Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Cell Res ; 1867(4): 118650, 2020 04.
Article in English | MEDLINE | ID: mdl-31953060

ABSTRACT

Transfection is a powerful tool that enables introducing foreign nucleic acids into living cells in order to study the function of a gene product. Ever since the discovery of transfection many side effects or artifacts caused by transfection reagents have been reported. Here, we show that the transfection reagent, JetPRIME alters the localization of the splicing protein SC35 widely used as a nuclear speckle marker. We demonstrate that transfection of plasmids with JetPRIME leads to enlarged SC35 speckles and SC35 cytoplasmic granules. By contrast, transfection of the same plasmid with Lipofectamine 3000 does not have any effect on SC35 localization. The formation of SC35 cytoplasmic granules by JetPRIME-mediated transfection is independent of exogenous expression by plasmid and although similar in morphology they are distinct from P-bodies and stress granules. This method of transfection affected only SC35 and phosphorylated SR proteins but not the nuclear speckles. The JetPRIME-mediated transfection also showed compromised transcription in cells with enlarged SC35 speckles. Our work indicates that the use of JetPRIME alters SC35 localization and can affect gene expression and alternative splicing. Therefore, caution should be exercised when interpreting results after the use of a transient transfection system, particularly when the subject of the study is the function of a protein in the control of gene expression or mRNA splicing.


Subject(s)
Artifacts , Serine-Arginine Splicing Factors/analysis , Transfection , Cell Line, Tumor , Cell Nucleus Structures/chemistry , Cytoplasmic Granules/chemistry , HeLa Cells , Humans , Indicators and Reagents , RNA Splicing , Transcription, Genetic
2.
Sci Transl Med ; 11(495)2019 06 05.
Article in English | MEDLINE | ID: mdl-31167929

ABSTRACT

Parkinson's disease (PD) is a neurological disorder characterized by the progressive accumulation of neuronal α-synuclein (αSyn) inclusions called Lewy bodies. It is believed that Lewy bodies spread throughout the nervous system due to the cell-to-cell propagation of αSyn via cycles of secretion and uptake. Here, we investigated the internalization and intracellular accumulation of exogenous αSyn, two key steps of Lewy body pathogenesis, amplification and spreading. We found that stable αSyn fibrils substantially accumulate in different cell lines upon internalization, whereas αSyn monomers, oligomers, and dissociable fibrils do not. Our data indicate that the uptake-mediated accumulation of αSyn in a human-derived neuroblastoma cell line triggered an adaptive response that involved proteins linked to ubiquitin ligases of the S-phase kinase-associated protein 1 (SKP1), cullin-1 (Cul1), and F-box domain-containing protein (SCF) family. We found that SKP1, Cul1, and the F-box/LRR repeat protein 5 (FBXL5) colocalized and physically interacted with internalized αSyn in cultured cells. Moreover, the SCF containing the F-box protein FBXL5 (SCFFBXL5) catalyzed αSyn ubiquitination in reconstitution experiments in vitro using recombinant proteins and in cultured cells. In the human brain, SKP1 and Cul1 were recruited into Lewy bodies from brainstem and neocortex of patients with PD and related neurological disorders. In both transgenic and nontransgenic mice, intracerebral administration of exogenous αSyn fibrils triggered a Lewy body-like pathology, which was amplified by SKP1 or FBXL5 loss of function. Our data thus indicate that SCFFXBL5 regulates αSyn in vivo and that SCF ligases may constitute targets for the treatment of PD and other α-synucleinopathies.


Subject(s)
Lewy Bodies/metabolism , Lewy Bodies/pathology , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/metabolism , Animals , Benzothiazoles/metabolism , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Humans , Mice , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/metabolism , Neurons/pathology , Parkinson Disease/metabolism , Proteome/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Ubiquitin/metabolism
3.
J Cell Sci ; 132(10)2019 05 16.
Article in English | MEDLINE | ID: mdl-31028180

ABSTRACT

During mitosis, the cell sequentially constructs two microtubule-based spindles to ensure faithful segregation of chromosomes. A bipolar spindle first pulls apart the sister chromatids, then a central spindle further separates them away. Although the assembly of the first spindle is well described, the assembly of the second remains poorly understood. We report here that the inhibition of Aurora A leads to an absence of the central spindle resulting from a lack of nucleation of microtubules in the midzone. In the absence of Aurora A, the HURP (also known as DLGAP5) and NEDD1 proteins that are involved in nucleation of microtubules fail to concentrate in the midzone. HURP is an effector of RanGTP, whereas NEDD1 serves as an anchor for the γ-tubulin ring complex (γTURC). Interestingly, Aurora A phosphorylates HURP and NEDD1 during assembly of the initial bipolar spindle. We show here that the expression of a NEDD1 isoform mimicking phosphorylation by Aurora A is sufficient to restore microtubule nucleation in the midzone under conditions of Aurora A inhibition. These results reveal a new control mechanism of microtubule nucleation by Aurora A during assembly of the central spindle.


Subject(s)
Aurora Kinase A/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Spindle Apparatus/metabolism , Anaphase/physiology , Aurora Kinase A/antagonists & inhibitors , Cell Line, Tumor , Cytokinesis/physiology , HeLa Cells , Humans , Neoplasm Proteins/metabolism , Phosphorylation , Serine/metabolism , Tubulin/metabolism
4.
J Cell Sci ; 131(7)2018 04 12.
Article in English | MEDLINE | ID: mdl-29555820

ABSTRACT

During the prometaphase stage of mitosis, the cell builds a bipolar spindle of microtubules that mechanically segregates sister chromatids between two daughter cells in anaphase. The spindle assembly checkpoint (SAC) is a quality control mechanism that monitors proper attachment of microtubules to chromosome kinetochores during prometaphase. Segregation occurs only when each chromosome is bi-oriented with each kinetochore pair attached to microtubules emanating from opposite spindle poles. Overexpression of the protein kinase Aurora A is a feature of various cancers and is thought to enable tumour cells to bypass the SAC, leading to aneuploidy. Here, we took advantage of a chemical and chemical-genetic approach to specifically inhibit Aurora A kinase activity in late prometaphase. We observed that a loss of Aurora A activity directly affects SAC function, that Aurora A is essential for maintaining the checkpoint protein Mad2 on unattached kinetochores and that inhibition of Aurora A leads to loss of the SAC, even in the presence of nocodazole or Taxol. This is a new finding that should affect the way Aurora A inhibitors are used in cancer treatments.This article has an associated First Person interview with the first authors of the paper.


Subject(s)
Aurora Kinase A/genetics , M Phase Cell Cycle Checkpoints/genetics , Mad2 Proteins/genetics , Prometaphase/genetics , Anaphase/genetics , Aurora Kinase A/antagonists & inhibitors , Azepines/pharmacology , Cell Line, Tumor , Chromatids/genetics , Chromosome Segregation/drug effects , Gene Expression Regulation, Enzymologic/genetics , Humans , Kinetochores/drug effects , Microtubules/drug effects , Mitosis/drug effects , Mitosis/genetics , Nocodazole/pharmacology , Paclitaxel/pharmacology , Prometaphase/drug effects , Pyrimidines/pharmacology , Spindle Apparatus/genetics
6.
Nat Commun ; 7: 12810, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27641145

ABSTRACT

Directed cell movement involves spatial and temporal regulation of the cortical microtubule (Mt) and actin networks to allow focal adhesions (FAs) to assemble at the cell front and disassemble at the rear. Mts are known to associate with FAs, but the mechanisms coordinating their dynamic interactions remain unknown. Here we show that the CRL3(KLHL21) E3 ubiquitin ligase promotes cell migration by controlling Mt and FA dynamics at the cell cortex. Indeed, KLHL21 localizes to FA structures preferentially at the leading edge, and in complex with Cul3, ubiquitylates EB1 within its microtubule-interacting CH-domain. Cells lacking CRL3(KLHL21) activity or expressing a non-ubiquitylatable EB1 mutant protein are unable to migrate and exhibit strong defects in FA dynamics, lamellipodia formation and cortical plasticity. Our study thus reveals an important mechanism to regulate cortical dynamics during cell migration that involves ubiquitylation of EB1 at focal adhesions.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Movement , Cytoskeletal Proteins/metabolism , Focal Adhesions/metabolism , Microtubules/metabolism , Ubiquitin-Protein Ligases/metabolism , HeLa Cells , Humans , Ubiquitination
7.
Nat Commun ; 6: 8015, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26259702

ABSTRACT

Although chromosome partitioning during mitosis is well studied, the molecular mechanisms that allow proper segregation of cytoplasmic organelles in human cells are poorly understood. Here we show that mitochondria interact with growing microtubule tips and are transported towards the daughter cell periphery at the end of mitosis. This phenomenon is promoted by the direct and cell cycle-dependent interaction of the mitochondrial protein Miro and the cytoskeletal-associated protein Cenp-F. Cenp-F is recruited to mitochondria by Miro at the time of cytokinesis and associates with microtubule growing tips. Cells devoid of Cenp-F or Miro show decreased spreading of the mitochondrial network as well as cytokinesis-specific defects in mitochondrial transport towards the cell periphery. Thus, Miro and Cenp-F promote anterograde mitochondrial movement and proper mitochondrial distribution in daughter cells.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Microfilament Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mitosis/physiology , rho GTP-Binding Proteins/metabolism , Amino Acid Sequence , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/genetics , Gene Expression Regulation/physiology , Humans , Microfilament Proteins/genetics , Microtubules/physiology , Mitochondrial Proteins/genetics , Molecular Sequence Data , Plasmids , rho GTP-Binding Proteins/genetics
8.
J Cell Biol ; 196(6): 757-74, 2012 Mar 19.
Article in English | MEDLINE | ID: mdl-22412019

ABSTRACT

In fission yeast, erroneous attachments of spindle microtubules to kinetochores are frequent in early mitosis. Most are corrected before anaphase onset by a mechanism involving the protein kinase Aurora B, which destabilizes kinetochore microtubules (ktMTs) in the absence of tension between sister chromatids. In this paper, we describe a minimal mathematical model of fission yeast chromosome segregation based on the stochastic attachment and detachment of ktMTs. The model accurately reproduces the timing of correct chromosome biorientation and segregation seen in fission yeast. Prevention of attachment defects requires both appropriate kinetochore orientation and an Aurora B-like activity. The model also reproduces abnormal chromosome segregation behavior (caused by, for example, inhibition of Aurora B). It predicts that, in metaphase, merotelic attachment is prevented by a kinetochore orientation effect and corrected by an Aurora B-like activity, whereas in anaphase, it is corrected through unbalanced forces applied to the kinetochore. These unbalanced forces are sufficient to prevent aneuploidy.


Subject(s)
Chromosome Segregation/physiology , Chromosomes, Fungal/metabolism , Kinetochores/metabolism , Microtubules/metabolism , Schizosaccharomyces/metabolism , Aurora Kinases , Chromatids/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Schizosaccharomyces/ultrastructure , Stochastic Processes
9.
Proc Natl Acad Sci U S A ; 107(30): 13330-5, 2010 Jul 27.
Article in English | MEDLINE | ID: mdl-20624975

ABSTRACT

The Dam1 complex is a kinetochore component that couples chromosomes to the dynamic ends of kinetochore microtubules (kMTs). Work in the budding yeast Saccharomyces cerevisiae has shown that the Dam1 complex forms a 16-unit ring encircling and tracking the tip of a MT in vitro, consistent with its cellular function as a coupler. Dam1 also forms smaller, nonring patches in vitro that track the dynamic ends of MTs. However, the identity of Dam1's functional form in vivo remains unknown. Here we report a comprehensive in vivo characterization of Dam1 in the fission yeast Schizosaccharomyces pombe. In addition to their dense localizations on kinetochores and spindle MTs during mitosis, we identify that Dam1 is also localized onto cytoplasmic MTs as discrete spots in interphase, providing the unique opportunity to analyze Dam1 oligomers at the single-particle resolution in live cells. Such analysis shows that each oligomer contains one to five copies of Dam1, and is able to "switch-rail" while moving along MTs, precluding the possibility of a 16-unit encircling structure. Dam1 patches track the plus ends of the shortening, but not the elongating, MTs and retard MT depolymerization. Together with Mal3, the EB1-like MT-interacting protein, cytoplasmic Dam1 plays an important role in maintaining proper cell shape. In mitosis, kinetochore-associated Dam1 appears to facilitate kMT depolymerization. Together, our findings suggest that patches, instead of rings, are the physiologically functional forms of Dam1 in pombe. Our findings help establish the benchmark parameters of the Dam1 coupler and elucidate the mechanism of its functions.


Subject(s)
Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Chromosome Segregation/physiology , Cytoplasm/metabolism , Gene Dosage , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Kinetics , Kinetochores/metabolism , Microscopy, Fluorescence , Microtubule-Associated Proteins/genetics , Mitosis/physiology , Mutation , Polymers/metabolism , Protein Binding , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics
10.
PLoS One ; 5(5): e10634, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20498706

ABSTRACT

The plus-end microtubule binding proteins (+TIPs) play an important role in the regulation of microtubule stability and cell polarity during interphase. In S. pombe, the CLIP-170 like protein Tip1, together with the kinesin Tea2, moves along the microtubules towards their plus ends. Tip1 also requires the EB1 homolog Mal3 to localize to the microtubule tips. Given the requirement for Tip1 for microtubule stability, we have investigated its role during spindle morphogenesis and chromosome movement. Loss of Tip1 affects metaphase plate formation and leads to the activation of the spindle assembly checkpoint. In the absence of Tip1 we also observed the appearance of lagging chromosomes, which do not influence the normal rate of spindle elongation. Our results suggest that S. pombe Tip1/CLIP170 is directly or indirectly required for correct chromosome poleward movement independently of Mal3/EB1.


Subject(s)
Cell Polarity , Chromosomes, Fungal/metabolism , Heat-Shock Proteins/metabolism , Intermediate Filament Proteins/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/cytology , Schizosaccharomyces/metabolism , Cell Polarity/drug effects , Gene Deletion , Kinetochores/drug effects , Kinetochores/metabolism , Metaphase/drug effects , Mitosis/drug effects , Phenotype , Protein Transport/drug effects , Schizosaccharomyces/drug effects , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Thiabendazole/pharmacology
11.
J Cell Biol ; 187(3): 399-412, 2009 Nov 02.
Article in English | MEDLINE | ID: mdl-19948483

ABSTRACT

Faithful segregation of sister chromatids requires the attachment of each kinetochore (Kt) to microtubules (MTs) that extend from opposite spindle poles. Merotelic Kt orientation is a Kt-MT misattachment in which a single Kt binds MTs from both spindle poles rather than just one. Genetic induction of merotelic Kt attachment during anaphase in fission yeast resulted in intra-Kt stretching followed by either correction or Kt disruption. Laser ablation of spindle MTs revealed that intra-Kt stretching and merotelic correction were dependent on MT forces. The presence of multiple merotelic chromosomes linearly antagonized the spindle elongation rate, and this phenomenon could be solved numerically using a simple force balance model. Based on the predictions of our mechanical model, we provide in vivo evidence that correction of merotelic attachment in anaphase is tension dependent and requires an Ase1/Prc1-dependent mechanism that prevents spindle collapse and thus asymmetric division and/or the appearance of the cut phenotype.


Subject(s)
Anaphase , Kinetochores/metabolism , Microtubule-Associated Proteins/physiology , Schizosaccharomyces pombe Proteins/physiology , Spindle Apparatus/physiology , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Microtubules/physiology , Models, Biological , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces/ultrastructure , Schizosaccharomyces pombe Proteins/metabolism , Spindle Apparatus/metabolism
12.
Mol Biol Cell ; 19(4): 1646-62, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18256284

ABSTRACT

In eukaryotic cells, proper formation of the spindle is necessary for successful cell division. We have studied chromosome recapture in the fission yeast Schizosaccharomyces pombe. We show by live cell analysis that lost kinetochores interact laterally with intranuclear microtubules (INMs) and that both microtubule depolymerization (end-on pulling) and minus-end-directed movement (microtubule sliding) contribute to chromosome retrieval to the spindle pole body (SPB). We find that the minus-end-directed motor Klp2 colocalizes with the kinetochore during its transport to the SPB and contributes to the effectiveness of retrieval by affecting both end-on pulling and lateral sliding. Furthermore, we provide in vivo evidence that Dam1, a component of the DASH complex, also colocalizes with the kinetochore during its transport and is essential for its retrieval by either of these mechanisms. Finally, we find that the position of the unattached kinetochore correlates with the size and orientation of the INMs, suggesting that chromosome recapture may not be a random process.


Subject(s)
Kinetochores/metabolism , Microtubule-Associated Proteins/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Biological Transport, Active , Dyneins/genetics , Dyneins/metabolism , Genes, Fungal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Models, Biological , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Multiprotein Complexes , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Sister Chromatid Exchange/genetics , Sister Chromatid Exchange/physiology , Spindle Apparatus/genetics , Spindle Apparatus/metabolism
13.
Biol Cell ; 99(11): 627-37, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17561805

ABSTRACT

BACKGROUND INFORMATION: In eukaryotic cells, proper formation of the spindle is necessary for successful cell division. For faithful segregation of sister chromatids, each sister kinetochore must attach to microtubules that extend to opposite poles (chromosome bi-orientation). At the metaphase-anaphase transition, cohesion between sister chromatids is removed, and each sister chromatid is pulled to opposite poles of the cell by microtubule-dependent forces. RESULTS: We have studied the role of the minus-end-directed motor protein dynein by analysing kinetochore dynamics in fission yeast cells deleted for the dynein heavy chain (Dhc1) or the light chain (Dlc1). In these mutants, we found an increased frequency of cells showing defects in chromosome segregation, which leads to the appearance of lagging chromosomes and an increased rate of chromosome loss. By following simultaneously kinetochore dynamics and localization of the checkpoint protein Mad2, we provide evidence that dynein function is not necessary for spindle-assembly checkpoint inactivation. Instead, we have demonstrated that loss of dynein function alters chromosome segregation and activates the Mad2-dependent spindle-assembly checkpoint. CONCLUSIONS: These results show an unexpected role for dynein in the control of chromosome segregation in fission yeast, most probably operating during the process of bi-orientation during early mitosis.


Subject(s)
Chromosome Segregation/physiology , Chromosomes, Fungal/metabolism , Dyneins/metabolism , Mitosis/physiology , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Anaphase/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatids/genetics , Chromatids/metabolism , Chromosomes, Fungal/genetics , Dyneins/genetics , Gene Deletion , Kinetochores/metabolism , Mad2 Proteins , Metaphase/physiology , Microtubules/genetics , Microtubules/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Schizosaccharomyces/cytology , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Spindle Apparatus/genetics , Spindle Apparatus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...