1
|
Zhang Y, Wang ZZ, Han AQ, Yang MY, Zhu LX, Pan FM, Wang Y. TuBG1 promotes hepatocellular carcinoma via ATR/P53-apoptosis and cycling pathways. Hepatobiliary Pancreat Dis Int 2024; 23:195-209. [PMID: 37806848 DOI: 10.1016/j.hbpd.2023.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 09/19/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND As reported, γ-tubulin (TuBG1) is related to the occurrence and development of various types of malignant tumors. However, its role in hepatocellular cancer (HCC) is not clear. The present study was to investigate the relationship between TuBG1 and clinical parameters and survival in HCC patients. METHODS The correlation between TuBG1 and clinical parameters and survival in HCC patients was explored by bioinformatics analysis. Immunohistochemistry was used for the verification. The molecular function of TuBG1 was measured using colony formation, scratch assay, trans-well assay and flow cytometry. Gene set enrichment analysis (GSEA) was used to pick up the enriched pathways, followed by investigating the target pathways using Western blotting. The tumor-immune system interactions and drug bank database (TISIDB) was used to evaluate TuBG1 and immunity. Based on the TuBG1-related immune genes, a prognostic model was constructed and was further validated internally and externally. RESULTS The bioinformatic analysis found high expressed TuBG1 in HCC tissue, which was confirmed using immunohistochemistry and Western blotting. After silencing the TuBG1 in HCC cell lines, more G1 arrested cells were found, cell proliferation and invasion were inhibited, and apoptosis was promoted. Furthermore, the silence of TuBG1 increased the expressions of Ataxia-Telangiectasia and Rad-3 (ATR), phospho-P38 mitogen-activated protein kinase (P-P38MAPK), phospho-P53 (P-P53), B-cell lymphoma-2 associated X protein (Bax), cleaved caspase 3 and P21; decreased the expressions of B-cell lymphoma-2 (Bcl-2), cyclin D1, cyclin E2, cyclin-dependent kinase 2 (CDK2) and CDK4. The correlation analysis of immunohistochemistry and clinical parameters and survival data revealed that TuBG1 was negatively correlated with the overall survival. The constructed immune prognosis model could effectively evaluate the prognosis. CONCLUSIONS The increased expression of TuBG1 in HCC is associated with poor prognosis, which might be involved in the occurrence and development of HCC.
Collapse
Affiliation(s)
- Yan Zhang
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Zhen-Zhen Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - An-Qi Han
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Ming-Ya Yang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Li-Xin Zhu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Fa-Ming Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei 230032, China
| | - Yong Wang
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei 230601, China.
| |
Collapse
|
2
|
Ramesh Babu PB. Prediction of anti-microtubular target proteins of tubulins and their interacting proteins using Gene Ontology tools. J Genet Eng Biotechnol 2023; 21:78. [PMID: 37466845 DOI: 10.1186/s43141-023-00531-8] [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: 07/29/2022] [Accepted: 07/01/2023] [Indexed: 07/20/2023]
Abstract
BACKGROUND Tubulins are highly conserved globular proteins involved in stabilization of cellular cytoskeletal microtubules during cell cycle. Different isoforms of tubulins are differentially expressed in various cell types, and their protein-protein interactions (PPIs) analysis will help in identifying the anti-microtubular drug targets for cancer and neurological disorders. Numerous web-based PPIs analysis methods are recently being used, and in this paper, I used Gene Ontology (GO) tools, e.g., Stringbase, ProteomeHD, GeneMANIA, and ShinyGO, to identify anti-microtubular target proteins by selecting strongly interacting proteins of tubulins. RESULTS I used 6 different human tubulin isoforms (two from each of α-, β-, and γ-tubulin) and found several thousands of node-to-node protein interactions (highest 4956 in GeneMANIA) and selected top 10 strongly interacting node-to-node interactions with highest score, which included 7 tubulin family protein and 6 non-tubulin family proteins (total 13). Functional enrichment analysis indicated a significant role of these 13 proteins in nucleation, polymerization or depolymerization of microtubules, membrane tethering and docking, dorsal root ganglion development, mitotic cycle, and cytoskeletal organization. I found γ-tubulins (TUBG1, TUBGCP4, and TUBBGCP6) were known to contribute majorly for tubulin-associated functions followed by α-tubulin (TUBA1A) and β-tubulins (TUBB AND TUBB3). In PPI results, I found several non-tubular proteins interacting with tubulins, and six of them (HTT, DPYSL2, SKI, UNC5C, NINL, and DDX41) were found closely associated with their functions. CONCLUSIONS Increasing number of regulatory proteins and subpopulation of tubulin proteins are being reported with poor understanding in their association with microtubule assembly and disassembly. The functional enrichment analysis of tubulin isoforms using recent GO tools resulted in identification of γ-tubulins playing a key role in microtubule functions and observed non-tubulin family of proteins HTT, DPYSL2, SKI, UNC5C, NINL, and DDX41 strongly interacting functional proteins of tubulins. The present study yields a promising model system using GO tools to narrow down tubulin-associated proteins as a drug target in cancer, Alzheimer's, neurological disorders, etc.
Collapse
Affiliation(s)
- Polani B Ramesh Babu
- Center for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Bharath Institute of Science and Technology, Selaiyur, Tambaram, Chennai, India.
| |
Collapse
|
3
|
Chowdhury SR, Koley T, Singh M, Samath EA, Kaur P. Association of Hsp90 with p53 and Fizzy related homolog (Fzr) synchronizing Anaphase Promoting Complex (APC/C): An unexplored ally towards oncogenic pathway. Biochim Biophys Acta Rev Cancer 2023; 1878:188883. [PMID: 36972769 DOI: 10.1016/j.bbcan.2023.188883] [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: 09/03/2022] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/29/2023]
Abstract
The intricate molecular interactions leading to the oncogenic pathway are the consequence of cell cycle modification controlled by a bunch of cell cycle regulatory proteins. The tumor suppressor and cell cycle regulatory proteins work in coordination to maintain a healthy cellular environment. The integrity of this cellular protein pool is perpetuated by heat shock proteins/chaperones, which assist in proper protein folding during normal and cellular stress conditions. Among these versatile groups of chaperone proteins, Hsp90 is one of the significant ATP-dependent chaperones that aid in stabilizing many tumor suppressors and cell cycle regulator protein targets. Recently, studies have revealed that in cancerous cell lines, Hsp90 stabilizes mutant p53, 'the guardian of the genome.' Hsp90 also has a significant impact on Fzr, an essential regulator of the cell cycle having an important role in the developmental process of various organisms, including Drosophila, yeast, Caenorhabditis elegans, and plants. During cell cycle progression, p53 and Fzr coordinately regulate the Anaphase Promoting Complex (APC/C) from metaphase to anaphase transition up to cell cycle exit. APC/C mediates proper centrosome function in the dividing cell. The centrosome acts as the microtubule organizing center for the correct segregation of the sister chromatids to ensure perfect cell division. This review examines the structure of Hsp90 and its co-chaperones, which work in synergy to stabilize proteins such as p53 and Fizzy-related homolog (Fzr) to synchronize the Anaphase Promoting Complex (APC/C). Dysfunction of this process activates the oncogenic pathway leading to the development of cancer. Additionally, an overview of current drugs targeting Hsp90 at various phases of clinical trials has been included.
Collapse
Affiliation(s)
- Sanghati Roy Chowdhury
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Tirthankar Koley
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Mandeep Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | | | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
4
|
Chen T, Zhang S, Zhou D, Lu P, Mo X, Tamrakar R, Yang X. Screening of co-pathogenic genes of non-alcoholic fatty liver disease and hepatocellular carcinoma. Front Oncol 2022; 12:911808. [PMID: 36033523 PMCID: PMC9410624 DOI: 10.3389/fonc.2022.911808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a risk factor for hepatocellular carcinoma (HCC). However, its carcinogenic mechanism is still unclear, looking for both diseases’ transcriptome levels, the same changes as we are looking for NAFLD may provide a potential mechanism of action of HCC. Thus, our study aimed to discover the coexisting pathogenic genes of NAFLD and HCC. Methods We performed a variance analysis with public data for both diseases. At the same time, weighted gene correlation network analysis (WGCNA) was used to find highly correlated gene modules in both diseases. The darkturquoise gene module was found to be highly correlated with both diseases. Based on the diagnosis related module genes and the differential genes of the two diseases, we constructed diagnostic and prognostic models by logistic regression, univariate Cox regression, and LASSO regression. Public datasets verified the results. Meanwhile, we built a competing endogenous RNA (ceRNA) network based on the model genes and explored the related pathways and immune correlation involved in the two diseases by using Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and gene set enrichment analyses. Immunohistochemistry was used to verify the different expression of ABCC5 and TUBG1 among the normal liver, NAFLD, and HCC tissues. Sodium palmitate/sodium oleate was used to establish high-fat cell models, and Real Time Quantitative Polymerase Chain Reaction (RT-qPCR) was used to verify the messenger RNA (mRNA) expression of ABCC5 in lipidization cells. Results A total of 26 upregulated genes and 87 downregulated genes were found using limma package identification analysis. According to WGCNA, the darkturquoise gene module was highly correlated with the prognosis of both diseases. The coexisting genes acquired by the two groups were only three central genes, that is, ABCC5, DHODH and TUBG1. The results indicated that the diagnostic and prognostic models constructed by ABCC5 and TUBG1 genes had high accuracy in both diseases. The results of immunohistochemistry showed that ABCC5 and TUBG1 were significantly overexpressed in NAFLD and HCC tissues compared with normal liver tissues. The Oil Red O staining and triglyceride identified the successful construction of HepG2 and LO2 high-fat models using PA/OA. The results of RT-qPCR showed that the lipidization of LO2 and HepG2 increased the mRNA expression of ABCC5. Conclusions The gene model constructed by ABCC5 and TUBG1 has high sensibility and veracity in the diagnosis of NAFLD as well as the diagnosis and prognosis of HCC. ABCC5 and TUBG1 may play an important role in the development of NAFLD to HCC. In addition, lipidization could upregulate the mRNA expression of ABCC5 in HCC.
Collapse
Affiliation(s)
- Ting Chen
- Department of Endocrinology, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Siwen Zhang
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Guangxi Medical University, Nanning, China
- *Correspondence: Xi Yang, ; Siwen Zhang,
| | - Dongmei Zhou
- Department of Endocrinology, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Peipei Lu
- Department of Geriatric Endocrinology and Metabolism, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Xianglai Mo
- Department of Geriatric Endocrinology and Metabolism, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Rashi Tamrakar
- Department of Endocrinology, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Xi Yang
- Department of Geriatric Endocrinology and Metabolism, First Affiliated Hospital, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, First Affiliated Hospital, Guangxi Medical University, Nanning, China
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, China
- *Correspondence: Xi Yang, ; Siwen Zhang,
| |
Collapse
|
5
|
Zhou J, Alvarado-Kristensson M. Optimization of production of recombinant gamma-tubulin in bacteria. MethodsX 2021; 8:101517. [PMID: 34754788 PMCID: PMC8563660 DOI: 10.1016/j.mex.2021.101517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/14/2021] [Indexed: 12/03/2022] Open
Abstract
Production of a protein of interest in bacteria and its purification from bacterial lysates are valuable tools for the purification of larger amounts of recombinant proteins. The low cost of culturing, and the rapid cell growth of bacteria make this host a good choice for protein production, but the folding and function of the purified protein might be altered due to the production of a eukaryotic protein in a prokaryotic host. Here, we provide a purification method for the purification of gamma (γ)-tubulin (TUBG) from soluble fractions of Escherichia (E.) coli lysates using affinity tags.This protocol describes a method that purifies soluble GST-TUBG1 from bacteria. Of the three tested induction conditions, the highest yield of recombinant GST-TUBG1 was obtained after the induction of E. coli with isopropyl-D-1-thiogalactopyranoside (IPTG) for 1 h at 37 °C followed by overnight incubation at room temperature. In comparison with other methodologies (Hoog et al., 2011), the technique described here retrieves larger amounts of recombinant TUBG1 from small-scale expression cultures.
Collapse
Affiliation(s)
- Jingkai Zhou
- Molecular Pathology, Department of Translational Medicine, Lund University, Jan Waldenströms gata 59, Malmö SE-205 02, Sweden
| | - Maria Alvarado-Kristensson
- Molecular Pathology, Department of Translational Medicine, Lund University, Jan Waldenströms gata 59, Malmö SE-205 02, Sweden
| |
Collapse
|
6
|
Sena P, Mancini S, Bertacchini J, Carnevale G, Pedroni M, Roncucci L. Autoimmunity Profiles as Prognostic Indicators in Patients with Colorectal Cancer versus Those with Cancer at Other Sites: A Prospective Study. Cancers (Basel) 2021; 13:3239. [PMID: 34209517 PMCID: PMC8269181 DOI: 10.3390/cancers13133239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/19/2021] [Accepted: 06/25/2021] [Indexed: 11/26/2022] Open
Abstract
Colorectal cancer represents a paradigmatic model of inflammatory carcinogenesis accompanied by the production of several kinds of tumor-associated autoantibodies (TAABs). The specific aim of this study is to define the clinical impact of the presence of non-specific circulating TAABs in a cohort of cancer patients and to establish whether significant differences were present between colorectal cancer and cancers at other sites. For this aim a prospective study was developed and a five-year survival analysis performed. Indirect immunofluorescence on rat tissues for non-organ specific autoantibodies (NOSAs: liver-kidney-stomach), on rat colon substrates (colon-related autoantibodies, CAAs) and on HEp-2 cell lines was performed. NOSA positivity was more frequent in patients with colorectal cancer than in those with cancer at other sites. Survival analysis demonstrated a significantly worse prognosis in cancer patients positive for TAABs. CAA positivity is a predictor of survival, independently from the presence of comorbidities, and HEp-2 reactivity was a strong predictor of survival in a stepwise Cox-regression model, including stage at diagnosis. Overall overproduction of TAABs is associated with advanced oncological disease, the presence of metastasis, and poorer prognosis of cancer patients.
Collapse
Affiliation(s)
- Paola Sena
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (P.S.); (J.B.); (G.C.)
| | - Stefano Mancini
- Department of Internal Medicine and Rehabilitation, Santa Maria Bianca Hospital, AUSL Modena, Via A. Fogazzaro 6, 41037 Mirandola, Italy;
| | - Jessika Bertacchini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (P.S.); (J.B.); (G.C.)
| | - Gianluca Carnevale
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (P.S.); (J.B.); (G.C.)
| | - Monica Pedroni
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy;
| | - Luca Roncucci
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy;
| |
Collapse
|
7
|
Nuclear Isoforms of Neurofibromin Are Required for Proper Spindle Organization and Chromosome Segregation. Cells 2020; 9:cells9112348. [PMID: 33114250 PMCID: PMC7690890 DOI: 10.3390/cells9112348] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 12/30/2022] Open
Abstract
Mitotic spindles are highly organized, microtubule (MT)-based, transient structures that serve the fundamental function of unerring chromosome segregation during cell division and thus of genomic stability during tissue morphogenesis and homeostasis. Hence, a multitude of MT-associated proteins (MAPs) regulates the dynamic assembly of MTs in preparation for mitosis. Some tumor suppressors, normally functioning to prevent tumor development, have now emerged as significant MAPs. Among those, neurofibromin, the product of the Neurofibromatosis-1 gene (NF1), a major Ras GTPase activating protein (RasGAP) in neural cells, controls also the critical function of chromosome congression in astrocytic cellular contexts. Cell type- and development-regulated splicings may lead to the inclusion or exclusion of NF1exon51, which bears a nuclear localization sequence (NLS) for nuclear import at G2; yet the functions of the produced NLS and ΔNLS neurofibromin isoforms have not been previously addressed. By using a lentiviral shRNA system, we have generated glioblastoma SF268 cell lines with conditional knockdown of NLS or ΔNLS transcripts. In dissecting the roles of NLS or ΔNLS neurofibromins, we found that NLS-neurofibromin knockdown led to increased density of cytosolic MTs but loss of MT intersections, anastral spindles featuring large hollows and abnormal chromosome positioning, and finally abnormal chromosome segregation and increased micronuclei frequency. Therefore, we propose that NLS neurofibromin isoforms exert prominent mitotic functions.
Collapse
|
8
|
Corvaisier M, Alvarado-Kristensson M. Non-Canonical Functions of the Gamma-Tubulin Meshwork in the Regulation of the Nuclear Architecture. Cancers (Basel) 2020; 12:cancers12113102. [PMID: 33114224 PMCID: PMC7690915 DOI: 10.3390/cancers12113102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/17/2020] [Accepted: 10/21/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The appearance of a cell is connected to its function. For example, the fusiform of smooth muscle cells is adapted to facilitate muscle contraction, the lobed nucleus in white blood cells assists with the migratory behavior of these immune cells, and the condensed nucleus in sperm aids in their swimming efficiency. Thus, changes in appearance have been used for decades by doctors as a diagnostic method for human cancers. Here, we summarize our knowledge of how a cell maintains the shape of the nuclear compartment. Specifically, we discuss the role of a novel protein meshwork, the gamma-tubulin meshwork, in the regulation of nuclear morphology and as a therapeutic target against cancer. Abstract The nuclear architecture describes the organization of the various compartments in the nucleus of eukaryotic cells, where a plethora of processes such as nucleocytoplasmic transport, gene expression, and assembly of ribosomal subunits occur in a dynamic manner. During the different phases of the cell cycle, in post-mitotic cells and after oncogenic transformation, rearrangements of the nuclear architecture take place, and, among other things, these alterations result in reorganization of the chromatin and changes in gene expression. A member of the tubulin family, γtubulin, was first identified as part of a multiprotein complex that allows nucleation of microtubules. However, more than a decade ago, γtubulin was also characterized as a nuclear protein that modulates several crucial processes that affect the architecture of the nucleus. This review presents the latest knowledge regarding changes that arise in the nuclear architecture of healthy cells and under pathological conditions and, more specifically, considers the particular involvement of γtubulin in the modulation of the biology of the nuclear compartment.
Collapse
|
9
|
Moonlighting in Mitosis: Analysis of the Mitotic Functions of Transcription and Splicing Factors. Cells 2020; 9:cells9061554. [PMID: 32604778 PMCID: PMC7348712 DOI: 10.3390/cells9061554] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
Moonlighting proteins can perform one or more additional functions besides their primary role. It has been posited that a protein can acquire a moonlighting function through a gradual evolutionary process, which is favored when the primary and secondary functions are exerted in different cellular compartments. Transcription factors (TFs) and splicing factors (SFs) control processes that occur in interphase nuclei and are strongly reduced during cell division, and are therefore in a favorable situation to evolve moonlighting mitotic functions. However, recently published moonlighting protein databases, which comprise almost 400 proteins, do not include TFs and SFs with secondary mitotic functions. We searched the literature and found several TFs and SFs with bona fide moonlighting mitotic functions, namely they localize to specific mitotic structure(s), interact with proteins enriched in the same structure(s), and are required for proper morphology and functioning of the structure(s). In addition, we describe TFs and SFs that localize to mitotic structures but cannot be classified as moonlighting proteins due to insufficient data on their biochemical interactions and mitotic roles. Nevertheless, we hypothesize that most TFs and SFs with specific mitotic localizations have either minor or redundant moonlighting functions, or are evolving towards the acquisition of these functions.
Collapse
|
10
|
Alvarado-Kristensson M. Choreography of the centrosome. Heliyon 2020; 6:e03238. [PMID: 31989056 PMCID: PMC6970175 DOI: 10.1016/j.heliyon.2020.e03238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/31/2022] Open
Abstract
More than a century ago, the centrosome was discovered and described as “the true division organ of the cell”. Electron microscopy revealed that a centrosome is an amorphous structure or pericentriolar protein matrix that surrounds a pair of well-organized centrioles. Today, the importance of the centrosome as a microtubule-organizing center and coordinator of the mitotic spindle is questioned, because centrioles are absent in up to half of all known eukaryotic species, and various mechanisms for acentrosomal microtubule nucleation have been described. This review recapitulates the known functions of centrosome movements in cellular homeostasis and discusses knowledge gaps in this field.
Collapse
Affiliation(s)
- Maria Alvarado-Kristensson
- Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, SE-20502, Sweden
| |
Collapse
|
11
|
Contadini C, Monteonofrio L, Virdia I, Prodosmo A, Valente D, Chessa L, Musio A, Fava LL, Rinaldo C, Di Rocco G, Soddu S. p53 mitotic centrosome localization preserves centrosome integrity and works as sensor for the mitotic surveillance pathway. Cell Death Dis 2019; 10:850. [PMID: 31699974 PMCID: PMC6838180 DOI: 10.1038/s41419-019-2076-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/02/2019] [Accepted: 10/16/2019] [Indexed: 12/22/2022]
Abstract
Centrosomal p53 has been described for three decades but its role is still unclear. We previously reported that, in proliferating human cells, p53 transiently moves to centrosomes at each mitosis. Such p53 mitotic centrosome localization (p53-MCL) occurs independently from DNA damage but requires ATM-mediated p53Ser15 phosphorylation (p53Ser15P) on discrete cytoplasmic p53 foci that, through MT dynamics, move to centrosomes during the mitotic spindle formation. Here, we show that inhibition of p53-MCL, obtained by p53 depletion or selective impairment of p53 centrosomal localization, induces centrosome fragmentation in human nontransformed cells. In contrast, tumor cells or mouse cells tolerate p53 depletion, as expected, and p53-MCL inhibition. Such tumor- and species-specific behavior of centrosomal p53 resembles that of the recently identified sensor of centrosome-loss, whose activation triggers the mitotic surveillance pathway in human nontransformed cells but not in tumor cells or mouse cells. The mitotic surveillance pathway prevents the growth of human cells with increased chance of making mitotic errors and accumulating numeral chromosome defects. Thus, we evaluated whether p53-MCL could work as a centrosome-loss sensor and contribute to the activation of the mitotic surveillance pathway. We provide evidence that centrosome-loss triggered by PLK4 inhibition makes p53 orphan of its mitotic dock and promotes accumulation of discrete p53Ser15P foci. These p53 foci are required for the recruitment of 53BP1, a key effector of the mitotic surveillance pathway. Consistently, cells from patients with constitutive impairment of p53-MCL, such as ATM- and PCNT-mutant carriers, accumulate numeral chromosome defects. These findings indicate that, in nontransformed human cells, centrosomal p53 contributes to safeguard genome integrity by working as sensor for the mitotic surveillance pathway.
Collapse
Affiliation(s)
- Claudia Contadini
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS-Regina Elena National Cancer Institute, Rome, Italy.,Department of Biology, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Laura Monteonofrio
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS-Regina Elena National Cancer Institute, Rome, Italy.,Laboratory of Cardiovascular Science, NIA/NIH Baltimore, Baltimore, MD, 21224, USA
| | - Ilaria Virdia
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Andrea Prodosmo
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS-Regina Elena National Cancer Institute, Rome, Italy.,GMP Biopharmaceutical Facility, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Davide Valente
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Luciana Chessa
- Department of Clinical and Molecular Medicine, Sapienza University, Rome, Italy
| | - Antonio Musio
- Institute of Genetics and Biomedical Research, National Research Council (CNR), Pisa, Italy
| | - Luca L Fava
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Povo, Italy
| | - Cinzia Rinaldo
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS-Regina Elena National Cancer Institute, Rome, Italy.,Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Sapienza University, Rome, Italy
| | - Giuliana Di Rocco
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Soddu
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS-Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
12
|
Abstract
Whole-genome and centrosome duplication as a consequence of cytokinesis failure can drive tumorigenesis in experimental model systems. However, whether cytokinesis failure is in fact an important cause of human cancers has remained unclear. In this Review, we summarize evidence that whole-genome-doubling events are frequently observed in human cancers and discuss the contribution that cytokinesis defects can make to tumorigenesis. We provide an overview of the potential causes of cytokinesis failure and discuss how tetraploid cells that are generated through cytokinesis defects are used in cancer as a transitory state on the route to aneuploidy. Finally, we discuss how cytokinesis defects can facilitate genetic diversification within the tumour to promote cancer development and could constitute the path of least resistance in tumour evolution.
Collapse
Affiliation(s)
- Susanne M A Lens
- Oncode Institute, Utrecht, Netherlands.
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.
| | - René H Medema
- Oncode Institute, Utrecht, Netherlands.
- Division of Cell Biology and Cancer Genomics Center, The Netherlands Cancer Institute, Amsterdam, Netherlands.
| |
Collapse
|
13
|
Alvarado-Kristensson M. γ-tubulin as a signal-transducing molecule and meshwork with therapeutic potential. Signal Transduct Target Ther 2018; 3:24. [PMID: 30221013 PMCID: PMC6137058 DOI: 10.1038/s41392-018-0021-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/23/2018] [Accepted: 05/06/2018] [Indexed: 01/05/2023] Open
Abstract
Knowledge of γ-tubulin is increasing with regard to the cellular functions of this protein beyond its participation in microtubule nucleation. γ-Tubulin expression is altered in various malignancies, and changes in the TUBG1 gene have been found in patients suffering from brain malformations. This review recapitulates the known functions of γ-tubulin in cellular homeostasis and discusses the possible influence of the protein on disease development and cancer.
Collapse
Affiliation(s)
- Maria Alvarado-Kristensson
- Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, 20502 Sweden
| |
Collapse
|
14
|
Park JE, Jang YL, Jang CY. The tobacco carcinogen NNK disturbs mitotic chromosome alignment by interrupting p53 targeting to the centrosome. Toxicol Lett 2017; 281:110-118. [PMID: 28964810 DOI: 10.1016/j.toxlet.2017.09.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/20/2017] [Accepted: 09/26/2017] [Indexed: 11/15/2022]
Abstract
The tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) is the most potent risk factor among tobacco-related carcinogens in lung cancer progression and outcomes. Although genetic mutations and chromosome instability have been detected in NNK-induced lung tumors, the oncogenic mechanisms of NNK are not fully understood. Here, we show that NNK increases chromosomal instability by disrupting spindle microtubule (MT) attachment to the kinetochore (KT) and spindle dynamics. Mechanistically, NNK blocks the targeting of p53 to the centrosome during mitosis, leading to chromosome alignment defects in metaphase. Therefore, lung cancer cells with wild-type p53, such as A594 and H226B, are more resistant to the NNK treatment than p53-mutant lung cancer cells, such as A1299 and H226Br. Although NNK does not affect the levels or transcriptional activity of p53, the reduction of the p53 level at the centrosome exacerbates the NNK-induced chromosome alignment defect in A549 and H226B cells. Therefore, p53 protects against NNK-induced chromosome instability by modulating the function of centrosome-localized p53 and not by modulating transcriptional activity. We conclude that NNK may increase the risk of lung cancer progression and poorer outcomes in patients with p53 mutations by perturbing proper mitotic progression and chromosome integrity.
Collapse
Affiliation(s)
- Ji Eun Park
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Yu Lim Jang
- Soongeui Girls' High School, Seoul 06944, Republic of Korea
| | - Chang-Young Jang
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea.
| |
Collapse
|
15
|
Mahathre MM, Rida PC, Aneja R. The more the messier: centrosome amplification as a novel biomarker for personalized treatment of colorectal cancers. J Biomed Res 2016; 30:441-451. [PMID: 27924065 PMCID: PMC5138576 DOI: 10.7555/jbr.30.20150109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 10/12/2015] [Indexed: 01/10/2023] Open
Abstract
Colon cancer is currently the third most common cancer and second most fatal cancer in the United States, resulting in approximately 600,000 deaths annually. Though colorectal cancer death rates are decreasing by about 3% every year, disease outcomes could be substantially improved with more research into the drivers of colon carcinogenesis, the determinants of aggressiveness in colorectal cancer and the identification of biomarkers that could enable choice of more optimal treatments. Colon carcinogenesis is notably a slow process that can take decades. Known factors that contribute to the development of colon cancer are mutational, epigenetic and environmental, and risk factors include age, history of polyps and family history of colon cancer. Colorectal cancers exhibit heterogeneity in their features and are often characterized by the presence of chromosomal instability, microscopic satellite instability, or CpG island methylator phenotype. In this review, we propose that centrosome amplification may be a widespread occurrence in colorectal cancers and could potently influence tumor biology. Moreover, the quantitation of this cancer-specific anomaly could offer valuable prognostic information and pave the way for further customization of treatment based on the organellar profile of patients. Patient stratification models that take into account centrosomal status could thus potentially reduce adverse side effects and result in improved outcomes for colorectal cancer patients.
Collapse
Affiliation(s)
- Monica M Mahathre
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Padmashree Cg Rida
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA.,Novazoi Theranostics Inc., Plano, TX 75025, USA
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA.,Institute of Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.,Center for Obesity Research, Georgia State University, Atlanta, GA 30303, USA;
| |
Collapse
|
16
|
Chun MJ, Hwang SK, Kim HG, Goh SH, Kim S, Lee CH. Aurora A kinase is required for activation of the Fanconi anemia/BRCA pathway upon DNA damage. FEBS Open Bio 2016; 6:782-90. [PMID: 27398318 PMCID: PMC4932458 DOI: 10.1002/2211-5463.12087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 05/09/2016] [Accepted: 05/19/2016] [Indexed: 12/23/2022] Open
Abstract
Previous studies have linked the DNA damage response to mitotic progression machinery. Mitotic kinases, such as Aurora A kinase and Polo‐like kinase, are involved in the phosphorylation of cell cycle regulators in response to DNA damage. Here, we investigated the potential involvement of Aurora A kinase in the activation of the Fanconi anemia (FA)/BRCA pathway, which participates in cellular response to DNA interstrand cross‐link lesions (ICL). Initially, we detected interactions between Aurora A kinase and FANCA protein, one of the components of the FA nuclear core complex. Silencing of Aurora A kinase led to inhibition of monoubiquitination of FANCD2 and formation of nuclear foci, the final consequences of FA/BRCA pathway activation upon ICL induction. An in vitro kinase assay revealed that Aurora A kinase phosphorylates S165 of FANCA. Moreover, this phosphorylation event was induced by the treatment with mitomycin C (MMC), an ICL‐inducing agent. In cells overexpressing S165A mutant FANCA, monoubiquitination of FANCD2 and nuclear foci formation was impaired and cellular sensitivity to MMC was enhanced. These results suggest that S165 phosphorylation by Aurora A kinase is required for proper activation of the FA/BRCA pathway in response to DNA damage.
Collapse
Affiliation(s)
- Min Jeong Chun
- Cancer Cell and Molecular Biology Branch Research Institute National Cancer Center Goyang Gyeonggi Korea
| | - Soo Kyung Hwang
- Cancer Cell and Molecular Biology Branch Research Institute National Cancer Center Goyang Gyeonggi Korea
| | - Hyoun Geun Kim
- Cancer Cell and Molecular Biology Branch Research Institute National Cancer Center Goyang Gyeonggi Korea
| | - Sung-Ho Goh
- Precision Medicine Branch Research Institute National Cancer Center Goyang Gyeonggi Korea
| | - Sunshin Kim
- Precision Medicine Branch Research Institute National Cancer Center Goyang Gyeonggi Korea
| | - Chang-Hun Lee
- Cancer Cell and Molecular Biology Branch Research Institute National Cancer Center Goyang Gyeonggi Korea
| |
Collapse
|
17
|
Kryukova E, Kryukov F, Hajek R. Centrosome amplification and clonal evolution in multiple myeloma: Short review. Crit Rev Oncol Hematol 2015; 98:116-21. [PMID: 26589397 DOI: 10.1016/j.critrevonc.2015.10.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 09/14/2015] [Accepted: 10/28/2015] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) is composed of an array of multiple clones, each potentially associated with different clinical behavior. Previous studies focused on clinical implication of centrosome amplification (CA) in MM show contradictory results. It seems that the role of CA as well as CA formation in MM differ from other malignancies. This has brought about a question about the role of CA positive clone which is--is it going to be a more aggressive clone evolutionally arising under pressure of negative conditions or can CA serve as a marker of cell abnormality and lead to cell death and further elimination of this damaged subpopulation? This current review is devoted to the discussion of the existence of MM subclones with centrosome amplification (CA), its evolutionary behaviour within intraclonal heterogeneity as well as its potential impact on the disease progression and MM treatment.
Collapse
Affiliation(s)
- Elena Kryukova
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Czech Republic; Department of Haematooncology, University Hospital Ostrava, Czech Republic
| | - Fedor Kryukov
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Czech Republic; Department of Haematooncology, University Hospital Ostrava, Czech Republic.
| | - Roman Hajek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Czech Republic; Department of Haematooncology, University Hospital Ostrava, Czech Republic
| |
Collapse
|
18
|
Turning the headlights on novel cancer biomarkers: Inspection of mechanics underlying intratumor heterogeneity. Mol Aspects Med 2015; 45:3-13. [PMID: 26024970 DOI: 10.1016/j.mam.2015.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/20/2015] [Indexed: 01/20/2023]
Abstract
Although the existence of intratumoral heterogeneity (ITH) in the expression of common biomarkers has been described by pathologists since the late 1890s, we have only recently begun to fathom the staggering extent and near ubiquity of this phenomenon. From the tumor's perspective, ITH provides a stabilizing diversity that allows for the evolution of aggressive cancer phenotypes. As the weight of the evidence correlating ITH to poor prognosis burgeons, it has become increasingly important to determine the mechanisms by which a tumor acquires ITH, find clinically-adaptable means to quantify ITH and design strategies to deal with the numerous profound clinical ramifications that ITH forces upon us. Elucidation of the drivers of ITH could enable development of novel biomarkers whose interrogation might permit quantitative evaluation of the ITH inherent in a tumor in order to predict the poor prognosis risk associated with that tumor. This review proposes centrosome amplification (CA), aided and abetted by centrosome clustering mechanisms, as a critical driver of chromosomal instability (CIN) that makes a key contribution to ITH generation. Herein we also evaluate how a tumor's inherent mitotic propensity, which reflects the cell cycling kinetics within the tumor's proliferative cells, functions as the indispensable engine underpinning CIN, and determines the rate of CIN. We thus expound how the forces of centrosome amplification and mitotic propensity collaborate to sculpt the genetic landscape of a tumor and spawn extensive subclonal diversity. As such, centrosome amplification and mitotic propensity profiles could serve as clinically facile and powerful prognostic biomarkers that would enable more accurate risk segmentation of patients and design of individualized therapies.
Collapse
|
19
|
Louwen F, Yuan J. Battle of the eternal rivals: restoring functional p53 and inhibiting Polo-like kinase 1 as cancer therapy. Oncotarget 2013; 4:958-71. [PMID: 23948487 PMCID: PMC3759674 DOI: 10.18632/oncotarget.1096] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 07/11/2013] [Indexed: 01/09/2023] Open
Abstract
Polo-like kinase 1, a pivotal regulator of mitosis and cytokinesis, is highly expressed in a broad spectrum of tumors and its expression correlates often with poor prognosis, suggesting its potential as a therapeutic target. p53, the guardian of the genome, is the most important tumor suppressor. In this review, we address the intertwined relationship of these two key molecules by fighting each other as eternal rivals in many signaling pathways. p53 represses the promoter of Polo-like kinase 1, whereas Polo-like kinase 1 inhibits p53 and its family members p63 and p73 in cancer cells lacking functional p53. Plk1 inhibitors target all rapidly dividing cells irrespective of tumor cells or non-transformed normal but proliferating cells. Upon treatment with Plk1 inhibitors, p53 in tumor cells is activated and induces strong apoptosis, whereas tumor cells with inactive p53 arrest in mitosis with DNA damage. Thus, inactive p53 is not associated with a susceptible cytotoxicity of Polo-like kinase 1 inhibition and could rather foster the induction of polyploidy/aneuploidy in surviving cells. In addition, compared to the mono-treatment, combination of Polo-like kinase 1 inhibition with anti-mitotic or DNA damaging agents boosts more severe mitotic defects, effectually triggers apoptosis and strongly inhibits proliferation of cancer cells with functional p53. In this regard, restoration of p53 in tumor cells with loss or mutation of p53 will reinforce the cytotoxicity of combined Polo-like kinase 1 therapy and provide a proficient strategy for combating relapse and metastasis of cancer.
Collapse
Affiliation(s)
- Frank Louwen
- Department of Gynecology and Obstetrics, School of Medicine, J. W. Goethe-University, Frankfurt, Germany
| | - Juping Yuan
- Department of Gynecology and Obstetrics, School of Medicine, J. W. Goethe-University, Frankfurt, Germany
| |
Collapse
|
20
|
Centrosome aberrations associated with cellular senescence and p53 localization at supernumerary centrosomes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2012; 2012:217594. [PMID: 23091651 PMCID: PMC3471474 DOI: 10.1155/2012/217594] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 08/27/2012] [Accepted: 09/11/2012] [Indexed: 12/12/2022]
Abstract
Centrosome overduplication or amplification has been observed in many human cancers and in premalignant tissue, but the mechanisms leading to such centrosome aberrations are not fully understood. We previously showed that abnormal mitotic cells with supernumerary centrosomes increase with replicative senescence in human fibroblasts, especially in a polyploid subpopulation. This study examines localization of p53 protein at centrosomes in mitotic cells, which is often observed in association with DNA damage response, to investigate a possible association between p53 localization and numerical centrosome aberrations induced by cellular senescence. Cultures at later passages or the 4th day after exposure to H(2)O(2) showed increased frequencies of mitotic cells with supernumerary centrosomes, especially in a polyploid subpopulation. Immunohistochemical analysis frequently showed p53-positive foci in mitotic cells, and some were localized at centrosomes. The number of p53-positive foci in mitotic cells and its localization to centrosomes increased with replicative and premature senescence. Supernumerary centrosomes showed higher frequencies of p53 localization compared to normally duplicated centrosomes. Centrosome-associated p53 protein was phosphorylated at Ser15. These data suggest a possible association between localization of p53 protein and numerical centrosome aberrations in replicatively or prematurely senescent cells.
Collapse
|
21
|
Siegel D, Kepa JK, Ross D. NAD(P)H:quinone oxidoreductase 1 (NQO1) localizes to the mitotic spindle in human cells. PLoS One 2012; 7:e44861. [PMID: 22984577 PMCID: PMC3439439 DOI: 10.1371/journal.pone.0044861] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 08/08/2012] [Indexed: 01/18/2023] Open
Abstract
NAD(P)H:quinone oxidoreductase 1 (NQO1) is an FAD containing quinone reductase that catalyzes the 2-electron reduction of a broad range of quinones. The 2-electron reduction of quinones to hydroquinones by NQO1 is believed to be a detoxification process since this reaction bypasses the formation of the highly reactive semiquinone. NQO1 is expressed at high levels in normal epithelium, endothelium and adipocytes as well as in many human solid tumors. In addition to its function as a quinone reductase NQO1 has been shown to reduce superoxide and regulate the 20 S proteasomal degradation of proteins including p53. Biochemical studies have indicated that NQO1 is primarily located in the cytosol, however, lower levels of NQO1 have also been found in the nucleus. In these studies we demonstrate using immunocytochemistry and confocal imaging that NQO1 was found associated with mitotic spindles in cells undergoing division. The association of NQO1 with the mitotic spindles was observed in many different human cell lines including nontransformed cells (astrocytes, HUVEC) immortalized cell lines (HBMEC, 16HBE) and cancer (pancreatic adenocarcinoma, BXPC3). Confocal analysis of double-labeling experiments demonstrated co-localization of NQO1with alpha-tubulin in mitotic spindles. In studies with BxPc-3 human pancreatic cancer cells the association of NQO1 with mitotic spindles appeared to be unchanged in the presence of NQO1 inhibitors ES936 or dicoumarol suggesting that NQO1 can associate with the mitotic spindle and still retain catalytic activity. Analysis of archival human squamous lung carcinoma tissue immunostained for NQO1 demonstrated positive staining for NQO1 in the spindles of mitotic cells. The purpose of this study is to demonstrate for the first time the association of the quinone reductase NQO1 with the mitotic spindle in human cells.
Collapse
Affiliation(s)
- David Siegel
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America.
| | | | | |
Collapse
|
22
|
p53: guardian of ploidy. Mol Oncol 2011; 5:315-23. [PMID: 21852209 DOI: 10.1016/j.molonc.2011.07.007] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 07/21/2011] [Accepted: 07/21/2011] [Indexed: 11/20/2022] Open
Abstract
Aneuploidy, often preceded by tetraploidy, is one of the hallmarks of solid tumors. Indeed, both aneuploidy and tetraploidy are oncogenic occurrences that are sufficient to drive neoplastic transformation and cancer progression. True to form, the tumor suppressor p53 obstructs propagation of these dangerous chromosomal events by either instigating irreversible cell cycle arrest or apoptosis. The tumor suppressor Lats2, along with other tumor inhibitory proteins such as BRCA1/2 and BubR1, are central to p53-dependent elimination of tetraploid cells. Not surprisingly, these proteins are frequently inactivated or downregulated in tumors, synergizing with p53 inactivation to establish an atmosphere of "tolerance" for a non-diploid state.
Collapse
|
23
|
Zeng F, Tian Y, Shi S, Wu Q, Liu S, Zheng H, Yue L, Li Y. Identification of mouse MARVELD1 as a microtubule associated protein that inhibits cell cycle progression and migration. Mol Cells 2011; 31:267-74. [PMID: 21347699 PMCID: PMC3932696 DOI: 10.1007/s10059-011-0037-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 12/15/2010] [Accepted: 12/24/2010] [Indexed: 12/20/2022] Open
Abstract
MARVEL domain-containing 1 (MARVELD1) is a newly identified nuclear protein; however its function has not been clear until now. Here, we report that mouse MARVELD1 (mMARVELD1), which is highly conserved between mice and humans, exhibits cell cycle-dependent cellular localization. In NIH3T3 cells, MARVELD1 was observed in the nucleus and at the perinuclear region during interphase, but was localized at the mitotic spindle and midbody at metaphase, and a significant fraction of mMARVELD1 translocated to the plasma membrane during anaphase. In addition, treatment of cells with colchicine, a microtubule-depolymerizing agent, resulted in translocation of mMARVELD1 to the plasma membrane, and association of mMARVELD1 and α-tubulin was confirmed by co-immunoprecipitation. Finally, overexpression of mMARVELD1 resulted in a remarkable inhibition of cell proliferation, G1-phase arrest, and reduced cell migration. These findings indicate that mMARVELD1 is a microtubule-associated protein that plays an important role in cell cycle progression and migration.
Collapse
Affiliation(s)
- Fanli Zeng
- Department of Life Science and Engineering, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| | - Yanyan Tian
- Department of Life Science and Engineering, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| | - Shuliang Shi
- Department of Life Science and Engineering, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| | - Qiong Wu
- Department of Life Science and Engineering, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| | - Shanshan Liu
- Department of Life Science and Engineering, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| | - Hongxia Zheng
- Department of Life Science and Engineering, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| | - Lei Yue
- The Academy of Fundamental and Interdisciplinary Science, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| | - Yu Li
- Department of Life Science and Engineering, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
- The Academy of Fundamental and Interdisciplinary Science, Harbin Institute of Technology, Harbin 150001, People’s Republic of China
| |
Collapse
|
24
|
Koledova Z, Krämer A, Kafkova LR, Divoky V. Cell-cycle regulation in embryonic stem cells: centrosomal decisions on self-renewal. Stem Cells Dev 2010; 19:1663-78. [PMID: 20594031 DOI: 10.1089/scd.2010.0136] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Embryonic stem cells seem to have the intriguing capacity to divide indefinitely while retaining their pluripotency. This self-renewal is accomplished by specialized mechanisms of cell-cycle control. In the last few years, several studies have provided evidence for a direct link between cell-cycle regulation and cell-fate decisions in stem cells. In this review, we discuss the peculiarities of embryonic stem cell-cycle control mechanisms, implicate their involvement in cell-fate decisions, and distinguish centrosomes as important players in the self-renewal versus differentiation roulette.
Collapse
Affiliation(s)
- Zuzana Koledova
- Department of Biology, Faculty of Medicine, Palacky University, Olomouc, Czech Republic.
| | | | | | | |
Collapse
|
25
|
Hernandez P, Tirnauer JS. Tumor suppressor interactions with microtubules: keeping cell polarity and cell division on track. Dis Model Mech 2010; 3:304-15. [DOI: 10.1242/dmm.004507] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tumor suppressor proteins protect cells and tissues from malignant transformation. Among their diverse actions, many of these proteins interact with the microtubule cytoskeleton. This review focuses on the interactions of several tumor suppressors with microtubules and speculates on how disruption of microtubule-dependent processes may contribute to cancer development and spread. We conclude that several tumor suppressors stabilize microtubules and organize microtubule arrays, functions that are likely to be important in preventing tumorigenesis. How tumor suppressors link microtubule stability with cell fate, and how their mutation affects the response of cancer cells to anti-microtubule chemotherapy drugs, remains unclear; these should prove fertile areas for future research.
Collapse
Affiliation(s)
- Paula Hernandez
- Center for Molecular Medicine and Neag Comprehensive Cancer Center, University of Connecticut Health Center, Farmington, CT 06030-3101, USA
| | - Jennifer S. Tirnauer
- Center for Molecular Medicine and Neag Comprehensive Cancer Center, University of Connecticut Health Center, Farmington, CT 06030-3101, USA
| |
Collapse
|
26
|
Abstract
The role of aneuploidy in tumorigenesis remains poorly understood, although the two have been known to be linked for more than 100 years. Recent studies indicate that aneuploidy can promote tumour cell growth and cell death and that the cellular outcome is dependent on the extent of aneuploidy induced. The mitotic checkpoint plays a pivotal role in the maintenance of genome stability and has been the focus of work investigating the distinct outcomes of aneuploidy. In the present article, we review the molecular mechanisms involved and discuss the potential of the mitotic checkpoint as a therapeutic target in cancer therapy.
Collapse
|
27
|
Localization of TEIF in the centrosome and its functional association with centrosome amplification in DNA damage, telomere dysfunction and human cancers. Oncogene 2009; 28:1549-60. [PMID: 19198626 DOI: 10.1038/onc.2008.503] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Centrosome amplification and telomere shortening, which are commonly detected in human cancers, have been implicated in the induction of chromosome instability in tumorigenesis. The functions of these two structures are closely related to DNA damage repair machinery, and some factors that operate in the maintenance of telomeres also take part in the regulation of centrosome status, suggesting they are functionally linked. We report that TEIF (telomerase transcriptional elements-interacting factor), a transactivator of the hTERT (human telomerase reverse transcriptase subunit) gene, is distributed in the centrosome throughout the cell cycle, but its transport into the centrosome is increased under some conditions, and its distribution is dependent on its C-terminal domain. Experimental modulation of TEIF expression through overexpression, polypeptide expression or depletion affected centrosome status and increased abnormalities of cell mitosis. Localization of TEIF to the centrosome was also stimulated by treatment with genotoxic agents and experimental telomere dysfunction, accompanying centrosome amplification. Moreover, we demonstrated that the expression level of TEIF is not only closely correlated with centrosome amplification in soft tissue sarcomas but it is also significantly related to tumor histologic grade. Our data confirmed TEIF functions as a centrosome regulator. Its participation in DNA damage response, including telomere dysfunction and tumorigenesis, indicates TEIF is likely to be a factor involved in linking centrosome amplification and telomere dysfunction in cancer development.
Collapse
|
28
|
Pascreau G, Eckerdt F, Lewellyn AL, Prigent C, Maller JL. Phosphorylation of p53 is regulated by TPX2-Aurora A in xenopus oocytes. J Biol Chem 2009; 284:5497-505. [PMID: 19121998 PMCID: PMC2645813 DOI: 10.1074/jbc.m805959200] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
p53 is an important tumor suppressor regulating the cell cycle at multiple
stages in higher vertebrates. The p53 gene is frequently deleted or mutated in
human cancers, resulting in loss of p53 activity. This leads to centrosome
amplification, aneuploidy, and tumorigenesis, three phenotypes also observed
after overexpression of the oncogenic kinase Aurora A. Accordingly, recent
studies have focused on the relationship between these two proteins. p53 and
Aurora A have been reported to interact in mammalian cells, but the function
of this interaction remains unclear. We recently reported that
Xenopus p53 can inhibit Aurora A activity in vitro but only
in the absence of TPX2. Here we investigate the interplay between
Xenopus Aurora A, TPX2, and p53 and show that newly synthesized TPX2
is required for nearly all Aurora A activation and for full p53 synthesis and
phosphorylation in vivo during oocyte maturation. In vitro,
phosphorylation mediated by Aurora A targets serines 129 and 190 within the
DNA binding domain of p53. Glutathione S-transferase pull-down
studies indicate that the interaction occurs via the p53 transactivation
domain and the Aurora A catalytic domain around the T-loop. Our studies
suggest that targeting of TPX2 might be an effective strategy for specifically
inhibiting the phosphorylation of Aurora A substrates, including p53.
Collapse
Affiliation(s)
- Gaetan Pascreau
- Howard Hughes Medical Institute and Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | | | | | | | | |
Collapse
|
29
|
Vakifahmetoglu H, Olsson M, Zhivotovsky B. Death through a tragedy: mitotic catastrophe. Cell Death Differ 2008; 15:1153-62. [PMID: 18404154 DOI: 10.1038/cdd.2008.47] [Citation(s) in RCA: 485] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Mitotic catastrophe (MC) has long been considered as a mode of cell death that results from premature or inappropriate entry of cells into mitosis and can be caused by chemical or physical stresses. Whereas it initially was depicted as the main form of cell death induced by ionizing radiation, it is today known to be triggered also by treatment with agents influencing the stability of microtubule, various anticancer drugs and mitotic failure caused by defective cell cycle checkpoints. Although various descriptions explaining MC exist, there is still no general accepted definition of this phenomenon. Here, we present evidences indicating that death-associated MC is not a separate mode of cell death, rather a process ('prestage') preceding cell death, which can occur through necrosis or apoptosis. The final outcome of MC depends on the molecular profile of the cell.
Collapse
Affiliation(s)
- H Vakifahmetoglu
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | | |
Collapse
|
30
|
Abstract
Chromosome instability, which is equated to mitotic defects and consequential chromosome segregation errors, provides a formidable basis for the acquisition of further malignant phenotypes during tumour progression. Centrosomes have a crucial role in the formation of bipolar mitotic spindles, which are essential for accurate chromosome segregation. Mutations of certain oncogenic and tumour-suppressor proteins directly induce chromosome instability by disrupting the normal function and numeral integrity of centrosomes. How these proteins control centrosome duplication and function, and how their mutational activation and/or inactivation results in numeral and functional centrosome abnormalities, is discussed in this Review.
Collapse
Affiliation(s)
- Kenji Fukasawa
- Molecular Oncology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA.
| |
Collapse
|
31
|
Hildrestrand GA, Rolseth V, Bjørås M, Luna L. Human NEIL1 localizes with the centrosomes and condensed chromosomes during mitosis. DNA Repair (Amst) 2007; 6:1425-33. [PMID: 17556049 DOI: 10.1016/j.dnarep.2007.04.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Revised: 03/22/2007] [Accepted: 04/03/2007] [Indexed: 01/01/2023]
Abstract
The DNA glycosylase hNEIL1 initiates base excision repair (BER) of a number of oxidized purines and pyrimidines in cellular DNA and is one of three mammalian orthologs of the Escherichia coli Nei/Fpg enzymes. Human NEIL1 has been purified and extensively characterized biochemically, however, not much is known about its intracellular distribution. In the present work, we have studied the cellular localization of hNEIL1 using both antibodies raised against the full-length recombinant protein and a stable HeLa cell line expressing hNEIL1 fused N-terminal to EGFP. The results presented reveal an intricate mitotic distribution of hNEIL1. Centrosomal localization of hNEIL1 was observed when mitotic HeLa cells were immunostained with hNEIL1 antibodies. This localization was confirmed when Western blots of isolated centrosomes from stably expressing hNEIL1-EGFP HeLa cells were probed with GFP or hNEIL1 antibodies, even though a fluorescent signal could not be detected in the centrosomes of these cells. Human NEIL1 was also shown to be associated with mitotic condensed chromosomes. Notably, the interaction of hNEIL1 with condensed chromatin was disrupted when cells were fixed with chemical fixatives that are regularly used in immunodetection techniques.
Collapse
Affiliation(s)
- Gunn A Hildrestrand
- Centre for Molecular Biology and Neuroscience and Institute of Medical Microbiology, Rikshospitalet-Radiumhospitalet HF, 0027 Oslo, Norway
| | | | | | | |
Collapse
|
32
|
Srsen V, Merdes A. The centrosome and cell proliferation. Cell Div 2006; 1:26. [PMID: 17109756 PMCID: PMC1654144 DOI: 10.1186/1747-1028-1-26] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2006] [Accepted: 11/16/2006] [Indexed: 11/15/2022] Open
Abstract
Centrosomes are frequently amplified in cancer cells. Increased numbers of centrosomes can give rise to multipolar spindles in mitosis, and thereby lead to the formation of aneuploid daughter cells. However, whether centrosome amplification is a cause or a consequence of cancer is unclear. In contrast, loss of a functional centrosome has been shown to lead to cell cycle arrest. In this review, the potential mechanisms underlying centrosome amplification and centrosome-dependent cell cycle regulation are discussed.
Collapse
Affiliation(s)
- Vlastimil Srsen
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, King's Buildings, Edinburgh EH9 3JR, UK
| | - Andreas Merdes
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, King's Buildings, Edinburgh EH9 3JR, UK
| |
Collapse
|
33
|
Shinmura K, Bennett RA, Tarapore P, Fukasawa K. Direct evidence for the role of centrosomally localized p53 in the regulation of centrosome duplication. Oncogene 2006; 26:2939-44. [PMID: 17072342 DOI: 10.1038/sj.onc.1210085] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Abnormal amplification of centrosomes is the major cause of mitotic defects and chromosome instability in cancer cells. Centrosomes duplicate once in each cell cycle, and abrogation of the regulatory mechanism underlying centrosome duplication leads to centrosome amplification. p53 tumor suppressor protein is involved in the regulation of centrosome duplication: loss of p53 as well as expression of certain p53 mutants result in deregulated centrosome duplication and centrosome amplification. p53 at least in part depends on its transactivation function to control centrosome duplication, primarily via upregulation of p21 cyclin-dependent kinase (CDK) inhibitor, which prevents untimely activation of CDK2/cyclin E, a key initiator of centrosome duplication. However, numerous studies have shown the presence of p53 at centrosomes, yet the role of the centrosomally localized p53 in the regulation of centrosome duplication had been enigmatic. Here, we comparatively examined wild-type p53 and p53 mutants that are transactivation(+)/centrosome-binding(-), transactivation(-)/centrosome-binding(+) and transactivation(-)/centrosome-binding(-) for their abilities to control centrosome duplication. We found that the transactivation(+)/centrosome-binding(-) and transactivation(-)/centrosome-binding(+) mutants suppress centrosome duplication only partially compared with wild-type p53. Moreover, the transactivation(-)/centrosome-binding(-) mutant almost completely lost the ability to suppress centrosome duplication. These observations provide direct evidence for the centrosomally localized p53 to participate in the regulation of centrosome duplication in a manner independent of its transactivation function in addition to its transactivation-dependent regulation of centrosome duplication.
Collapse
Affiliation(s)
- K Shinmura
- Department of Cell Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | | | | | | |
Collapse
|
34
|
Srsen V, Gnadt N, Dammermann A, Merdes A. Inhibition of centrosome protein assembly leads to p53-dependent exit from the cell cycle. ACTA ACUST UNITED AC 2006; 174:625-30. [PMID: 16943179 PMCID: PMC2064305 DOI: 10.1083/jcb.200606051] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous evidence has indicated that an intact centrosome is essential for cell cycle progress and that elimination of the centrosome or depletion of individual centrosome proteins prevents the entry into S phase. To investigate the molecular mechanisms of centrosome-dependent cell cycle progress, we performed RNA silencing experiments of two centrosome-associated proteins, pericentriolar material 1 (PCM-1) and pericentrin, in primary human fibroblasts. We found that cells depleted of PCM-1 or pericentrin show lower levels of markers for S phase and cell proliferation, including cyclin A, Ki-67, proliferating cell nuclear antigen, minichromosome maintenance deficient 3, and phosphorylated retinoblastoma protein. Also, the percentage of cells undergoing DNA replication was reduced by >50%. At the same time, levels of p53 and p21 increased in these cells, and cells were predisposed to undergo senescence. Conversely, depletion of centrosome proteins in cells lacking p53 did not cause any cell cycle arrest. Inhibition of p38 mitogen-activated protein kinase rescued cell cycle activity after centrosome protein depletion, indicating that p53 is activated by the p38 stress pathway.
Collapse
Affiliation(s)
- Vlastimil Srsen
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3JR, Scotland, UK
| | | | | | | |
Collapse
|
35
|
Ma Z, Izumi H, Kanai M, Kabuyama Y, Ahn NG, Fukasawa K. Mortalin controls centrosome duplication via modulating centrosomal localization of p53. Oncogene 2006; 25:5377-90. [PMID: 16619038 DOI: 10.1038/sj.onc.1209543] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Abnormal amplification of centrosomes, commonly found in human cancer, is the major cause of mitotic defects and chromosome instability in cancer cells. Like DNA, centrosomes duplicate once in each cell cycle, hence the defect in the mechanism that ensures centrosome duplication to occur once and only once in each cell cycle results in abnormal amplification of centrosomes and mitotic defects. Centrosomes are non-membranous organelles, and undergo dynamic changes in its constituents during the centrosome duplication cycle. Through a comparative mass spectrometric analysis of unduplicated and duplicated centrosomes, we identified mortalin, a member of heat shock protein family, as a protein that associates preferentially with duplicated centrosomes. Further analysis revealed that mortalin localized to centrosomes in late G1 before centrosome duplication, remained at centrosomes during S and G2, and dissociated from centrosomes during mitosis. Overexpression of mortalin overrides the p53-dependent suppression of centrosome duplication, and mortalin-driven centrosome duplication requires physical interaction between mortalin and p53. Moreover, mortalin promotes dissociation of p53 from centrosomes through physical interaction. The p53 mutant that lacks the ability to bind to mortalin remains at centrosomes, and suppresses centrosome duplication in a transactivation function-independent manner. Thus, our present findings not only identify mortalin as an upstream molecule of p53 but also provide evidence for the involvement of centrosomally localized p53 in the regulation of centrosome duplication.
Collapse
Affiliation(s)
- Z Ma
- Department of Cell Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | | | | | | | | | | |
Collapse
|
36
|
Zhao Y, Zhang N, Kong Q. Does the cell-brain theory work in explaining carcinogenesis? Med Hypotheses 2006; 65:708-15. [PMID: 15975733 DOI: 10.1016/j.mehy.2005.04.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2005] [Revised: 03/25/2005] [Accepted: 04/12/2005] [Indexed: 10/25/2022]
Abstract
As a major microtubule-organizing center, the centrosome, together with the embedded centrioles and connecting filaments (or microtubules), has lately been proposed to be the "brain" of a cell. Although there are a lot of works to be done to test this hypothesis, emerging data have suggested that this centrosome-centered "cell brain" is playing increasingly important roles in cell control. Genes seem not to tell the whole story, despite the commonly held view that genetic alteration is the cause of most medical problems including cancer development. Although the mechanisms through which gene expression and protein synthesis are regulated remain to be studied, current advances in our understanding of the roles of the centrosome in the regulation of DNA synthesis, DNA repair, cell cycle, apoptosis and in the maintenance of genetic stability are challenging our tradition thoughts. Genetic alterations may be repaired by the centrosome-centered "cell brain"-mediated self-defense, but the cell brain defects intend to cause genetic alterations, which, in turn, may result in cancer development. Further understanding of the roles of the centrosome/cell brain in these and other new aspects are becoming very helpful in comprehending why and how medical problems including tumors develop. Meanwhile, it suggests that great attention should be given to the centrosome/cell brain, instead of gene alone when treating medical problems, which is discussed in this paper on the basis of cell brain theory and may prove helpful in shedding light on the often paradoxical observations seen in cell control, particularly in cancer development.
Collapse
Affiliation(s)
- Yunfeng Zhao
- Cell Brain Research Center, School of Life Science, Shandong University, Room 128, Biology Building, Jinan, Shandong Province 250100, China
| | | | | |
Collapse
|
37
|
Zhang Z, Yang Y, Gong A, Wang C, Liang Y, Chen Y. Localization of NGF and TrkA at mitotic apparatus in human glioma cell line U251. Biochem Biophys Res Commun 2005; 337:68-74. [PMID: 16181609 DOI: 10.1016/j.bbrc.2005.08.265] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2005] [Accepted: 08/30/2005] [Indexed: 10/25/2022]
Abstract
It has previously been implicated that nerve growth factor (NGF) with its high-affinity receptor tyrosine kinase A (TrkA) could play an important role in the growth modulation of human tumor cells, such as glioblastoma multiform cell lines and human breast cancer cell lines. However, the direct mitogenic effects of NGF and TrkA in these tumor cells still remain to be elucidated. Herein we show, by immunofluorescence staining, that NGF was colocalized with gamma-tubulin at the centrosomes or the spindle poles throughout the cell cycle and phosphorylated TrkA was colocalized with alpha-tubulin at mitotic spindle in the glioma cell line U251. The results suggest that NGF concentrated to centrosome can recruit its receptor TrkA there and cause phosphorylation of the latter. The phosphorylated TrkA with the tyrosine kinase activity may phosphorylate the tubulin and promote the mitotic spindle assembly. By these mechanisms, NGF can modulate the mitosis of human glioma cells.
Collapse
Affiliation(s)
- Zhijian Zhang
- School of Medicine, Jiangsu University, Jiangsu Zhenjiang 212001, China
| | | | | | | | | | | |
Collapse
|
38
|
Duensing A, Duensing S. Guilt by association? p53 and the development of aneuploidy in cancer. Biochem Biophys Res Commun 2005; 331:694-700. [PMID: 15865924 DOI: 10.1016/j.bbrc.2005.03.157] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2005] [Indexed: 02/07/2023]
Abstract
Aneuploidy is one of the most frequent genetic alterations in solid tumors. It is commonly caused by cell division errors that are induced by oncogene activation or loss of tumor suppressor functions. In addition, certain viral oncoproteins have been implicated in the induction of chromosome copy number changes. Aneuploidy and inactivation of p53 frequently coincide in human cancers but there is increasing evidence that loss of p53 by itself is not a primary cause of aneuploidy. Nonetheless, p53 inactivation synergizes with additional oncogenic events to promote aneuploidy and may facilitate chromosomal imbalances through indirect mechanisms. This review summarizes the current knowledge about the association between aneuploidy and p53, and discusses two of the most controversial mechanisms that have been implicated in genomic instability associated with loss of p53: subversion of ploidy control and aberrant centrosome duplication.
Collapse
Affiliation(s)
- Anette Duensing
- Molecular Virology Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
39
|
Vogel C, Kienitz A, Hofmann I, Müller R, Bastians H. Crosstalk of the mitotic spindle assembly checkpoint with p53 to prevent polyploidy. Oncogene 2004; 23:6845-53. [PMID: 15286707 DOI: 10.1038/sj.onc.1207860] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Treatment of cells with microtubule inhibitors results in activation of the mitotic spindle assembly checkpoint, leading to mitotic arrest before anaphase. Upon prolonged treatment, however, cells can adapt and exit mitosis aberrantly, resulting in the occurrence of tetraploid cells in G1. Those cells subsequently arrest in postmitotic G1 due to the activation of a p53-dependent G1 checkpoint. Failure of the G1 checkpoint leads to endoreduplication and further polyploidization. Using HCT116 and isogenic p53-deficient or spindle checkpoint compromised derivatives, we show here that not only p53 but also a functional spindle assembly checkpoint is required for postmitotic G1 checkpoint function. During transient mitotic arrest, p53 stabilization and activation is triggered by a pathway independent of ATM/ATR, Chk1 and Chk2. We further show that a prolonged spindle checkpoint-mediated mitotic arrest is required for proper postmitotic G1 checkpoint function. In addition, we demonstrate that polyploid cells are inhibited to re-enter mitosis by an additional checkpoint acting in G2. Thus, during a normal cell cycle, polyploidization and subsequent aneuploidization is prevented by the function of the mitotic spindle checkpoint, a p53-dependent G1 checkpoint and an additional G2 checkpoint.
Collapse
Affiliation(s)
- Celia Vogel
- Institute for Molecular Biology and Tumor Research, Philipps University Marburg, Emil-Mannkopff-Strasse 2, D-35037 Marburg, Germany
| | | | | | | | | |
Collapse
|
40
|
Wang Q, Hirohashi Y, Furuuchi K, Zhao H, Liu Q, Zhang H, Murali R, Berezov A, Du X, Li B, Greene MI. The Centrosome in Normal and Transformed Cells. DNA Cell Biol 2004; 23:475-89. [PMID: 15307950 DOI: 10.1089/1044549041562276] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The centrosome is a unique organelle that functions as the microtubule organizing center in most animal cells. During cell division, the centrosomes form the poles of the bipolar mitotic spindle. In addition, the centrosomes are also needed for cytokinesis. Each mammalian somatic cell typically contains one centrosome, which is duplicated in coordination with DNA replication. Just like the chromosomes, the centrosome is precisely reproduced once and only once during each cell cycle. However, it remains a mystery how this protein-based structure undergoes accurate duplication in a semiconservative manner. Intriguingly, amplification of the centrosome has been found in numerous forms of cancers. Cells with multiple centrosomes tend to form multipolar spindles, which result in abnormal chromosome segregation during mitosis. It has therefore been postulated that centrosome aberration may compromise the fidelity of cell division and cause chromosome instability. Here we review the current understanding of how the centrosome is assembled and duplicated. We also discuss the possible mechanisms by which centrosome abnormality contributes to the development of malignant phenotype.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Reini K, Uitto L, Perera D, Moens PB, Freire R, Syväoja JE. TopBP1 localises to centrosomes in mitosis and to chromosome cores in meiosis. Chromosoma 2004; 112:323-30. [PMID: 15138768 DOI: 10.1007/s00412-004-0277-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2003] [Accepted: 03/08/2004] [Indexed: 01/24/2023]
Abstract
Topoisomerase IIbeta binding protein 1 (TopBP1), previously shown to localise to sites of DNA damage and to stalled replication forks, has been implicated in DNA replication and in DNA damage response. In this work we showed that TopBP1 was localised in structures other than stalled replication forks. In late mitosis TopBP1 localises to centrosomes in a manner similar to other DNA damage response proteins such as BRCA1 and p53. Spindle checkpoint activation does not affect this centrosomal localisation. Moreover, in the testis, we detected high levels of TopBP1 associated with meiotic prophase chromosome cores and the X-Y pair. Together, these data suggest a direct role of TopBP1 during both mitosis and meiotic prophase I.
Collapse
Affiliation(s)
- Kaarina Reini
- Biocenter Oulu and Department of Biochemistry, P.O. Box 3000, 90014, University of Oulu, Finland
| | | | | | | | | | | |
Collapse
|
42
|
Garriga-Canut M, Orkin SH. Transforming acidic coiled-coil protein 3 (TACC3) controls friend of GATA-1 (FOG-1) subcellular localization and regulates the association between GATA-1 and FOG-1 during hematopoiesis. J Biol Chem 2004; 279:23597-605. [PMID: 15037632 DOI: 10.1074/jbc.m313987200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Physical association between the transcription factor GATA-1 and the cofactor, Friend of GATA-1 (FOG-1), is essential for the differentiation of two blood cell types, erythroid cells and megakaryocytes. However, little is known regarding the mechanisms that modulate their interaction within cells. In the present study, we have identified TACC3 as a FOG-1-interacting protein. Transforming acidic coiled-coil protein 3 (TACC3), a protein that is highly expressed in hematopoietic cells, has been reported to have a critical role in the expansion of immature hematopoietic progenitors. We show that TACC3 affects FOG-1 nuclear localization, altering the interaction between GATA-1 and FOG-1. However, GATA-1 competes with TACC3 in the interaction with FOG-1. We observe that high levels of TACC3 inhibit the function of FOG-1 as a transcriptional cofactor of GATA-1. Furthermore, forced expression of TACC3 to levels similar to those found in progenitor cells delays terminal maturation of MEL and G1ER cells, two cell models of erythroid cell development. We suggest a role for TACC3 in regulating the cellular distribution of FOG-1 and thus the direct interaction of GATA-1 and FOG-1 as a mechanism to control the transition between expansion of multipotential progenitor cell populations and final stages of erythroid maturation.
Collapse
Affiliation(s)
- Mireia Garriga-Canut
- Division of Hematology/Oncology, Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School and Howard Hughes Medical Institute, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
43
|
Ou Y, Rattner JB. The Centrosome in Higher Organisms: Structure, Composition, and Duplication. INTERNATIONAL REVIEW OF CYTOLOGY 2004; 238:119-82. [PMID: 15364198 DOI: 10.1016/s0074-7696(04)38003-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The centrosome found in higher organisms is an organelle with a complex and dynamic architecture and composition. This organelle not only functions as a microtubule-organizing center, but also is integrated with or impacts a number of cellular processes. Defects associated with this organelle have been linked to a variety of human diseases including several forms of cancer. Here we review the emerging picture of how the structure, composition, duplication, and function of the centrosome found in higher organisms are interrelated.
Collapse
Affiliation(s)
- Young Ou
- Department of Cell Biology and Anatomy, University of Calgary 3330 Hospital Drive NW, Calgary, Alberta, Canada
| | | |
Collapse
|
44
|
Abstract
p53 regulates a number of genes through transcriptional activation and repression. p53-dependent mitotic checkpoint has been described, but the underlying mechanism is still obscure. Here we examined the effect of p53 on the expression of a human mitotic checkpoint protein, Mitosis Arrest Deficiency 1 (MAD1), in cultured human cells. The expression of MAD1 was reduced when the cells were overexpressing exogenously introduced wild-type p53. The same reduction was also observed when the cells were treated with anticancer agents 5-fluorouracil and cisplatin or were irradiated with UV. Consistently, MAD1 promoter activity diminished in a dose-dependent manner when induced by p53, indicating that p53 repressed MAD1 at a transcriptional level. Intriguingly, several tumor hot spot mutations in p53 (V143A, R175H, R248W, and R273H) did not abolish the ability of p53 to repress MAD1 expression. By serial truncation of the MAD1 promoter, we confined the p53-responsive element to a 38-bp region that represents a novel sequence distinct from the known p53 consensus binding site. Trichostatin A, a histone deacetylase inhibitor, relieved the p53 transrepression activity on MAD1. Chromatin immunoprecipitation assay revealed that p53, histone deacetylase 1, and co-repressor mSin3a associated with the MAD1 promoter in vivo. Taken together, our findings suggest a regulatory mechanism for the mitotic checkpoint in which MAD1 is inhibited by p53.
Collapse
Affiliation(s)
- Abel C S Chun
- Department of Biochemistry, the University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
45
|
Abstract
Apoptosis (or programmed cell death) is one of the central cellular processes in development, stress response, aging, carcinogenesis, and disease in multi-cellular eukaryotes. Although great effort has been made, the detailed mechanism through which apoptosis is initiated is yet unclear. Previously, the centrosome, or more explicitly the complex comprising the centrosome, centrioles, and connecting filaments, was reported to be required for apoptosis. It may be through this 'cell brain', reminiscent of the long known brain of animals (or humans), that complicated cellular processes, including apoptosis, are precisely coordinated. In this paper, the latest data to support this contention are scrutinized.
Collapse
Affiliation(s)
- Q Kong
- Cell Brain Research Center, Shandong University, Jinan, China.
| |
Collapse
|
46
|
Abstract
Cancer has long been regarded as a genetic disease. Therefore, current theories on cancer development focus on genetic alterations affecting oncogenes or tumor suppressor genes. However, the mechanisms through which genetic alterations are induced are largely unknown. In this paper a theory will be developed which interprets cancer as a cell brain illness rather than a genetic disease. The complex comprising the centrosome, normally two centrioles and connecting filaments, was recently termed the 'cell brain', and was found to determine a cell's fate. It is through the cell brain, instead of the genes, that genetic stability and expression are maintained and regulated. Accordingly, the nucleus is regarded as a safe storage for inheriting materials (genes) that primarily act as manufacturing templates. Therefore, cancer should be regarded as a 'brain illness' of a cell, instead of a genetic disease, which is strongly supported by the latest evidence, as discussed in this paper. Such a theory serves to better clarify the confusing observations in cancer development accumulated over the last decades.
Collapse
Affiliation(s)
- Q Kong
- Cell Brain Research Center, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
47
|
Shih JY, Lee YCG, Yang SC, Hong TM, Huang CYF, Yang PC. Collapsin response mediator protein-1: a novel invasion-suppressor gene. Clin Exp Metastasis 2003; 20:69-76. [PMID: 12650609 DOI: 10.1023/a:1022598604565] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Numerous genetic changes are associated with metastasis of cancer cells. Previously, we used microarray to identify that collapsin response mediator protein-1 (CRMP-1) was involved in cancer invasion and metastasis. We further characterized that CRMP-1 was a novel invasion-suppression gene. Members of the CRMP gene family are intracellular phosphoproteins involved in the mediation of semaphorin induced F-actin depolymerization and growth cone collapse. The precise mechanism by which CRMP-I inhibits invasion is not yet clear. However, CRMP-1 transfected cells had fewer filopodia and less Matrigel-invasion abilities. A low expression of CRMP-I mRNA in lung cancer tissue was significantly associated with advanced disease, lymph node metastasis, early post-operative relapse, and shorter survival. In this article, we reviewed the functions of CRMPs and semaphorins and analyzed the structure and motifs of CRMP-1 by bioinformatics. As such, we hoped to shed further light on the mechanism by which CRMP-1 suppresses the invasion of cancer cells.
Collapse
Affiliation(s)
- Jin-Yuan Shih
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
48
|
Kanai M, Tong WM, Sugihara E, Wang ZQ, Fukasawa K, Miwa M. Involvement of poly(ADP-Ribose) polymerase 1 and poly(ADP-Ribosyl)ation in regulation of centrosome function. Mol Cell Biol 2003; 23:2451-62. [PMID: 12640128 PMCID: PMC150716 DOI: 10.1128/mcb.23.7.2451-2462.2003] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The regulatory mechanism of centrosome function is crucial to the accurate transmission of chromosomes to the daughter cells in mitosis. Recent findings on the posttranslational modifications of many centrosomal proteins led us to speculate that these modifications might be involved in centrosome behavior. Poly(ADP-ribose) polymerase 1 (PARP-1) catalyzes poly(ADP-ribosyl)ation to various proteins. We show here that PARP-1 localizes to centrosomes and catalyzes poly(ADP-ribosyl)ation of centrosomal proteins. Moreover, centrosome hyperamplification is frequently observed with PARP inhibitor, as well as in PARP-1-null cells. Thus, it is possible that chromosomal instability known in PARP-1-null cells can be attributed to the centrosomal dysfunction. P53 tumor suppressor protein has been also shown to be localized at centrosomes and to be involved in the regulation of centrosome duplication and monitoring of the chromosomal stability. We found that centrosomal p53 is poly(ADP-ribosyl)ated in vivo and centrosomal PARP-1 directly catalyzes poly(ADP-ribosyl)ation of p53 in vitro. These results indicate that PARP-1 and PARP-1-mediated poly(ADP-ribosyl)ation of centrosomal proteins are involved in the regulation of centrosome function.
Collapse
Affiliation(s)
- Masayuki Kanai
- Department of Biochemistry and Molecular Oncology, Institute of Basic Medical Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | | | | | | | | | | |
Collapse
|
49
|
Sakai H, Urano T, Ookata K, Kim MH, Hirai Y, Saito M, Nojima Y, Ishikawa F. MBD3 and HDAC1, two components of the NuRD complex, are localized at Aurora-A-positive centrosomes in M phase. J Biol Chem 2002; 277:48714-23. [PMID: 12354758 DOI: 10.1074/jbc.m208461200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
MBD3, a component of the histone deacetylase NuRD complex, contains the methyl-CpG-binding domain (MBD), yet does not possess appreciable mCpG-specific binding activity. The functional significance of MBD3 in the NuRD complex remains enigmatic, partly because of the limited availability of biochemical approaches, such as immunoprecipitation, to analyze MBD3. In this study, we stably expressed the FLAG-tagged version of MBD3 in HeLa cells. We found that MBD3-FLAG was incorporated into the NuRD complex, and the MBD3-FLAG-containing NuRD complex was efficiently immunoprecipitated by anti-FLAG antibodies. By exploiting this system, we found that MBD3 is phosphorylated in vivo in the late G(2) and early M phases. Moreover, we found that Aurora-A, a serine/threonine kinase active specifically in the late G(2) and early M phases, phosphorylates MBD3 in vitro, physically associates with MBD3 in vivo, and co-localizes with MBD3 at the centrosomes in the early M phase. Interestingly, HDAC1 is distributed at the centrosomes in a manner similar to MBD3. These results suggest the highly dynamic nature of the temporal and spatial distributions, as well as the biochemical modification, of the NuRD complex in M phase, probably through an interaction with kinases, including Aurora-A. These observations will contribute significantly to the elucidation of the yet-uncharacterized cell cycle-controlled functions of the NuRD complex.
Collapse
Affiliation(s)
- Hirotaka Sakai
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama 226-8501, Japan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Centrosomes are microtubule organising centres that act as spindle poles during mitosis. Recent work implicates centrosomes in many other processes, and shows that centrosome defects can cause genetic instability. Many regulators of mammalian centrosome function were predicted from studies of model systems. Surprisingly, some well-known tumour suppressors have recently been found at centrosomes, where they influence centrosome duplication and function, suggesting that control of centrosome function is central to genetic stability.
Collapse
Affiliation(s)
- Harold A Fisk
- Molecular, Cellular and Developmental Biology, UCB347, University of Colorado, Boulder, CO 80309-0347, USA
| | | | | |
Collapse
|