Markus Kellner
- Department of Molecular Epigenetics; Helmholtz Center Munich; Center for Integrated Protein Science Munich (CIPSM); Munich, Germany
| | - Michaela Rohrmoser
- Department of Molecular Epigenetics; Helmholtz Center Munich; Center for Integrated Protein Science Munich (CIPSM); Munich, Germany
| | - Anita Gruber-Eber
- Department of Molecular Epigenetics; Helmholtz Center Munich; Center for Integrated Protein Science Munich (CIPSM); Munich, Germany
| | - Lukas Windhager
- Institute for Informatics, Teaching and Research Unit Bioinformatics; Ludwig-Maximilians-University Munich; Munich, Germany
| | - Caroline C Friedel
- Institute for Informatics, Teaching and Research Unit Bioinformatics; Ludwig-Maximilians-University Munich; Munich, Germany
| | - Lars Dölken
- Department of Medicine; Addenbrooke's Hospital; University of Cambridge; Cambridge, UK
| | - Dirk Eick
- Department of Molecular Epigenetics; Helmholtz Center Munich; Center for Integrated Protein Science Munich (CIPSM); Munich, Germany
| |
Collapse
|
44
|
Proteasome activity influences UV-mediated subnuclear localization changes of NPM. PLoS One 2013; 8:e59096. [PMID: 23554979 PMCID: PMC3595268 DOI: 10.1371/journal.pone.0059096] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 02/12/2013] [Indexed: 01/08/2023] Open
Abstract
UV damage activates cellular stress signaling pathways, causes DNA helix distortions and inhibits transcription by RNA polymerases I and II. In particular, the nucleolus, which is the site of RNA polymerase I transcription and ribosome biogenesis, disintegrates following UV damage. The disintegration is characterized by reorganization of the subnucleolar structures and change of localization of many nucleolar proteins. Here we have queried the basis of localization change of nucleophosmin (NPM), a nucleolar granular component protein, which is increasingly detected in the nucleoplasm following UV radiation. Using photobleaching experiments of NPM-fluorescent fusion protein in live human cells we show that NPM mobility increases after UV damage. However, we show that the increase in NPM nucleoplasmic abundance after UV is independent of UV-activated cellular stress and DNA damage signaling pathways. Unexpectedly, we find that proteasome activity affects NPM redistribution. NPM nucleolar expression was maintained when the UV-treated cells were exposed to proteasome inhibitors or when the expression of proteasome subunits was inhibited using RNAi. However, there was no evidence of increased NPM turnover in the UV damaged cells, or that ubiquitin or ubiquitin recycling affected NPM localization. These findings suggest that proteasome activity couples to nucleolar protein localizations in UV damage stress.
Collapse
|
45
|
Yamada K, Ono M, Perkins ND, Rocha S, Lamond AI. Identification and functional characterization of FMN2, a regulator of the cyclin-dependent kinase inhibitor p21. Mol Cell 2013; 49:922-33. [PMID: 23375502 PMCID: PMC3594747 DOI: 10.1016/j.molcel.2012.12.023] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 10/17/2012] [Accepted: 12/26/2012] [Indexed: 01/06/2023]
Abstract
The ARF tumor suppressor is a central component of the cellular defense against oncogene activation in mammals. p14ARF activates p53 by binding and inhibiting HDM2, resulting, inter alia, in increased transcription and expression of the cyclin-dependent kinase inhibitor p21 and consequent cell-cycle arrest. We analyzed the effect of p14ARF induction on nucleolar protein dynamics using SILAC mass spectrometry and have identified the human Formin-2 (FMN2) protein as a component of the p14ARF tumor suppressor pathway. We show that FMN2 is increased upon p14ARF induction at both the mRNA and the protein level via a NF-κB-dependent mechanism that is independent of p53. FMN2 enhances expression of the cell-cycle inhibitor p21 by preventing its degradation. FMN2 is also induced by activation of other oncogenes, hypoxia, and DNA damage. These results identify FMN2 as a crucial component in the regulation of p21 and consequent oncogene/stress-induced cell-cycle arrest in human cells.
Collapse
Affiliation(s)
- Kayo Yamada
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee, UK
| | | | | | | | | |
Collapse
|
46
|
Berger G, Turpin J, Cordeil S, Tartour K, Nguyen XN, Mahieux R, Cimarelli A. Functional analysis of the relationship between Vpx and the restriction factor SAMHD1. J Biol Chem 2012; 287:41210-7. [PMID: 23076149 PMCID: PMC3510820 DOI: 10.1074/jbc.m112.403816] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 09/27/2012] [Indexed: 11/06/2022] Open
Abstract
SAMHD1 is a newly identified restriction factor that targets lentiviruses in myeloid cells and is countered by the SIV(SM)/HIV-2 Vpx protein. By analyzing a large panel of Vpx mutants, we identify several residues throughout the 3-helix bundle predicted for Vpx that impair both its functionality and its ability to degrade SAMHD1. We determine that SAMHD1 is a strictly non-shuttling nuclear protein and that as expected WT Vpx localizes with it in the nucleus. However, we also identify a functional Vpx mutant with predominant cytoplasmic distribution that colocalizes with SAMHD1 in this location, suggesting that Vpx may also retain SAMHD1 in the cell cytoplasm, prior to its entry into the nucleus. Several mutations in Vpx were shown to affect the stability of Vpx, as well as Vpx:Vpx interactions. However, no strict correlation was observed between these parameters and the functionality of Vpx, implying that neither properties is absolutely required for this function and indicating that even unstable Vpx mutants may be very efficient in inducing SAMHD1 degradation. Overall, our analysis identifies several Vpx residues required for SAMHD1 degradation and points to a very efficient and plastic mechanism through which Vpx depletes this restriction factor.
Collapse
Affiliation(s)
- Gregory Berger
- From the Department of Human Virology, ENS-L
- INSERM, U758
- University of Lyon, Lyon I, and
- UMS3444/US8, Lyon, F69364, France
| | - Jocelyn Turpin
- From the Department of Human Virology, ENS-L
- INSERM, U758
- University of Lyon, Lyon I, and
- UMS3444/US8, Lyon, F69364, France
| | - Stéphanie Cordeil
- From the Department of Human Virology, ENS-L
- INSERM, U758
- University of Lyon, Lyon I, and
- UMS3444/US8, Lyon, F69364, France
| | - Kevin Tartour
- From the Department of Human Virology, ENS-L
- INSERM, U758
- University of Lyon, Lyon I, and
- UMS3444/US8, Lyon, F69364, France
| | - Xuan-Nhi Nguyen
- From the Department of Human Virology, ENS-L
- INSERM, U758
- University of Lyon, Lyon I, and
- UMS3444/US8, Lyon, F69364, France
| | - Renaud Mahieux
- From the Department of Human Virology, ENS-L
- INSERM, U758
- University of Lyon, Lyon I, and
- UMS3444/US8, Lyon, F69364, France
| | - Andrea Cimarelli
- From the Department of Human Virology, ENS-L
- INSERM, U758
- University of Lyon, Lyon I, and
- UMS3444/US8, Lyon, F69364, France
| |
Collapse
|
47
|
Holmberg Olausson K, Nistér M, Lindström MS. p53 -Dependent and -Independent Nucleolar Stress Responses. Cells 2012; 1:774-98. [PMID: 24710530 PMCID: PMC3901145 DOI: 10.3390/cells1040774] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 09/28/2012] [Accepted: 10/01/2012] [Indexed: 12/27/2022] Open
Abstract
The nucleolus has emerged as a cellular stress sensor and key regulator of p53-dependent and -independent stress responses. A variety of abnormal metabolic conditions, cytotoxic compounds, and physical insults induce alterations in nucleolar structure and function, a situation known as nucleolar or ribosomal stress. Ribosomal proteins, including RPL11 and RPL5, become increasingly bound to the p53 regulatory protein MDM2 following nucleolar stress. Ribosomal protein binding to MDM2 blocks its E3 ligase function leading to stabilization and activation of p53. In this review we focus on a number of novel regulators of the RPL5/RPL11-MDM2-p53 complex including PICT1 (GLTSCR2), MYBBP1A, PML and NEDD8. p53-independent pathways mediating the nucleolar stress response are also emerging and in particular the negative control that RPL11 exerts on Myc oncoprotein is of importance, given the role of Myc as a master regulator of ribosome biogenesis. We also briefly discuss the potential of chemotherapeutic drugs that specifically target RNA polymerase I to induce nucleolar stress.
Collapse
Affiliation(s)
- Karl Holmberg Olausson
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska R8:05, Karolinska University Hospital in Solna, SE-17176, Stockholm, Sweden.
| | - Monica Nistér
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska R8:05, Karolinska University Hospital in Solna, SE-17176, Stockholm, Sweden.
| | - Mikael S Lindström
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska R8:05, Karolinska University Hospital in Solna, SE-17176, Stockholm, Sweden.
| |
Collapse
|
48
|
Vartak-Sharma N, Ghorpade A. Astrocyte elevated gene-1 regulates astrocyte responses to neural injury: implications for reactive astrogliosis and neurodegeneration. J Neuroinflammation 2012; 9:195. [PMID: 22884085 PMCID: PMC3488579 DOI: 10.1186/1742-2094-9-195] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 07/16/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Reactive astrogliosis is a ubiquitous but poorly understood hallmark of central nervous system pathologies such as trauma and neurodegenerative diseases. In vitro and in vivo studies have identified proinflammatory cytokines and chemokines as mediators of astrogliosis during injury and disease; however, the molecular mechanism remains unclear. In this study, we identify astrocyte elevated gene-1 (AEG-1), a human immunodeficiency virus 1 or tumor necrosis factor α-inducible oncogene, as a novel modulator of reactive astrogliosis. AEG-1 has engendered tremendous interest in the field of cancer research as a therapeutic target for aggressive tumors. However, little is known of its role in astrocytes and astrocyte-mediated diseases. Based on its oncogenic role in several cancers, here we investigate the AEG-1-mediated regulation of astrocyte migration and proliferation during reactive astrogliosis. METHODS An in vivo brain injury mouse model was utilized to show AEG-1 induction following reactive astrogliosis. In vitro wound healing and cell migration assays following AEG-1 knockdown were performed to analyze the role of AEG-1 in astrocyte migration. AEG-1-mediated regulation of astrocyte proliferation was assayed by quantifying the levels of cell proliferation markers, Ki67 and proliferation cell nuclear antigen, using immunocytochemistry. Confocal microscopy was used to evaluate nucleolar localization of AEG-1 in cultured astrocytes following injury. RESULTS The in vivo mouse model for brain injury showed reactive astrocytes with increased glial fibrillary acidic protein and AEG-1 colocalization at the wound site. AEG-1 knockdown in cultured human astrocytes significantly reduced astrocyte migration into the wound site and cell proliferation. Confocal analysis showed colocalization of AEG-1 to the nucleolus of injured cultured human astrocytes. CONCLUSIONS The present findings report for the first time the novel role of AEG-1 in mediating reactive astrogliosis and in regulating astrocyte responses to injury. We also report the nucleolar localization of AEG-1 in human astrocytes in response to injury. Future studies may be directed towards elucidating the molecular mechanism of AEG-1 action in astrocytes during reactive astrogliosis.
Collapse
Affiliation(s)
- Neha Vartak-Sharma
- University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, USA
| | | |
Collapse
|
49
|
Dundr M. Nuclear bodies: multifunctional companions of the genome. Curr Opin Cell Biol 2012; 24:415-22. [PMID: 22541757 DOI: 10.1016/j.ceb.2012.03.010] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 03/16/2012] [Accepted: 03/29/2012] [Indexed: 01/22/2023]
Abstract
It has become increasingly apparent that gene expression is regulated by the functional interplay between spatial genome organization and nuclear architecture. Within the nuclear environment a variety of distinct nuclear bodies exist. They are dynamic, self-organizing structures that do not assemble as pre-formed entities but rather emerge as a direct reflection of specific activities associated with gene expression and genome maintenance. Here I summarize recent findings on functions of some of the most prominent nuclear bodies, including the nucleolus, Cajal body, PML nuclear body, Polycomb group body and the 53BP1 nuclear body. The emerging view is that their organization is orchestrated by similar principles, and they function in fundamental cellular processes involved in homeostasis, differentiation, development and disease.
Collapse
Affiliation(s)
- Miroslav Dundr
- Department of Cell Biology, Rosalind Franklin University of Medicine & Science, North Chicago, IL 60064, United States.
| |
Collapse
|
50
|
Cornelissen B, Waller A, Target C, Kersemans V, Smart S, Vallis KA. 111In-BnDTPA-F3: an Auger electron-emitting radiotherapeutic agent that targets nucleolin. EJNMMI Res 2012; 2:9. [PMID: 22348532 PMCID: PMC3298710 DOI: 10.1186/2191-219x-2-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 02/20/2012] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION The F3 peptide (KDEPQRRSARLSAKPAPPKPEPKPKKAPAKK), a fragment of the human high mobility group protein 2, binds nucleolin. Nucleolin is expressed in the nuclei of normal cells but is also expressed on the membrane of some cancer cells. The goal was to investigate the use of 111In-labeled F3 peptide for Auger electron-targeted radiotherapy. METHODS F3 was labeled with fluorescein isothiocyanate (FITC) for confocal microscopy and conjugated to p-SCN-benzyl-diethylenetriaminepentaacetic acid (BnDTPA) for labeling with 111In to form 111In-BnDTPA-F3. MDA-MB-231-H2N (231-H2N) human breast cancer cells were exposed to 111In-BnDTPA-F3 and used in cell fractionation, γH2AX immunostaining (a marker of DNA double-strand breaks), and clonogenic assays. In vivo, biodistribution studies of 111In-BnDTPA-F3 were performed in 231-H2N xenograft-bearing mice. In tumor growth delay studies, 111In-BnDTPA-F3 (3 μg, 6 MBq/μg) was administered intravenously to 231-H2N xenograft-bearing mice once weekly for 3 weeks. RESULTS Membrane-binding of FITC-F3 was observed in 231-H2N cells, and there was co-localization of FITC-F3 with nucleolin in the nuclei. After exposure of 231-H2N cells to 111In-BnDTPA-F3 for 2 h, 1.7% of 111In added to the medium was membrane-bound. Of the bound 111In, 15% was internalized, and of this, 37% was localized in the nucleus. Exposure of 231-H2N cells to 111In-BnDTPA-F3 (1 μM, 6 MBq/μg) resulted in a dose-dependent increase in γH2AX foci and in a significant reduction of clonogenic survival compared to untreated cells or cells exposed to unlabeled BnDTPA-F3 (46 ± 4.1%, 100 ± 1.8%, and 132 ± 7.7%, respectively). In vivo, tumor uptake of 111In-BnDTPA-F3 (3 μg, 6 MBq/μg) at 3-h post-injection was 1% of the injected dose per gram (%ID/g), and muscle uptake was 0.5%ID/g. In tumor growth delay studies, tumor growth rate was reduced 19-fold compared to untreated or unlabeled BnDTPA-F3-treated mice (p = 0.023). CONCLUSION 111In-BnDTPA-F3 is internalized into 231-H2N cells and translocates to the nucleus. 111In-BnDTPA-F3 has a potent cytotoxic effect in vitro and an anti-tumor effect in mice bearing 231-H2N xenografts despite modest total tumor accumulation.
Collapse
Affiliation(s)
- Bart Cornelissen
- Department of Oncology, Cancer Research UK/Medical Research Council Gray Institute for Radiation Oncology and Biology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Andrew Waller
- Department of Oncology, Cancer Research UK/Medical Research Council Gray Institute for Radiation Oncology and Biology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Carol Target
- Department of Oncology, Cancer Research UK/Medical Research Council Gray Institute for Radiation Oncology and Biology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Veerle Kersemans
- Department of Oncology, Cancer Research UK/Medical Research Council Gray Institute for Radiation Oncology and Biology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Sean Smart
- Department of Oncology, Cancer Research UK/Medical Research Council Gray Institute for Radiation Oncology and Biology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Katherine A Vallis
- Department of Oncology, Cancer Research UK/Medical Research Council Gray Institute for Radiation Oncology and Biology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford, OX3 7DQ, UK
| |
Collapse
|