Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Development ; 142(22): 3869-78, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26395483

ABSTRACT

Yolk provides an important source of nutrients during the early development of oviparous organisms. It is composed mainly of vitellogenin proteins packed into membrane-bound compartments called yolk platelets. Catabolism of yolk is initiated by acidification of the yolk platelet, leading to the activation of Cathepsin-like proteinases, but it is unknown how this process is triggered. Yolk catabolism initiates at cellularization in Drosophila melanogaster embryos. Using maternal shRNA technology we found that yolk catabolism depends on the Tor pathway and on the autophagy-initiating kinase Atg1. Whereas Atg1 was required for a burst of spatially regulated autophagy during late cellularization, autophagy was not required for initiating yolk catabolism. We propose that the conserved Tor metabolic sensing pathway regulates yolk catabolism, similar to Tor-dependent metabolic regulation on the lysosome.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/embryology , Egg Yolk/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Autophagy-Related Protein-1 Homolog , Blotting, Western , In Situ Nick-End Labeling , Microscopy, Electron , Real-Time Polymerase Chain Reaction , Rosaniline Dyes , Signal Transduction/genetics
2.
J Cell Biol ; 203(5): 801-14, 2013 Dec 09.
Article in English | MEDLINE | ID: mdl-24297752

ABSTRACT

In metazoans the endoplasmic reticulum (ER) changes during the cell cycle, with the nuclear envelope (NE) disassembling and reassembling during mitosis and the peripheral ER undergoing extensive remodeling. Here we address how ER morphology is generated during the cell cycle using crude and fractionated Xenopus laevis egg extracts. We show that in interphase the ER is concentrated at the microtubule (MT)-organizing center by dynein and is spread by outward extension of ER tubules through their association with plus ends of growing MTs. Fusion of membranes into an ER network is dependent on the guanosine triphosphatase atlastin (ATL). NE assembly requires fusion by both ATL and ER-soluble N-ethyl-maleimide-sensitive factor adaptor protein receptors. In mitotic extracts, the ER converts into a network of sheets connected by ER tubules and loses most of its interactions with MTs. Together, these results indicate that fusion of ER membranes by ATL and interaction of ER with growing MT ends and dynein cooperate to generate distinct ER morphologies during the cell cycle.


Subject(s)
Cell Cycle , Endoplasmic Reticulum/ultrastructure , Animals , Cell Fractionation , Dyneins/analysis , Dyneins/metabolism , Dyneins/physiology , Endoplasmic Reticulum/metabolism , GTP Phosphohydrolases/physiology , Interphase , Intracellular Membranes/metabolism , Intracellular Membranes/ultrastructure , Membrane Fusion , Microtubule-Organizing Center/metabolism , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Xenopus laevis
3.
Nat Commun ; 4: 1504, 2013.
Article in English | MEDLINE | ID: mdl-23422672

ABSTRACT

Pharmacokinetic analysis at the organ level provides insight into how drugs distribute throughout the body, but cannot explain how drugs work at the cellular level. Here we demonstrate in vivo single-cell pharmacokinetic imaging of PARP-1 inhibitors and model drug behaviour under varying conditions. We visualize intracellular kinetics of the PARP-1 inhibitor distribution in real time, showing that PARP-1 inhibitors reach their cellular target compartment, the nucleus, within minutes in vivo both in cancer and normal cells in various cancer models. We also use these data to validate predictive finite element modelling. Our theoretical and experimental data indicate that tumour cells are exposed to sufficiently high PARP-1 inhibitor concentrations in vivo and suggest that drug inefficiency is likely related to proteomic heterogeneity or insensitivity of cancer cells to DNA-repair inhibition. This suggests that single-cell pharmacokinetic imaging and derived modelling improve our understanding of drug action at single-cell resolution in vivo.


Subject(s)
Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/pharmacokinetics , Molecular Imaging/methods , Single-Cell Analysis/methods , Animals , Cell Line, Tumor , Computer Simulation , Enzyme Inhibitors/chemistry , Finite Element Analysis , Humans , Mice , Models, Biological , Neoplasms/blood supply , Neoplasms/pathology , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Reproducibility of Results , Small Molecule Libraries/pharmacology , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Xenograft Model Antitumor Assays
4.
ACS Chem Biol ; 7(1): 185-96, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-21992004

ABSTRACT

The members of the Aurora kinase family play critical roles in the regulation of the cell cycle and mitotic spindle assembly and have been intensively investigated as potential targets for a new class of anticancer drugs. We describe a new highly potent and selective class of Aurora kinase inhibitors discovered using a phenotypic cellular screen. Optimized inhibitors display many of the hallmarks of Aurora inhibition including endoreduplication, polyploidy, and loss of cell viability in cancer cells. Structure-activity relationships with respect to kinome-wide selectivity and guided by an Aurora B co-crystal structure resulted in the identification of key selectivity determinants and discovery of a subseries with selectivity toward Aurora A. A direct comparison of biochemical and cellular profiles with respect to published Aurora inhibitors including VX-680, AZD1152, MLN8054, and a pyrimidine-based compound from Genentech demonstrates that compounds 1 and 3 will become valuable additional pharmacological probes of Aurora-dependent functions.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , M Phase Cell Cycle Checkpoints/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/chemistry , Aurora Kinase B , Aurora Kinases , Benzazepines/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Models, Molecular , Organophosphates/pharmacology , Paclitaxel/pharmacology , Piperazines/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Pyrimidines/pharmacology , Quinazolines/pharmacology , Structure-Activity Relationship , Xenopus laevis
5.
Nat Rev Mol Cell Biol ; 12(10): 669-74, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21941276

ABSTRACT

Nature Reviews Molecular Cell Biology celebrated its 10-year anniversary during this past year with a series of specially commissioned articles. To complement this, here we have asked researchers from across the field for their insights into how molecular cell biology research has evolved during this past decade, the key concepts that have emerged and the most promising interfaces that have developed. Their comments highlight the broad impact that particular advances have had, some of the basic understanding that we still require, and the collaborative approaches that will be essential for driving the field forward.


Subject(s)
Cell Biology/history , Molecular Biology/history , Molecular Biology/trends , Cell Biology/trends , History, 20th Century , History, 21st Century , Molecular Biology/methods
6.
Science ; 333(6039): 222-5, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21636745

ABSTRACT

Rod-shaped bacteria elongate by the action of cell wall synthesis complexes linked to underlying dynamic MreB filaments. To understand how the movements of these filaments relate to cell wall synthesis, we characterized the dynamics of MreB and the cell wall elongation machinery using high-precision particle tracking in Bacillus subtilis. We found that MreB and the elongation machinery moved circumferentially around the cell, perpendicular to its length, with nearby synthesis complexes and MreB filaments moving independently in both directions. Inhibition of cell wall synthesis by various methods blocked the movement of MreB. Thus, bacteria elongate by the uncoordinated, circumferential movements of synthetic complexes that insert radial hoops of new peptidoglycan during their transit, possibly driving the motion of the underlying MreB filaments.


Subject(s)
Bacillus subtilis/growth & development , Bacillus subtilis/metabolism , Bacterial Proteins/metabolism , Cell Wall/metabolism , Peptidoglycan/metabolism , Anti-Bacterial Agents/pharmacology , Bacillus subtilis/drug effects , Bacillus subtilis/ultrastructure , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Models, Biological , Morphogenesis , Motion , Mutation , Peptidoglycan/chemistry , Polymerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
7.
Mol Biol Cell ; 21(2): 323-33, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19940016

ABSTRACT

Metaphase spindles are steady-state ensembles of microtubules that turn over rapidly and slide poleward in some systems. Since the discovery of dynamic instability in the mid-1980s, models for spindle morphogenesis have proposed that microtubules are stabilized by the spindle environment. We used single molecule imaging to measure tubulin turnover in spindles, and nonspindle assemblies, in Xenopus laevis egg extracts. We observed many events where tubulin molecules spend only a few seconds in polymer and thus are difficult to reconcile with standard models of polymerization dynamics. Our data can be quantitatively explained by a simple, phenomenological model-with only one adjustable parameter-in which the growing and shrinking of microtubule ends is approximated as a biased random walk. Microtubule turnover kinetics did not vary with position in the spindle and were the same in spindles and nonspindle ensembles nucleated by Tetrahymena pellicles. These results argue that the high density of microtubules in spindles compared with bulk cytoplasm is caused by local enhancement of nucleation and not by local stabilization. It follows that the key to understanding spindle morphogenesis will be to elucidate how nucleation is spatially controlled.


Subject(s)
Meiosis , Microtubules/metabolism , Molecular Imaging/methods , Xenopus laevis/metabolism , Animals , Biopolymers/metabolism , Cell Extracts , Cyclopropanes/metabolism , Kinetics , Ovum/cytology , Photobleaching , Pyridines/metabolism , Thiazoles/metabolism , Time Factors , Tubulin/metabolism
8.
Cancer Res ; 68(9): 3269-76, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451153

ABSTRACT

To improve cancer chemotherapy, we need to understand the mechanisms that determine drug sensitivity in cancer and normal cells. Here, we investigate this question across a panel of 11 cell lines at a phenotypic and molecular level for three antimitotic drugs: paclitaxel, nocodazole, and an inhibitor of kinesin-5 (also known as KSP, Eg5, Kif11). Using automated microscopy with markers for mitosis and apoptosis (high content screening), we find that the mitotic arrest response shows relatively little variation between cell types, whereas the tendency to undergo apoptosis shows large variation. We found no correlation between levels of mitotic arrest and apoptosis. Apoptosis depended on entry into mitosis and occurred both from within mitosis and after exit. Response to the three drugs strongly correlated, although paclitaxel caused more apoptosis in some cell lines at similar levels of mitotic arrest. Molecular investigations showed that sensitivity to apoptosis correlated with loss of an antiapoptotic protein, XIAP, during the drug response, but not its preresponse levels, and to some extent also correlated with activation of the p38 and c-Jun NH(2) kinase pathways. We conclude that variation in sensitivity to antimitotic drugs in drug-naive cell lines is governed more by differences in apoptotic signaling than by differences in mitotic spindle or spindle assembly checkpoint proteins and that antimitotics with different mechanisms trigger very similar, but not identical, responses.


Subject(s)
Antimitotic Agents/therapeutic use , Drug Resistance, Neoplasm/physiology , Kinesins/antagonists & inhibitors , Microtubules/drug effects , Neoplasms/classification , Neoplasms/drug therapy , Antimitotic Agents/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Dose-Response Relationship, Drug , Drug Delivery Systems , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , HeLa Cells , Humans , Neoplasms/genetics , Neoplasms/metabolism , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Tumor Cells, Cultured
9.
J Cell Biol ; 161(2): 349-58, 2003 Apr 28.
Article in English | MEDLINE | ID: mdl-12719474

ABSTRACT

Microtubules (MTs) polymerized with GMPCPP, a slowly hydrolyzable GTP analogue, are stable in buffer but are rapidly depolymerized in Xenopus egg extracts. This depolymerization is independent of three previously identified MT destabilizers (Op18, katanin, and XKCM1/KinI). We purified the factor responsible for this novel depolymerizing activity using biochemical fractionation and a visual activity assay and identified it as XMAP215, previously identified as a prominent MT growth-promoting protein in Xenopus extracts. Consistent with the purification results, we find that XMAP215 is necessary for GMPCPP-MT destabilization in extracts and that recombinant full-length XMAP215 as well as an NH2-terminal fragment have depolymerizing activity in vitro. Stimulation of depolymerization is specific for the MT plus end. These results provide evidence for a robust MT-destabilizing activity intrinsic to this microtubule-associated protein and suggest that destabilization may be part of its essential biochemical functions. We propose that the substrate in our assay, GMPCPP-stabilized MTs, serves as a model for the pause state of MT ends and that the multiple activities of XMAP215 are unified by a mechanism of antagonizing MT pauses.


Subject(s)
Cell Extracts/chemistry , Cell Extracts/pharmacology , Guanosine Triphosphate/analogs & derivatives , Microtubule-Associated Proteins/isolation & purification , Microtubules/metabolism , Oocytes/metabolism , Xenopus Proteins , Xenopus laevis/metabolism , Animals , Biological Assay , Female , Guanosine Triphosphate/pharmacology , Microtubule Proteins/biosynthesis , Microtubule Proteins/drug effects , Microtubule Proteins/genetics , Microtubule-Associated Proteins/pharmacology , Microtubules/drug effects , Models, Biological , Oocytes/drug effects , Protein Structure, Tertiary/physiology , Recombinant Fusion Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...