Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(1): e2213846120, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36574677

ABSTRACT

Piezo1 and 2 are evolutionarily conserved mechanosensory cation channels known to function on the cell surface by responding to external pressure and transducing a mechanically activated Ca2+ current. Here we show that both Piezo1 and 2 also exhibit concentrated intracellular localization at centrosomes. Both Piezo1 and 2 loss-of-function and Piezo1 activation by the small molecule Yoda1 result in supernumerary centrosomes, premature centriole disengagement, multi-polar spindles, and mitotic delay. By using a GFP, Calmodulin and M13 Protein fusion (GCaMP) Ca2+-sensitive reporter, we show that perturbations in Piezo modulate Ca2+ flux at centrosomes. Moreover, the inhibition of Polo-like-kinase 1 eliminates Yoda1-induced centriole disengagement. Because previous studies have implicated force generation by microtubules as essential for maintaining centrosomal integrity, we propose that mechanotransduction by Piezo maintains pericentrosomal Ca2+ within a defined range, possibly through sensing cell intrinsic forces from microtubules.


Subject(s)
Centrosome , Mechanotransduction, Cellular , Centrosome/metabolism , Centrioles , Microtubules
2.
PLoS One ; 17(6): e0268857, 2022.
Article in English | MEDLINE | ID: mdl-35704642

ABSTRACT

Factor quinolinone inhibitors (FQIs), a first-in-class set of small molecule inhibitors targeted to the transcription factor LSF (TFCP2), exhibit promising cancer chemotherapeutic properties. FQI1, the initial lead compound identified, unexpectedly induced a concentration-dependent delay in mitotic progression. Here, we show that FQI1 can rapidly and reversibly lead to mitotic arrest, even when added directly to mitotic cells, implying that FQI1-mediated mitotic defects are not transcriptionally based. Furthermore, treatment with FQIs resulted in a striking, concentration-dependent diminishment of spindle microtubules, accompanied by a concentration-dependent increase in multi-aster formation. Aberrant γ-tubulin localization was also observed. These phenotypes suggest that perturbation of spindle microtubules is the primary event leading to the mitotic delays upon FQI1 treatment. Previously, FQIs were shown to specifically inhibit not only LSF DNA-binding activity, which requires LSF oligomerization to tetramers, but also other specific LSF-protein interactions. Other transcription factors participate in mitosis through non-transcriptional means, and we recently reported that LSF directly binds α-tubulin and is present in purified cellular tubulin preparations. Consistent with a microtubule role for LSF, here we show that LSF enhanced the rate of tubulin polymerization in vitro, and FQI1 inhibited such polymerization. To probe whether the FQI1-mediated spindle abnormalities could result from inhibition of mitotic LSF-protein interactions, mass spectrometry was performed using as bait an inducible, tagged form of LSF that is biotinylated by endogenous enzymes. The global proteomics analysis yielded expected associations for a transcription factor, notably with RNA processing machinery, but also to nontranscriptional components. In particular, and consistent with spindle disruption due to FQI treatment, mitotic, FQI1-sensitive interactions were identified between the biotinylated LSF and microtubule-associated proteins that regulate spindle assembly, positioning, and dynamics, as well as centrosome-associated proteins. Probing the mitotic LSF interactome using small molecule inhibitors therefore supported a non-transcriptional role for LSF in mediating progression through mitosis.


Subject(s)
Microtubule-Associated Proteins , Quinolones , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Mitosis , Quinolones/metabolism , Quinolones/pharmacology , Spindle Apparatus/metabolism , Transcription Factors/metabolism , Tubulin/metabolism
3.
Sci Data ; 6(1): 92, 2019 06 14.
Article in English | MEDLINE | ID: mdl-31201317

ABSTRACT

Kidney fibrosis represents an urgent unmet clinical need due to the lack of effective therapies and an inadequate understanding of the molecular pathogenesis. We have generated a comprehensive and combined multi-omics dataset (proteomics, mRNA and small RNA transcriptomics) of fibrotic kidneys that is searchable through a user-friendly web application: http://hbcreports.med.harvard.edu/fmm/ . Two commonly used mouse models were utilized: a reversible chemical-induced injury model (folic acid (FA) induced nephropathy) and an irreversible surgically-induced fibrosis model (unilateral ureteral obstruction (UUO)). mRNA and small RNA sequencing, as well as 10-plex tandem mass tag (TMT) proteomics were performed with kidney samples from different time points over the course of fibrosis development. The bioinformatics workflow used to process, technically validate, and combine the single omics data will be described. In summary, we present temporal multi-omics data from fibrotic mouse kidneys that are accessible through an interrogation tool (Mouse Kidney Fibromics browser) to provide a searchable transcriptome and proteome for kidney fibrosis researchers.


Subject(s)
Disease Models, Animal , Kidney Diseases/genetics , MicroRNAs/genetics , Proteome , RNA, Messenger/genetics , Animals , Fibrosis , Mice , Proteomics , Ureteral Obstruction
4.
J Cell Biol ; 212(7): 767-76, 2016 Mar 28.
Article in English | MEDLINE | ID: mdl-27002163

ABSTRACT

Accurate chromosome segregation depends on proper kinetochore-microtubule attachment. Upon microtubule interaction, kinetochores are subjected to forces generated by the microtubules. In this work, we used laser ablation to sever microtubules attached to a merotelic kinetochore, which is laterally stretched by opposing pulling forces exerted by microtubules, and inferred the mechanical response of the kinetochore from its length change. In both mammalian PtK1 cells and in the fission yeast Schizosaccharomyces pombe, kinetochores shortened after microtubule severing. Interestingly, the inner kinetochore-centromere relaxed faster than the outer kinetochore. Whereas in fission yeast all kinetochores relaxed to a similar length, in PtK1 cells the more stretched kinetochores remained more stretched. Simple models suggest that these differences arise because the mechanical structure of the mammalian kinetochore is more complex. Our study establishes merotelic kinetochores as an experimental model for studying the mechanical response of the kinetochore in live cells and reveals a viscoelastic behavior of the kinetochore that is conserved in yeast and mammalian cells.


Subject(s)
Chromosome Segregation , Kinetochores/metabolism , Laser Therapy , Mechanotransduction, Cellular , Microsurgery , Microtubules/physiology , Schizosaccharomyces/physiology , Cell Line , Cytoskeletal Proteins , Elasticity , Humans , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Microscopy, Confocal , Microscopy, Video , Microtubules/metabolism , Models, Biological , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Stress, Mechanical , Time-Lapse Imaging , Transfection , Viscosity , Mitogen-Activated Protein Kinase Kinase Kinase 11
6.
Proc Natl Acad Sci U S A ; 113(5): 1267-72, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26764383

ABSTRACT

Chemotaxis, the directional migration of cells in a chemical gradient, is robust to fluctuations associated with low chemical concentrations and dynamically changing gradients as well as high saturating chemical concentrations. Although a number of reports have identified cellular behavior consistent with a directional memory that could account for behavior in these complex environments, the quantitative and molecular details of such a memory process remain unknown. Using microfluidics to confine cellular motion to a 1D channel and control chemoattractant exposure, we observed directional memory in chemotactic neutrophil-like cells. We modeled this directional memory as a long-lived intracellular asymmetry that decays slower than observed membrane phospholipid signaling. Measurements of intracellular dynamics revealed that moesin at the cell rear is a long-lived element that when inhibited, results in a reduction of memory. Inhibition of ROCK (Rho-associated protein kinase), downstream of RhoA (Ras homolog gene family, member A), stabilized moesin and directional memory while depolymerization of microtubules (MTs) disoriented moesin deposition and also reduced directional memory. Our study reveals that long-lived polarized cytoskeletal structures, specifically moesin, actomyosin, and MTs, provide a directional memory in neutrophil-like cells even as they respond on short time scales to external chemical cues.


Subject(s)
Cell Polarity , Chemotaxis , Cytoskeleton/metabolism , Immunologic Memory , HL-60 Cells , Humans
7.
FASEB J ; 30(3): 1155-70, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26644349

ABSTRACT

Group IVA phospholipase A2 [cytosolic phospholipase A2α (cPLA2α)] is a key mediator of inflammation and tumorigenesis. In this study, by using a combination of chemical inhibition and genetic approaches in zebrafish and murine cells, we identify a mechanism by which cPLA2α promotes cell proliferation. We identified 2 cpla2α genes in zebrafish, cpla2αa and cpla2αb, with conserved phospholipase activity. In zebrafish, loss of cpla2α expression or inhibition of cpla2α activity diminished G1 progression through the cell cycle. This phenotype was also seen in both mouse embryonic fibroblasts and mesangial cells. G1 progression was rescued by the addition of arachidonic acid or prostaglandin E2 (PGE2), indicating a phospholipase-dependent mechanism. We further show that PGE2, through PI3K/AKT activation, promoted Forkhead box protein O1 (FOXO1) phosphorylation and FOXO1 nuclear export. This led to up-regulation of cyclin D1 and down-regulation of p27(Kip1), thus promoting G1 progression. Finally, using pharmacologic inhibitors, we show that cPLA2α, rapidly accelerated fibrosarcoma (RAF)/MEK/ERK, and PI3K/AKT signaling pathways cooperatively regulate G1 progression in response to platelet-derived growth factor stimulation. In summary, these data indicate that cPLA2α, through its phospholipase activity, is a critical effector of G1 phase progression through the cell cycle and suggest that pharmacological targeting of this enzyme may have important therapeutic benefits in disease mechanisms that involve excessive cell proliferation, in particular, cancer and proliferative glomerulopathies.


Subject(s)
Forkhead Transcription Factors/metabolism , G1 Phase/physiology , Group IV Phospholipases A2/metabolism , Animals , Arachidonic Acid/pharmacology , Cell Division/drug effects , Cell Line , Cell Proliferation/drug effects , Dinoprostone/pharmacology , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , G1 Phase/drug effects , Group IV Phospholipases A2/genetics , HEK293 Cells , Humans , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , Zebrafish
9.
Nat Commun ; 6: 8325, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26446488

ABSTRACT

Leukemia/lymphoma-related factor (LRF) is a POZ/BTB and Krüppel (POK) transcriptional repressor characterized by context-dependent key roles in cell fate decision and tumorigenesis. Here we demonstrate an unexpected transcription-independent function for LRF in the classical non-homologous end joining (cNHEJ) pathway of double-strand break (DSB) repair. We find that LRF loss in cell lines and mouse tissues results in defective cNHEJ, genomic instability and hypersensitivity to ionizing radiation. Mechanistically, we show that LRF binds and stabilizes DNA-PKcs on DSBs, in turn favouring DNA-PK activity. Importantly, LRF loss restores ionizing radiation sensitivity to p53 null cells, making LRF an attractive biomarker to direct p53-null LRF-deficient tumours towards therapeutic treatments based on genotoxic agents or PARP inhibitors following a synthetic lethal strategy.


Subject(s)
DNA Repair/physiology , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation/physiology , Genome , Mice , Protein Array Analysis , Transcription Factors/genetics
10.
Nat Cell Biol ; 17(10): 1356-69, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26389662

ABSTRACT

The AAA-ATPase VCP (also known as p97 or CDC48) uses ATP hydrolysis to 'segregate' ubiquitylated proteins from their binding partners. VCP acts through UBX-domain-containing adaptors that provide target specificity, but the targets and functions of UBXD proteins remain poorly understood. Through systematic proteomic analysis of UBXD proteins in human cells, we reveal a network of over 195 interacting proteins, implicating VCP in diverse cellular pathways. We have explored one such complex between an unstudied adaptor UBXN10 and the intraflagellar transport B (IFT-B) complex, which regulates anterograde transport into cilia. UBXN10 localizes to cilia in a VCP-dependent manner and both VCP and UBXN10 are required for ciliogenesis. Pharmacological inhibition of VCP destabilized the IFT-B complex and increased trafficking rates. Depletion of UBXN10 in zebrafish embryos causes defects in left-right asymmetry, which depends on functional cilia. This study provides a resource for exploring the landscape of UBXD proteins in biology and identifies an unexpected requirement for VCP-UBXN10 in ciliogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Cilia/metabolism , Protein Interaction Maps , Proteomics/methods , Adaptor Proteins, Signal Transducing/genetics , Adenosine Triphosphatases/genetics , Animals , Cell Cycle Proteins/genetics , Cilia/physiology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , LLC-PK1 Cells , Microscopy, Confocal , Microscopy, Fluorescence , Morphogenesis/physiology , Protein Binding , Protein Interaction Mapping/methods , RNA Interference , Swine , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Valosin Containing Protein
11.
Mol Cell Biol ; 35(21): 3768-84, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26303530

ABSTRACT

The G2-to-M transition (or prophase) checkpoint of the cell cycle is a critical regulator of mitotic entry. SIRT2, a tumor suppressor gene, contributes to the control of this checkpoint by blocking mitotic entry under cellular stress. However, the mechanism underlying both SIRT2 activation and regulation of the G2-to-M transition remains largely unknown. Here, we report the formation of a multiprotein complex at the G2-to-M transition in vitro and in vivo. Group IVA cytosolic phospholipase A2 (cPLA2α) acts as a bridge in this complex to promote binding of SIRT2 to cyclin A-Cdk2. Cyclin A-Cdk2 then phosphorylates SIRT2 at Ser331. This phosphorylation reduces SIRT2 catalytic activity and its binding affinity to centrosomes and mitotic spindles, promoting G2-to-M transition. We show that the inhibitory effect of cPLA2α on SIRT2 activity impacts various cellular processes, including cellular levels of histone H4 acetylated at K16 (Ac-H4K16) and Ac-α-tubulin. This regulatory effect of cPLA2α on SIRT2 defines a novel function of cPLA2α independent of its phospholipase activity and may have implications for the impact of SIRT2-related effects on tumorigenesis and age-related diseases.


Subject(s)
Cell Division , G2 Phase , Group IV Phospholipases A2/metabolism , Sirtuin 2/metabolism , Animals , Cell Line , Gene Deletion , Group IV Phospholipases A2/analysis , Group IV Phospholipases A2/genetics , HEK293 Cells , Humans , Male , Mice , Mitosis , Phosphorylation , Protein Interaction Maps , Sirtuin 2/analysis
12.
ACS Chem Neurosci ; 6(9): 1503-8, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-26212450

ABSTRACT

This work describes the use of fluorescence correlation spectroscopy (FCS) and a novel amyloid-binding fluorescent probe, ARCAM 1, to monitor the aggregation of the Alzheimer's disease-associated amyloid ß-peptide (Aß). ARCAM 1 exhibits a large increase in fluorescence emission upon binding to Aß assemblies, making it an excellent candidate for probe enhancement FCS (PE-FCS). ARCAM 1 binding does not change Aß aggregation kinetics. It also exhibits greater dynamic range as a probe in reporting aggregate size by FCS in Aß, when compared to thioflavin T (ThT) or an Aß peptide modified with a fluorophore. Using fluorescent burst analysis (via PE-FCS) to follow aggregation of Aß, we detected soluble aggregates at significantly earlier time points compared to typical bulk fluorescence measurements. Autocorrelation analysis revealed the size of these early Aß assemblies. These results indicate that PE-FCS/ARCAM 1 based assays can detect and provide size characterization of small Aß aggregation intermediates during the assembly process, which could enable monitoring and study of such aggregates that transiently accumulate in biofluids of patients with Alzheimer's and other neurodegenerative diseases.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Spectrometry, Fluorescence/methods , Acrylamides/chemical synthesis , Acrylamides/chemistry , Amyloid beta-Peptides/chemistry , Benzothiazoles , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Kinetics , Molecular Structure , Naphthalenes/chemical synthesis , Naphthalenes/chemistry , Peptide Fragments/chemistry , Protein Aggregates , Solubility , Thiazoles/chemistry , Thiazoles/metabolism
13.
Mol Biol Cell ; 26(11): 2054-66, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25877871

ABSTRACT

We report an improved variant of mKeima, a monomeric long Stokes shift red fluorescent protein, hmKeima8.5. The increased intracellular brightness and large Stokes shift (∼180 nm) make it an excellent partner with teal fluorescent protein (mTFP1) for multiphoton, multicolor applications. Excitation of this pair by a single multiphoton excitation wavelength (MPE, 850 nm) yields well-separable emission peaks (∼120-nm separation). Using this pair, we measure homo- and hetero-oligomerization interactions in living cells via multiphoton excitation fluorescence correlation spectroscopy (MPE-FCS). Using tandem dimer proteins and small-molecule inducible dimerization domains, we demonstrate robust and quantitative detection of intracellular protein-protein interactions. We also use MPE-FCCS to detect drug-protein interactions in the intracellular environment using a Coumarin 343 (C343)-conjugated drug and hmKeima8.5 as a fluorescence pair. The mTFP1/hmKeima8.5 and C343/hmKeima8.5 combinations, together with our calibration constructs, provide a practical and broadly applicable toolbox for the investigation of molecular interactions in the cytoplasm of living cells.


Subject(s)
Fluorescent Dyes , Green Fluorescent Proteins , Luminescent Proteins , Protein Multimerization , Spectrometry, Fluorescence/methods , Amino Acid Sequence , Animals , Cytoplasm , HEK293 Cells , Humans , Microscopy, Fluorescence, Multiphoton , Molecular Sequence Data , Potoroidae , Protein Binding , Sequence Alignment , Red Fluorescent Protein
14.
PLoS One ; 10(2): e0117523, 2015.
Article in English | MEDLINE | ID: mdl-25671541

ABSTRACT

The microtubule-targeting maytansinoids accumulate in cells and induce mitotic arrest at 250- to 1000-fold lower concentrations than those required for their association with tubulin or microtubules. To identify the mechanisms of this intracellular accumulation and exceptional cytotoxicity of maytansinoids we studied interaction of a highly cytotoxic maytansinoid, S-methyl DM1 and several other maytansinoids with cells. S-methyl DM1 accumulated inside the cells with a markedly higher apparent affinity than to tubulin or microtubules. The apparent affinities of maytansinoids correlated with their cytotoxicities. The number of intracellular binding sites for S-methyl DM1 in MCF7 cells was comparable to the number of tubulin molecules per cell (~ 4-6 × 10(7) copies). Efflux of 3[H]-S-methyl DM1 from cells was enhanced in the presence of an excess of non-labeled S-methyl DM1, indicating that re-binding of 3 [H]-S-methyl DM1 to intracellular binding sites contributed to its intracellular retention. Liposomes loaded with non-polymerized tubulin recapitulated the apparent high-affinity association of S-methyl DM1 to cells. We propose a model for the intracellular accumulation of maytansinoids in which molecules of the compounds diffuse into a cell and associate with tubulin. Affinities of maytansinoids for individual tubulin molecules are weak, but the high intracellular concentration of tubulin favors, after dissociation of a compound-tubulin complex, their re-binding to a tubulin molecule, or to a tip of a microtubule in the same cell, over their efflux. As a result, a significant fraction of microtubule tips is occupied with a maytansinoid when added to cells at sub-nanomolar concentrations, inducing mitotic arrest and cell death.


Subject(s)
Maytansine/metabolism , Tubulin/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Liposomes , Maytansine/pharmacology , Microtubules/drug effects , Microtubules/metabolism , Models, Biological , Protein Binding
15.
Nat Commun ; 6: 6023, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25599650

ABSTRACT

The ciliary kinase NEK8 plays a critical role in situs determination and cystic kidney disease, yet its exact function remains unknown. In this study, we identify ANKS6 as a target and activator of NEK8. ANKS6 requires NEK8 for localizing to the ciliary inversin compartment (IC) and activates NEK8 by binding to its kinase domain. Here we demonstrate the functional importance of this interaction through the analysis of two novel mouse mutations, Anks6(Streaker) and Nek8(Roc). Both display heterotaxy, cardiopulmonary malformations and cystic kidneys, a syndrome also characteristic of mutations in Invs and Nphp3, the other known components of the IC. The Anks6(Strkr) mutation decreases ANKS6 interaction with NEK8, precluding NEK8 activation. The Nek8(Roc) mutation inactivates NEK8 kinase function while preserving ANKS6 localization to the IC. Together, these data reveal the crucial role of NEK8 kinase activation within the IC, promoting proper left-right patterning, cardiopulmonary development and renal morphogenesis.


Subject(s)
Body Patterning/physiology , Carrier Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Blotting, Western , Body Patterning/genetics , Carrier Proteins/genetics , Cell Line , Female , Humans , Kinesins/genetics , Kinesins/metabolism , Male , Mice , Morphogenesis/genetics , Morphogenesis/physiology , Mutation , NIMA-Related Kinases , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Rats , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Proc Natl Acad Sci U S A ; 111(33): 12085-90, 2014 Aug 19.
Article in English | MEDLINE | ID: mdl-25092318

ABSTRACT

Expansions of preexisting polyglutamine (polyQ) tracts in at least nine different proteins cause devastating neurodegenerative diseases. There are many unique features to these pathologies, but there must also be unifying mechanisms underlying polyQ toxicity. Using a polyQ-expanded fragment of huntingtin exon-1 (Htt103Q), the causal protein in Huntington disease, we and others have created tractable models for investigating polyQ toxicity in yeast cells. These models recapitulate key pathological features of human diseases and provide access to an unrivalled genetic toolbox. To identify toxicity modifiers, we performed an unbiased overexpression screen of virtually every protein encoded by the yeast genome. Surprisingly, there was no overlap between our modifiers and those from a conceptually identical screen reported recently, a discrepancy we attribute to an artifact of their overexpression plasmid. The suppressors of Htt103Q toxicity recovered in our screen were strongly enriched for glutamine- and asparagine-rich prion-like proteins. Separated from the rest of the protein, the prion-like sequences of these proteins were themselves potent suppressors of polyQ-expanded huntingtin exon-1 toxicity, in both yeast and human cells. Replacing the glutamines in these sequences with asparagines abolished suppression and converted them to enhancers of toxicity. Replacing asparagines with glutamines created stronger suppressors. The suppressors (but not the enhancers) coaggregated with Htt103Q, forming large foci at the insoluble protein deposit in which proteins were highly immobile. Cells possessing foci had fewer (if any) small diffusible oligomers of Htt103Q. Until such foci were lost, cells were protected from death. We discuss the therapeutic implications of these findings.


Subject(s)
Exons , Nerve Tissue Proteins/genetics , Prions/physiology , GPI-Linked Proteins/physiology , Humans , Huntingtin Protein , Microscopy, Confocal
17.
Elife ; 3: e02137, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-25027692

ABSTRACT

The centromere, responsible for chromosome segregation during mitosis, is epigenetically defined by CENP-A containing chromatin. The amount of centromeric CENP-A has direct implications for both the architecture and epigenetic inheritance of centromeres. Using complementary strategies, we determined that typical human centromeres contain ∼400 molecules of CENP-A, which is controlled by a mass-action mechanism. This number, despite representing only ∼4% of all centromeric nucleosomes, forms a ∼50-fold enrichment to the overall genome. In addition, although pre-assembled CENP-A is randomly segregated during cell division, this amount of CENP-A is sufficient to prevent stochastic loss of centromere function and identity. Finally, we produced a statistical map of CENP-A occupancy at a human neocentromere and identified nucleosome positions that feature CENP-A in a majority of cells. In summary, we present a quantitative view of the centromere that provides a mechanistic framework for both robust epigenetic inheritance of centromeres and the paucity of neocentromere formation.DOI: http://dx.doi.org/10.7554/eLife.02137.001.


Subject(s)
Centromere , Chromatin/chemistry , Alleles , Autoantigens/genetics , Autoantigens/metabolism , Centromere Protein A , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Diploidy , Gene Dosage , Humans , Stochastic Processes
18.
J Biol Chem ; 289(33): 22771-22784, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24982423

ABSTRACT

Chromosome ends contain nucleoprotein structures known as telomeres. Damage to chromosome ends during interphase elicits a DNA damage response (DDR) resulting in cell cycle arrest. However, little is known regarding the signaling from damaged chromosome ends (designated here as "TIPs") during mitosis. In the present study, we investigated the consequences of DNA damage induced at a single TIP in mitosis. We used laser microirradiation to damage mitotic TIPs or chromosome arms (non-TIPs) in PtK2 kidney epithelial cells. We found that damage to a single TIP, but not a non-TIP, delays anaphase onset. This TIP-specific checkpoint response is accompanied by differential recruitment of DDR proteins. Although phosphorylation of H2AX and the recruitment of several repair factors, such as Ku70-Ku80, occur in a comparable manner at both TIP and non-TIP damage sites, DDR factors such as ataxia telangiectasia mutated (ATM), MDC1, WRN, and FANCD2 are specifically recruited to TIPs but not to non-TIPs. In addition, Nbs1, BRCA1, and ubiquitin accumulate at damaged TIPs more rapidly than at damaged non-TIPs. ATR and 53BP1 are not detected at either TIPs or non-TIPs in mitosis. The observed delay in anaphase onset is dependent on the activity of DDR kinases ATM and Chk1, and the spindle assembly checkpoint kinase Mps1. Cells damaged at a single TIP or non-TIP eventually exit mitosis with unrepaired lesions. Damaged TIPs are segregated into micronuclei at a significantly higher frequency than damaged non-TIPs. Together, these findings reveal a mitosis-specific DDR uniquely associated with chromosome ends.


Subject(s)
Anaphase , Chromosomes, Mammalian/metabolism , DNA Damage , Epithelial Cells/metabolism , Kidney/metabolism , Lasers/adverse effects , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line , Checkpoint Kinase 1 , Epithelial Cells/cytology , Exodeoxyribonucleases/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Focal Adhesion Kinase 2/metabolism , Histones/metabolism , Kidney/cytology , Phosphorylation , Potoroidae , Protein Kinases/metabolism
19.
Bioconjug Chem ; 25(6): 1043-51, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24797834

ABSTRACT

Small molecule fluorophores are indispensable tools for modern biomedical imaging techniques. In this report, we present the development of a new class of BODIPY dyes based on an alkoxy-fluoro-boron-dipyrromethene core. These novel fluorescent dyes, which we term MayaFluors, are characterized by good aqueous solubility and favorable in vitro physicochemical properties. MayaFluors are readily accessible in good yields in a one-pot, two-step approach starting from well-established BODIPY dyes, and allow for facile modification with functional groups of relevance to bioconjugate chemistry and bioorthogonal labeling. Biological profiling in living cells demonstrates excellent membrane permeability, low nonspecific binding, and lack of cytotoxicity.


Subject(s)
Boron Compounds/analysis , Fluorescent Dyes/analysis , Molecular Imaging/methods , Boron Compounds/chemical synthesis , Boron Compounds/chemistry , Cell Membrane Permeability , Cell Survival , Female , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , MCF-7 Cells , Molecular Structure , Solubility , Tumor Cells, Cultured
20.
Cell ; 156(5): 1017-31, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24581499

ABSTRACT

The spindle assembly checkpoint (SAC) delays anaphase until all chromosomes are bioriented on the mitotic spindle. Under current models, unattached kinetochores transduce the SAC by catalyzing the intramitotic production of a diffusible inhibitor of APC/C(Cdc20) (the anaphase-promoting complex/cyclosome and its coactivator Cdc20, a large ubiquitin ligase). Here we show that nuclear pore complexes (NPCs) in interphase cells also function as scaffolds for anaphase-inhibitory signaling. This role is mediated by Mad1-Mad2 complexes tethered to the nuclear basket, which activate soluble Mad2 as a binding partner and inhibitor of Cdc20 in the cytoplasm. Displacing Mad1-Mad2 from nuclear pores accelerated anaphase onset, prevented effective correction of merotelic errors, and increased the threshold of kinetochore-dependent signaling needed to halt mitosis in response to spindle poisons. A heterologous Mad1-NPC tether restored Cdc20 inhibitor production and normal M phase control. We conclude that nuclear pores and kinetochores both emit "wait anaphase" signals that preserve genome integrity.


Subject(s)
Anaphase , Cell Cycle Proteins/metabolism , M Phase Cell Cycle Checkpoints , Mad2 Proteins/metabolism , Nuclear Pore/metabolism , Nuclear Proteins/metabolism , Active Transport, Cell Nucleus , Cell Cycle Proteins/genetics , Dimerization , HCT116 Cells , HeLa Cells , Humans , Interphase , Kinetochores/metabolism , Mitosis , Nuclear Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL