1
|
Safieddine A, Benassy MN, Bonte T, Slimani F, Pourcelot O, Kress M, Ernoult-Lange M, Courel M, Coleno E, Imbert A, Laine A, Godebert AM, Vinit A, Blugeon C, Chevreux G, Gautheret D, Walter T, Bertrand E, Bénard M, Weil D. Cell-cycle-dependent mRNA localization in P-bodies. Mol Cell 2024; 84:4191-4208.e7. [PMID: 39368464 DOI: 10.1016/j.molcel.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 07/05/2024] [Accepted: 09/10/2024] [Indexed: 10/07/2024]
Abstract
Understanding the dynamics of RNA targeting to membraneless organelles is essential to disentangle their functions. Here, we investigate how P-bodies (PBs) evolve during cell-cycle progression in HEK293 cells. PB purification across the cell cycle uncovers widespread changes in their RNA content, partly uncoupled from cell-cycle-dependent changes in RNA expression. Single-molecule fluorescence in situ hybridization (FISH) shows various mRNA localization patterns in PBs peaking in G1, S, or G2, with examples illustrating the timely capture of mRNAs in PBs when their encoded protein becomes dispensable. Rather than directly reflecting absence of translation, cyclic mRNA localization in PBs can be controlled by RBPs, such as HuR in G2, and by RNA features. Indeed, while PB mRNAs are AU rich at all cell-cycle phases, they are specifically longer in G1, possibly related to post-mitotic PB reassembly. Altogether, our study supports a model where PBs are more than a default location for excess untranslated mRNAs.
Collapse
Affiliation(s)
- Adham Safieddine
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine (IBPS), Laboratoire de Biologie du Développement, 75005 Paris, France.
| | - Marie-Noëlle Benassy
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine (IBPS), Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Thomas Bonte
- Centre for Computational Biology (CBIO), Mines Paris, PSL University, 75006 Paris, France; Institut Curie, PSL University, 75005 Paris, France; INSERM, U900, 75005 Paris, France
| | - Floric Slimani
- Institut de Génétique Humaine, University of Montpellier, CNRS, 34090 Montpellier, France
| | - Oriane Pourcelot
- Institut de Génétique Humaine, University of Montpellier, CNRS, 34090 Montpellier, France
| | - Michel Kress
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine (IBPS), Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Michèle Ernoult-Lange
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine (IBPS), Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Maïté Courel
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine (IBPS), Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Emeline Coleno
- Institut de Génétique Humaine, University of Montpellier, CNRS, 34090 Montpellier, France
| | - Arthur Imbert
- Centre for Computational Biology (CBIO), Mines Paris, PSL University, 75006 Paris, France
| | - Antoine Laine
- Institute for Integrative Biology of the Cell, UMR 9198, CEA, CNRS, Université Paris-Saclay, 91190 Gif-Sur-Yvette, France
| | - Annie Munier Godebert
- Research Center Saint-Antoine (CRSA), CISA Flow Cytometry Facility, UMRS 938, Sorbonne University, F-75012 Paris, France
| | - Angelique Vinit
- Research Center Saint-Antoine (CRSA), CISA Flow Cytometry Facility, UMRS 938, Sorbonne University, F-75012 Paris, France
| | - Corinne Blugeon
- GenomiqueENS, Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Guillaume Chevreux
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Daniel Gautheret
- Institute for Integrative Biology of the Cell, UMR 9198, CEA, CNRS, Université Paris-Saclay, 91190 Gif-Sur-Yvette, France
| | - Thomas Walter
- Centre for Computational Biology (CBIO), Mines Paris, PSL University, 75006 Paris, France; Institut Curie, PSL University, 75005 Paris, France; INSERM, U900, 75005 Paris, France
| | - Edouard Bertrand
- Institut de Génétique Humaine, University of Montpellier, CNRS, 34090 Montpellier, France
| | - Marianne Bénard
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine (IBPS), Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Dominique Weil
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine (IBPS), Laboratoire de Biologie du Développement, 75005 Paris, France.
| |
Collapse
|
2
|
Mitchell DK, Burgess B, White EE, Smith AE, Potchanant EAS, Mang H, Hickey BE, Lu Q, Qian S, Bessler W, Li X, Jiang L, Brewster K, Temm C, Horvai A, Albright EA, Fishel ML, Pratilas CA, Angus SP, Clapp DW, Rhodes SD. Spatial Gene-Expression Profiling Unveils Immuno-oncogenic Programs of NF1-Associated Peripheral Nerve Sheath Tumor Progression. Clin Cancer Res 2024; 30:1038-1053. [PMID: 38127282 PMCID: PMC11095977 DOI: 10.1158/1078-0432.ccr-23-2548] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/25/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE Plexiform neurofibromas (PNF) are benign peripheral nerve sheath tumors (PNST) associated with neurofibromatosis type 1 (NF1). Despite similar histologic appearance, these neoplasms exhibit diverse evolutionary trajectories, with a subset progressing to malignant peripheral nerve sheath tumor (MPNST), the leading cause of premature death in individuals with NF1. Malignant transformation of PNF often occurs through the development of atypical neurofibroma (ANF) precursor lesions characterized by distinct histopathologic features and CDKN2A copy-number loss. Although genomic studies have uncovered key driver events promoting tumor progression, the transcriptional changes preceding malignant transformation remain poorly defined. EXPERIMENTAL DESIGN Here we resolve gene-expression profiles in PNST across the neurofibroma-to-MPNST continuum in NF1 patients and mouse models, revealing early molecular features associated with neurofibroma evolution and transformation. RESULTS Our findings demonstrate that ANF exhibit enhanced signatures of antigen presentation and immune response, which are suppressed as malignant transformation ensues. MPNST further displayed deregulated survival and mitotic fidelity pathways, and targeting key mediators of these pathways, CENPF and BIRC5, disrupted the growth and viability of human MPNST cell lines and primary murine Nf1-Cdkn2a-mutant Schwann cell precursors. Finally, neurofibromas contiguous with MPNST manifested distinct alterations in core oncogenic and immune surveillance programs, suggesting that early molecular events driving disease progression may precede histopathologic evidence of malignancy. CONCLUSIONS If validated prospectively in future studies, these signatures may serve as molecular diagnostic tools to augment conventional histopathologic diagnosis by identifying neurofibromas at high risk of undergoing malignant transformation, facilitating risk-adapted care.
Collapse
Affiliation(s)
- Dana K. Mitchell
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Breanne Burgess
- Medical Scientist Training Program, Indiana University School of Medicine
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine
| | - Emily E. White
- Medical Scientist Training Program, Indiana University School of Medicine
- Department of Medical and Molecular Genetics, Indiana University School of Medicine
| | - Abbi E. Smith
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | | | - Henry Mang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Brooke E. Hickey
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Qingbo Lu
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Shaomin Qian
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Waylan Bessler
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Xiaohong Li
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Li Jiang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Kylee Brewster
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Constance Temm
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Andrew Horvai
- Department of Pathology and Laboratory Medicine, University of California San Francisco
| | - Eric A. Albright
- Department of Clinical Pathology and Laboratory Medicine, Indiana University School of Medicine
| | - Melissa L. Fishel
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
- Department of Pharmacology and Toxicology, Indiana University School of Medicine
| | - Christine A. Pratilas
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine
| | - Steven P. Angus
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
- Department of Pharmacology and Toxicology, Indiana University School of Medicine
- IU Simon Comprehensive Cancer Center, Indiana University School of Medicine
| | - D. Wade Clapp
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine
- Department of Medical and Molecular Genetics, Indiana University School of Medicine
- IU Simon Comprehensive Cancer Center, Indiana University School of Medicine
| | - Steven D. Rhodes
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
- Department of Medical and Molecular Genetics, Indiana University School of Medicine
- Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Indiana University School of Medicine
- IU Simon Comprehensive Cancer Center, Indiana University School of Medicine
| |
Collapse
|
3
|
Abstract
The microtubule minus-end-directed motility of cytoplasmic dynein 1 (dynein), arguably the most complex and versatile cytoskeletal motor, is harnessed for diverse functions, such as long-range organelle transport in neuronal axons and spindle assembly in dividing cells. The versatility of dynein raises a number of intriguing questions, including how is dynein recruited to its diverse cargo, how is recruitment coupled to activation of the motor, how is motility regulated to meet different requirements for force production and how does dynein coordinate its activity with that of other microtubule-associated proteins (MAPs) present on the same cargo. Here, these questions will be discussed in the context of dynein at the kinetochore, the supramolecular protein structure that connects segregating chromosomes to spindle microtubules in dividing cells. As the first kinetochore-localized MAP described, dynein has intrigued cell biologists for more than three decades. The first part of this Review summarizes current knowledge about how kinetochore dynein contributes to efficient and accurate spindle assembly, and the second part describes the underlying molecular mechanisms and highlights emerging commonalities with dynein regulation at other subcellular sites.
Collapse
Affiliation(s)
- Reto Gassmann
- Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, 4200-135 Porto, Portugal.,Instituto de Biologia Molecular e Celular - IBMC, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
4
|
Gao J, Xu W, Zeng T, Tian Y, Wu C, Liu S, Zhao Y, Zhou S, Lin X, Cao H, Lu L. Genome-Wide Association Study of Egg-Laying Traits and Egg Quality in LingKun Chickens. Front Vet Sci 2022; 9:877739. [PMID: 35795788 PMCID: PMC9251537 DOI: 10.3389/fvets.2022.877739] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/23/2022] [Indexed: 11/23/2022] Open
Abstract
Egg production is the most important trait of laying hens. To identify molecular markers and candidate genes associated with egg production and quality, such as body weight at first oviposition (BWF), the number of eggs produced in 500 days (EN500), egg weight (EW), egg shell thickness (EST), egg shell strength (ESS), and Haugh unit (HU), a genome-wide analysis was performed in 266 LingKun Chickens. The results showed that thirty-seven single nucleotide polymorphisms (SNPs) were associated with all traits (p < 9.47 × 10−8, Bonferroni correction). These SNPs were located in close proximity to or within the sequence of the thirteen candidate genes, such as Galanin And GMAP Prepropeptide (GAL), Centromere Protein (CENPF), Glypican 2 (GPC2), Phosphatidylethanolamine N-Methyltransferase (PEMT), Transcription Factor AP-2 Delta (TFAP2D), and Carboxypeptidase Q (CPQ) gene related to egg-laying and Solute Carrier Family 5 Member 7 (SLC5A7), Neurocalcin Delta (NCALD), Proteasome 20S Subunit Beta 2 (PSMB2), Slit Guidance Ligand 3 (SLIT3), and Tubulin Tyrosine Ligase Like 7 (TTLL7) genes related to egg quality. Interestingly, one of the genes involved in bone formation (SLIT3) was identified as a candidate gene for ESS. Our candidate genes and SNPs associated with egg-laying traits were significant for molecular breeding of egg-laying traits and egg quality in LingKun chickens.
Collapse
Affiliation(s)
- Jinfeng Gao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Science and Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Wenwu Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Science and Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Tao Zeng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Science and Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Yong Tian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Science and Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Chunqin Wu
- Wenzhou Vocational College of Science and Technology, Wenzhou, China
| | - Suzhen Liu
- Wenzhou Vocational College of Science and Technology, Wenzhou, China
| | - Yan Zhao
- Wenzhou Vocational College of Science and Technology, Wenzhou, China
| | - Shuhe Zhou
- Wenzhou Golden Land Agricultural Development Co., Ltd., Wenzhou, China
| | - Xinqin Lin
- Wenzhou Golden Land Agricultural Development Co., Ltd., Wenzhou, China
| | - Hongguo Cao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
- Hongguo Cao
| | - Lizhi Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Science and Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
- *Correspondence: Lizhi Lu
| |
Collapse
|
5
|
Li M, Zhao J, Yang R, Cai R, Liu X, Xie J, Shu B, Qi S. CENPF as an independent prognostic and metastasis biomarker corresponding to CD4+ memory T cells in cutaneous melanoma. Cancer Sci 2022; 113:1220-1234. [PMID: 35189004 PMCID: PMC8990861 DOI: 10.1111/cas.15303] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/02/2022] [Accepted: 02/09/2022] [Indexed: 12/01/2022] Open
Abstract
Owing to recent advances in immunotherapies, the overall survival of patients with skin cutaneous melanoma (SKCM) has increased; however, the 5-year survival rate of metastatic patients remains poor. Skin cutaneous melanoma-upregulated genes were screened via analysis of differentially expressed genes (GSE3189 and GSE46517), and metastasis-related oncogenes were identified via weighted gene coexpression network analysis of the GSE46517 dataset. As confirmed by the Tumor Immune Estimation Resource, we found highly expressed centromere protein F (CENPF) in SKCM and its metastases. Immunostaining of human melanoma tissues demonstrated high CENPF expression. According to the Kaplan-Meier survival curve log-rank test, receiver-operating characteristic curve, and univariate and multivariate analyses, the Cancer Genome Atlas (TCGA) database suggested CENPF be a typical independent predictor of SKCM. The CIBERSORT algorithm classified the types of the immune cells from GSE46517 and showed higher proportion of CD4+ memory-activated T cells in metastatic melanoma. Single-sample gene set enrichment analysis of TCGA data confirmed the correlation between CENPF and activated CD4+ T cells. Centromere protein F was positively correlated with tumor mutational burden and CD4+ memory T cell markers (interleukin [IL]-23A, CD28, and CD62L), negatively associated with memory T cell maintenance factors (IL-7 and IL-15) by correlation analysis. Moreover, immunofluorescence showed high coexpression of CENPF and IL23A, CD4 in melanoma. Upregulated CENPF might lead to premature depletion of CD4+ memory T cells and immunosuppression. Nomogram indicated CENPF clinical predictive value for 1-, 3-, 5-, and 7-year melanoma overall survival. Therefore, CENPF plays a vital role in the progression and metastasis of melanoma and can be an effective therapeutic target.
Collapse
Affiliation(s)
- Mengzhi Li
- Department of BurnsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Jingling Zhao
- Department of BurnsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Ronghua Yang
- Department of Burn SurgeryThe First People’s Hospital of FoshanFoshanChina
| | - Ruizhao Cai
- Department of BurnsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Xusheng Liu
- Department of BurnsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Julin Xie
- Department of BurnsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Bin Shu
- Department of BurnsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Shaohai Qi
- Department of BurnsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
6
|
The Mitotic Apparatus and Kinetochores in Microcephaly and Neurodevelopmental Diseases. Cells 2019; 9:cells9010049. [PMID: 31878213 PMCID: PMC7016623 DOI: 10.3390/cells9010049] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/18/2019] [Accepted: 12/21/2019] [Indexed: 12/15/2022] Open
Abstract
Regulators of mitotic division, when dysfunctional or expressed in a deregulated manner (over- or underexpressed) in somatic cells, cause chromosome instability, which is a predisposing condition to cancer that is associated with unrestricted proliferation. Genes encoding mitotic regulators are growingly implicated in neurodevelopmental diseases. Here, we briefly summarize existing knowledge on how microcephaly-related mitotic genes operate in the control of chromosome segregation during mitosis in somatic cells, with a special focus on the role of kinetochore factors. Then, we review evidence implicating mitotic apparatus- and kinetochore-resident factors in the origin of congenital microcephaly. We discuss data emerging from these works, which suggest a critical role of correct mitotic division in controlling neuronal cell proliferation and shaping the architecture of the central nervous system.
Collapse
|
7
|
Smirnova L, Seregin A, Boksha I, Dmitrieva E, Simutkin G, Kornetova E, Savushkina O, Letova A, Bokhan N, Ivanova S, Zgoda V. The difference in serum proteomes in schizophrenia and bipolar disorder. BMC Genomics 2019; 20:535. [PMID: 31291891 PMCID: PMC6620192 DOI: 10.1186/s12864-019-5848-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Purpose of study is revealing significant differences in serum proteomes in schizophrenia and bipolar disorder (BD). RESULTS Quantitative mass-spectrometry based proteomic analysis was used to quantify proteins in the blood serum samples after the depletion of six major blood proteins. Comparison of proteome profiles of different groups revealed 27 proteins being specific for schizophrenia, and 18 - for BD. Protein set in schizophrenia was mostly associated with immune response, cell communication, cell growth and maintenance, protein metabolism and regulation of nucleic acid metabolism. Protein set in BD was mostly associated with immune response, regulating transport processes across cell membrane and cell communication, development of neurons and oligodendrocytes and cell growth. Concentrations of ankyrin repeat domain-containing protein 12 (ANKRD12) and cadherin 5 in serum samples were determined by ELISA. Significant difference between three groups was revealed in ANKRD12 concentration (p = 0.02), with maximum elevation of ANKRD12 concentration (median level) in schizophrenia followed by BD. Cadherin 5 concentration differed significantly (p = 0.035) between schizophrenic patients with prevailing positive symptoms (4.78 [2.71, 7.12] ng/ml) and those with prevailing negative symptoms (1.86 [0.001, 4.11] ng/ml). CONCLUSIONS Our results are presumably useful for discovering the new pathways involved in endogenous psychotic disorders.
Collapse
Affiliation(s)
- Liudmila Smirnova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Alexander Seregin
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | | | - Elena Dmitrieva
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
- Siberian State Medical University, Tomsk, Russia
| | - German Simutkin
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Elena Kornetova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
- Siberian State Medical University, Tomsk, Russia
| | | | | | - Nikolay Bokhan
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Svetlana Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
- Siberian State Medical University, Tomsk, Russia
| | - Victor Zgoda
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
8
|
Monda JK, Cheeseman IM. Dynamic regulation of dynein localization revealed by small molecule inhibitors of ubiquitination enzymes. Open Biol 2018; 8:rsob.180095. [PMID: 30257893 PMCID: PMC6170511 DOI: 10.1098/rsob.180095] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/29/2018] [Indexed: 11/27/2022] Open
Abstract
Cytoplasmic dynein is a minus-end-directed microtubule-based motor that acts at diverse subcellular sites. During mitosis, dynein localizes simultaneously to the mitotic spindle, spindle poles, kinetochores and the cell cortex. However, it is unclear what controls the relative targeting of dynein to these locations. As dynein is heavily post-translationally modified, we sought to test a role for these modifications in regulating dynein localization. We find that dynein rapidly and strongly accumulates at mitotic spindle poles following treatment with NSC697923, a small molecule that inhibits the ubiquitin E2 enzyme, Ubc13, or treatment with PYR-41, a ubiquitin E1 inhibitor. Subsets of dynein regulators such as Lis1, ZW10 and Spindly accumulate at the spindle poles, whereas others do not, suggesting that NSC697923 differentially affects specific dynein populations. We additionally find that dynein relocalization induced by NSC697923 or PYR-41 can be suppressed by simultaneous treatment with the non-selective deubiquitinase inhibitor, PR-619. However, we did not observe altered dynein localization following treatment with the selective E1 inhibitor, TAK-243. Although it is possible that off-target effects of NSC697923 and PYR-41 are responsible for the observed changes in dynein localization, the rapid relocalization upon drug treatment highlights the highly dynamic nature of dynein regulation during mitosis.
Collapse
Affiliation(s)
- Julie K Monda
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142, USA .,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| |
Collapse
|
9
|
Loss of CENP-F Results in Dilated Cardiomyopathy with Severe Disruption of Cardiac Myocyte Architecture. Sci Rep 2018; 8:7546. [PMID: 29765066 PMCID: PMC5953941 DOI: 10.1038/s41598-018-25774-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 04/27/2018] [Indexed: 12/14/2022] Open
Abstract
Centromere-binding protein F (CENP-F) is a very large and complex protein with many and varied binding partners including components of the microtubule network. Numerous CENP-F functions impacting diverse cellular behaviors have been identified. Importantly, emerging data have shown that CENP-F loss- or gain-of-function has critical effects on human development and disease. Still, it must be noted that data at the single cardiac myocyte level examining the impact of CENP-F loss-of-function on fundamental cellular behavior is missing. To address this gap in our knowledge, we analyzed basic cell structure and function in cardiac myocytes devoid of CENP-F. We found many diverse structural abnormalities including disruption of the microtubule network impacting critical characteristics of the cardiac myocyte. This is the first report linking microtubule network malfunction to cardiomyopathy. Importantly, we also present data demonstrating a direct link between a CENP-F single nucleotide polymorphism (snp) and human cardiac disease. In a proximate sense, these data examining CENP-F function explain the cellular basis underlying heart disease in this genetic model and, in a larger sense, they will hopefully provide a platform upon which the field can explore diverse cellular outcomes in wide-ranging areas of research on this critical protein.
Collapse
|
10
|
Cui H, Loftus KM, Noell CR, Solmaz SR. Identification of Cyclin-dependent Kinase 1 Specific Phosphorylation Sites by an In Vitro Kinase Assay. J Vis Exp 2018. [PMID: 29782014 DOI: 10.3791/57674] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Cyclin-dependent kinase 1 (Cdk1) is a master controller for the cell cycle in all eukaryotes and phosphorylates an estimated 8 - 13% of the proteome; however, the number of identified targets for Cdk1, particularly in human cells is still low. The identification of Cdk1-specific phosphorylation sites is important, as they provide mechanistic insights into how Cdk1 controls the cell cycle. Cell cycle regulation is critical for faithful chromosome segregation, and defects in this complicated process lead to chromosomal aberrations and cancer. Here, we describe an in vitro kinase assay that is used to identify Cdk1-specific phosphorylation sites. In this assay, a purified protein is phosphorylated in vitro by commercially available human Cdk1/cyclin B. Successful phosphorylation is confirmed by SDS-PAGE, and phosphorylation sites are subsequently identified by mass spectrometry. We also describe purification protocols that yield highly pure and homogeneous protein preparations suitable for the kinase assay, and a binding assay for the functional verification of the identified phosphorylation sites, which probes the interaction between a classical nuclear localization signal (cNLS) and its nuclear transport receptor karyopherin α. To aid with experimental design, we review approaches for the prediction of Cdk1-specific phosphorylation sites from protein sequences. Together these protocols present a very powerful approach that yields Cdk1-specific phosphorylation sites and enables mechanistic studies into how Cdk1 controls the cell cycle. Since this method relies on purified proteins, it can be applied to any model organism and yields reliable results, especially when combined with cell functional studies.
Collapse
Affiliation(s)
- Heying Cui
- Department of Chemistry, State University of New York at Binghamton
| | - Kyle M Loftus
- Department of Chemistry, State University of New York at Binghamton
| | - Crystal R Noell
- Department of Chemistry, State University of New York at Binghamton
| | - Sozanne R Solmaz
- Department of Chemistry, State University of New York at Binghamton;
| |
Collapse
|
11
|
A transcriptomic study of myogenic differentiation under the overexpression of PPARγ by RNA-Seq. Sci Rep 2017; 7:15308. [PMID: 29127356 PMCID: PMC5681552 DOI: 10.1038/s41598-017-14275-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/06/2017] [Indexed: 12/18/2022] Open
Abstract
To study the cellular and molecular function of peroxisome proliferator-activated receptor γ (PPARγ) in skeletal muscle differentiation, we have generated inducible gain-of-function to overexpress PPARγ in C2C12 myoblasts. In order to identify PPARγ targets, RNA sequencing (RNA-seq) was used to evaluate and quantify the transcriptomes and expression patterns during myogenic differentiation under the overexpression of PPARγ. The formation of myotubes and the expression of muscle-specific myogenic genes such as MyoD and MyoG may be inhibited by PPARγ overexpression. Multiple genes and pathways were significantly involved in this process, including 11 genes such as Fndc9 and Slc14a1 with fundamental change of regulation modes, 9 genes of which were validated by the data of qRT-PCR. Our studies demonstrate that PPARγ would play critical roles on myoblasts differentiation, mediating crosstalk among several pathways and transcription factors. Our data is available in the Gene Expression Omnibus (GEO) database with the accession number as GSE99399.
Collapse
|
12
|
Loftus KM, Cui H, Coutavas E, King DS, Ceravolo A, Pereiras D, Solmaz SR. Mechanism for G2 phase-specific nuclear export of the kinetochore protein CENP-F. Cell Cycle 2017; 16:1414-1429. [PMID: 28723232 DOI: 10.1080/15384101.2017.1338218] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Centromere protein F (CENP-F) is a component of the kinetochore and a regulator of cell cycle progression. CENP-F recruits the dynein transport machinery and orchestrates several cell cycle-specific transport events, including transport of the nucleus, mitochondria and chromosomes. A key regulatory step for several of these functions is likely the G2 phase-specific export of CENP-F from the nucleus to the cytosol, where the cytoplasmic dynein transport machinery resides; however, the molecular mechanism of this process is elusive. Here, we have identified 3 phosphorylation sites within the bipartite classical nuclear localization signal (cNLS) of CENP-F. These sites are specific for cyclin-dependent kinase 1 (Cdk1), which is active in G2 phase. Phosphomimetic mutations of these residues strongly diminish the interaction of the CENP-F cNLS with its nuclear transport receptor karyopherin α. These mutations also diminish nuclear localization of the CENP-F cNLS in cells. Notably, the cNLS is phosphorylated in the -1 position, which is important to orient the adjacent major motif for binding into its pocket on karyopherin α. We propose that localization of CENP-F is regulated by a cNLS, and a nuclear export pathway, resulting in nuclear localization during most of interphase. In G2 phase, the cNLS is weakened by phosphorylation through Cdk1, likely resulting in nuclear export of CENP-F via the still active nuclear export pathway. Once CENP-F resides in the cytosol, it can engage in pathways that are important for cell cycle progression, kinetochore assembly and the faithful segregation of chromosomes into daughter cells.
Collapse
Affiliation(s)
- Kyle M Loftus
- a Department of Chemistry , State University of New York at Binghamton , New York , NY , USA
| | - Heying Cui
- a Department of Chemistry , State University of New York at Binghamton , New York , NY , USA
| | - Elias Coutavas
- b Laboratory of Cell Biology , The Rockefeller University , New York , NY , USA
| | - David S King
- c Howard Hughes Medical Institute, Mass Spectrometry Laboratory , University of California at Berkeley , Berkeley , CA , USA
| | - Amanda Ceravolo
- a Department of Chemistry , State University of New York at Binghamton , New York , NY , USA
| | - Dylan Pereiras
- a Department of Chemistry , State University of New York at Binghamton , New York , NY , USA
| | - Sozanne R Solmaz
- a Department of Chemistry , State University of New York at Binghamton , New York , NY , USA
| |
Collapse
|
13
|
Ozkinay F, Atik T, Isik E, Gormez Z, Sagiroglu M, Sahin OA, Corduk N, Onay H. A further family of Stromme syndrome carrying CENPF
mutation. Am J Med Genet A 2017; 173:1668-1672. [DOI: 10.1002/ajmg.a.38173] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 01/16/2017] [Accepted: 01/18/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Ferda Ozkinay
- Division of Pediatric Genetics, Department of Pediatrics, Faculty of Medicine; Ege University; Izmir Turkey
| | - Tahir Atik
- Division of Pediatric Genetics, Department of Pediatrics, Faculty of Medicine; Ege University; Izmir Turkey
| | - Esra Isik
- Division of Pediatric Genetics, Department of Pediatrics, Faculty of Medicine; Ege University; Izmir Turkey
| | - Zeliha Gormez
- Advanced Genomics and Bioinformatics Research Center; TUBITAK-BILGEM; Kocaeli Turkey
| | - Mahmut Sagiroglu
- Advanced Genomics and Bioinformatics Research Center; TUBITAK-BILGEM; Kocaeli Turkey
| | - Ozlem Atan Sahin
- Biochemistry and Molecular Biology, Institude of Health Sciences; Acibadem University; Istanbul Turkey
| | - Nergul Corduk
- Department of Pediatric Surgery, Faculty of Medicine; Pamukkale University; Denizli Turkey
| | - Huseyin Onay
- Department of Medical Genetics, Faculty of Medicine; Ege University; Izmir Turkey
| |
Collapse
|
14
|
Qu YH, Fu JC, Liu K, Zuo ZY, Jia HN, Ma Y, Luo HL. Screening of α-Tocopherol Transfer Protein Sensitive Genes in Human Hepatoma Cells (HepG2). Int J Mol Sci 2016; 17:ijms17071016. [PMID: 27355945 PMCID: PMC4964392 DOI: 10.3390/ijms17071016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 06/19/2016] [Accepted: 06/21/2016] [Indexed: 01/18/2023] Open
Abstract
α-Tocopherol transfer protein (α-TTP) is a ~32 kDa protein expressed mainly in hepatocytes. The major function of the protein is to bind specifically to α-tocopherol and, together, the complex transfers from late lysosomes to the cell membrane. A previous study indicated that some factors might be required in the transferring process. However, there is little information available about the potential transferring factors. In addition, there remains much to learn about other physiological processes which α-TTP might participate in. Thus, in this study a human α-TTP eukaryotic expression vector was successfully constructed and expressed in human hepatoma cells (HepG2). The sensitive genes related to α-TTP were then screened by microarray technology. Results showed that expression of the vector in HepG2 cells led to the identification of 323 genes showing differential expression. The differentially expressed transcripts were divided into four main categories, including (1) cell inflammation; (2) cell cycle and cell apoptosis; (3) cell signaling and gene regulation; and (4) cellular movement. A few cellular movement related transcripts were selected and verified by quantitative real-time PCR. Expressions of some were significantly increased in α-TTP-expressed group, which indicated that these factors were likely to play a role in the transferring process.
Collapse
Affiliation(s)
- Yang-Hua Qu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Jun-Cai Fu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Kun Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Zhao-Yun Zuo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Hui-Na Jia
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yong Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Hai-Ling Luo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
15
|
Abstract
During mitosis, cells undergo massive deformation and reorganization, impacting on all cellular structures. Mitochondria, in particular, are highly dynamic organelles, which constantly undergo events of fission, fusion and cytoskeleton-based transport. This plasticity ensures the proper distribution of the metabolism, and the proper inheritance of functional organelles. During cell cycle, mitochondria undergo dramatic changes in distribution. In this review, we focus on the dynamic events that target mitochondria during mitosis. We describe how the cell-cycle-dependent microtubule-associated protein centromeric protein F (Cenp-F) is recruited to mitochondria by the mitochondrial Rho GTPase (Miro) to promote mitochondrial transport and re-distribution following cell division.
Collapse
|
16
|
Morris-Rosendahl DJ, Kaindl AM. What next-generation sequencing (NGS) technology has enabled us to learn about primary autosomal recessive microcephaly (MCPH). Mol Cell Probes 2015; 29:271-81. [PMID: 26050940 DOI: 10.1016/j.mcp.2015.05.015] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 11/27/2022]
Abstract
The impact that next-generation sequencing technology (NGS) is having on many aspects of molecular and cell biology, is becoming increasingly apparent. One of the most noticeable outcomes of the new technology in human genetics, has been the accelerated rate of identification of disease-causing genes. Especially for rare, heterogeneous disorders, such as autosomal recessive primary microcephaly (MCPH), the handful of genes previously known to harbour disease-causing mutations, has grown at an unprecedented rate within a few years. Knowledge of new genes mutated in MCPH over the last four years has contributed to our understanding of the disorder at both the clinical and cellular levels. The functions of proteins such as WDR62, CASC5, PHC1, CDK6, CENP-E, CENP-F, CEP63, ZNF335, PLK4 and TUBGPC, have been added to the complex network of critical cellular processes known to be involved in brain growth and size. In addition to the importance of mitotic spindle assembly and structure, centrosome and centriole function and DNA repair and damage response, new mechanisms involving kinetochore-associated proteins and chromatin remodelling complexes have been elucidated. Two of the major contributions to our clinical knowledge are the realisation that primary microcephaly caused by mutations in genes at the MCPH loci is seldom an isolated clinical feature and is often accompanied either by additional cortical malformations or primordial dwarfism. Gene-phenotype correlations are being revisited, with a new dimension of locus heterogeneity and phenotypic variability being revealed.
Collapse
Affiliation(s)
- Deborah J Morris-Rosendahl
- Clinical Genetics and Genomics, Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| | - Angela M Kaindl
- Department of Pediatric Neurology, Charité University Medicine, Berlin, Germany; Institute of Cell Biology and Neurobiology, Charité University Medicine, Berlin, Germany
| |
Collapse
|
17
|
Waters AM, Asfahani R, Carroll P, Bicknell L, Lescai F, Bright A, Chanudet E, Brooks A, Christou-Savina S, Osman G, Walsh P, Bacchelli C, Chapgier A, Vernay B, Bader DM, Deshpande C, O' Sullivan M, Ocaka L, Stanescu H, Stewart HS, Hildebrandt F, Otto E, Johnson CA, Szymanska K, Katsanis N, Davis E, Kleta R, Hubank M, Doxsey S, Jackson A, Stupka E, Winey M, Beales PL. The kinetochore protein, CENPF, is mutated in human ciliopathy and microcephaly phenotypes. J Med Genet 2015; 52:147-56. [PMID: 25564561 PMCID: PMC4345935 DOI: 10.1136/jmedgenet-2014-102691] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 11/26/2014] [Accepted: 11/27/2014] [Indexed: 12/02/2022]
Abstract
BACKGROUND Mutations in microtubule-regulating genes are associated with disorders of neuronal migration and microcephaly. Regulation of centriole length has been shown to underlie the pathogenesis of certain ciliopathy phenotypes. Using a next-generation sequencing approach, we identified mutations in a novel centriolar disease gene in a kindred with an embryonic lethal ciliopathy phenotype and in a patient with primary microcephaly. METHODS AND RESULTS Whole exome sequencing data from a non-consanguineous Caucasian kindred exhibiting mid-gestation lethality and ciliopathic malformations revealed two novel non-synonymous variants in CENPF, a microtubule-regulating gene. All four affected fetuses showed segregation for two mutated alleles [IVS5-2A>C, predicted to abolish the consensus splice-acceptor site from exon 6; c.1744G>T, p.E582X]. In a second unrelated patient exhibiting microcephaly, we identified two CENPF mutations [c.1744G>T, p.E582X; c.8692 C>T, p.R2898X] by whole exome sequencing. We found that CENP-F colocalised with Ninein at the subdistal appendages of the mother centriole in mouse inner medullary collecting duct cells. Intraflagellar transport protein-88 (IFT-88) colocalised with CENP-F along the ciliary axonemes of renal epithelial cells in age-matched control human fetuses but did not in truncated cilia of mutant CENPF kidneys. Pairwise co-immunoprecipitation assays of mitotic and serum-starved HEKT293 cells confirmed that IFT88 precipitates with endogenous CENP-F. CONCLUSIONS Our data identify CENPF as a new centriolar disease gene implicated in severe human ciliopathy and microcephaly related phenotypes. CENP-F has a novel putative function in ciliogenesis and cortical neurogenesis.
Collapse
Affiliation(s)
- Aoife M Waters
- Institute of Child Health, University College London, London, UK Department of Nephrology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Rowan Asfahani
- Institute of Child Health, University College London, London, UK
| | - Paula Carroll
- Institute of Genetics & Molecular Medicine, Edinburgh, UK
| | | | - Francesco Lescai
- Institute of Child Health, University College London, London, UK
| | | | - Estelle Chanudet
- Institute of Child Health, University College London, London, UK
| | - Anthony Brooks
- Institute of Child Health, University College London, London, UK
| | | | - Guled Osman
- Institute of Child Health, University College London, London, UK
| | - Patrick Walsh
- Institute of Child Health, University College London, London, UK
| | - Chiara Bacchelli
- Institute of Child Health, University College London, London, UK
| | - Ariane Chapgier
- Institute of Child Health, University College London, London, UK
| | - Bertrand Vernay
- Institute of Child Health, University College London, London, UK
| | - David M Bader
- Department of Cell and Developmental Biology, Vanderbilt University, USA
| | - Charu Deshpande
- Department of Clinical Genetics, Evelina Children's Hospital, London, UK
| | - Mary O' Sullivan
- Institute of Child Health, University College London, London, UK
| | - Louise Ocaka
- Institute of Child Health, University College London, London, UK
| | - Horia Stanescu
- Centre for Nephrology, Royal Free Hospital, University College London, London, UK
| | - Helen S Stewart
- Department of Clinical Genetics, Oxford Radcliffe Hospitals NHS Trust, Churchill Hospital, Oxford, UK
| | - Friedhelm Hildebrandt
- Department of Medicine, Boston Children's Hospital and Harvard Medical School, Boston, USA
| | - Edgar Otto
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Colin A Johnson
- Department of Pediatrics, Leeds Institute of Biomedical and Clinical Sciences, Leeds, UK
| | - Katarzyna Szymanska
- Department of Pediatrics, Leeds Institute of Biomedical and Clinical Sciences, Leeds, UK
| | - Nicholas Katsanis
- Center for Human Disease Modeling, Department of Cell Biology, Duke University Medical Center
| | - Erica Davis
- Center for Human Disease Modeling, Department of Cell Biology, Duke University Medical Center
| | - Robert Kleta
- Centre for Nephrology, Royal Free Hospital, University College London, London, UK
| | - Mike Hubank
- Institute of Child Health, University College London, London, UK
| | | | - Andrew Jackson
- Institute of Genetics & Molecular Medicine, Edinburgh, UK MRC Human Genetics, University of Edinburgh, Edinburgh, UK
| | - Elia Stupka
- Institute of Child Health, University College London, London, UK
| | - Mark Winey
- Molecular, Ceullular, and Developmental Biology, University of Colorado at Boulder, Boulder, CO 80309, USA
| | - Philip L Beales
- Institute of Child Health, University College London, London, UK
| |
Collapse
|
18
|
Tan T, Chen Z, Lei Y, Zhu Y, Liang Q. A regulatory effect of INMAP on centromere proteins: antisense INMAP induces CENP-B variation and centromeric halo. PLoS One 2014; 9:e91937. [PMID: 24633075 PMCID: PMC3954832 DOI: 10.1371/journal.pone.0091937] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 02/18/2014] [Indexed: 11/19/2022] Open
Abstract
CENP-B is a highly conserved protein that facilitates the assembly of specific centromere structures both in interphase nuclei and on mitotic chromosomes. INMAP is a conserved protein that localizes at nucleus in interphase cells and at mitotic apparatus in mitotic cells. Our previous results showed that INMAP over-expression leads to spindle defects, mitotic arrest and formation of polycentrosomal and multinuclear cells, indicating that INMAP may modulate the function of (a) key protein(s) in mitotic apparatus. In this study, we demonstrate that INMAP interacts with CENP-B and promotes cleavage of the N-terminal DNA binding domain from CENP-B. The cleaved CENP-B cannot associate with centromeres and thus lose its centromere-related functions. Consistent with these results, CENP-B in INMAP knockdown cells becomes more diffused around kinetochores. Although INMAP knockdown cells do not exhibit gross defects in mitotic spindle formation, these cells go through mitosis, especially prophase and metaphase, with different relative timing, indicating subtle abnormality. These results identify INMAP as a model regulator of CENP-B and support the notion that INMAP regulates mitosis through modulating CENP-B-mediated centromere organization.
Collapse
Affiliation(s)
- Tan Tan
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Zhe Chen
- Beijing Key Laboratory of Gene Resource and Molecular Development / Beijing Key Laboratory of Gene Engineering Drugs & Biological Technology, Beijing Normal University, Beijing, China
| | - Yan Lei
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yan Zhu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Qianjin Liang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Gene Resource and Molecular Development / Beijing Key Laboratory of Gene Engineering Drugs & Biological Technology, Beijing Normal University, Beijing, China
- * E-mail:
| |
Collapse
|
19
|
Welner S, Trier NH, Frisch M, Locht H, Hansen PR, Houen G. Correlation between centromere protein-F autoantibodies and cancer analyzed by enzyme-linked immunosorbent assay. Mol Cancer 2013; 12:95. [PMID: 23978088 PMCID: PMC3844405 DOI: 10.1186/1476-4598-12-95] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 08/22/2013] [Indexed: 11/10/2022] Open
Abstract
Background Centromere protein-F (CENP-F) is a large nuclear protein of 367 kDa, which is involved in multiple mitosis-related events such as proper assembly of the kinetochores, stabilization of heterochromatin, chromosome alignment and mitotic checkpoint signaling. Several studies have shown a correlation between CENP-F and cancer, e.g. the expression of CENP-F has been described to be upregulated in cancer cells. Furthermore, several studies have described a significant correlation between the expression of autoantibodies to CENP-F and cancer. Methods Autoantibodies to CENP-F were detected in a small number of samples during routine indirect immunofluorescence (IIF) analysis for anti-nuclear antibodies (ANA) using HEp-2 cells as substrate. Using overlapping synthetic peptides covering a predicted structural maintenance of chromosomes (SMC) domain, we developed an enzyme-linked immunosorbent assay (ELISA) for detection of CENP-F antibodies. Results Analyzing the reactivity of the sera positive in IIF for CENP-F antibodies to overlapping CENP-F peptides, we showed that autoantibodies to several peptides correlate with the presence of antibodies to CENP-F and a diagnosis of cancer, as increased CENP-F antibody expression specific for malignant cancer patients to five peptides was found (A9, A12, A14, A16, A27). These antibodies to CENP-F in clinical samples submitted for ANA analysis were found to have a positive predictive value for cancer of 50%. Furthermore, the expression of cancer-correlated CENP-F antibodies seemed to increase as a function of time from diagnosis. Conclusion These results conform to previous findings that approximately 50% of those patients clinically tested for ANA analyses who express CENP-F antibodies are diagnosed with cancer, confirming that these antibodies may function as circulating tumor markers. Thus, a peptide-based CENP-F ELISA focused on the SMC domain may aid in identifying individuals with a potential cancer.
Collapse
Affiliation(s)
- Simon Welner
- Department of Clinical Biochemistry, Immunology and Genetics, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen S, Denmark.
| | | | | | | | | | | |
Collapse
|
20
|
Richmond D, Rizkallah R, Liang F, Hurt MM, Wang Y. Slk19 clusters kinetochores and facilitates chromosome bipolar attachment. Mol Biol Cell 2013; 24:566-77. [PMID: 23283988 PMCID: PMC3583661 DOI: 10.1091/mbc.e12-07-0552] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Yeast kinetochore protein Slk19 is required for kinetochore clustering, and nocodazole exposure to slk19 mutant cells causes impaired kinetochore capture and delayed chromosome bipolar attachment after nocodazole washout. In all eukaryotic cells, DNA is packaged into multiple chromosomes that are linked to microtubules through a large protein complex called a kinetochore. Previous data show that the kinetochores are clustered together during most of the cell cycle, but the mechanism and the biological significance of kinetochore clustering are unknown. As a kinetochore protein in budding yeast, the role of Slk19 in the stability of the anaphase spindle has been well studied, but its function in chromosome segregation has remained elusive. Here we show that Slk19 is required for kinetochore clustering when yeast cells are treated with the microtubule-depolymerizing agent nocodazole. We further find that slk19Δ mutant cells exhibit delayed kinetochore capture and chromosome bipolar attachment after the disruption of the kinetochore–microtubule interaction by nocodazole, which is likely attributed to defective kinetochore clustering. In addition, we show that Slk19 interacts with itself, suggesting that the dimerization of Slk19 may mediate the interaction between kinetochores for clustering. Therefore Slk19 likely acts as kinetochore glue that clusters kinetochores to facilitate efficient and faithful chromosome segregation.
Collapse
Affiliation(s)
- Daniel Richmond
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | | | | | | | | |
Collapse
|
21
|
Dees E, Miller PM, Moynihan KL, Pooley RD, Hunt RP, Galindo CL, Rottman JN, Bader DM. Cardiac-specific deletion of the microtubule-binding protein CENP-F causes dilated cardiomyopathy. Dis Model Mech 2012; 5:468-80. [PMID: 22563055 PMCID: PMC3380710 DOI: 10.1242/dmm.008680] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
CENP-F is a large multifunctional protein with demonstrated regulatory roles in cell proliferation, vesicular transport and cell shape through its association with the microtubule (MT) network. Until now, analysis of CENP-F has been limited to in vitro analysis. Here, using a Cre-loxP system, we report the in vivo disruption of CENP-F gene function in murine cardiomyocytes, a cell type displaying high levels of CENP-F expression. Loss of CENP-F function in developing myocytes leads to decreased cell division, blunting of trabeculation and an initially smaller, thin-walled heart. Still, embryos are born at predicted mendelian ratios on an outbred background. After birth, hearts lacking CENP-F display disruption of their intercalated discs and loss of MT integrity particularly at the costamere; these two structures are essential for cell coupling/electrical conduction and force transduction in the heart. Inhibition of myocyte proliferation and cell coupling as well as loss of MT maintenance is consistent with previous reports of generalized CENP-F function in isolated cells. One hundred percent of these animals develop progressive dilated cardiomyopathy with heart block and scarring, and there is a 20% mortality rate. Importantly, although it has long been postulated that the MT cytoskeleton plays a role in the development of heart disease, this study is the first to reveal a direct genetic link between disruption of this network and cardiomyopathy. Finally, this study has broad implications for development and disease because CENP-F loss of function affects a diverse array of cell-type-specific activities in other organs.
Collapse
Affiliation(s)
- Ellen Dees
- Department of Pediatrics, Vanderbilt University, Nashville, TN 37232-6300, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Misfolded Gβ is recruited to cytoplasmic dynein by Nudel for efficient clearance. Cell Res 2012; 22:1140-54. [PMID: 22430153 DOI: 10.1038/cr.2012.41] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The Gβγ heterodimer is an important signal transducer. Gβ, however, is prone to misfolding due to its requirement for Gγ and chaperones for proper folding. How cells dispose of misfolded Gβ (mfGβ) is not clear. Here, we showed that mfGβ was able to be polyubiquitinated and subsequently degraded by the proteasome. It was sequestered in aggresomes after the inhibition of the proteasome activity with MG132. Sustained activation of Gβγ signaling further elevated cellular levels of the ubiquitinated Gβ. Moreover, Nudel, a regulator of cytoplasmic dynein, the microtubule minus end-directed motor, directly interacted with both the unubiquitinated and ubiquitinated mfGβ. Increasing the levels of both mfGβ and Nudel promoted the association of Gβ with both Nudel and dynein, resulting in robust aggresome formation in a dynein-dependent manner. Depletion of Nudel by RNAi reduced the dynein-associated mfGβ, impaired the MG132-induced aggresome formation, and markedly prolonged the half-life of nascent Gβ. Therefore, cytosolic mfGβ is recruited to dynein by Nudel and transported to the centrosome for rapid sequestration and degradation. Such a process not only eliminates mfGβ efficiently for the control of protein quality, but may also help to terminate the Gβγ signaling.
Collapse
|
23
|
Famulski JK, Vos LJ, Rattner JB, Chan GK. Dynein/Dynactin-mediated transport of kinetochore components off kinetochores and onto spindle poles induced by nordihydroguaiaretic acid. PLoS One 2011; 6:e16494. [PMID: 21305043 PMCID: PMC3030593 DOI: 10.1371/journal.pone.0016494] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 12/20/2010] [Indexed: 01/10/2023] Open
Abstract
The mitotic checkpoint functions to ensure accurate chromosome segregation by regulating the progression from metaphase to anaphase. Once the checkpoint has been satisfied, it is inactivated in order to allow the cell to proceed into anaphase and complete the cell cycle. The minus end-directed microtubule motor dynein/dynactin has been implicated in the silencing of the mitotic checkpoint by "stripping" checkpoint proteins off kinetochores. A recent study suggested that Nordihydroguaiaretic acid (NDGA) stimulates dynein/dynactin-mediated transport of its cargo including ZW10 (Zeste White 10). We analyzed the effects of NDGA on dynein/dynactin dependent transport of the RZZ (Zeste White 10, Roughdeal, Zwilch) complex as well as other kinetochore components from kinetochores to spindle poles. Through this approach we have catalogued several kinetochore and centromere components as dynein/dynactin cargo. These include hZW10, hZwilch, hROD, hSpindly, hMad1, hMad2, hCENP-E, hCdc27, cyclin-B and hMps1. Furthermore, we found that treatment with NDGA induced a robust accumulation and complete stabilization of hZW10 at spindle poles. This finding suggests that NDGA may not induce dynein/dynactin transport but rather interfere with cargo release. Lastly, we determined that NDGA induced accumulation of checkpoint proteins at the poles requires dynein/dynactin-mediated transport, hZW10 kinetochore localization and kinetochore-microtubule attachments but not tension or Aurora B kinase activity.
Collapse
Affiliation(s)
- Jakub K. Famulski
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Larissa J. Vos
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Jerome B. Rattner
- Department of Anatomy and Cell Biology, University of Calgary, Calgary, Alberta, Canada
| | - Gordon K. Chan
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
- Experimental Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada
- School of Cancer, Engineering and Imaging Sciences, University of Alberta, Edmonton, Alberta, Canada
- * E-mail:
| |
Collapse
|
24
|
Du J, Zhang Y, Liu Y, Li Y, Zhu X. Involvement of Cenp-F in interphase chromatin organization possibly through association with DNA-dependent protein kinase. Acta Biochim Biophys Sin (Shanghai) 2010; 42:839-46. [PMID: 20978035 DOI: 10.1093/abbs/gmq095] [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] [Indexed: 11/15/2022] Open
Abstract
Cenp-F (also named mitosin) is a 350-kDa human kinetochore protein important for the mitotic progression. It is also a nuclear matrix protein in interphase cells. Here, we showed that overexpression of N-terminal deletion mutants of Cenp-F containing the C-terminal 112 residues induced chromatin condensation into numerous aggregates of varying sizes in interphase nucleus, colocalizing with the exogenous proteins. In situ hybridization using whole chromosome painting probes indicated that the chromatin aggregates were not prematurely condensed individual chromosomes. Neither were they due to apoptosis. We provided evidence showing association of Cenp-F with certain regions of interphase chromatin fibers. Cenp-F associated with the DNA-dependent protein kinase (DNA-PK), a trimeric protein complex critical for genome homeostasis. Moreover, the DNA-PK association activity of Cenp-F mutants correlated with their ability to induce chromatin aggregation. These results imply a role of Cenp-F in organization of interphase chromatin through association and possibly regulation of DNA-PK.
Collapse
Affiliation(s)
- Juan Du
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China
| | | | | | | | | |
Collapse
|
25
|
Du J, Li Y, Zhu X. Involvement of CENP-F in histone methylation. Acta Biochim Biophys Sin (Shanghai) 2010; 42:173-6. [PMID: 20213041 DOI: 10.1093/abbs/gmq001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
CENP-F (also named mitosin) is a multifunctional protein of 350 kDa. In interphase, it is a nuclear protein, whereas in M phase it localizes to the kinetochore, the major microtubule-binding structure on chromosomes essential for chromosome segregation. CENP-F is also critical for myocyte differentiation through the interaction with Rb. It binds to ATF4 and negatively regulates the transcriptional activity of ATF4. It is also important for mitotic progression. Here we show that depletion of CENP-F by RNAi markedly downregulated the methylation of histone H3 at K4 and K9. Consistently, association of HP1a with mitotic chromosomes was largely decreased. These results uncover a novel role of CENP-F in regulation of epigenetic modification on histone H3.
Collapse
Affiliation(s)
- Juan Du
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | |
Collapse
|
26
|
|
27
|
Li Y, Yu W, Liang Y, Zhu X. Kinetochore dynein generates a poleward pulling force to facilitate congression and full chromosome alignment. Cell Res 2007; 17:701-12. [PMID: 17680027 DOI: 10.1038/cr.2007.65] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
For proper chromosome segregation, all kinetochores must achieve bipolar microtubule (MT) attachment and subsequently align at the spindle equator before anaphase onset. The MT minus end-directed motor dynein/dynactin binds kinetochores in prometaphase and has long been implicated in chromosome congression. Unfortunately, inactivation of dynein usually disturbs spindle organization, thus hampering evaluation of its kinetochore roles. Here we specifically eliminated kinetochore dynein/dynactin by RNAi-mediated depletion of ZW10, a protein essential for kinetochore localization of the motor. Time-lapse microscopy indicated markedly-reduced congression efficiency, though congressing chromosomes displayed similar velocities as in control cells. Moreover, cells frequently failed to achieve full chromosome alignment, despite their normal spindles. Confocal microcopy revealed that the misaligned kinetochores were monooriented or unattached and mostly lying outside the spindle, suggesting a difficulty to capture MTs from the opposite pole. Kinetochores on monoastral spindles were dispersed farther away from the pole and exhibited only mild oscillation. Furthermore, inactivating dynein by other means generated similar phenotypes. Therefore, kinetochore dynein produces on monooriented kinetochores a poleward pulling force, which may contribute to efficient bipolar attachment by facilitating their proper microtubule captures to promote congression as well as full chromosome alignment.
Collapse
Affiliation(s)
- Yan Li
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | | | | |
Collapse
|
28
|
Taxanes, microtubules and chemoresistant breast cancer. Biochim Biophys Acta Rev Cancer 2007; 1785:96-132. [PMID: 18068131 DOI: 10.1016/j.bbcan.2007.10.004] [Citation(s) in RCA: 226] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2007] [Revised: 10/23/2007] [Accepted: 10/28/2007] [Indexed: 01/09/2023]
Abstract
The taxanes, paclitaxel and docetaxel are microtubule-stabilizing agents that function primarily by interfering with spindle microtubule dynamics causing cell cycle arrest and apoptosis. However, the mechanisms underlying their action have yet to be fully elucidated. These agents have become widely recognized as active chemotherapeutic agents in the treatment of metastatic breast cancer and early-stage breast cancer with benefits gained in terms of overall survival (OS) and disease-free survival (DFS). However, even with response to taxane treatment the time to progression (TTP) is relatively short, prolonging life for a matter of months, with studies showing that patients treated with taxanes eventually relapse. This review focuses on chemoresistance to taxane treatment particularly in relation to the spindle assembly checkpoint (SAC) and dysfunctional regulation of apoptotic signaling. Since spindle microtubules are the primary drug targets for taxanes, important SAC proteins such as MAD2, BUBR1, Synuclein-gamma and Aurora A have emerged as potentially important predictive markers of taxane resistance, as have specific checkpoint proteins such as BRCA1. Moreover, overexpression of the drug efflux pump MDR-1/P-gp, altered expression of microtubule-associated proteins (MAPs) including tau, stathmin and MAP4 may help to identify those patients who are most at risk of recurrence and those patients most likely to benefit from taxane treatment.
Collapse
|
29
|
Liang Y, Yu W, Li Y, Yu L, Zhang Q, Wang F, Yang Z, Du J, Huang Q, Yao X, Zhu X. Nudel modulates kinetochore association and function of cytoplasmic dynein in M phase. Mol Biol Cell 2007; 18:2656-66. [PMID: 17494871 PMCID: PMC1924840 DOI: 10.1091/mbc.e06-04-0345] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The microtubule-based motor cytoplasmic dynein/dynactin is a force generator at the kinetochore. It also transports proteins away from kinetochores to spindle poles. Regulation of such diverse functions, however, is poorly understood. We have previously shown that Nudel is critical for dynein-mediated protein transport, whereas mitosin, a kinetochore protein that binds Nudel, is involved in retention of kinetochore dynein/dynactin against microtubule-dependent stripping. Here we demonstrate that Nudel is required for robust localization of dynein/dynactin at the kinetochore. It localizes to kinetochores after nuclear envelope breakdown, depending mostly ( approximately 78%) on mitosin and slightly on dynein/dynactin. Depletion of Nudel by RNA interference (RNAi) or overexpression of its mutant incapable of binding either Lis1 or dynein heavy chain abolishes the kinetochore protein transport and mitotic progression. Similar to mitosin RNAi, Nudel RNAi also leads to increased stripping of kinetochore dynein/dynactin in the presence of microtubules. Taking together, our results suggest a dual role of kinetochore Nudel: it activates dynein-mediated protein transport and, when interacting with both mitosin and dynein, stabilizes kinetochore dynein/dynactin against microtubule-dependent stripping to facilitate the force generation function of the motor.
Collapse
Affiliation(s)
- Yun Liang
- *Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Wei Yu
- *Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Yan Li
- *Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Lihou Yu
- *Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Qiangge Zhang
- *Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Fubin Wang
- *Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Zhenye Yang
- *Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Juan Du
- *Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Qiongping Huang
- *Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Xuebiao Yao
- Hefei National Laboratory for Physical Sciences and the School of Life Sciences, University of Science and Technology of China, Anhui 230027, China
| | - Xueliang Zhu
- *Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| |
Collapse
|
30
|
Evans HJ, Edwards L, Goodwin RL. Conserved C-terminal domains of mCenp-F (LEK1) regulate subcellular localization and mitotic checkpoint delay. Exp Cell Res 2007; 313:2427-37. [PMID: 17498689 PMCID: PMC3991481 DOI: 10.1016/j.yexcr.2007.03.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Revised: 03/16/2007] [Accepted: 03/19/2007] [Indexed: 11/24/2022]
Abstract
Centromeric Protein-F (Cenp-F) family members have been identified in organisms from yeast to human. Cenp-F proteins are a component of kinetochores during mitosis, bind to the Rb family of tumor suppressors, and have regulatory effects on the cell cycle and differentiation; however, their role in these processes has not been resolved. Here, we provide evidence that the role of murine Cenp-F (mCenp-F, also known as LEK1) remains largely conserved and that the domains within the C-terminus collectively function to regulate the G2/M cell cycle checkpoint. Overexpression of the C-terminal domain of mCenp-F decreases DNA synthesis. Analyses of deletion mutants of mCenp-F reveal that the complete C-terminal domain is required to delay cell cycle progression at G2/M. Signal transduction pathway profiling experiments indicate that the mCenp-F-mediated cell cycle delay does not involve transcriptional activity of key cell cycle regulators such as Rb, E2F, p53, or Myc. However, endogenous mCenp-F colocalizes with pRb and p107, which demonstrates in vivo protein-protein interaction during cell division. These observations suggest that the domains of the C-terminus of mCenp-F have a conserved function in control of mitotic progression through protein-protein interaction with pocket proteins, thus providing a direct connection between cell cycle regulation and mitotic progression.
Collapse
Affiliation(s)
- Heather J Evans
- Department of Cell and Developmental Biology and Anatomy, University of South Carolina School of Medicine, SC, USA
| | | | | |
Collapse
|
31
|
Dees E, Robertson JB, Zhu T, Bader D. Specific deletion of CMF1 nuclear localization domain causes incomplete cell cycle withdrawal and impaired differentiation in avian skeletal myoblasts. Exp Cell Res 2006; 312:3000-14. [PMID: 16904105 DOI: 10.1016/j.yexcr.2006.06.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2006] [Revised: 05/31/2006] [Accepted: 06/01/2006] [Indexed: 11/17/2022]
Abstract
CMF1 is a protein expressed in embryonic striated muscle with onset of expression preceding that of contractile proteins. Disruption of CMF1 in myoblasts disrupts muscle-specific protein expression. Preliminary studies indicate both nuclear and cytoplasmic distribution of CMF1 protein, suggesting functional roles in both cellular compartments. Here we examine the nuclear function of CMF1, using a newly characterized antibody generated against the CMF1 nuclear localization domain and a CMF1 nuclear localization domain-deleted stable myocyte line. The antibody demonstrates nuclear distribution of the CMF1 protein both in vivo and in cell lines, with clustering of CMF1 protein around chromatin during mitosis. In more differentiated myocytes, the protein shifts to the cytoplasm. The CMF1 NLS-deleted cell lines have markedly impaired capacity to differentiate. Specifically, these cells express less contractile protein than wild-type or full-length CMF1 stably transfected cells, and do not fuse properly into multinucleate syncytia with linear nuclear alignment. In response to low serum medium, a signal to differentiate, CMF1 NLS-deleted cells enter G0, but continue to express proliferation markers and will reenter the cell cycle when stimulated by restoring growth medium. These data suggest that CMF1 is involved in regulation the transition from proliferation to differentiation in embryonic muscle.
Collapse
Affiliation(s)
- Ellen Dees
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | | | | | | |
Collapse
|
32
|
Varis A, Salmela AL, Kallio MJ. Cenp-F (mitosin) is more than a mitotic marker. Chromosoma 2006; 115:288-95. [PMID: 16565862 DOI: 10.1007/s00412-005-0046-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2005] [Revised: 12/02/2005] [Accepted: 12/15/2005] [Indexed: 12/18/2022]
Abstract
Cenp-F (mitosin) is a large coiled-coil protein whose function has remained obscure since its identification a decade ago. It has been suggested that the protein plays a role in the kinetochore-mediated mitotic functions but until recently there was little evidence to support this postulation. Recent results from five laboratories have given insights on how Cenp-F may participate in the regulation of cell division. In this mini-review, we will summarize the current data regarding the mitotic tasks of Cenp-F as well as discuss how it is used as a proliferation marker of malignant cell growth in the clinic. Also, the protein's post-translational modification by farnesylation and potential contribution to cell cycle effects of farnesyl transferase inhibitors will be addressed.
Collapse
Affiliation(s)
- Asta Varis
- Cancer Biology and Cell Screening Department, VTT Medical Biotechnology, Itäinen Pitkäkatu 4A, 20521, Turku, Finland
| | | | | |
Collapse
|
33
|
Ma L, Zhao X, Zhu X. Mitosin/CENP-F in mitosis, transcriptional control, and differentiation. J Biomed Sci 2006; 13:205-13. [PMID: 16456711 DOI: 10.1007/s11373-005-9057-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2005] [Accepted: 12/22/2005] [Indexed: 01/03/2023] Open
Abstract
Mitosin/CENP-F is a large nuclear/kinetochore protein containing multiple leucine zipper motifs potentially for protein interactions. Its expression levels and subcellular localization patterns are regulated in a cell cycle-dependent manner. Recently, accumulating lines of evidence have suggested it a multifunctional protein involved in mitotic control, microtubule dynamics, transcriptional regulation, and muscle cell differentiation. Consistently, it is shown to interact directly with a variety of proteins including CENP-E, NudE/Nudel, ATF4, and Rb. Here we review the current progress and discuss possible mechanisms through which mitosin may function.
Collapse
Affiliation(s)
- Li Ma
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | | | | |
Collapse
|
34
|
Shigeishi H, Mizuta K, Higashikawa K, Yoneda S, Ono S, Kamata N. Correlation of CENP-F gene expression with tumor-proliferating activity in human salivary gland tumors. Oral Oncol 2006; 41:716-22. [PMID: 15927522 DOI: 10.1016/j.oraloncology.2005.03.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2005] [Accepted: 03/20/2005] [Indexed: 12/25/2022]
Abstract
We examined the expression of Centromere protein F (CENP-F) mRNA in 26 human salivary gland tumors (seven pleomorphic adenomas, three Warthin tumors, seven mucoepidermoid carcinomas, four adenoid cystic carcinomas, four acinic cell carcinomas and one malignant myoepithelioma) and four normal submandibular glands using the real time quantitative reverse transcription-polymerase chain reaction (RT-PCR). The mean expression level of CENP-F mRNA was higher in malignant tumors (1.05+/-0.32) than normal submandibular glands (0.11+/-0.05) and benign tumors (0.46+/-0.16). We found a significant association between the level of CENP-F mRNA expression and clinical stage in 16 malignant tumors (Mann-Whitney U test, p=0.027). We also found a significant correlation between the Ki-67 labeling index and CENP-F expression in malignant tumors (Spearmans correlation coefficient by rank test, p=0.029). These results indicate that human CENP-F mRNA is closely linked to the increased or abnormal cell proliferation in malignant conditions.
Collapse
Affiliation(s)
- Hideo Shigeishi
- Department of Oral and Maxillofacial Surgery, Division of Cervico-Gnathostomatology, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | | | | | | | | | | |
Collapse
|
35
|
Guo J, Yang Z, Song W, Chen Q, Wang F, Zhang Q, Zhu X. Nudel contributes to microtubule anchoring at the mother centriole and is involved in both dynein-dependent and -independent centrosomal protein assembly. Mol Biol Cell 2005; 17:680-9. [PMID: 16291865 PMCID: PMC1356579 DOI: 10.1091/mbc.e05-04-0360] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The centrosome is the major microtubule-organizing center in animal cells. Although the cytoplasmic dynein regulator Nudel interacts with centrosomes, its role herein remains unclear. Here, we show that in Cos7 cells Nudel is a mother centriole protein with rapid turnover independent of dynein activity. During centriole duplication, Nudel targets to the new mother centriole later than ninein but earlier than dynactin. Its centrosome localization requires a C-terminal region that is essential for associations with dynein, dynactin, pericentriolar material (PCM)-1, pericentrin, and gamma-tubulin. Overexpression of a mutant Nudel lacking this region, a treatment previously shown to inactivate dynein, dislocates centrosomal Lis1, dynactin, and PCM-1, with little influence on pericentrin and gamma-tubulin in Cos7 and HeLa cells. Silencing Nudel in HeLa cells markedly decreases centrosomal targeting of all the aforementioned proteins. Silencing Nudel also represses centrosomal MT nucleation and anchoring. Furthermore, Nudel can interact with pericentrin independently of dynein. Our current results suggest that Nudel plays a role in both dynein-mediated centripetal transport of dynactin, Lis1, and PCM-1 as well as in dynein-independent centrosomal targeting of pericentrin and gamma-tubulin. Moreover, Nudel seems to tether dynactin and dynein to the mother centriole for MT anchoring.
Collapse
Affiliation(s)
- Jing Guo
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, and Graduate School, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | |
Collapse
|
36
|
Yuen KWY, Montpetit B, Hieter P. The kinetochore and cancer: what's the connection? Curr Opin Cell Biol 2005; 17:576-82. [PMID: 16233975 DOI: 10.1016/j.ceb.2005.09.012] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2005] [Accepted: 09/29/2005] [Indexed: 12/19/2022]
Abstract
The molecular mechanisms ensuring accurate chromosome segregation during meiosis and mitosis are critical to the conservation of euploidy (normal chromosome number) in eukaryotic cells. A dysfunctional kinetochore represents one possible source for chromosome instability (CIN) and the generation of aneuploidy. The kinetochore is a large complex of proteins and associated centromeric DNA that is responsible for mediating the segregation of sister chromatids to daughter cells via its interactions with the mitotic spindle. Continued identification of conserved kinetochore components in model systems such as yeast has provided a rich resource of candidate genes that may be mutated or misregulated in human cancers. Systematic mutational testing and transcriptional profiling of CIN candidate kinetochore genes should shed light on the kinetochore's role in tumorigenesis, and on the general role CIN plays in cancer development.
Collapse
Affiliation(s)
- Karen W Y Yuen
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, British Columbia, Canada V6T 1Z4
| | | | | |
Collapse
|
37
|
Yang Z, Guo J, Chen Q, Ding C, Du J, Zhu X. Silencing mitosin induces misaligned chromosomes, premature chromosome decondensation before anaphase onset, and mitotic cell death. Mol Cell Biol 2005; 25:4062-74. [PMID: 15870278 PMCID: PMC1087709 DOI: 10.1128/mcb.25.10.4062-4074.2005] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Mitosin (also named CENP-F) is a large human nuclear protein transiently associated with the outer kinetochore plate in M phase. Using RNA interference and fluorescence microscopy, we showed that mitosin depletion attenuated chromosome congression and led to metaphase arrest with misaligned polar chromosomes whose kinetochores showed few cold-stable microtubules. Kinetochores of fully aligned chromosomes often failed to show orientation in the direction of the spindle long axis. Moreover, tension across their sister kinetochores was decreased by 53% on average. These phenotypes collectively imply defects in motor functions in mitosin-depleted cells and are similar to those of CENP-E depletion. Consistently, the intensities of CENP-E and cytoplasmic dynein and dynactin, which are motors controlling microtubule attachment and chromosome movement, were reduced at the kinetochore in a microtubule-dependent manner. In addition, after being arrested in pseudometaphase for approximately 2 h, mitosin-depleted cells died before anaphase initiation through apoptosis. The dying cells exhibited progressive chromosome arm decondensation, while the centromeres were still associated with spindles. Mitosin is therefore essential for full chromosome alignment, possibly by promoting proper kinetochore attachments through modulating CENP-E and dynein functions. Its depletion also prematurely triggers chromosome decondensation, a process that normally occurs from telophase for the nucleus reassembly, thus resulting in apoptosis.
Collapse
Affiliation(s)
- Zhenye Yang
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | | | | | | | | | | |
Collapse
|
38
|
Buffin E, Lefebvre C, Huang J, Gagou ME, Karess RE. Recruitment of Mad2 to the kinetochore requires the Rod/Zw10 complex. Curr Biol 2005; 15:856-61. [PMID: 15886105 DOI: 10.1016/j.cub.2005.03.052] [Citation(s) in RCA: 168] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2005] [Revised: 03/15/2005] [Accepted: 03/30/2005] [Indexed: 11/24/2022]
Abstract
Compromising the activity of the spindle checkpoint permits mitotic exit in the presence of unattached kinetochores and, consequently, greatly increases the rate of aneuploidy in the daughter cells. The metazoan checkpoint mechanism is more complex than in yeast in that it requires additional proteins and activities besides the classical Mads and Bubs. Among these are Rod, Zw10, and Zwilch, components of a 700 Kdal complex (Rod/Zw10) that is required for recruitment of dynein/dynactin to kinetochores but whose role in the checkpoint is poorly understood. The dynamics of Rod and Mad2, examined in different organisms, show intriguing similarities as well as apparent differences. Here we simultaneously follow GFP-Mad2 and RFP-Rod and find they are in fact closely associated throughout early mitosis. They accumulate simultaneously on kinetochores and are shed together along microtubule fibers after attachment. Their behavior and position within attached kinetochores is distinct from that of BubR1; Mad2 and Rod colocalize to the outermost kinetochore region (the corona), whereas BubR1 is slightly more interior. Moreover, Mad2, but not BubR1, Bub1, Bub3, or Mps1, requires Rod/Zw10 for its accumulation on unattached kinetochores. Rod/Zw10 thus contributes to checkpoint activation by promoting Mad2 recruitment and to checkpoint inactivation by recruiting dynein/dynactin that subsequently removes Mad2 from attached kinetochores.
Collapse
Affiliation(s)
- Eulalie Buffin
- Centre National de la Recherche Scientifique, Centre de Génétique Moléculaire, Avenue de la Terrasse, 91198 Gif sur Yvette, France
| | | | | | | | | |
Collapse
|
39
|
Zhou X, Wang R, Fan L, Li Y, Ma L, Yang Z, Yu W, Jing N, Zhu X. Mitosin/CENP-F as a negative regulator of activating transcription factor-4. J Biol Chem 2005; 280:13973-7. [PMID: 15677469 DOI: 10.1074/jbc.m414310200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Mitosin/CENP-F is a human nuclear matrix protein with multiple leucine zipper motifs. Its accumulation in S-G2 phases and transient kinetochore localization in mitosis suggest a multifunctional protein for cell proliferation. Moreover, its murine and avian orthologs are implicated in myocyte differentiation. Here we report its interaction with activating transcription factor-4 (ATF4), a ubiquitous basic leucine zipper transcription factor important for proliferation, differentiation, and stress response. The C-terminal portion of mitosin between residues 2488 and 3113 bound to ATF4 through two distinct domains, one of which was a leucine zipper motif. Mitosin mutants containing these domains were able to either supershift or disrupt the ATF4-DNA complex. On the other hand, ATF4, but not ATF1-3 or ATF6, interacted with mitosin through a region containing the basic leucine zipper motif. Moreover, overexpression of full-length mitosin repressed the transactivation activity of ATF4 in dual luciferase-based reporter assays, while knocking down mitosin expression manifested the opposite effects. These findings suggest mitosin to be a negative regulator of ATF4 in interphase through direct interaction.
Collapse
Affiliation(s)
- Xubin Zhou
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Liang Y, Yu W, Li Y, Yang Z, Yan X, Huang Q, Zhu X. Nudel functions in membrane traffic mainly through association with Lis1 and cytoplasmic dynein. ACTA ACUST UNITED AC 2004; 164:557-66. [PMID: 14970193 PMCID: PMC2171987 DOI: 10.1083/jcb.200308058] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Nudel and Lis1 appear to regulate cytoplasmic dynein in neuronal migration and mitosis through direct interactions. However, whether or not they regulate other functions of dynein remains elusive. Herein, overexpression of a Nudel mutant defective in association with either Lis1 or dynein heavy chain is shown to cause dispersions of membranous organelles whose trafficking depends on dynein. In contrast, the wild-type Nudel and the double mutant that binds to neither protein are much less effective. Time-lapse microscopy for lysosomes reveals significant reduction in both frequencies and velocities of their minus end–directed motions in cells expressing the dynein-binding defective mutant, whereas neither the durations of movement nor the plus end–directed motility is considerably altered. Moreover, silencing Nudel expression by RNA interference results in Golgi apparatus fragmentation and cell death. Together, it is concluded that Nudel is critical for dynein motor activity in membrane transport and possibly other cellular activities through interactions with both Lis1 and dynein heavy chain.
Collapse
Affiliation(s)
- Yun Liang
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China
| | | | | | | | | | | | | |
Collapse
|
41
|
Prasanth SG, Prasanth KV, Siddiqui K, Spector DL, Stillman B. Human Orc2 localizes to centrosomes, centromeres and heterochromatin during chromosome inheritance. EMBO J 2004; 23:2651-63. [PMID: 15215892 PMCID: PMC449767 DOI: 10.1038/sj.emboj.7600255] [Citation(s) in RCA: 215] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2004] [Accepted: 05/05/2004] [Indexed: 01/04/2023] Open
Abstract
The initiation of DNA replication in S phase requires the prior assembly of an origin recognition complex (ORC)-dependent pre-replicative complex on chromatin during G1 phase of the cell division cycle. In human cells, the Orc2 subunit localized to the nucleus as expected, but it also localized to centrosomes throughout the entire cell cycle. Furthermore, Orc2 was tightly bound to heterochromatin and heterochromatin protein 1alpha (HP1alpha) and HP1beta in G1 and early S phase, but during late S, G2 and M phases tight chromatin association was restricted to centromeres. Depletion of Orc2 by siRNA caused multiple phenotypes. A population of cells showed an S-phase defect with little proliferating cell nuclear antigen (PCNA) on chromatin, although MCM proteins remained. Orc2 depletion also disrupted HP1 localization, but not histone-H3-lysine-9 methylation at prominent heterochromatic foci. Another subset of Orc2-depleted cells containing replicated DNA arrested with abnormally condensed chromosomes, failed chromosome congression and multiple centrosomes. These results implicate Orc2 protein in chromosome duplication, chromosome structure and centrosome copy number control, suggesting that it coordinates all stages of the chromosome inheritance cycle.
Collapse
Affiliation(s)
| | | | | | | | - Bruce Stillman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA. Tel.: +1 516 367 8383; Fax: +1 516 367 8879; E-mail:
| |
Collapse
|