101
|
Abstract
The mitotic checkpoint is a specialized signal transduction pathway that contributes to the fidelity of chromosome segregation. The signaling of the checkpoint originates from defective kinetochore-microtubule interactions and leads to formation of the mitotic checkpoint complex (MCC), a highly potent inhibitor of the Anaphase Promoting Complex/Cyclosome (APC/C)—the E3 ubiquitin ligase essential for anaphase onset. Many important questions concerning the MCC and its interaction with APC/C have been intensively investigated and debated in the past 15 years, such as the exact composition of the MCC, how it is assembled during a cell cycle, how it inhibits APC/C, and how the MCC is disassembled to allow APC/C activation. These efforts have culminated in recently reported structure models for human MCC:APC/C supra-complexes at near-atomic resolution that shed light on multiple aspects of the mitotic checkpoint mechanisms. However, confusing statements regarding the MCC are still scattered in the literature, making it difficult for students and scientists alike to obtain a clear picture of MCC composition, structure, function and dynamics. This review will comb through some of the most popular concepts or misconceptions about the MCC, discuss our current understandings, present a synthesized model on regulation of CDC20 ubiquitination, and suggest a few future endeavors and cautions for next phase of MCC research.
Collapse
Affiliation(s)
- Song-Tao Liu
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA
| | - Hang Zhang
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA
| |
Collapse
|
102
|
Manic G, Corradi F, Sistigu A, Siteni S, Vitale I. Molecular Regulation of the Spindle Assembly Checkpoint by Kinases and Phosphatases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 328:105-161. [PMID: 28069132 DOI: 10.1016/bs.ircmb.2016.08.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The spindle assembly checkpoint (SAC) is a surveillance mechanism contributing to the preservation of genomic stability by monitoring the microtubule attachment to, and/or the tension status of, each kinetochore during mitosis. The SAC halts metaphase to anaphase transition in the presence of unattached and/or untensed kinetochore(s) by releasing the mitotic checkpoint complex (MCC) from these improperly-oriented kinetochores to inhibit the anaphase-promoting complex/cyclosome (APC/C). The reversible phosphorylation of a variety of substrates at the kinetochore by antagonistic kinases and phosphatases is one major signaling mechanism for promptly turning on or turning off the SAC. In such a complex network, some kinases act at the apex of the SAC cascade by either generating (monopolar spindle 1, MPS1/TTK and likely polo-like kinase 1, PLK1), or contributing to generate (Aurora kinase B) kinetochore phospho-docking sites for the hierarchical recruitment of the SAC proteins. Aurora kinase B, MPS1 and budding uninhibited by benzimidazoles 1 (BUB1) also promote sister chromatid biorientation by modulating kinetochore microtubule stability. Moreover, MPS1, BUB1, and PLK1 seem to play key roles in APC/C inhibition by mechanisms dependent and/or independent on MCC assembly. The protein phosphatase 1 and 2A (PP1 and PP2A) are recruited to kinetochores to oppose kinase activity. These phosphatases reverse the phosphorylation of kinetochore targets promoting the microtubule attachment stabilization, sister kinetochore biorientation and SAC silencing. The kinase-phosphatase network is crucial as it renders the SAC a dynamic, graded-signaling, high responsive, and robust process thereby ensuring timely anaphase onset and preventing the generation of proneoplastic aneuploidy.
Collapse
Affiliation(s)
- G Manic
- Regina Elena National Cancer Institute, Rome, Italy.
| | - F Corradi
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - A Sistigu
- Regina Elena National Cancer Institute, Rome, Italy
| | - S Siteni
- Regina Elena National Cancer Institute, Rome, Italy; Department of Biology, University of Rome "Roma Tre", Rome, Italy
| | - I Vitale
- Regina Elena National Cancer Institute, Rome, Italy; Department of Biology, University of Rome "Tor Vergata", Rome, Italy.
| |
Collapse
|
103
|
Abstract
The centrosome is the main microtubule organizing center of animal cells. It contributes to spindle assembly and orientation during mitosis and to ciliogenesis in interphase. Numerical and structural defects in this organelle are known to be associated with developmental disorders such as dwarfism and microcephaly, but only recently, the molecular mechanisms linking centrosome aberrations to altered physiology are being elucidated. Defects in centrosome number or structure have also been described in cancer. These opposite clinical outcomes--arising from reduced proliferation and overproliferation respectively--can be explained in light of the tissue- and developmental-specific requirements for centrosome functions. The pathological outcomes of centrosome deficiencies have become clearer when considering its consequences. Among them, there are genetic instability (mainly aneuploidy, a defect in chromosome number), defects in the symmetry of cell division (important for cell fate specification and tissue architecture) and impaired ciliogenesis. In this review, we discuss the origins and the consequences of centrosome flaws, with particular attention on how they contribute to developmental diseases.
Collapse
Affiliation(s)
- Maddalena Nano
- Institut Curie, PSL Research University, CNRS UMR144, 12 rue Lhomond, 75005, Paris, France
| | - Renata Basto
- Institut Curie, PSL Research University, CNRS UMR144, 12 rue Lhomond, 75005, Paris, France.
| |
Collapse
|
104
|
Cosenza MR, Krämer A. Centrosome amplification, chromosomal instability and cancer: mechanistic, clinical and therapeutic issues. Chromosome Res 2016; 24:105-26. [PMID: 26645976 DOI: 10.1007/s10577-015-9505-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Centrosomes, the main microtubule-organizing centers in most animal cells, are of crucial importance for the assembly of a bipolar mitotic spindle and subsequent faithful segregation of chromosomes into two daughter cells. Centrosome abnormalities can be found in virtually all cancer types and have been linked to chromosomal instability (CIN) and tumorigenesis. Although our knowledge on centrosome structure, replication, and amplification has greatly increased within recent years, still only very little is known on nature, causes, and consequences of centrosome aberrations in primary tumor tissues. In this review, we summarize our current insights into the mechanistic link between centrosome aberrations, aneuploidy, CIN and tumorigenesis. Mechanisms of induction and cellular consequences of aneuploidy, tetraploidization and CIN, as well as origin and effects of supernumerary centrosomes will be discussed. In addition, animal models for both CIN and centrosome amplification will be outlined. Finally, we describe approaches to exploit centrosome amplification, aneuploidy and CIN for novel and specific anticancer treatment strategies based on the modulation of chromosome missegregation rates.
Collapse
Affiliation(s)
- Marco Raffaele Cosenza
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
105
|
Bhat A, Wu Z, Maher VM, McCormick JJ, Xiao W. Rev7/Mad2B plays a critical role in the assembly of a functional mitotic spindle. Cell Cycle 2016; 14:3929-38. [PMID: 26697843 DOI: 10.1080/15384101.2015.1120922] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The spindle assembly checkpoint (SAC) acts as a guardian against cellular threats that may lead to chromosomal missegregation and aneuploidy. Mad2, an anaphase-promoting complex/cyclosome-Cdc20 (APC/C(Cdc20)) inhibitor, has an additional homolog in mammals known as Mad2B, Mad2L2 or Rev7. Apart from its role in Polζ-mediated translesion DNA synthesis and double-strand break repair, Rev7 is also believed to inhibit APC/C by negatively regulating Cdh1. Here we report yet another function of Rev7 in cultured human cells. Rev7, as predicted earlier, is involved in the formation of a functional spindle and maintenance of chromosome segregation. In the absence of Rev7, cells tend to arrest in G2/M-phase and display increased monoastral and abnormal spindles with misaligned chromosomes. Furthermore, Rev7-depleted cells show Mad2 localization at the kinetochores of metaphase cells, an indicator of activated SAC, coupled with increased levels of Cyclin B1, an APC(Cdc20) substrate. Surprisingly unlike Mad2, depletion of Rev7 in several cultured human cell lines did not compromise SAC activity. Our data therefore suggest that besides its role in APC/C(Cdh1) inhibition, Rev7 is also required for mitotic spindle organization and faithful chromosome segregation most probably through its physical interaction with RAN.
Collapse
Affiliation(s)
- Audesh Bhat
- a Department of Microbiology and Immunology ; University of Saskatchewan ; Saskatchewan , Canada
| | - Zhaojia Wu
- a Department of Microbiology and Immunology ; University of Saskatchewan ; Saskatchewan , Canada
| | - Veronica M Maher
- b Carcinogenesis Laboratory; Michigan State University ; East Lansing , MI USA
| | - J Justin McCormick
- b Carcinogenesis Laboratory; Michigan State University ; East Lansing , MI USA
| | - Wei Xiao
- a Department of Microbiology and Immunology ; University of Saskatchewan ; Saskatchewan , Canada.,c College of Life Sciences; Capital Normal University ; Beijing , China
| |
Collapse
|
106
|
Cunningham CE, Li S, Vizeacoumar FS, Bhanumathy KK, Lee JS, Parameswaran S, Furber L, Abuhussein O, Paul JM, McDonald M, Templeton SD, Shukla H, El Zawily AM, Boyd F, Alli N, Mousseau DD, Geyer R, Bonham K, Anderson DH, Yan J, Yu-Lee LY, Weaver BA, Uppalapati M, Ruppin E, Sablina A, Freywald A, Vizeacoumar FJ. Therapeutic relevance of the protein phosphatase 2A in cancer. Oncotarget 2016; 7:61544-61561. [PMID: 27557495 PMCID: PMC5308671 DOI: 10.18632/oncotarget.11399] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 08/10/2016] [Indexed: 11/25/2022] Open
Abstract
Chromosomal Instability (CIN) is regarded as a unifying feature of heterogeneous tumor populations, driving intratumoral heterogeneity. Polo-Like Kinase 1 (PLK1), a serine-threonine kinase that is often overexpressed across multiple tumor types, is one of the key regulators of CIN and is considered as a potential therapeutic target. However, targeting PLK1 has remained a challenge due to the off-target effects caused by the inhibition of other members of the polo-like family. Here we use synthetic dosage lethality (SDL), where the overexpression of PLK1 is lethal only when another, normally non-lethal, mutation or deletion is present. Rather than directly inhibiting PLK1, we found that inhibition of PP2A causes selective lethality to PLK1-overexpressing breast, pancreatic, ovarian, glioblastoma, and prostate cancer cells. As PP2A is widely regarded as a tumor suppressor, we resorted to gene expression datasets from cancer patients to functionally dissect its therapeutic relevance. We identified two major classes of PP2A subunits that negatively correlated with each other. Interestingly, most mitotic regulators, including PLK1, exhibited SDL interactions with only one class of PP2A subunits (PPP2R1A, PPP2R2D, PPP2R3B, PPP2R5B and PPP2R5D). Validation studies and other functional cell-based assays showed that inhibition of PPP2R5D affects both levels of phospho-Rb as well as sister chromatid cohesion in PLK1-overexpressing cells. Finally, analysis of clinical data revealed that patients with high expression of mitotic regulators and low expression of Class I subunits of PP2A improved survival. Overall, these observations point to a context-dependent role of PP2A that warrants further exploration for therapeutic benefits.
Collapse
Affiliation(s)
- Chelsea E. Cunningham
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Shuangshuang Li
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Frederick S. Vizeacoumar
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | | | - Joo Sang Lee
- Center for Bioinformatics and Computational Biology, Department of Computer Science, University of Maryland, Maryland, MD 20742, USA
| | - Sreejit Parameswaran
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Levi Furber
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Omar Abuhussein
- College of Pharmacy, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 2Z4, Canada
| | - James M. Paul
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Megan McDonald
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Shaina D. Templeton
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Hersh Shukla
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Amr M. El Zawily
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Frederick Boyd
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Nezeka Alli
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Darrell D. Mousseau
- Cell Signaling Laboratory, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Ron Geyer
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Keith Bonham
- Cancer Research, Saskatchewan Cancer Agency, Saskatoon, Saskatchewan, S7N 5E5, Canada
| | - Deborah H. Anderson
- Cancer Research, Saskatchewan Cancer Agency, Saskatoon, Saskatchewan, S7N 5E5, Canada
| | - Jiong Yan
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Li-Yuan Yu-Lee
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Beth A. Weaver
- Department of Cell and Regenerative Biology and Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705-2275, USA
| | - Maruti Uppalapati
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Eytan Ruppin
- Center for Bioinformatics and Computational Biology, Department of Computer Science, University of Maryland, Maryland, MD 20742, USA
| | - Anna Sablina
- VIB Center for the Biology of Disease, VIB, 3000 Leuven, Belgium
| | - Andrew Freywald
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Franco J. Vizeacoumar
- Department of Pathology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
- College of Pharmacy, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 2Z4, Canada
- Cancer Research, Saskatchewan Cancer Agency, Saskatoon, Saskatchewan, S7N 5E5, Canada
| |
Collapse
|
107
|
Rao CV, Asch AS, Yamada HY. Emerging links among Chromosome Instability (CIN), cancer, and aging. Mol Carcinog 2016; 56:791-803. [PMID: 27533343 DOI: 10.1002/mc.22539] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/14/2016] [Accepted: 08/15/2016] [Indexed: 12/15/2022]
Abstract
Aneuploidy was predicted to cause cancer. To test the prediction, various Chromosome Instability (CIN) mice models that carry transgenic mutations in mitotic regulators have been created. The availability of these mice has aided researchers in discovering connections between CIN, cancer, and aging. This review will focus on recent interdisciplinary findings regarding how CIN and aneuploidy affect carcinogenesis, immune dysfunction, and aging. High CIN can be generated in vivo by various intrinsic alterations (e.g., gene mutation, epigenetic modification) and extrinsic/environmental challenges (e.g., biological, chemical, biophysical), while immune surveillance, cell death, and natural turnover can remove cells with CIN. CIN itself is mutagenic and may cause further cellular mutations, which can be carcinogenic. Mitotically damaged cells can activate senescence-related tumor suppressors (e.g., p21WAF1 , p27KIP1 , p16INK4A ), which may lead to tissue-level senescence/aging through inflammatory paracrine mechanisms called Senescence-Associated Secretory Phenotype (SASP) and Senescence Inflammatory Response (SIR). Organs with high CIN show altered gene expressions in both organ-specific and non-specific manners. Organ-specific gene expression signatures include activation of oncogenic pathways. Non-organ-specific gene expression signatures include metabolic changes and downregulations in immune functions. Immune surveillance normally targets senescent cells and tetraploid cells, a form of aneuploidy, for elimination. However, with partial immune dysfunction, immune surveillance is weakened with systemic CIN. In this case, more senescent cells and aneuploid cells survive, which further leads to an inflammatory, pro-tumorigenic, and senescent/aging microenvironment. We also discuss how we may intervene in this sequence of events to prevent CIN- or age-related carcinogenesis and/or some aspects of tissue aging. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Chinthalapally V Rao
- Department of Medicine, Center for Cancer Prevention and Drug Development, Hematology/Oncology Section, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma.,Stephenson Cancer Center, Hematology/Oncology, University of Oklahoma, Oklahoma City, Oklahoma
| | - Adam S Asch
- Stephenson Cancer Center, Hematology/Oncology, University of Oklahoma, Oklahoma City, Oklahoma
| | - Hiroshi Y Yamada
- Department of Medicine, Center for Cancer Prevention and Drug Development, Hematology/Oncology Section, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma.,Stephenson Cancer Center, Hematology/Oncology, University of Oklahoma, Oklahoma City, Oklahoma
| |
Collapse
|
108
|
Weaver RL, Limzerwala JF, Naylor RM, Jeganathan KB, Baker DJ, van Deursen JM. BubR1 alterations that reinforce mitotic surveillance act against aneuploidy and cancer. eLife 2016; 5. [PMID: 27528194 PMCID: PMC4987139 DOI: 10.7554/elife.16620] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 07/19/2016] [Indexed: 12/21/2022] Open
Abstract
BubR1 is a key component of the spindle assembly checkpoint (SAC). Mutations that reduce BubR1 abundance cause aneuploidization and tumorigenesis in humans and mice, whereas BubR1 overexpression protects against these. However, how supranormal BubR1 expression exerts these beneficial physiological impacts is poorly understood. Here, we used Bub1b mutant transgenic mice to explore the role of the amino-terminal (BubR1N) and internal (BubR1I) Cdc20-binding domains of BubR1 in preventing aneuploidy and safeguarding against cancer. BubR1N was necessary, but not sufficient to protect against aneuploidy and cancer. In contrast, BubR1 lacking the internal Cdc20-binding domain provided protection against both, which coincided with improved microtubule-kinetochore attachment error correction and SAC activity. Maximal SAC reinforcement occurred when both the Phe- and D-box of BubR1I were disrupted. Thus, while under- or overexpression of most mitotic regulators impairs chromosome segregation fidelity, certain manipulations of BubR1 can positively impact this process and therefore be therapeutically exploited. DOI:http://dx.doi.org/10.7554/eLife.16620.001 Human DNA is organized into 46 chromosomes, which must be duplicated before a cell divides and are then shared equally between the two new cells. When this process goes awry, the new cells either have too many or too few chromosomes. This situation – known as aneuploidy – frequently occurs in cancer cells, and is thought to cause cells to gain extra copies or lose copies of genes that promote or prevent cancer, respectively. Cells have several ways to prevent aneuploidy. One of these safeguards, known as the spindle assembly checkpoint (SAC), involves a protein called BubR1, which acts at the stage when the duplicated chromosomes need to be equally divided into each daughter cell. Mouse models show that low levels of the BubR1 protein result in aneuploidy and increased predisposition to cancer. High levels of BubR1, on the other hand, allow the mice to stay healthier for longer and can stop tumors from forming. However, it was not known exactly how high amounts of BubR1 protect against cancer. To address this question, Weaver et al. set out to determine which parts, or domains, of the BubR1 protein protect against cancer. Mice with high levels of the full-length BubR1 protein were compared with mice that made mutant versions of BubR1 lacking certain domains. These experiments revealed that a small portion of the beginning of the protein was necessary to protect against tumor formation, but removing a large region in the middle of BubR1 still protected mice against lung cancer and aneuploidy. Additional experiments performed on mouse cells grown in the laboratory revealed that whole BubR1 protein and the mutant protein lacking the middle region might prevent aneuploidy in multiple ways. For example, both systems had stronger SAC signaling, which could serve to make segregating the chromosomes more accurate. In the future, it will be important to find out whether BubR1 acts in the same way in human cells and cancers. Lastly, since it is not possible to over-produce BubR1 in humans, other methods will need to be investigated to use this knowledge to treat cancer. DOI:http://dx.doi.org/10.7554/eLife.16620.002
Collapse
Affiliation(s)
- Robbyn L Weaver
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Jazeel F Limzerwala
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Ryan M Naylor
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Karthik B Jeganathan
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, United States
| | - Darren J Baker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States.,Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, United States
| | - Jan M van Deursen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States.,Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, United States
| |
Collapse
|
109
|
Restall IJ, Parolin DAE, Daneshmand M, Hanson JEL, Simard MA, Fitzpatrick ME, Kumar R, Lavictoire SJ, Lorimer IAJ. PKCι depletion initiates mitotic slippage-induced senescence in glioblastoma. Cell Cycle 2016. [PMID: 26208522 PMCID: PMC4825548 DOI: 10.1080/15384101.2015.1071744] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Cellular senescence is a tumor suppressor mechanism where cells enter a permanent growth arrest following cellular stress. Oncogene-induced senescence (OIS) is induced in non-malignant cells following the expression of an oncogene or inactivation of a tumor suppressor. Previously, we have shown that protein kinase C iota (PKCι) depletion induces cellular senescence in glioblastoma cells in the absence of a detectable DNA damage response. Here we demonstrate that senescent glioblastoma cells exhibit an aberrant centrosome morphology. This was observed in basal levels of senescence, in p21-induced senescence, and in PKCι depletion-induced senescence. In addition, senescent glioblastoma cells are polyploid, Ki-67 negative and arrest at the G1/S checkpoint, as determined by expression of cell cycle regulatory proteins. These markers are all consistent with cells that have undergone mitotic slippage. Failure of the spindle assembly checkpoint to function properly can lead to mitotic slippage, resulting in the premature exit of mitotic cells into the G1 phase of the cell cycle. Although in G1, these cells have the replicated DNA and centrosomal phenotype of a cell that has entered mitosis and failed to divide. Overall, we demonstrate that PKCι depletion initiates mitotic slippage-induced senescence in glioblastoma cells. To our knowledge, this is the first evidence of markers of mitotic slippage directly in senescent cells by co-staining for senescence-associated β-galactosidase and immunofluorescence markers in the same cell population. We suggest that markers of mitotic slippage be assessed in future studies of senescence to determine the extent of mitotic slippage in the induction of cellular senescence.
Collapse
Affiliation(s)
- Ian J Restall
- a Centre for Cancer Therapeutics; Ottawa Hospital Research Institute ; Ottawa , ON Canada.,b Department of Biochemistry ; Microbiology and Immunology; University of Ottawa ; Ottawa , ON Canada
| | - Doris A E Parolin
- a Centre for Cancer Therapeutics; Ottawa Hospital Research Institute ; Ottawa , ON Canada
| | - Manijeh Daneshmand
- a Centre for Cancer Therapeutics; Ottawa Hospital Research Institute ; Ottawa , ON Canada
| | - Jennifer E L Hanson
- a Centre for Cancer Therapeutics; Ottawa Hospital Research Institute ; Ottawa , ON Canada
| | - Manon A Simard
- a Centre for Cancer Therapeutics; Ottawa Hospital Research Institute ; Ottawa , ON Canada.,b Department of Biochemistry ; Microbiology and Immunology; University of Ottawa ; Ottawa , ON Canada
| | - Megan E Fitzpatrick
- a Centre for Cancer Therapeutics; Ottawa Hospital Research Institute ; Ottawa , ON Canada.,b Department of Biochemistry ; Microbiology and Immunology; University of Ottawa ; Ottawa , ON Canada
| | - Ritesh Kumar
- a Centre for Cancer Therapeutics; Ottawa Hospital Research Institute ; Ottawa , ON Canada.,b Department of Biochemistry ; Microbiology and Immunology; University of Ottawa ; Ottawa , ON Canada
| | - Sylvie J Lavictoire
- a Centre for Cancer Therapeutics; Ottawa Hospital Research Institute ; Ottawa , ON Canada
| | - Ian A J Lorimer
- a Centre for Cancer Therapeutics; Ottawa Hospital Research Institute ; Ottawa , ON Canada.,b Department of Biochemistry ; Microbiology and Immunology; University of Ottawa ; Ottawa , ON Canada.,c Department of Medicine ; University of Ottawa ; Ottawa , ON Canada
| |
Collapse
|
110
|
Ibrahim B. In silico spatial simulations reveal that MCC formation and excess BubR1 are required for tight inhibition of the anaphase-promoting complex. MOLECULAR BIOSYSTEMS 2016; 11:2867-77. [PMID: 26256776 DOI: 10.1039/c5mb00395d] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In response to the activation of the mitotic spindle assembly checkpoint (SAC), distinct inhibitory pathways control the activity of the anaphase-promoting complex (APC/C). It remains unclear whether the different regulatory mechanisms function in separate pathways or as part of an integrated signalling system. Here, five variant models of APC/C regulation were constructed and analysed. The simulations showed that all variant models were able to reproduce the wild type behaviour of the APC. However, only one model, which included both the mitotic checkpoint complex (MCC) as well as BubR1 as direct inhibitors of the APC/C, was able to reproduce both wild and mutant type behaviour of APC/C regulation. Interestingly, in this model, the MCC as well as the BubR1 binding rate to the APC/C was comparable to the known Cdc20-Mad2 binding rate and could not be made higher. Mad2 active transport towards the spindle mid-zone accelerated the inhibition speed of the APC/C but not its concentration level. The presented study highlights the principle that a systems biology approach is critical for the SAC mechanism and could also be used for predicting hypotheses to design future experiments. The presented work has successfully distinguished between five potent inhibitors of the APC/C using a systems biology approach. Here, the favoured model contains both BubR1 and MCC as direct inhibitors of the APC/C.
Collapse
Affiliation(s)
- Bashar Ibrahim
- Bio System Analysis Group, Friedrich-Schiller-University Jena, and Jena Centre for Bioinformatics (JCB), 07743 Jena, Germany.
| |
Collapse
|
111
|
Zhou Z, He M, Shah AA, Wan Y. Insights into APC/C: from cellular function to diseases and therapeutics. Cell Div 2016; 11:9. [PMID: 27418942 PMCID: PMC4944252 DOI: 10.1186/s13008-016-0021-6] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/27/2016] [Indexed: 02/07/2023] Open
Abstract
Anaphase-promoting complex/cyclosome (APC/C) is a multifunctional ubiquitin-protein ligase that targets different substrates for ubiquitylation and therefore regulates a variety of cellular processes such as cell division, differentiation, genome stability, energy metabolism, cell death, autophagy as well as carcinogenesis. Activity of APC/C is principally governed by two WD-40 domain proteins, Cdc20 and Cdh1, in and beyond cell cycle. In the past decade, the results based on numerous biochemical, 3D structural, mouse genetic and small molecule inhibitor studies have largely attracted our attention into the emerging role of APC/C and its regulation in biological function, human diseases and potential therapeutics. This review will aim to summarize some recently reported insights into APC/C in regulating cellular function, connection of its dysfunction with human diseases and its implication of therapeutics.
Collapse
Affiliation(s)
- Zhuan Zhou
- Department of Cell Biology, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Hillman Cancer Center, HCC2.6c, Pittsburgh, PA 15213 USA
| | - Mingjing He
- Department of Cell Biology, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Hillman Cancer Center, HCC2.6c, Pittsburgh, PA 15213 USA ; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan People's Republic of China
| | - Anil A Shah
- Department of Cell Biology, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Hillman Cancer Center, HCC2.6c, Pittsburgh, PA 15213 USA
| | - Yong Wan
- Department of Cell Biology, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Hillman Cancer Center, HCC2.6c, Pittsburgh, PA 15213 USA
| |
Collapse
|
112
|
Zhang X, Ling Y, Guo Y, Bai Y, Shi X, Gong F, Tan P, Zhang Y, Wei C, He X, Ramirez A, Liu X, Cao C, Zhong H, Xu Q, Ma RZ. Mps1 kinase regulates tumor cell viability via its novel role in mitochondria. Cell Death Dis 2016; 7:e2292. [PMID: 27383047 PMCID: PMC4973343 DOI: 10.1038/cddis.2016.193] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 06/01/2016] [Accepted: 06/03/2016] [Indexed: 12/22/2022]
Abstract
Targeting mitotic kinase monopolar spindle 1 (Mps1) for tumor therapy has been investigated for many years. Although it was suggested that Mps1 regulates cell viability through its role in spindle assembly checkpoint (SAC), the underlying mechanism remains less defined. In an endeavor to reveal the role of high levels of mitotic kinase Mps1 in the development of colon cancer, we unexpectedly found the amount of Mps1 required for cell survival far exceeds that of maintaining SAC in aneuploid cell lines. This suggests that other functions of Mps1 besides SAC are also employed to maintain cell viability. Mps1 regulates cell viability independent of its role in cytokinesis as the genetic depletion of Mps1 spanning from metaphase to cytokinesis affects neither cytokinesis nor cell viability. Furthermore, we developed a single-cycle inhibition strategy that allows disruption of Mps1 function only in mitosis. Using this strategy, we found the functions of Mps1 in mitosis are vital for cell viability as short-term treatment of mitotic colon cancer cell lines with Mps1 inhibitors is sufficient to cause cell death. Interestingly, Mps1 inhibitors synergize with microtubule depolymerizing drug in promoting polyploidization but not in tumor cell growth inhibition. Finally, we found that Mps1 can be recruited to mitochondria by binding to voltage-dependent anion channel 1 (VDAC1) via its C-terminal fragment. This interaction is essential for cell viability as Mps1 mutant defective for interaction fails to main cell viability, causing the release of cytochrome c. Meanwhile, deprivation of VDAC1 can make tumor cells refractory to loss of Mps1-induced cell death. Collectively, we conclude that inhibition of the novel mitochondrial function Mps1 is sufficient to kill tumor cells.
Collapse
Affiliation(s)
- X Zhang
- State Key Laboratory for Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Y Ling
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Y Guo
- State Key Laboratory for Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.,Graduate School, University of the Chinese Academy of Sciences, Beijing 100149, China
| | - Y Bai
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - X Shi
- State Key Laboratory for Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.,Graduate School, University of the Chinese Academy of Sciences, Beijing 100149, China
| | - F Gong
- State Key Laboratory for Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.,Graduate School, University of the Chinese Academy of Sciences, Beijing 100149, China
| | - P Tan
- State Key Laboratory for Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Y Zhang
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - C Wei
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - X He
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - A Ramirez
- University of Colorado at Boulder, Boulder, CO 80302, USA
| | - X Liu
- University of Colorado at Boulder, Boulder, CO 80302, USA
| | - C Cao
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - H Zhong
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Q Xu
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - R Z Ma
- State Key Laboratory for Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.,Graduate School, University of the Chinese Academy of Sciences, Beijing 100149, China
| |
Collapse
|
113
|
López-Saavedra A, Ramírez-Otero M, Díaz-Chávez J, Cáceres-Gutiérrez R, Justo-Garrido M, Andonegui MA, Mendoza J, Downie-Ruíz Á, Cortés-González C, Reynoso N, Castro-Hernández C, Domínguez-Gómez G, Santibáñez M, Fabián-Morales E, Pruefer F, Luna-Maldonado F, González-Barrios R, Herrera LA. MAD2γ, a novel MAD2 isoform, reduces mitotic arrest and is associated with resistance in testicular germ cell tumors. Cell Cycle 2016; 15:2066-76. [PMID: 27315568 PMCID: PMC4968973 DOI: 10.1080/15384101.2016.1198863] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Background: Prolonged mitotic arrest in response to anti-cancer chemotherapeutics, such as DNA-damaging agents, induces apoptosis, mitotic catastrophe, and senescence. Disruptions in mitotic checkpoints contribute resistance to DNA-damaging agents in cancer. MAD2 has been associated with checkpoint failure and chemotherapy response. In this study, a novel splice variant of MAD2, designated MAD2γ, was identified, and its association with the DNA damage response was investigated. Methods: Endogenous expression of MAD2γ and full-length MAD2 (MAD2α) was measured using RT-PCR in cancer cell lines, normal foreskin fibroblasts, and tumor samples collected from patients with testicular germ cell tumors (TGCTs). A plasmid expressing MAD2γ was transfected into HCT116 cells, and its intracellular localization and checkpoint function were evaluated according to immunofluorescence and mitotic index. Results: MAD2γ was expressed in several cancer cell lines and non-cancerous fibroblasts. Ectopically expressed MAD2γ localized to the nucleus and reduced the mitotic index, suggesting checkpoint impairment. In patients with TGCTs, the overexpression of endogenous MAD2γ, but not MAD2α, was associated with resistance to cisplatin-based chemotherapy. Likewise, cisplatin induced the overexpression of endogenous MAD2γ, but not MAD2α, in HCT116 cells. Conclusions: Overexpression of MAD2γ may play a role in checkpoint disruption and is associated with resistance to cisplatin-based chemotherapy in TGCTs.
Collapse
Affiliation(s)
- Alejandro López-Saavedra
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Miguel Ramírez-Otero
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - José Díaz-Chávez
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Rodrigo Cáceres-Gutiérrez
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Monserrat Justo-Garrido
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Marco A Andonegui
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Julia Mendoza
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Ángela Downie-Ruíz
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Carlo Cortés-González
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Nancy Reynoso
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Clementina Castro-Hernández
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Guadalupe Domínguez-Gómez
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Miguel Santibáñez
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Eunice Fabián-Morales
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Franz Pruefer
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Fernando Luna-Maldonado
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Rodrigo González-Barrios
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| | - Luis A Herrera
- a Unidad de Investigación Biomédica en Cáncer , Instituto Nacional de Cancerología (INCan) - Instituto de Investigaciones Biomédicas - Universidad Nacional Autónoma de México (UNAM) , Del. Tlalpan , Mexico D.F
| |
Collapse
|
114
|
Aurora kinase-induced phosphorylation excludes transcription factor RUNX from the chromatin to facilitate proper mitotic progression. Proc Natl Acad Sci U S A 2016; 113:6490-5. [PMID: 27217562 DOI: 10.1073/pnas.1523157113] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The Runt-related transcription factors (RUNX) are master regulators of development and major players in tumorigenesis. Interestingly, unlike most transcription factors, RUNX proteins are detected on the mitotic chromatin and apparatus, suggesting that they are functionally active in mitosis. Here, we identify key sites of RUNX phosphorylation in mitosis. We show that the phosphorylation of threonine 173 (T173) residue within the Runt domain of RUNX3 disrupts RUNX DNA binding activity during mitotic entry to facilitate the recruitment of RUNX proteins to mitotic structures. Moreover, knockdown of RUNX3 delays mitotic entry. RUNX3 phosphorylation is therefore a regulatory mechanism for mitotic entry. Cancer-associated mutations of RUNX3 T173 and its equivalent in RUNX1 further corroborate the role of RUNX phosphorylation in regulating proper mitotic progression and genomic integrity.
Collapse
|
115
|
Abstract
The terms 'haploid' and 'diploid' that describe single (n) and double (2n) chromosome sets in cells were coined by the Polish-German botanist Eduard Strasburger and originate from the Greek terms haplóos meaning 'single' and diplóos meaning 'double'. The term 'ploidy' was subsequently derived to describe the total chromosome content of cells. Consequently, the term 'euploid' refers to a chromosome complement that is an exact multiple of the haploid number. Therefore, haploids and diploids are both cases of normal euploidy. Euploid types that have more than two sets of chromosomes are 'polyploid' such as 'triploid' (3n), 'tetraploid' (4n), 'pentaploid' (5n), and so forth. There are various natural euploid states with some organisms existing as haploids (fungi), diploids (most mammals), and polyploids (plants).
Collapse
Affiliation(s)
- Bernardo Orr
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Norris Cotton Cancer Center, Lebanon, NH, USA
| | - Kristina M Godek
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Norris Cotton Cancer Center, Lebanon, NH, USA
| | - Duane Compton
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Norris Cotton Cancer Center, Lebanon, NH, USA.
| |
Collapse
|
116
|
Lee JK, Choi YL, Kwon M, Park PJ. Mechanisms and Consequences of Cancer Genome Instability: Lessons from Genome Sequencing Studies. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 11:283-312. [PMID: 26907526 DOI: 10.1146/annurev-pathol-012615-044446] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During tumor evolution, cancer cells can accumulate numerous genetic alterations, ranging from single nucleotide mutations to whole-chromosomal changes. Although a great deal of progress has been made in the past decades in characterizing genomic alterations, recent cancer genome sequencing studies have provided a wealth of information on the detailed molecular profiles of such alterations in various types of cancers. Here, we review our current understanding of the mechanisms and consequences of cancer genome instability, focusing on the findings uncovered through analysis of exome and whole-genome sequencing data. These analyses have shown that most cancers have evidence of genome instability, and the degree of instability is variable within and between cancer types. Importantly, we describe some recent evidence supporting the idea that chromosomal instability could be a major driving force in tumorigenesis and cancer evolution, actively shaping the genomes of cancer cells to maximize their survival advantage.
Collapse
Affiliation(s)
- June-Koo Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea;
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute of Health Sciences and Technology (SAIHST), Sungkyunkwan University School of Medicine, Seoul 06351, South Korea;
| | - Mijung Kwon
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115;
| | - Peter J Park
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts 02115;
| |
Collapse
|
117
|
p31comet-Induced Cell Death Is Mediated by Binding and Inactivation of Mad2. PLoS One 2015; 10:e0141523. [PMID: 26544187 PMCID: PMC4636321 DOI: 10.1371/journal.pone.0141523] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 10/10/2015] [Indexed: 11/20/2022] Open
Abstract
Mad2, a key component of the spindle checkpoint, is closely associated with chromosomal instability and poor prognosis in cancer. p31comet is a Mad2-interacting protein that serves as a spindle checkpoint silencer at mitosis. In this study, we showed that p31comet-induced apoptosis and senescence occur via counteraction of Mad2 activity. Upon retroviral transduction of p31comet, the majority of human cancer cell lines tested lost the ability to form colonies in a low-density seeding assay. Cancer cells with p31comet overexpression underwent distinct apoptosis and/or senescence, irrespective of p53 status, confirming the cytotoxicity of p31comet. Interestingly, both cytotoxic and Mad2 binding activities were eliminated upon deletion of the C-terminal 30 amino acids of p31comet. Point mutation or deletion of the region affecting Mad2 binding additionally abolished cytotoxic activity. Consistently, wild-type Mad2 interacting with p31comet, but not its non-binding mutant, inhibited cell death, indicating that the mechanism of p31comet-induced cell death involves Mad2 inactivation. Our results clearly suggest that the regions of p31comet affecting interactions with Mad2, including the C-terminus, are essential for induction of cell death. The finding that p31comet-induced cell death is mediated by interactions with Mad2 that lead to its inactivation is potentially applicable in anticancer therapy.
Collapse
|
118
|
Elliott EN, Kaestner KH. Epigenetic regulation of the intestinal epithelium. Cell Mol Life Sci 2015; 72:4139-56. [PMID: 26220502 PMCID: PMC4607638 DOI: 10.1007/s00018-015-1997-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 07/09/2015] [Accepted: 07/17/2015] [Indexed: 12/12/2022]
Abstract
The intestinal epithelium is an ideal model system for the study of normal and pathological differentiation processes. The mammalian intestinal epithelium is a single cell layer comprising proliferative crypts and differentiated villi. The crypts contain both proliferating and quiescent stem cell populations that self-renew and produce all the differentiated cell types, which are replaced every 3-5 days. The genetics of intestinal development, homeostasis, and disease are well defined, but less is known about the contribution of epigenetics in modulating these processes. Epigenetics refers to heritable phenotypic traits, including gene expression, which are independent of mutations in the DNA sequence. We have known for several decades that human colorectal cancers contain hypomethylated DNA, but the causes and consequences of this phenomenon are not fully understood. In contrast, tumor suppressor gene promoters are often hypermethylated in colorectal cancer, resulting in decreased expression of the associated gene. In this review, we describe the role that epigenetics plays in intestinal homeostasis and disease, with an emphasis on results from mouse models. We highlight the importance of producing and analyzing next-generation sequencing data detailing the epigenome from intestinal stem cell to differentiated intestinal villus cell.
Collapse
Affiliation(s)
- Ellen N Elliott
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, 12-126 Translational Research Center, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, 12-126 Translational Research Center, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA.
| |
Collapse
|
119
|
Veneziano L, Barra V, Lentini L, Spatafora S, Di Leonardo A. p14ARFPrevents Proliferation of Aneuploid Cells by Inducing p53-Dependent Apoptosis. J Cell Physiol 2015; 231:336-44. [DOI: 10.1002/jcp.24976] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 02/24/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Lorena Veneziano
- Department of Biological; Chemical and Pharmaceutical Sciences and Technologies; University of Palermo; Palermo Italy
| | - Viviana Barra
- Department of Biological; Chemical and Pharmaceutical Sciences and Technologies; University of Palermo; Palermo Italy
| | - Laura Lentini
- Department of Biological; Chemical and Pharmaceutical Sciences and Technologies; University of Palermo; Palermo Italy
| | - Sergio Spatafora
- Department of Biological; Chemical and Pharmaceutical Sciences and Technologies; University of Palermo; Palermo Italy
| | - Aldo Di Leonardo
- Department of Biological; Chemical and Pharmaceutical Sciences and Technologies; University of Palermo; Palermo Italy
- Centro di OncoBiologia Sperimentale (COBS) via San Lorenzo Colli; Palermo Italy
| |
Collapse
|
120
|
Abstract
The evolutionary conserved chromosomal passenger complex (CPC) is essential for faithful transmission of the genome during cell division. Perturbation of this complex in cultured cells gives rise to chromosome segregation errors and cytokinesis failure and as a consequence the ploidy status of the next generation of cells is changed. Aneuploidy and chromosomal instability (CIN) is observed in many human cancers, but whether this may be caused by deregulation of the CPC is unknown. In the present review, we discuss if and how a dysfunctional CPC could contribute to CIN in cancer.
Collapse
|
121
|
Fachinetti D, Han JS, McMahon MA, Ly P, Abdullah A, Wong AJ, Cleveland DW. DNA Sequence-Specific Binding of CENP-B Enhances the Fidelity of Human Centromere Function. Dev Cell 2015; 33:314-27. [PMID: 25942623 DOI: 10.1016/j.devcel.2015.03.020] [Citation(s) in RCA: 194] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 02/09/2015] [Accepted: 03/25/2015] [Indexed: 12/31/2022]
Abstract
Human centromeres are specified by a stably inherited epigenetic mark that maintains centromere position and function through a two-step mechanism relying on self-templating centromeric chromatin assembled with the histone H3 variant CENP-A, followed by CENP-A-dependent nucleation of kinetochore assembly. Nevertheless, natural human centromeres are positioned within specific megabase chromosomal regions containing α-satellite DNA repeats, which contain binding sites for the DNA sequence-specific binding protein CENP-B. We now demonstrate that CENP-B directly binds both CENP-A's amino-terminal tail and CENP-C, a key nucleator of kinetochore assembly. DNA sequence-dependent binding of CENP-B within α-satellite repeats is required to stabilize optimal centromeric levels of CENP-C. Chromosomes bearing centromeres without bound CENP-B, including the human Y chromosome, are shown to mis-segregate in cells at rates several-fold higher than chromosomes with CENP-B-containing centromeres. These data demonstrate a DNA sequence-specific enhancement by CENP-B of the fidelity of epigenetically defined human centromere function.
Collapse
Affiliation(s)
- Daniele Fachinetti
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA.
| | - Joo Seok Han
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Moira A McMahon
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Peter Ly
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Amira Abdullah
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Alex J Wong
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Don W Cleveland
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
122
|
Bargiela-Iparraguirre J, Prado-Marchal L, Pajuelo-Lozano N, Jiménez B, Perona R, Sánchez-Pérez I. Mad2 and BubR1 modulates tumourigenesis and paclitaxel response in MKN45 gastric cancer cells. Cell Cycle 2015; 13:3590-601. [PMID: 25483095 DOI: 10.4161/15384101.2014.962952] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Aneuploidy and chromosomal instability (CIN) are common features of gastric cancer (GC), but their contribution to carcinogenesis and antitumour therapy response is still poorly understood. Failures in the mitotic checkpoint induced by changes in expression levels of the spindle assembly checkpoint (SAC) proteins cause the missegregation of chromosomes in mitosis as well as aneuploidy. To evaluate the possible contribution of SAC to GC, we analyzed the expression levels of proteins of the mitotic checkpoint complex in a cohort of GC cell lines. We found that the central SAC proteins, Mad2 and BubR1, were the more prominently expressed members in disseminated GC cell lines. Silencing of Mad2 and BubR1 in MKN45 and ST2957 cells decreased their cell proliferation, migration and invasion abilities, indicating that Mad2 and BubR1 could contribute to cellular transformation and tumor progression in GC. We next evaluated whether silencing of SAC proteins could affect the response to microtubule poisons. We discovered that paclitaxel treatment increased cell survival in MKN45 cells interfered for Mad2 or BubR1 expression. However, apoptosis (assessed by caspase-3 activation, PARP proteolysis and levels of antiapoptotic Bcl 2-family members), the DNA damage response (assessed by H2Ax phosphorylation) and exit from mitosis (assessed by Cyclin B degradation and Cdk1 regulation) were activated equally between cells, independently of Mad2 or BubR1-protein levels. In contrast, we observed that the silencing of Mad2 or BubR1 in MKN45 cells showed the induction of a senescence-like phenotype accompanied by cell enlargement, increased senescence-associated β-galactosidase activity and increased IL-6 and IL-8 expression. In addition, the senescent phenotype is highly increased after treatment with PTX, indicating that senescence could prevent tumorigenesis in GC. In conclusion, the results presented here suggest that Mad2 and BubR1 could be used as prognostic markers of tumor progression and new pharmacological targets in the treatment for GC.
Collapse
Key Words
- BMC, bleomycin
- BubR1
- BubR1, budding uninhibited by benzimidazoles 1 homolog B protein (gene BUB1B)
- CDDP, cisplatin
- CIN, chromosome instability
- DDR, DNA damage response
- Mad2
- Mad2, mitotic arrest deficient-like-1 protein (gene Mad2L1)
- Monopolar Spindle kinase, MPS1
- PTX, paclitaxel
- SAC, spindle assembly checkpoint
- SASP, senescence associate secretory phenotype
- apoptosis
- gastric cancer
- mitosis
- paclitaxel
- senescence
- γH2AX, phosphorylated H2AX
Collapse
|
123
|
Wang Z, Katsaros D, Shen Y, Fu Y, Canuto EM, Benedetto C, Lu L, Chu WM, Risch HA, Yu H. Biological and Clinical Significance of MAD2L1 and BUB1, Genes Frequently Appearing in Expression Signatures for Breast Cancer Prognosis. PLoS One 2015; 10:e0136246. [PMID: 26287798 PMCID: PMC4546117 DOI: 10.1371/journal.pone.0136246] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 07/31/2015] [Indexed: 12/15/2022] Open
Abstract
To investigate the biologic relevance and clinical implication of genes involved in multiple gene expression signatures for breast cancer prognosis, we identified 16 published gene expression signatures, and selected two genes, MAD2L1 and BUB1. These genes appeared in 5 signatures and were involved in cell-cycle regulation. We analyzed the expression of these genes in relation to tumor features and disease outcomes. In vitro experiments were also performed in two breast cancer cell lines, MDA-MB-231 and MDA-MB-468, to assess cell proliferation, migration and invasion after knocking down the expression of these genes. High expression of these genes was found to be associated with aggressive tumors and poor disease-free survival of 203 breast cancer patients in our study, and the association with survival was confirmed in an online database consisting of 914 patients. In vitro experiments demonstrated that lowering the expression of these genes by siRNAs reduced tumor cell growth and inhibited cell migration and invasion. Our investigation suggests that MAD2L1 and BUB1 may play important roles in breast cancer progression, and measuring the expression of these genes may assist the prediction of breast cancer prognosis.
Collapse
Affiliation(s)
- Zhanwei Wang
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Dionyssios Katsaros
- Department of Surgical Sciences, Gynecologic Oncology, Azienda Ospedaliero-Universitaria Città della Salute, Turin, Italy
| | - Yi Shen
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Yuanyuan Fu
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Emilie Marion Canuto
- Department of Surgical Sciences, Gynecologic Oncology, Azienda Ospedaliero-Universitaria Città della Salute, Turin, Italy
| | - Chiara Benedetto
- Department of Surgical Sciences, Gynecologic Oncology, Azienda Ospedaliero-Universitaria Città della Salute, Turin, Italy
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Wen-Ming Chu
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Harvey A. Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Herbert Yu
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
- * E-mail:
| |
Collapse
|
124
|
Touati SA, Wassmann K. How oocytes try to get it right: spindle checkpoint control in meiosis. Chromosoma 2015; 125:321-35. [PMID: 26255654 DOI: 10.1007/s00412-015-0536-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 07/09/2015] [Accepted: 07/20/2015] [Indexed: 11/27/2022]
Abstract
The generation of a viable, diploid organism depends on the formation of haploid gametes, oocytes, and spermatocytes, with the correct number of chromosomes. Halving the genome requires the execution of two consecutive specialized cell divisions named meiosis I and II. Unfortunately, and in contrast to male meiosis, chromosome segregation in oocytes is error prone, with human oocytes being extraordinarily "meiotically challenged". Aneuploid oocytes, that are with the wrong number of chromosomes, give rise to aneuploid embryos when fertilized. In humans, most aneuploidies are lethal and result in spontaneous abortions. However, some trisomies survive to birth or even adulthood, such as the well-known trisomy 21, which gives rise to Down syndrome (Nagaoka et al. in Nat Rev Genet 13:493-504, 2012). A staggering 20-25 % of oocytes ready to be fertilized are aneuploid in humans. If this were not bad enough, there is an additional increase in meiotic missegregations as women get closer to menopause. A woman above 40 has a risk of more than 30 % of getting pregnant with a trisomic child. Worse still, in industrialized western societies, child birth is delayed, with women getting their first child later in life than ever. This trend has led to an increase of trisomic pregnancies by 70 % in the last 30 years (Nagaoka et al. in Nat Rev Genet 13:493-504, 2012; Schmidt et al. in Hum Reprod Update 18:29-43, 2012). To understand why errors occur so frequently during the meiotic divisions in oocytes, we review here the molecular mechanisms at works to control chromosome segregation during meiosis. An important mitotic control mechanism, namely the spindle assembly checkpoint or SAC, has been adapted to the special requirements of the meiotic divisions, and this review will focus on our current knowledge of SAC control in mammalian oocytes. Knowledge on how chromosome segregation is controlled in mammalian oocytes may help to identify risk factors important for questions related to human reproductive health.
Collapse
Affiliation(s)
- Sandra A Touati
- Institut de Biologie Paris Seine (IBPS), UMR7622, Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, France.,Chromosome Segregation Laboratory, Lincoln's Inn Fields Laboratory, The Francis Crick Institute, London, UK
| | - Katja Wassmann
- Institut de Biologie Paris Seine (IBPS), UMR7622, Sorbonne Universités, UPMC Univ Paris 06, Paris, France. .,CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, France.
| |
Collapse
|
125
|
Corkery DP, Holly AC, Lahsaee S, Dellaire G. Connecting the speckles: Splicing kinases and their role in tumorigenesis and treatment response. Nucleus 2015; 6:279-88. [PMID: 26098145 PMCID: PMC4615201 DOI: 10.1080/19491034.2015.1062194] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Alternative pre-mRNA splicing in higher eukaryotes enhances transcriptome complexity and proteome diversity. Its regulation is mediated by a complex RNA-protein network that is essential for the maintenance of cellular and tissue homeostasis. Disruptions to this regulatory network underlie a host of human diseases and contribute to cancer development and progression. The splicing kinases are an important family of pre-mRNA splicing regulators, , which includes the CDC-like kinases (CLKs), the SRSF protein kinases (SRPKs) and pre-mRNA splicing 4 kinase (PRP4K/PRPF4B). These splicing kinases regulate pre-mRNA splicing via phosphorylation of spliceosomal components and serine-arginine (SR) proteins, affecting both their nuclear localization within nuclear speckle domains as well as their nucleo-cytoplasmic shuttling. Here we summarize the emerging evidence that splicing kinases are dysregulated in cancer and play important roles in both tumorigenesis as well as therapeutic response to radiation and chemotherapy.
Collapse
Affiliation(s)
- Dale P Corkery
- a Department of Biochemistry & Molecular Biology ; Dalhousie University ; Halifax , Nova Scotia , Canada
| | | | | | | |
Collapse
|
126
|
Giam M, Rancati G. Aneuploidy and chromosomal instability in cancer: a jackpot to chaos. Cell Div 2015; 10:3. [PMID: 26015801 PMCID: PMC4443636 DOI: 10.1186/s13008-015-0009-7] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 05/08/2015] [Indexed: 12/12/2022] Open
Abstract
Genomic instability (GIN) is a hallmark of cancer cells that facilitates the acquisition of mutations conferring aggressive or drug-resistant phenotypes during cancer evolution. Chromosomal instability (CIN) is a form of GIN that involves frequent cytogenetic changes leading to changes in chromosome copy number (aneuploidy). While both CIN and aneuploidy are common characteristics of cancer cells, their roles in tumor initiation and progression are unclear. On the one hand, CIN and aneuploidy are known to provide genetic variation to allow cells to adapt in changing environments such as nutrient fluctuations and hypoxia. Patients with constitutive aneuploidies are more susceptible to certain types of cancers, suggesting that changes in chromosome copy number could positively contribute to cancer evolution. On the other hand, chromosomal imbalances have been observed to have detrimental effects on cellular fitness and might trigger cell cycle arrest or apoptosis. Furthermore, mouse models for CIN have led to conflicting results. Taken together these findings suggest that the relationship between CIN, aneuploidy and cancer is more complex than what was previously anticipated. Here we review what is known about this complex ménage à trois, discuss recent evidence suggesting that aneuploidy, CIN and GIN together promote a vicious cycle of genome chaos. Lastly, we propose a working hypothesis to reconcile the conflicting observations regarding the role of aneuploidy and CIN in tumorigenesis.
Collapse
Affiliation(s)
- Maybelline Giam
- Institute for Medical Biology (IMB), Agency for Science, Technology and Research (ASTAR), Singapore, 138648 Singapore
| | - Giulia Rancati
- Institute for Medical Biology (IMB), Agency for Science, Technology and Research (ASTAR), Singapore, 138648 Singapore ; School of Biological Sciences, Nanyang Technological University, Singapore, 637551 Singapore ; Department of Biochemistry, Yong Loo Lin School of Medicine, NUS, Singapore, 117597 Singapore
| |
Collapse
|
127
|
Kollu S, Abou-Khalil R, Shen C, Brack AS. The Spindle Assembly Checkpoint Safeguards Genomic Integrity of Skeletal Muscle Satellite Cells. Stem Cell Reports 2015; 4:1061-74. [PMID: 25960061 PMCID: PMC4471836 DOI: 10.1016/j.stemcr.2015.04.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 04/09/2015] [Accepted: 04/10/2015] [Indexed: 01/29/2023] Open
Abstract
To ensure accurate genomic segregation, cells evolved the spindle assembly checkpoint (SAC), whose role in adult stem cells remains unknown. Inducible perturbation of a SAC kinase, Mps1, and its downstream effector, Mad2, in skeletal muscle stem cells shows the SAC to be critical for normal muscle growth, repair, and self-renewal of the stem cell pool. SAC-deficient muscle stem cells arrest in G1 phase of the cell cycle with elevated aneuploidy, resisting differentiation even under inductive conditions. p21(CIP1) is responsible for these SAC-deficient phenotypes. Despite aneuploidy's correlation with aging, we find that aged proliferating muscle stem cells display robust SAC activity without elevated aneuploidy. Thus, muscle stem cells have a two-step mechanism to safeguard their genomic integrity. The SAC prevents chromosome missegregation and, if it fails, p21(CIP1)-dependent G1 arrest limits cellular propagation and tissue integration. These mechanisms ensure that muscle stem cells with compromised genomes do not contribute to tissue homeostasis.
Collapse
Affiliation(s)
- Swapna Kollu
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Boston, MA 02114, USA
| | - Rana Abou-Khalil
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Boston, MA 02114, USA
| | - Carl Shen
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Boston, MA 02114, USA
| | - Andrew S Brack
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
128
|
How C, Bruce J, So J, Pintilie M, Haibe-Kains B, Hui A, Clarke BA, Hedley DW, Hill RP, Milosevic M, Fyles A, Liu FF. Chromosomal instability as a prognostic marker in cervical cancer. BMC Cancer 2015; 15:361. [PMID: 25944123 PMCID: PMC4433070 DOI: 10.1186/s12885-015-1372-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/27/2015] [Indexed: 01/10/2023] Open
Abstract
Background Cervical cancer is the third most common cancer in women globally, and despite treatment, distant metastasis and nodal recurrence will still develop in approximately 30% of patients. The ability to predict which patients are likely to experience distant relapse would allow clinicians to better tailor treatment. Previous studies have investigated the role of chromosomal instability (CIN) in cancer, which can promote tumour initiation and growth; a hallmark of human malignancies. In this study, we sought to examine the published CIN70 gene signature in a cohort of cervical cancer patients treated at the Princess Margaret (PM) Cancer Centre and an independent cohort of The Cancer Genome Atlas (TCGA) cervical cancer patients, to determine if this CIN signature associated with patient outcome. Methods Cervical cancer samples were collected from 79 patients, treated between 2000–2007 at the PM, prior to undergoing curative chemo-radiation. Total RNA was extracted from each patient sample and analyzed using the GeneChip Human Genome U133 Plus 2.0 array (Affymetrix). Results High CIN70 scores were significantly related to increased chromosomal alterations in TCGA cervical cancer patients, including a higher percentage of genome altered and a higher number of copy number alterations. In addition, this same CIN70 signature was shown to be predictive of para-aortic nodal relapse in the PM Cancer Centre cohort. Conclusions These findings demonstrate that chromosomal instability plays an important role in cervical cancer, and is significantly associated with patient outcome. For the first time, this CIN70 gene signature provided prognostic value for patients with cervical cancer.
Collapse
Affiliation(s)
- Christine How
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| | - Jeff Bruce
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| | - Jonathan So
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| | - Melania Pintilie
- Division of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Medical Biophysics Department, University of Toronto, Toronto, ON, Canada.
| | - Angela Hui
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| | - Blaise A Clarke
- Department of Pathology, University Health Network, Toronto, ON, Canada.
| | - David W Hedley
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Division of Medical Oncology, Princess Margaret Cancer Centre, Toronto, ON, Canada.
| | - Richard P Hill
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| | - Michael Milosevic
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| | - Anthony Fyles
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| | - Fei-Fei Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
129
|
Matrka MC, Hennigan RF, Kappes F, DeLay ML, Lambert PF, Aronow BJ, Wells SI. DEK over-expression promotes mitotic defects and micronucleus formation. Cell Cycle 2015; 14:3939-53. [PMID: 25945971 PMCID: PMC4825741 DOI: 10.1080/15384101.2015.1044177] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/18/2015] [Indexed: 10/23/2022] Open
Abstract
The DEK gene encodes a nuclear protein that binds chromatin and is involved in various fundamental nuclear processes including transcription, RNA splicing, DNA replication and DNA repair. Several cancer types characteristically over-express DEK at the earliest stages of transformation. In order to explore relevant mechanisms whereby DEK supports oncogenicity, we utilized cancer databases to identify gene transcripts whose expression patterns are tightly correlated with that of DEK. We identified an enrichment of genes involved in mitosis and thus investigated the regulation and possible function of DEK in cell division. Immunofluorescence analyses revealed that DEK dissociates from DNA in early prophase and re-associates with DNA during telophase in human keratinocytes. Mitotic cell populations displayed a sharp reduction in DEK protein levels compared to the corresponding interphase population, suggesting DEK may be degraded or otherwise removed from the cell prior to mitosis. Interestingly, DEK overexpression stimulated its own aberrant association with chromatin throughout mitosis. Furthermore, DEK co-localized with anaphase bridges, chromosome fragments, and micronuclei, suggesting a specific association with mitotically defective chromosomes. We found that DEK over-expression in both non-transformed and transformed cells is sufficient to stimulate micronucleus formation. These data support a model wherein normal chromosomal clearance of DEK is required for maintenance of high fidelity cell division and chromosomal integrity. Therefore, the overexpression of DEK and its incomplete removal from mitotic chromosomes promotes genomic instability through the generation of genetically abnormal daughter cells. Consequently, DEK over-expression may be involved in the initial steps of developing oncogenic mutations in cells leading to cancer initiation.
Collapse
Affiliation(s)
- Marie C Matrka
- Cancer and Blood Diseases Institute; Cincinnati Children's Hospital Medical Center and University of Cincinnati; Cincinnati, OH USA
| | - Robert F Hennigan
- Cancer and Blood Diseases Institute; Cincinnati Children's Hospital Medical Center and University of Cincinnati; Cincinnati, OH USA
| | - Ferdinand Kappes
- Department of Biological Sciences; Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu Province, China
- Institute of Biochemistry and Molecular Biology; Medical School; RWTH Aachen University; Aachen, Germany
| | - Monica L DeLay
- Division of Rheumatology; Cincinnati Children's Hospital Medical Center; Cincinnati, OH USA
| | - Paul F Lambert
- McArdle Laboratory for Cancer Research; University of Wisconsin-Madison School of Medicine and Public Health; Madison, WI USA
| | - Bruce J Aronow
- Biomedical Informatics; Cincinnati Children's Hospital Medical Center; Cincinnati, OH USA
| | - Susanne I Wells
- Cancer and Blood Diseases Institute; Cincinnati Children's Hospital Medical Center and University of Cincinnati; Cincinnati, OH USA
| |
Collapse
|
130
|
Giovinazzi S, Sirleto P, Aksenova V, Morozov VM, Zori R, Reinhold WC, Ishov AM. Usp7 protects genomic stability by regulating Bub3. Oncotarget 2015; 5:3728-42. [PMID: 25003721 PMCID: PMC4116516 DOI: 10.18632/oncotarget.1989] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
USP7 (Ubiquitin Specific processing Protease-7) is a deubiquitinase which, over the past decade emerged as a critical regulator of cellular processes. Deregulation of USP7 activity has been linked to cancer, making USP7 inhibition an appealing anti-cancer strategy. The identification of novel USP7 substrates and additional USP7-dependent cellular activities will broaden our knowledge towards potential clinical application of USP7 inhibitors. Results presented in this study uncover a novel and pivotal function of USP7 in the maintenance of genomic stability. Upon USP7 depletion we observed prolonged mitosis and mitotic abnormalities including micronuclei accumulation, lagging chromosomes and karyotype instability. Inhibition of USP7 with small molecule inhibitors stabilizes cyclin B and causes mitotic abnormalities. Our results suggest that these USP7-dependent effects are mediated by decreased levels of spindle assembly checkpoint (SAC) component Bub3, which we characterized as an interacting partner and substrate of USP7. In silico analysis across the NCI-60 panels of cell lines supports our results where lower levels of USP7 strongly correlate with genomic instability. In conclusion, we identified a novel role of USP7 as regulator of the SAC component Bub3 and genomic stability.
Collapse
Affiliation(s)
- Serena Giovinazzi
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL; University of Florida Health Cancer Center, Gainesville, FL
| | | | | | | | | | | | - Alexander M Ishov
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL; University of Florida Health Cancer Center, Gainesville, FL
| |
Collapse
|
131
|
Lawrence KS, Chau T, Engebrecht J. DNA damage response and spindle assembly checkpoint function throughout the cell cycle to ensure genomic integrity. PLoS Genet 2015; 11:e1005150. [PMID: 25898113 PMCID: PMC4405263 DOI: 10.1371/journal.pgen.1005150] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 03/17/2015] [Indexed: 11/21/2022] Open
Abstract
Errors in replication or segregation lead to DNA damage, mutations, and aneuploidies. Consequently, cells monitor these events and delay progression through the cell cycle so repair precedes division. The DNA damage response (DDR), which monitors DNA integrity, and the spindle assembly checkpoint (SAC), which responds to defects in spindle attachment/tension during metaphase of mitosis and meiosis, are critical for preventing genome instability. Here we show that the DDR and SAC function together throughout the cell cycle to ensure genome integrity in C. elegans germ cells. Metaphase defects result in enrichment of SAC and DDR components to chromatin, and both SAC and DDR are required for metaphase delays. During persistent metaphase arrest following establishment of bi-oriented chromosomes, stability of the metaphase plate is compromised in the absence of DDR kinases ATR or CHK1 or SAC components, MAD1/MAD2, suggesting SAC functions in metaphase beyond its interactions with APC activator CDC20. In response to DNA damage, MAD2 and the histone variant CENPA become enriched at the nuclear periphery in a DDR-dependent manner. Further, depletion of either MAD1 or CENPA results in loss of peripherally associated damaged DNA. In contrast to a SAC-insensitive CDC20 mutant, germ cells deficient for SAC or CENPA cannot efficiently repair DNA damage, suggesting that SAC mediates DNA repair through CENPA interactions with the nuclear periphery. We also show that replication perturbations result in relocalization of MAD1/MAD2 in human cells, suggesting that the role of SAC in DNA repair is conserved. Checkpoints are surveillance pathways that monitor and correct cellular errors to ensure that the genome is transmitted intact through cell division; defects in checkpoints lead to human disease such as cancer. Two major checkpoint pathways that have been extensively studied are the DNA damage response and the spindle assembly checkpoint. As their names imply, they have been thought to monitor distinct chromosomal events during the cell cycle. Here, we used C. elegans proliferating germ cells and human cells to investigate the role of these checkpoints when either DNA is damaged or the spindle is perturbed. We discovered that these checkpoints function together in response to these different perturbations to ensure genome integrity. Our studies have important implications for cancer treatments, as many cancer chemotherapies target one of these checkpoint pathways without consideration for the effect on the other pathway.
Collapse
Affiliation(s)
- Katherine S. Lawrence
- Department of Molecular and Cellular Biology; Biochemistry, Molecular Cellular and Developmental Biology Graduate Group, University of California, Davis, Davis, California, United States of America
| | - Thinh Chau
- Department of Molecular and Cellular Biology; Biochemistry, Molecular Cellular and Developmental Biology Graduate Group, University of California, Davis, Davis, California, United States of America
| | - JoAnne Engebrecht
- Department of Molecular and Cellular Biology; Biochemistry, Molecular Cellular and Developmental Biology Graduate Group, University of California, Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
132
|
Ibrahim B. Systems Biology Modeling of Five Pathways for Regulation and Potent Inhibition of the Anaphase-Promoting Complex (APC/C): Pivotal Roles for MCC and BubR1. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2015; 19:294-305. [PMID: 25871779 DOI: 10.1089/omi.2015.0027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Correct DNA segregation is a fundamental process that ensures the precise and reliable inheritance of genomic information for the propagation of cell life. Eukaryotic cells have evolved a conserved surveillance control mechanism for DNA segregation named the Spindle Assembly Checkpoint (SAC).The SAC ensures that the sister chromatids of the duplicated genome are not separated and distributed to the spindle poles before all chromosomes have been properly linked to the microtubules of the mitotic spindle. Biochemically, the SAC delays cell cycle progression by preventing activation of the anaphase-promoting complex (APC/C) or cyclosome whose activation by Cdc20 is required for sister-chromatid separation; this marks the transition into anaphase. In response to activation of the checkpoint, various species control the activity of both APC/C and Cdc20. However, the underlying regulatory pathways remain largely elusive. In this study, five possible model variants of APC/C regulation were constructed, namely BubR1, Mad2, MCC, MCF2, and an all-pathways model variant. These models were validated with experimental data from the literature. A wide range of parameter values has been tested to find the critical values of the APC/C binding rate. The results show that all variants are able to capture the wild-type behavior of the APC/C. However, only one model variant, which included both MCC as well as BubR1 as potent inhibitors of the APC/C, was able to reproduce both wild-type and mutant type behavior of APC/C regulation. In conclusion, the presented work informs the regulation of fundamental processes such as SAC and APC/C in cell biology and has successfully distinguished between five competing dynamical models using a systems biology approach. The results attest that systems-level approaches are vital for molecular and cell biology.
Collapse
Affiliation(s)
- Bashar Ibrahim
- 1 Bio System Analysis Group, Friedrich-Schiller-University Jena , and Jena Centre for Bioinformatics (JCB), Jena, Germany
| |
Collapse
|
133
|
Ibrahim B. Spindle assembly checkpoint is sufficient for complete Cdc20 sequestering in mitotic control. Comput Struct Biotechnol J 2015; 13:320-8. [PMID: 25977749 PMCID: PMC4430708 DOI: 10.1016/j.csbj.2015.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 03/26/2015] [Accepted: 03/31/2015] [Indexed: 11/05/2022] Open
Abstract
The spindle checkpoint assembly (SAC) ensures genome fidelity by temporarily delaying anaphase onset, until all chromosomes are properly attached to the mitotic spindle. The SAC delays mitotic progression by preventing activation of the ubiquitin ligase anaphase-promoting complex (APC/C) or cyclosome; whose activation by Cdc20 is required for sister-chromatid separation marking the transition into anaphase. The mitotic checkpoint complex (MCC), which contains Cdc20 as a subunit, binds stably to the APC/C. Compelling evidence by Izawa and Pines (Nature 2014; 10.1038/nature13911) indicates that the MCC can inhibit a second Cdc20 that has already bound and activated the APC/C. Whether or not MCC per se is sufficient to fully sequester Cdc20 and inhibit APC/C remains unclear. Here, a dynamic model for SAC regulation in which the MCC binds a second Cdc20 was constructed. This model is compared to the MCC, and the MCC-and-BubR1 (dual inhibition of APC) core model variants and subsequently validated with experimental data from the literature. By using ordinary nonlinear differential equations and spatial simulations, it is shown that the SAC works sufficiently to fully sequester Cdc20 and completely inhibit APC/C activity. This study highlights the principle that a systems biology approach is vital for molecular biology and could also be used for creating hypotheses to design future experiments.
Collapse
Affiliation(s)
- Bashar Ibrahim
- Bio System Analysis Group, Friedrich-Schiller-University Jena, and Jena Centre for Bioinformatics (JCB), 07743 Jena, Germany
| |
Collapse
|
134
|
Karsli-Ceppioglu S, Ngollo M, Adjakly M, Dagdemir A, Judes G, Lebert A, Boiteux JP, Penault-LLorca F, Bignon YJ, Guy L, Bernard-Gallon D. Genome-wide DNA methylation modified by soy phytoestrogens: role for epigenetic therapeutics in prostate cancer? OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2015; 19:209-19. [PMID: 25831061 DOI: 10.1089/omi.2014.0142] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In prostate cancer, DNA methylation is significantly associated with tumor initiation, progression, and metastasis. Previous studies have suggested that soy phytoestrogens might regulate DNA methylation at individual candidate gene loci and that they play a crucial role as potential therapeutic agents for prostate cancer. The purpose of our study was to examine the modulation effects of phytoestrogens on a genome-wide scale in regards to DNA methylation in prostate cancer. Prostate cancer cell lines DU-145 and LNCaP were treated with 40 μM of genistein and 110 μM of daidzein. DNMT inhibitor 5-azacytidine (2 μM) and the methylating agent budesonide (2 μM) were used to compare their demethylation/methylation effects with phytoestrogens. The regulatory effects of phytoestrogens on DNA methylation were analyzed by using a methyl-DNA immunoprecipitation method coupled with Human DNA Methylation Microarrays (MeDIP-chip). We observed that the methylation profiles of 58 genes were altered by genistein and daidzein treatments in DU-145 and LNCaP prostate cancer cells. In addition, the methylation frequencies of the MAD1L1, TRAF7, KDM4B, and hTERT genes were remarkably modified by genistein treatment. Our results suggest that the modulation effects of phytoestrogens on DNA methylation essentially lead to inhibition of cell growth and induction of apoptosis. Genome-wide methylation profiling reported here suggests that epigenetic regulation mechanisms and, by extension, epigenetics-driven novel therapeutic candidates warrant further consideration in future "omics" studies of prostate cancer.
Collapse
Affiliation(s)
- Seher Karsli-Ceppioglu
- 1 Department of Oncogenetics, Centre Jean Perrin-CBRV , Dunant, Clermont-Ferrand, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Shi F, Zhang Q, Wang P, Sun H, Wang J, Rong X, Chen M, Ju C, Reinhard F, Chen H, Wrachtrup J, Wang J, Du J. Single-protein spin resonance spectroscopy under ambient conditions. Science 2015; 347:1135-8. [DOI: 10.1126/science.aaa2253] [Citation(s) in RCA: 217] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
136
|
Abstract
The majority of human cancer cells are highly aneuploid harboring chromosome numbers deviating from the modal number of 46. In cancer, aneuploidy is a consequence of an increased rate of whole chromosome missegregation during mitosis, a process known as chromosomal instability (CIN). In fact, CIN is a hallmark of human cancer and is thought to contribute to tumorigenesis, tumor progression, and the development of therapy resistance by providing a high genetic variability that might foster rapid adaptation processes. However, the molecular mechanisms that cause chromosome missegregation in cancer cells are still poorly understood. So far, several mechanisms underlying CIN have been proposed and some of them are indeed detectable in human cancer cells exhibiting CIN. Examples include, for instance, weakened spindle checkpoint signaling, supernumerary centrosomes, defects in chromatid cohesion, abnormal kinetochore-microtubule attachments and increased spindle microtubule dynamics. Here, the mechanisms leading to CIN in human cancer cells are summarized.
Collapse
Affiliation(s)
- Holger Bastians
- Goettingen Center for Molecular Biosciences (GZMB), University Medical Center, Institute of Molecular Oncology, Section for Cellular Oncology, Georg-August University Goettingen, Grisebachstrasse 8, 37077, Goettingen, Germany.
| |
Collapse
|
137
|
Kurkalang S, Banerjee A, Dkhar H, Nongrum HB, Ganguly B, Islam M, Rangad GM, Chatterjee A. Precocious anaphase and expression of Securin and p53 genes as candidate biomarkers for the early detection in areca nut-induced carcinogenesis. Mutagenesis 2014; 30:381-9. [DOI: 10.1093/mutage/geu083] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
138
|
Abstract
FAT10 (HLA-F-adjacent transcript 10) is a ubiquitin-like modifier that is commonly overexpressed in various tumors. It was found to play a role in mitotic regulation through its interaction with mitotic arrest-deficient 2 (MAD2). Overexpression of FAT10 promotes tumor growth and malignancy. Here, we identified the MAD2-binding interface of FAT10 to be located on its first ubiquitin-like domain whose NMR structure thus was determined. We further proceeded to demonstrate that disruption of the FAT10-MAD2 interaction through mutation of specific MAD2-binding residues did not interfere with the interaction of FAT10 with its other known interacting partners. Significantly, ablation of the FAT10-MAD2 interaction dramatically limited the promalignant capacity of FAT10, including promoting tumor growth in vivo and inducing aneuploidy, proliferation, migration, invasion, and resistance to apoptosis in vitro. Our results strongly suggest that the interaction of FAT10 with MAD2 is a key mechanism underlying the promalignant property of FAT10 and offer prospects for the development of anticancer strategies.
Collapse
|
139
|
Schmid S, Gillessen S, Binet I, Brändle M, Engeler D, Greiner J, Hader C, Heinimann K, Kloos P, Krek W, Krull I, Stoeckli SJ, Sulz MC, van Leyen K, Weber J, Rothermundt C, Hundsberger T. Management of von hippel-lindau disease: an interdisciplinary review. Oncol Res Treat 2014; 37:761-71. [PMID: 25531723 DOI: 10.1159/000369362] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 10/22/2014] [Indexed: 12/11/2022]
Abstract
Von Hippel-Lindau (VHL) disease is an autosomal dominantly inherited tumour predisposition syndrome with an incidence of 1:36,000 newborns, the estimated prevalence in Europe is about 1-9/100,000. It is associated with an increased risk of developing various benign and malignant tumours, thus affecting multiple organs at different time points in the life of a patient. Disease severity and diversity as well as age at first symptoms vary considerably, and diagnostic delay due to failure of recognition is a relevant issue. The identification of a disease-causing VHL germline mutation subsequently allows family members at risk to undergo predictive genetic testing after genetic counselling. Clinical management of patients and families should optimally be offered as an interdisciplinary approach. Prophylactic screening programs are a cornerstone of care, and have markedly improved median overall survival of affected patients. The aim of this review is to give an overview of the heterogeneous manifestations of the VHL syndrome and to highlight the diagnostic and therapeutic challenges characteristic for this orphan disease. A comprehensive update of the underlying genetic and molecular principles is additionally provided. We also describe how the St. Gallen VHL multidisciplinary group is organised as an example of interdisciplinary cooperation in a tertiary hospital in Switzerland.
Collapse
Affiliation(s)
- Sabine Schmid
- Division of Haematology and Oncology, Cantonal Hospital St. Gallen, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Chapard C, Meraldi P, Gleich T, Bachmann D, Hohl D, Huber M. TRAIP is a regulator of the spindle assembly checkpoint. J Cell Sci 2014; 127:5149-56. [PMID: 25335891 DOI: 10.1242/jcs.152579] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Accurate chromosome segregation during mitosis is temporally and spatially coordinated by fidelity-monitoring checkpoint systems. Deficiencies in these checkpoint systems can lead to chromosome segregation errors and aneuploidy, and promote tumorigenesis. Here, we report that the TRAF-interacting protein (TRAIP), a ubiquitously expressed nucleolar E3 ubiquitin ligase important for cellular proliferation, is localized close to mitotic chromosomes. Its knockdown in HeLa cells by RNA interference (RNAi) decreased the time of early mitosis progression from nuclear envelope breakdown (NEB) to anaphase onset and increased the percentages of chromosome alignment defects in metaphase and lagging chromosomes in anaphase compared with those of control cells. The decrease in progression time was corrected by the expression of wild-type but not a ubiquitin-ligase-deficient form of TRAIP. TRAIP-depleted cells bypassed taxol-induced mitotic arrest and displayed significantly reduced kinetochore levels of MAD2 (also known as MAD2L1) but not of other spindle checkpoint proteins in the presence of nocodazole. These results imply that TRAIP regulates the spindle assembly checkpoint, MAD2 abundance at kinetochores and the accurate cellular distribution of chromosomes. The TRAIP ubiquitin ligase activity is functionally required for the spindle assembly checkpoint control.
Collapse
Affiliation(s)
- Christophe Chapard
- Service of Dermatology, Lausanne University Hospital, CHUV, 1011 Lausanne, Switzerland
| | - Patrick Meraldi
- Department of Cell Physiology and Metabolism, University of Geneva, 1211 Geneva, Switzerland
| | - Tobias Gleich
- Service of Dermatology, Lausanne University Hospital, CHUV, 1011 Lausanne, Switzerland
| | - Daniel Bachmann
- Service of Dermatology, Lausanne University Hospital, CHUV, 1011 Lausanne, Switzerland
| | - Daniel Hohl
- Service of Dermatology, Lausanne University Hospital, CHUV, 1011 Lausanne, Switzerland
| | - Marcel Huber
- Service of Dermatology, Lausanne University Hospital, CHUV, 1011 Lausanne, Switzerland
| |
Collapse
|
141
|
Ye B, Li C, Yang Z, Wang Y, Hao J, Wang L, Li Y, Du Y, Hao L, Liu B, Wang S, Xia P, Huang G, Sun L, Tian Y, Fan Z. Cytosolic carboxypeptidase CCP6 is required for megakaryopoiesis by modulating Mad2 polyglutamylation. ACTA ACUST UNITED AC 2014; 211:2439-54. [PMID: 25332286 PMCID: PMC4235637 DOI: 10.1084/jem.20141123] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Ye et al. identify cytosolic carboxypeptidase CCP6 as a protein required for the regulation of bone marrow megakaryopoiesis in mice. The authors find that Mad2 (a core component of spindle checkpoint in mitosis) is a substrate of CCP6 in megakaryocytes and is polyglutamylated by proteins TTLL6 and TTLL4, subsequently affecting the activity of Aurora B kinase. Mad2 is thus additionally implicated in megakaryopoiesis regulation. Bone marrow progenitor cells develop into mature megakaryocytes (MKs) to produce platelets for hemostasis and other physiological functions. However, the molecular mechanisms underlying megakaryopoiesis are not completely defined. We show that cytosolic carboxypeptidase (CCP) 6 deficiency in mice causes enlarged spleens and increased platelet counts with underdeveloped MKs and dysfunctional platelets. The prominent phenotypes of CCP6 deficiency are different from those of CCP1-deficient mice. We found that CCP6 and tubulin tyrosine ligase-like family (TTLL) members TTLL4 and TTLL6 are highly expressed in MKs. We identify Mad2 (mitotic arrest deficient 2) as a novel substrate for CCP6 and not CCP1. Mad2 can be polyglutamylated by TTLL4 and TTLL6 to modulate the maturation of MKs. CCP6 deficiency causes hyperglutamylation of Mad2 to promote activation of Aurora B, leading to suppression of MK maturation. We reveal that Mad2 polyglutamylation plays a critical role in the regulation of megakaryopoiesis.
Collapse
Affiliation(s)
- Buqing Ye
- Key Laboratory of Infection and Immunity of CAS, Center for Laboratory Animal Research, Center for Biological Imaging, Key Laboratory of RNA Biology and Beijing Noncoding RNA Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Chong Li
- Key Laboratory of Infection and Immunity of CAS, Center for Laboratory Animal Research, Center for Biological Imaging, Key Laboratory of RNA Biology and Beijing Noncoding RNA Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhao Yang
- Key Laboratory of Infection and Immunity of CAS, Center for Laboratory Animal Research, Center for Biological Imaging, Key Laboratory of RNA Biology and Beijing Noncoding RNA Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanying Wang
- Key Laboratory of Infection and Immunity of CAS, Center for Laboratory Animal Research, Center for Biological Imaging, Key Laboratory of RNA Biology and Beijing Noncoding RNA Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Junfeng Hao
- Key Laboratory of Infection and Immunity of CAS, Center for Laboratory Animal Research, Center for Biological Imaging, Key Laboratory of RNA Biology and Beijing Noncoding RNA Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Li Wang
- Key Laboratory of Infection and Immunity of CAS, Center for Laboratory Animal Research, Center for Biological Imaging, Key Laboratory of RNA Biology and Beijing Noncoding RNA Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, China
| | - Ying Du
- Key Laboratory of Infection and Immunity of CAS, Center for Laboratory Animal Research, Center for Biological Imaging, Key Laboratory of RNA Biology and Beijing Noncoding RNA Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Lu Hao
- Key Laboratory of Infection and Immunity of CAS, Center for Laboratory Animal Research, Center for Biological Imaging, Key Laboratory of RNA Biology and Beijing Noncoding RNA Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Benyu Liu
- Key Laboratory of Infection and Immunity of CAS, Center for Laboratory Animal Research, Center for Biological Imaging, Key Laboratory of RNA Biology and Beijing Noncoding RNA Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Shuo Wang
- Key Laboratory of Infection and Immunity of CAS, Center for Laboratory Animal Research, Center for Biological Imaging, Key Laboratory of RNA Biology and Beijing Noncoding RNA Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Pengyan Xia
- Key Laboratory of Infection and Immunity of CAS, Center for Laboratory Animal Research, Center for Biological Imaging, Key Laboratory of RNA Biology and Beijing Noncoding RNA Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Guanling Huang
- Key Laboratory of Infection and Immunity of CAS, Center for Laboratory Animal Research, Center for Biological Imaging, Key Laboratory of RNA Biology and Beijing Noncoding RNA Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Lei Sun
- Key Laboratory of Infection and Immunity of CAS, Center for Laboratory Animal Research, Center for Biological Imaging, Key Laboratory of RNA Biology and Beijing Noncoding RNA Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yong Tian
- Key Laboratory of Infection and Immunity of CAS, Center for Laboratory Animal Research, Center for Biological Imaging, Key Laboratory of RNA Biology and Beijing Noncoding RNA Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zusen Fan
- Key Laboratory of Infection and Immunity of CAS, Center for Laboratory Animal Research, Center for Biological Imaging, Key Laboratory of RNA Biology and Beijing Noncoding RNA Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
142
|
Lu D, Hsiao JY, Davey NE, Van Voorhis VA, Foster SA, Tang C, Morgan DO. Multiple mechanisms determine the order of APC/C substrate degradation in mitosis. ACTA ACUST UNITED AC 2014; 207:23-39. [PMID: 25287299 PMCID: PMC4195823 DOI: 10.1083/jcb.201402041] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
To ensure proper mitotic progression, robust ordering of the destruction of APC/CCdc20 substrates is driven by the integration of molecular mechanisms ranging from phosphorylation-dependent interaction with substrates to sensing of the status of the spindle assembly checkpoint. The ubiquitin protein ligase anaphase-promoting complex or cyclosome (APC/C) controls mitosis by promoting ordered degradation of securin, cyclins, and other proteins. The mechanisms underlying the timing of APC/C substrate degradation are poorly understood. We explored these mechanisms using quantitative fluorescence microscopy of GFP-tagged APC/CCdc20 substrates in living budding yeast cells. Degradation of the S cyclin, Clb5, begins early in mitosis, followed 6 min later by the degradation of securin and Dbf4. Anaphase begins when less than half of securin is degraded. The spindle assembly checkpoint delays the onset of Clb5 degradation but does not influence securin degradation. Early Clb5 degradation depends on its interaction with the Cdk1–Cks1 complex and the presence of a Cdc20-binding “ABBA motif” in its N-terminal region. The degradation of securin and Dbf4 is delayed by Cdk1-dependent phosphorylation near their Cdc20-binding sites. Thus, a remarkably diverse array of mechanisms generates robust ordering of APC/CCdc20 substrate destruction.
Collapse
Affiliation(s)
- Dan Lu
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Jennifer Y Hsiao
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Norman E Davey
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Vanessa A Van Voorhis
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Scott A Foster
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Chao Tang
- Center for Quantitative Biology and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - David O Morgan
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| |
Collapse
|
143
|
Nascimento AV, Singh A, Bousbaa H, Ferreira D, Sarmento B, Amiji MM. Mad2 checkpoint gene silencing using epidermal growth factor receptor-targeted chitosan nanoparticles in non-small cell lung cancer model. Mol Pharm 2014; 11:3515-27. [PMID: 25256346 PMCID: PMC4186685 DOI: 10.1021/mp5002894] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
RNA
interference has emerged as a powerful strategy in cancer therapy
because it allows silencing of specific genes associated with tumor
progression and resistance. Mad2 is an essential mitotic checkpoint
component required for accurate chromosome segregation during mitosis,
and its complete abolition leads to cell death. We have developed
an epidermal growth factor receptor (EGFR)-targeted chitosan system
for silencing the Mad2 gene as a strategy to efficiently
induce cell death in EGFR overexpressing human A549 non-small cell
lung cancer cells. Control and EGFR-targeted chitosan nanoparticles
loaded with small interfering RNAs (siRNAs) against Mad2 were formulated
and characterized for size, charge, morphology, and encapsulation
efficiency. Qualitative and quantitative intracellular uptake studies
by confocal imaging and flow cytometry, respectively, showed time-dependent
enhanced and selective intracellular internalization of EGFR-targeted
nanoparticles compared to nontargeted system. Targeted nanoparticles
showed nearly complete depletion of Mad2 expression in A549 cells
contrasting with the partial depletion in the nontargeted system.
Accordingly, Mad2-silencing-induced apoptotic cell death was confirmed
by cytotoxicity assay and flow cytometry. Our results demonstrate
that EGFR-targeted chitosan loaded with Mad2 siRNAs
is a potent delivery system for selective killing of cancer cells.
Collapse
Affiliation(s)
- Ana Vanessa Nascimento
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, IINFACTS, Rua Central de Gandra 1317, 4585-116 Gandra PRD, Portugal
| | | | | | | | | | | |
Collapse
|
144
|
Varetti G, Pellman D, Gordon DJ. Aurea mediocritas: the importance of a balanced genome. Cold Spring Harb Perspect Biol 2014; 6:a015842. [PMID: 25237130 DOI: 10.1101/cshperspect.a015842] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Aneuploidy, defined as an abnormal number of chromosomes, is a hallmark of cancer. Paradoxically, aneuploidy generally has a negative impact on cell growth and fitness in nontransformed cells. In this work, we review recent progress in identifying how aneuploidy leads to genomic and chromosomal instability, how cells can adapt to the deleterious effects of aneuploidy, and how aneuploidy contributes to tumorigenesis in different genetic contexts. Finally, we also discuss how aneuploidy might be a target for anticancer therapies.
Collapse
Affiliation(s)
- Gianluca Varetti
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115 Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - David Pellman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115 Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115 Howard Hughes Medical Institute, Chevy Chase, Maryland 20815-6789
| | - David J Gordon
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115
| |
Collapse
|
145
|
Britigan EMC, Wan J, Zasadil LM, Ryan SD, Weaver BA. The ARF tumor suppressor prevents chromosomal instability and ensures mitotic checkpoint fidelity through regulation of Aurora B. Mol Biol Cell 2014; 25:2761-73. [PMID: 25057018 PMCID: PMC4161511 DOI: 10.1091/mbc.e14-05-0966] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The ARF tumor suppressor is best known for its role in stabilizing p53. This study identifies p53-independent functions of ARF in chromosome segregation and the mitotic checkpoint. Mitotic defects caused by loss of ARF are recapitulated by Aurora B overexpression and rescued by partial depletion of Aurora B. The ARF tumor suppressor is part of the CDKN2A locus and is mutated or undetectable in numerous cancers. The best-characterized role for ARF is in stabilizing p53 in response to cellular stress. However, ARF has tumor suppressive functions outside this pathway that have not been fully defined. Primary mouse embryonic fibroblasts (MEFs) lacking the ARF tumor suppressor contain abnormal numbers of chromosomes. However, no role for ARF in cell division has previously been proposed. Here we demonstrate a novel, p53-independent role for ARF in the mitotic checkpoint. Consistent with this, loss of ARF results in aneuploidy in vitro and in vivo. ARF−/− MEFs exhibit mitotic defects including misaligned and lagging chromosomes, multipolar spindles, and increased tetraploidy. ARF−/− cells exhibit overexpression of Mad2, BubR1, and Aurora B, but only overexpression of Aurora B phenocopies mitotic defects observed in ARF−/− MEFs. Restoring Aurora B to near-normal levels rescues mitotic phenotypes in cells lacking ARF. Our results define an unexpected role for ARF in chromosome segregation and mitotic checkpoint function. They further establish maintenance of chromosomal stability as one of the additional tumor-suppressive functions of ARF and offer a molecular explanation for the common up-regulation of Aurora B in human cancers.
Collapse
Affiliation(s)
- Eric M C Britigan
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53705 Molecular and Cellular Pharmacology Training Program, University of Wisconsin, Madison, WI 53705
| | - Jun Wan
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53705 Physiology Training Program, University of Wisconsin, Madison, WI 53705
| | - Lauren M Zasadil
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53705 Molecular and Cellular Pharmacology Training Program, University of Wisconsin, Madison, WI 53705
| | - Sean D Ryan
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53705
| | - Beth A Weaver
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53705 Carbone Cancer Center, University of Wisconsin, Madison, WI 53705
| |
Collapse
|
146
|
Vicente JJ, Cande WZ. Mad2, Bub3, and Mps1 regulate chromosome segregation and mitotic synchrony in Giardia intestinalis, a binucleate protist lacking an anaphase-promoting complex. Mol Biol Cell 2014; 25:2774-87. [PMID: 25057014 PMCID: PMC4161512 DOI: 10.1091/mbc.e14-05-0975] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The binucleate pathogen Giardia intestinalis is a highly divergent eukaryote with a semiopen mitosis, lacking an anaphase-promoting complex/cyclosome (APC/C) and many of the mitotic checkpoint complex (MCC) proteins. However, Giardia has some MCC components (Bub3, Mad2, and Mps1) and proteins from the cohesin system (Smc1 and Smc3). Mad2 localizes to the cytoplasm, but Bub3 and Mps1 are either located on chromosomes or in the cytoplasm, depending on the cell cycle stage. Depletion of Bub3, Mad2, or Mps1 resulted in a lowered mitotic index, errors in chromosome segregation (including lagging chromosomes), and abnormalities in spindle morphology. During interphase, MCC knockdown cells have an abnormal number of nuclei, either one nucleus usually on the left-hand side of the cell or two nuclei with one mislocalized. These results suggest that the minimal set of MCC proteins in Giardia play a major role in regulating many aspects of mitosis, including chromosome segregation, coordination of mitosis between the two nuclei, and subsequent nuclear positioning. The critical importance of MCC proteins in an organism that lacks their canonical target, the APC/C, suggests a broader role for these proteins and hints at new pathways to be discovered.
Collapse
Affiliation(s)
- Juan-Jesus Vicente
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - W Zacheus Cande
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
147
|
Ling Y, Zhang X, Bai Y, Li P, Wei C, Song T, Zheng Z, Guan K, Zhang Y, Zhang B, Liu X, Ma RZ, Cao C, Zhong H, Xu Q. Overexpression of Mps1 in colon cancer cells attenuates the spindle assembly checkpoint and increases aneuploidy. Biochem Biophys Res Commun 2014; 450:1690-5. [PMID: 25063032 DOI: 10.1016/j.bbrc.2014.07.071] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 07/15/2014] [Indexed: 12/25/2022]
Abstract
The spindle assembly checkpoint kinase Mps1 is highly expressed in several types of cancers, but its cellular involvement in tumorigenesis is less defined. Herein, we confirm that Mps1 is overexpressed in colon cancer tissues. Further, we find that forced expression of Mps1 in the colon cancer cell line SW480 enables cells to become resistant to both Mps1 inhibition-induced checkpoint depletion and cell death. Overexpression of Mps1 also increases genome instability in tumor cells owing to a weakened spindle assembly checkpoint. Collectively, our findings suggest that high levels of Mps1 contribute to tumorigenesis by attenuating the spindle assembly checkpoint.
Collapse
Affiliation(s)
- Youguo Ling
- Department of Life Science, Anhui University, Hefei, China; Beijing Institute of Biotechnology, Beijing, China
| | - Xiaojuan Zhang
- Center for Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yuanyuan Bai
- Beijing Institute of Biotechnology, Beijing, China
| | - Ping Li
- Beijing Institute of Biotechnology, Beijing, China
| | - Congwen Wei
- Beijing Institute of Biotechnology, Beijing, China
| | - Ting Song
- Beijing Institute of Biotechnology, Beijing, China
| | - Zirui Zheng
- Beijing Institute of Biotechnology, Beijing, China
| | - Kai Guan
- Beijing Institute of Biotechnology, Beijing, China
| | | | - Buchang Zhang
- Department of Life Science, Anhui University, Hefei, China
| | - Xuedong Liu
- University of Colorado at Boulder, Boulder, CO, USA
| | - Runlin Z Ma
- Center for Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Cheng Cao
- Beijing Institute of Biotechnology, Beijing, China.
| | - Hui Zhong
- Beijing Institute of Biotechnology, Beijing, China.
| | - Quanbin Xu
- Beijing Institute of Biotechnology, Beijing, China.
| |
Collapse
|
148
|
Kamthan M, Nalla VK, Ruhela D, Kamthan A, Maiti P, Datta A. Characterization of a putative spindle assembly checkpoint kinase Mps1, suggests its involvement in cell division, morphogenesis and oxidative stress tolerance in Candida albicans. PLoS One 2014; 9:e101517. [PMID: 25025778 PMCID: PMC4098995 DOI: 10.1371/journal.pone.0101517] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 06/09/2014] [Indexed: 01/14/2023] Open
Abstract
In Saccharomyces cerevisiae MPS1 is one of the major protein kinase that governs the spindle checkpoint pathway. The S. cerevisiae structural homolog of opportunistic pathogen Candida albicans CaMPS1, is indispensable for the cell viability. The essentiality of Mps1 was confirmed by Homozygote Trisome test. To determine its biological function in this pathogen conditional mutant was generated through regulatable MET3 promoter. Examination of heterozygous and conditional (+Met/Cys) mps1 mutants revealed a mitosis specific arrest phenotype, where mutants showed large buds with undivided nuclei. Flowcytometry analysis revealed abnormal ploidy levels in mps1mutant. In presence of anti-microtubule drug Nocodazole, mps1 mutant showed a dramatic loss of viability suggesting a role of Mps1 in Spindle Assembly Checkpoint (SAC) activation. These mutants were also defective in microtubule organization. Moreover, heterozygous mutant showed defective in-vitro yeast to hyphae morphological transition. Growth defect in heterozygous mutant suggest haploinsufficiency of this gene. qRT PCR analysis showed around 3 fold upregulation of MPS1 in presence of serum. This expression of MPS1 is dependent on Efg1and is independent of other hyphal regulators like Ras1 and Tpk2. Furthermore, mps1 mutants were also sensitive to oxidative stress. Heterozygous mps1 mutant did not undergo morphological transition and showed 5-Fold reduction in colony forming units in response to macrophage. Thus, the vital checkpoint kinase, Mps1 besides cell division also has a role in morphogenesis and oxidative stress tolerance, in this pathogenic fungus.
Collapse
Affiliation(s)
- Mohan Kamthan
- National Institute of Plant Genome Research, New Delhi, India
| | - Vijaya Kumar Nalla
- Department of Microbiology, Bhaskaracharya college, University of Delhi, New Delhi, India
| | - Deepa Ruhela
- National Institute of Plant Genome Research, New Delhi, India
| | - Ayushi Kamthan
- National Institute of Plant Genome Research, New Delhi, India
| | - Protiti Maiti
- National Institute of Plant Genome Research, New Delhi, India
| | - Asis Datta
- National Institute of Plant Genome Research, New Delhi, India
- * E-mail:
| |
Collapse
|
149
|
Nagai M, Ushimaru T. Cdh1 is an antagonist of the spindle assembly checkpoint. Cell Signal 2014; 26:2217-22. [PMID: 25025567 DOI: 10.1016/j.cellsig.2014.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 07/08/2014] [Indexed: 10/25/2022]
Abstract
The spindle assembly checkpoint (SAC) monitors unsatisfied connections of microtubules to kinetochores and prevents anaphase onset by inhibition of the ubiquitin ligase E3 anaphase-promoting complex or cyclosome (APC/C) in association with the activator Cdc20. Another APC/C activator, Cdh1, exists permanently throughout the cell cycle but it becomes active from telophase to G1. Here, we show that Cdh1 is partially active and mediates securin degradation even in SAC-active metaphase cells. Additionally, Cdh1 mediates Cdc20 degradation in metaphase, promoting formation of the APC/C-Cdh1. These results indicate that Cdh1 opposes the SAC and promotes anaphase transition.
Collapse
Affiliation(s)
- Masayoshi Nagai
- Department of Biological Science, Faculty of Science, Shizuoka University, Shizuoka 422-8529, Japan
| | - Takashi Ushimaru
- Department of Biological Science, Faculty of Science, Shizuoka University, Shizuoka 422-8529, Japan.
| |
Collapse
|
150
|
Schmid M, Steinlein C, Tian Q, Hanlon Newell AE, Gessler M, Olson SB, Rosenwald A, Kneitz B, Fedorov LM. Mosaic variegated aneuploidy in mouse BubR1 deficient embryos and pregnancy loss in human. Chromosome Res 2014; 22:375-92. [DOI: 10.1007/s10577-014-9432-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 06/13/2014] [Accepted: 06/17/2014] [Indexed: 12/23/2022]
|