Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Elife ; 122023 02 15.
Article in English | MEDLINE | ID: mdl-36790165

ABSTRACT

Centrosomes are orbited by centriolar satellites, dynamic multiprotein assemblies nucleated by Pericentriolar material 1 (PCM1). To study the requirement for centriolar satellites, we generated mice lacking PCM1, a crucial component of satellites. Pcm1-/- mice display partially penetrant perinatal lethality with survivors exhibiting hydrocephalus, oligospermia, and cerebellar hypoplasia, and variably expressive phenotypes such as hydronephrosis. As many of these phenotypes have been observed in human ciliopathies and satellites are implicated in cilia biology, we investigated whether cilia were affected. PCM1 was dispensable for ciliogenesis in many cell types, whereas Pcm1-/- multiciliated ependymal cells and human PCM1-/- retinal pigmented epithelial 1 (RPE1) cells showed reduced ciliogenesis. PCM1-/- RPE1 cells displayed reduced docking of the mother centriole to the ciliary vesicle and removal of CP110 and CEP97 from the distal mother centriole, indicating compromised early ciliogenesis. Similarly, Pcm1-/- ependymal cells exhibited reduced removal of CP110 from basal bodies in vivo. We propose that PCM1 and centriolar satellites facilitate efficient trafficking of proteins to and from centrioles, including the departure of CP110 and CEP97 to initiate ciliogenesis, and that the threshold to trigger ciliogenesis differs between cell types.


Subject(s)
Centrioles , Cilia , Animals , Female , Humans , Mice , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Centrioles/metabolism , Centrosome/metabolism , Cilia/metabolism , Cytoskeletal Proteins/metabolism
2.
Nat Cell Biol ; 24(4): 483-496, 2022 04.
Article in English | MEDLINE | ID: mdl-35411088

ABSTRACT

Protein degradation is critical to maintaining cellular homeostasis, and perturbation of the ubiquitin proteasome system leads to the accumulation of protein aggregates. These aggregates are either directed towards autophagy for destruction or sequestered into an inclusion, termed the aggresome, at the centrosome. Utilizing high-resolution quantitative analysis, here, we define aggresome assembly at the centrosome in human cells. Centriolar satellites are proteinaceous granules implicated in the trafficking of proteins to the centrosome. During aggresome assembly, satellites were required for the growth of the aggresomal structure from an initial ring of phosphorylated HSP27 deposited around the centrioles. The seeding of this phosphorylated HSP27 ring depended on the centrosomal proteins CP110, CEP97 and CEP290. Owing to limiting amounts of CP110, senescent cells, which are characterized by the accumulation of protein aggregates, were defective in aggresome formation. Furthermore, satellites and CP110-CEP97-CEP290 were required for the aggregation of mutant huntingtin. Together, these data reveal roles for CP110-CEP97-CEP290 and satellites in the control of cellular proteostasis and the aggregation of disease-relevant proteins.


Subject(s)
Centrioles , Protein Aggregates , Antigens, Neoplasm/metabolism , Cell Cycle Proteins/metabolism , Centrioles/metabolism , Centrosome/metabolism , Cilia/metabolism , Cytoskeletal Proteins/metabolism , HSP27 Heat-Shock Proteins/metabolism , Humans , Microtubule-Associated Proteins/metabolism
3.
Nat Commun ; 12(1): 1405, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33658502

ABSTRACT

Population scale sweeps of viral pathogens, such as SARS-CoV-2, require high intensity testing for effective management. Here, we describe "Systematic Parallel Analysis of RNA coupled to Sequencing for Covid-19 screening" (C19-SPAR-Seq), a multiplexed, scalable, readily automated platform for SARS-CoV-2 detection that is capable of analyzing tens of thousands of patient samples in a single run. To address strict requirements for control of assay parameters and output demanded by clinical diagnostics, we employ a control-based Precision-Recall and Receiver Operator Characteristics (coPR) analysis to assign run-specific quality control metrics. C19-SPAR-Seq coupled to coPR on a trial cohort of several hundred patients performs with a specificity of 100% and sensitivity of 91% on samples with low viral loads, and a sensitivity of >95% on high viral loads associated with disease onset and peak transmissibility. This study establishes the feasibility of employing C19-SPAR-Seq for the large-scale monitoring of SARS-CoV-2 and other pathogens.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , SARS-CoV-2/growth & development , SARS-CoV-2/pathogenicity , COVID-19/genetics , COVID-19/immunology , COVID-19/virology , Humans , Molecular Diagnostic Techniques , Nucleic Acid Amplification Techniques , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Viral Load
4.
Curr Opin Struct Biol ; 66: 32-40, 2021 02.
Article in English | MEDLINE | ID: mdl-33130249

ABSTRACT

The centrosome and its associated structures of the primary cilium and centriolar satellites have been established as central players in a plethora of cellular processes ranging from cell division to cellular signaling. Consequently, defects in the structure or function of these organelles are linked to a diverse range of human diseases, including cancer, microcephaly, ciliopathies, and neurodegeneration. To understand the molecular mechanisms underpinning these diseases, the biology of centrosomes, cilia, and centriolar satellites has to be elucidated. Central to solving this conundrum is the identification, localization, and functional analysis of all the proteins that reside and interact with these organelles. In this review, we discuss the technological breakthroughs that are dissecting the molecular players of these enigmatic organelles with unprecedented spatial and temporal resolution.


Subject(s)
Cilia , Neoplasms , Centrioles , Centrosome , Humans , Organelles
5.
J Cell Sci ; 133(1)2020 01 02.
Article in English | MEDLINE | ID: mdl-31896603

ABSTRACT

Centriolar satellites are non-membranous cytoplasmic granules that concentrate in the vicinity of the centrosome, the major microtubule-organizing centre (MTOC) in animal cells. Originally assigned as conduits for the transport of proteins towards the centrosome and primary cilium, the complexity of satellites is starting to become apparent. Recent studies defined the satellite proteome and interactomes, placing hundreds of proteins from diverse pathways in association with satellites. In addition, studies on cells lacking satellites have revealed that the centrosome can assemble in their absence, whereas studies on acentriolar cells have demonstrated that satellite assembly is independent from an intact MTOC. A role for satellites in ciliogenesis is well established; however, their contribution to other cellular functions is poorly understood. In this Review, we discuss the developments in our understanding of centriolar satellite assembly and function, and why satellites are rapidly becoming established as governors of multiple cellular processes. We highlight the composition and biogenesis of satellites and what is known about the regulation of these aspects. Furthermore, we discuss the evolution from thinking of satellites as mere facilitators of protein trafficking to the centrosome to thinking of them being key regulators of protein localization and cellular proteostasis for a diverse set of pathways, making them of broader interest to fields beyond those focused on centrosomes and ciliogenesis.


Subject(s)
Centrioles/metabolism , Vertebrates/metabolism , Animals , Humans , Organelle Biogenesis
6.
J Cell Sci ; 132(19)2019 10 03.
Article in English | MEDLINE | ID: mdl-31492759

ABSTRACT

Centrin 2 is a small conserved calcium-binding protein that localizes to the centriolar distal lumen in human cells. It is required for efficient primary ciliogenesis and nucleotide excision repair (NER). Centrin 2 forms part of the xeroderma pigmentosum group C protein complex. To explore how centrin 2 contributes to these distinct processes, we mutated the four calcium-binding EF-hand domains of human centrin 2. Centrin 2 in which all four EF-hands had been mutated to ablate calcium binding (4DA mutant) was capable of supporting in vitro NER and was as effective as the wild-type protein in rescuing the UV sensitivity of centrin 2-null cells. However, we found that mutation of any of the EF-hand domains impaired primary ciliogenesis in human TERT-RPE1 cells to the same extent as deletion of centrin 2. Phenotypic analysis of the 4DA mutant revealed defects in centrosome localization, centriole satellite assembly, ciliary assembly and function and in interactions with POC5 and SFI1. These observations indicate that centrin 2 requires calcium-binding capacity for its primary ciliogenesis functions, but not for NER, and suggest that these functions require centrin 2 to be capable of forming complexes with partner proteins.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Cell Cycle Proteins/metabolism , DNA Repair/physiology , Calcium-Binding Proteins/genetics , Cell Cycle Proteins/genetics , Cell Line , Centrioles/metabolism , DNA Damage/genetics , DNA Damage/physiology , DNA Repair/genetics , DNA, Complementary/metabolism , Humans , Immunoblotting , Immunoprecipitation , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
7.
EMBO J ; 38(14): e101109, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31304627

ABSTRACT

Centriolar satellites are small electron-dense granules that cluster in the vicinity of centrosomes. Satellites have been implicated in multiple critical cellular functions including centriole duplication, centrosome maturation, and ciliogenesis, but their precise composition and assembly properties have remained poorly explored. Here, we perform in vivo proximity-dependent biotin identification (BioID) on 22 human satellite proteins, to identify 2,113 high-confidence interactions among 660 unique polypeptides. Mining this network, we validate six additional satellite components. Analysis of the satellite interactome, combined with subdiffraction imaging, reveals the existence of multiple unique microscopically resolvable satellite populations that display distinct protein interaction profiles. We further show that loss of satellites in PCM1-depleted cells results in a dramatic change in the satellite interaction landscape. Finally, we demonstrate that satellite composition is largely unaffected by centriole depletion or disruption of microtubules, indicating that satellite assembly is centrosome-independent. Together, our work offers the first systematic spatial and proteomic profiling of human centriolar satellites and paves the way for future studies aimed at better understanding the biogenesis and function(s) of these enigmatic structures.


Subject(s)
Autoantigens/genetics , Cell Cycle Proteins/genetics , Centrioles/metabolism , Proteomics/methods , Autoantigens/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Gene Deletion , Humans , Microtubule-Associated Proteins/metabolism , Protein Interaction Maps , Tandem Mass Spectrometry
8.
Nat Rev Mol Cell Biol ; 18(3): 187-201, 2017 03.
Article in English | MEDLINE | ID: mdl-28174430

ABSTRACT

The mitotic spindle has a crucial role in ensuring the accurate segregation of chromosomes into the two daughter cells during cell division, which is paramount for maintaining genome integrity. It is a self-organized and dynamic macromolecular structure that is constructed from microtubules, microtubule-associated proteins and motor proteins. Thirty years of research have led to the identification of centrosome-, chromatin- and microtubule-mediated microtubule nucleation pathways that each contribute to mitotic spindle assembly. Far from being redundant pathways, data are now emerging regarding how they function together to ensure the timely completion of mitosis. We are also beginning to comprehend the multiple mechanisms by which cells regulate spindle scaling. Together, this research has increased our understanding of how cells coordinate hundreds of proteins to assemble the dynamic, precise and robust structure that is the mitotic spindle.


Subject(s)
Centrosome/metabolism , Microtubules/metabolism , Spindle Apparatus/physiology , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/genetics , Chromatin/metabolism , Humans , Kinetochores/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/genetics
9.
Mol Cell Oncol ; 3(3): e1062952, 2016 May.
Article in English | MEDLINE | ID: mdl-27314078

ABSTRACT

The mitotic spindle is the apparatus upon which chromosomes are segregated during cell division. We have discovered new roles for two members of the NIMA-related kinase (NEK) family in different molecular processes of spindle assembly. Moreover, loss of these proteins leads to segregation errors that drive cancer progression.

11.
Curr Biol ; 25(15): R656-9, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26241140

ABSTRACT

As a microtubule-organizing center, the centrosome undergoes a dramatic increase in size - via expansion of the pericentriolar material - during mitosis. Recent work reveals shared assembly properties of a protein scaffold that facilitates and supports this expansion, a process critical to spindle assembly.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Cell Cycle Proteins/genetics , Centrosome/metabolism , Drosophila/genetics , Animals
12.
J Cell Biol ; 209(3): 339-48, 2015 May 11.
Article in English | MEDLINE | ID: mdl-25963817

ABSTRACT

Nek5 is a poorly characterized member of the NIMA-related kinase family, other members of which play roles in cell cycle progression and primary cilia function. Here, we show that Nek5, similar to Nek2, localizes to the proximal ends of centrioles. Depletion of Nek5 or overexpression of kinase-inactive Nek5 caused unscheduled separation of centrosomes in interphase, a phenotype also observed upon overexpression of active Nek2. However, separated centrosomes that resulted from Nek5 depletion remained relatively close together, exhibited excess recruitment of the centrosome linker protein rootletin, and had reduced levels of Nek2. In addition, Nek5 depletion led to loss of PCM components, including γ-tubulin, pericentrin, and Cdk5Rap2, with centrosomes exhibiting reduced microtubule nucleation. Upon mitotic entry, Nek5-depleted cells inappropriately retained centrosome linker components and exhibited delayed centrosome separation and defective chromosome segregation. Hence, Nek5 is required for the loss of centrosome linker proteins and enhanced microtubule nucleation that lead to timely centrosome separation and bipolar spindle formation in mitosis.


Subject(s)
Centrosome/metabolism , Interphase/physiology , Protein Kinases/metabolism , Antigens/genetics , Antigens/metabolism , Base Sequence , Cell Cycle Proteins , Cytoskeletal Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Microtubules/genetics , Microtubules/metabolism , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Kinases/genetics , Spindle Apparatus/genetics , Spindle Apparatus/metabolism , Tubulin/genetics , Tubulin/metabolism
13.
J Cell Biol ; 208(6): 693-701, 2015 Mar 16.
Article in English | MEDLINE | ID: mdl-25753040

ABSTRACT

Primary cilia are antenna-like sensory microtubule structures that extend from basal bodies, plasma membrane-docked mother centrioles. Cellular quiescence potentiates ciliogenesis, but the regulation of basal body formation is not fully understood. We used reverse genetics to test the role of the small calcium-binding protein, centrin2, in ciliogenesis. Primary cilia arise in most cell types but have not been described in lymphocytes. We show here that serum starvation of transformed, cultured B and T cells caused primary ciliogenesis. Efficient ciliogenesis in chicken DT40 B lymphocytes required centrin2. We disrupted CETN2 in human retinal pigmented epithelial cells, and despite having intact centrioles, they were unable to make cilia upon serum starvation, showing abnormal localization of distal appendage proteins and failing to remove the ciliation inhibitor CP110. Knockdown of CP110 rescued ciliation in CETN2-deficient cells. Thus, centrin2 regulates primary ciliogenesis through controlling CP110 levels.


Subject(s)
Calcium-Binding Proteins/physiology , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Cilia/physiology , Microtubule-Associated Proteins/metabolism , Phosphoproteins/metabolism , Amino Acid Sequence , Animals , B-Lymphocytes/ultrastructure , Base Sequence , Centrioles/metabolism , Chickens , Humans , Jurkat Cells , Molecular Sequence Data , Protein Transport , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/ultrastructure
14.
Cell Cycle ; 13(17): 2773-9, 2014.
Article in English | MEDLINE | ID: mdl-25486364

ABSTRACT

Somatic cells senesce in culture after a finite number of divisions indefinitely arresting their proliferation. DNA damage and senescence increase the cellular number of centrosomes, the 2 microtubule organizing centers that ensure bipolar mitotic spindles. Centrosomes also provide the basal body from which primary cilia extend to sense and transduce various extracellular signals, notably Hedgehog. Primary cilium formation is facilitated by cellular quiescence a temporary cell cycle exit, but the impact of senescence on cilia is unknown. We found that senescent human fibroblasts have increased frequency and length of primary cilia. Levels of the negative ciliary regulator CP110 were reduced in senescent cells, as were levels of key elements of the Hedgehog pathway. Hedgehog inhibition reduced proliferation in young cells with increased cilium length accompanying cell cycle arrest suggesting a regulatory function for Hedgehog in primary ciliation. Depletion of CP110 in young cell populations increased ciliation frequencies and reduced cell proliferation. These data suggest that primary cilia are potentially novel determinants of the reduced cellular proliferation that initiates senescence.


Subject(s)
Cellular Senescence , Cilia/pathology , Fibroblasts/pathology , Cell Cycle Proteins/metabolism , Cell Proliferation , Cells, Cultured , Cilia/metabolism , Fibroblasts/metabolism , Hedgehog Proteins/metabolism , Humans , Microtubule-Associated Proteins/metabolism , Phosphoproteins/metabolism
15.
Nucleus ; 5(5): 462-73, 2014.
Article in English | MEDLINE | ID: mdl-25482198

ABSTRACT

At the onset mitosis in higher eukaryotes, the nuclear envelope (NE) undergoes dramatic deconstruction to allow separation of duplicated chromosomes. Studies have shown that during this process of nuclear envelope breakdown (NEBD), the extensive protein networks of the nuclear lamina are disassembled through phosphorylation of lamins and several inner nuclear membrane (INM) proteins. The LINC complex, composed of SUN and nesprin proteins, is involved in multiple interactions at the NE and plays vital roles in nuclear and cellular mechanics by connecting the nucleus to the cytoskeleton. Here, we show that SUN1, located in the INM, undergoes mitosis-specific phosphorylation on at least 3 sites within its nucleoplasmic N-terminus. We further identify Cdk1 as the kinase responsible for serine 48 and 333 phosphorylation, while serine 138 is phosphorylated by Plk1. In mitotic cells, SUN1 loses its interaction with N-terminal domain binding partners lamin A/C, emerin, and short nesprin-2 isoforms. Furthermore, a triple phosphomimetic SUN1 mutant displays increased solubility and reduced retention at the NE. In contrast, the central LINC complex interaction between the SUN1 C-terminus and the KASH domain of nesprin-2 is maintained during mitosis. Together, these data support a model whereby mitotic phosphorylation of SUN1 disrupts interactions with nucleoplasmic binding partners, promoting disassembly of the nuclear lamina and, potentially, its chromatin interactions. At the same time, our data add to an emerging picture that the core LINC complex plays an active role in NEBD.


Subject(s)
Lamin Type A/metabolism , Membrane Proteins/genetics , Microtubule-Associated Proteins/genetics , Mitosis/genetics , Nuclear Envelope/genetics , Nuclear Proteins/genetics , CDC2 Protein Kinase/metabolism , Cell Nucleus/genetics , Chromatin/genetics , HeLa Cells , Humans , Lamin Type A/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Lamina/genetics , Nuclear Proteins/metabolism , Phosphorylation
16.
J Cell Sci ; 127(Pt 11): 2493-506, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24695856

ABSTRACT

During mitotic entry, centrosomes separate to establish the bipolar spindle. Delays in centrosome separation can perturb chromosome segregation and promote genetic instability. However, interphase centrosomes are physically tethered by a proteinaceous linker composed of C-Nap1 (also known as CEP250) and the filamentous protein rootletin. Linker disassembly occurs at the onset of mitosis in a process known as centrosome disjunction and is triggered by the Nek2-dependent phosphorylation of C-Nap1. However, the mechanistic consequences of C-Nap1 phosphorylation are unknown. Here, we demonstrate that Nek2 phosphorylates multiple residues within the C-terminal domain of C-Nap1 and, collectively, these phosphorylation events lead to loss of oligomerization and centrosome association. Mutations in non-phosphorylatable residues that make the domain more acidic are sufficient to release C-Nap1 from the centrosome, suggesting that it is an increase in overall negative charge that is required for this process. Importantly, phosphorylation of C-Nap1 also perturbs interaction with the core centriolar protein, Cep135, and interaction of endogenous C-Nap1 and Cep135 proteins is specifically lost in mitosis. We therefore propose that multisite phosphorylation of C-Nap1 by Nek2 perturbs both oligomerization and Cep135 interaction, and this precipitates centrosome disjunction at the onset of mitosis.


Subject(s)
Autoantigens/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Centrioles/metabolism , Centrosome/physiology , Spindle Apparatus/metabolism , Autoantigens/genetics , Cell Cycle Proteins/genetics , Chromosome Segregation/genetics , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Genomic Instability , HeLa Cells , Humans , Mitosis , Mutation/genetics , NIMA-Related Kinases , Phosphorylation , Protein Binding/genetics , Protein Engineering , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics
17.
J Cell Sci ; 125(Pt 22): 5353-68, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22956538

ABSTRACT

Centrosome duplication is licensed by the disengagement, or 'uncoupling', of centrioles during late mitosis. However, arrest of cells in G2 can trigger premature centriole disengagement. Here, we show that premature disengagement results from untimely activation of the anaphase-promoting complex (APC/C), leading to securin degradation and release of active separase. Although APC/C activation during G2 arrest is dependent on polo-like kinase 1 (Plk1)-mediated degradation of the APC/C inhibitor, early mitotic inhibitor 1 (Emi1), Plk1 also has a second APC/C-independent role in promoting disengagement. Importantly, APC/C and Plk1 activity also stimulates centriole disengagement in response to hydroxyurea or DNA damage-induced cell-cycle arrest and this leads to centrosome amplification. However, the reduplication of disengaged centrioles is dependent on cyclin-dependent kinase 2 (Cdk2) activity and Cdk2 activation coincides with a subsequent inactivation of the APC/C and re-accumulation of cyclin A. Although release from these arrests leads to mitotic entry, the presence of disengaged and/or amplified centrosomes results in the formation of abnormal mitotic spindles that lead to chromosome mis-segregation. Thus, oscillation of APC/C activity during cell cycle arrest promotes both centrosome amplification and genome instability.


Subject(s)
Cell Cycle Checkpoints , Centrosome/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Cell Cycle Proteins/metabolism , Centrioles/drug effects , Centrioles/metabolism , Centrioles/radiation effects , Centrosome/drug effects , Centrosome/radiation effects , Endopeptidases/metabolism , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , HeLa Cells , Humans , Hydroxyurea/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Radiation, Ionizing , Separase , Signal Transduction/drug effects , Signal Transduction/radiation effects , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Spindle Apparatus/radiation effects , Polo-Like Kinase 1
18.
J Cell Sci ; 124(Pt 4): 600-12, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21266464

ABSTRACT

Ciliopathies are caused by mutations in genes encoding proteins required for cilia organization or function. We show through colocalization with PCM-1, that OFD1 (the product of the gene mutated in oral-facial-digital syndrome 1) as well as BBS4 and CEP290 (proteins encoded by other ciliopathy genes) are primarily components of centriolar satellites, the particles surrounding centrosomes and basal bodies. RNA interference experiments reveal that satellite integrity is mutually dependent upon each of these proteins. Upon satellite dispersal, through mitosis or forced microtubule depolymerization, OFD1 and CEP290 remain centrosomal, whereas BBS4 and PCM-1 do not. OFD1 interacts via its fifth coiled-coil motif with the N-terminal coiled-coil domain of PCM-1, which itself interacts via its C-terminal non-coiled-coil region with BBS4. OFD1 localization to satellites requires its N-terminal region, encompassing the LisH motif, whereas expression of OFD1 C-terminal constructs causes PCM-1 and CEP290 mislocalization. Moreover, in embryonic zebrafish, OFD1 and BBS4 functionally synergize, determining morphogenesis. Our observation that satellites are assembly points for several mutually dependent ciliopathy proteins provides a further possible explanation as to why the clinical spectrum of OFD1, Bardet-Biedl and Joubert syndromes overlap. Furthermore, definition of how OFD1 and PCM-1 interact helps explain why different OFD1 mutations lead to clinically variable phenotypes.


Subject(s)
Centrioles/metabolism , Orofaciodigital Syndromes/metabolism , Proteins/metabolism , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Autoantigens/genetics , Autoantigens/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Centrioles/genetics , Cytoskeletal Proteins , Humans , Microtubule-Associated Proteins , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Orofaciodigital Syndromes/embryology , Orofaciodigital Syndromes/genetics , Protein Binding , Proteins/genetics , Zebrafish
19.
Methods Mol Biol ; 545: 165-83, 2009.
Article in English | MEDLINE | ID: mdl-19475388

ABSTRACT

The formation of a bipolar spindle is essential for the equal segregation of duplicated DNA into two daughter cells during mitosis. Spindle bipolarity is largely dependent on the mitotic cell possessing two centrosomes that can each establish one spindle pole. The centrosome is also now known to regulate many other aspects of cell cycle progression, including G1/S progression, spindle orientation and symmetry, cytokinesis, and checkpoint signalling. As a result, defects in centrosome arrangement or number can lead to loss of cell polarity, defective cell division, and abnormal chromosome segregation, all events that are typical of cancer cells. Indeed, cancer cells often exhibit overduplicated centrosomes and multipolar spindles. Here, we outline a number of fluorescence imaging methodologies that can be used to study events of the centrosome duplication cycle, as well as the dynamics of individual centrosome proteins. Specifically, we discuss the generation and imaging of cell lines with fluorescently labelled centrosomes, the use of photobleaching methods to measure the dynamics of centrosome proteins, and assays for observing centrosome overduplication and centrosome separation in fixed and live cells. These experimental approaches can provide important information on the regulation of centrosomes, their role in normal cell cycle progression and how their deregulation might contribute to the deleterious phenotypes of malignant cancer cells.


Subject(s)
Cell Cycle/physiology , Centrosome/physiology , Animals , Cell Line, Tumor , Centrosome/ultrastructure , Chromosome Segregation , Cricetinae , Cricetulus , Humans , Microscopy, Fluorescence , Spindle Apparatus/physiology , Spindle Apparatus/ultrastructure
20.
Mol Cell Biol ; 29(7): 1760-73, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19139275

ABSTRACT

Cancer cells frequently exhibit overduplicated centrosomes that lead to formation of multipolar spindles, chromosome missegregation, and aneuploidy. However, the molecular events involved in centrosome overduplication remain largely unknown. Experimentally, centrosome overduplication is observed in p53-deficient cells arrested in S phase with hydroxyurea. Using this assay, we have identified distinct roles for Cdk2, microtubules, dynein, and Hsp90 in the overduplication of functional centrosomes in mammalian cells and show that Cdk2 is also required for the generation of centriolar satellites. Moreover, we demonstrate that nuclear export is required for centriolar satellite formation and centrosome overduplication, with export inhibitors causing a Cdk-dependent accumulation of nuclear centrin granules. Hence, we propose that centrosome precursors may arise in the nucleus, providing a novel mechanistic explanation for how nuclear Cdk2 can promote centrosome overduplication in the cytoplasm. Furthermore, this study defines a molecular pathway that may be targeted to prevent centrosome overduplication in S-phase-arrested cancer cells.


Subject(s)
Centrosome/metabolism , S Phase , Active Transport, Cell Nucleus , Animals , CHO Cells , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Centrioles/metabolism , Centrosome/ultrastructure , Cricetinae , Cricetulus , Cyclin-Dependent Kinase 2/metabolism , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Dyneins/metabolism , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice , Microtubules/metabolism , Microtubules/ultrastructure , Protein Binding , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...