Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Nat Commun ; 12(1): 2412, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33893303

ABSTRACT

The development of a widely adopted cryopreservation method remains a major challenge in Drosophila research. Here we report a robust and easily implemented cryopreservation protocol of Drosophila melanogaster embryos. We present innovations for embryo permeabilization, cryoprotectant agent loading, and rewarming. We show that the protocol is broadly applicable, successfully implemented in 25 distinct strains from different sources. We demonstrate that for most strains, >50% embryos hatch and >25% of the resulting larvae develop into adults after cryopreservation. We determine that survival can be significantly improved by outcrossing to mitigate the effect of genetic background for strains with low survival after cryopreservation. We show that flies retain normal sex ratio, fertility, and original mutation after successive cryopreservation of 5 generations and 6-month storage in liquid nitrogen. Lastly, we find that non-specialists are able to use this protocol to obtain consistent results, demonstrating potential for wide adoption.


Subject(s)
Cryopreservation/methods , Drosophila melanogaster/embryology , Embryo, Nonmammalian/embryology , Rewarming/methods , Vitrification , Animals , Cryoprotective Agents/pharmacology , Drosophila melanogaster/genetics , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/ultrastructure , Female , Fertility/genetics , Larva/genetics , Larva/metabolism , Microscopy, Electron , Permeability/drug effects , Temperature , Time Factors
2.
IEEE ASME Trans Mechatron ; 23(4): 1532-1542, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30595643

ABSTRACT

Optical tweezers have enabled important insights into intracellular transport through the investigation of motor proteins, with their ability to manipulate particles at the microscale, affording femto newton force resolution. Its use to realize a constant force clamp has enabled vital insights into the behavior of motor proteins under different load conditions. However, the varying nature of disturbances and the effect of thermal noise pose key challenges to force regulation. Furthermore, often the main aim of many studies is to determine the motion of the motor and the statistics related to the motion, which can be at odds with the force regulation objective. In this article, we propose a mixed objective H 2 /H ∞ optimization framework using a model-based design, that achieves the dual goals of force regulation and real time motion estimation with quantifiable guarantees. Here, we minimize the H ∞ norm for the force regulation and error in step estimation while maintaining the H 2 norm of the noise on step estimate within user specified bounds. We demonstrate the efficacy of the framework through extensive simulations and an experimental implementation using an optical tweezer setup with live samples of the motor protein 'kinesin'; where regulation of forces below 1 piconewton with errors below 10% is obtained while simultaneously providing real time estimates of motor motion.

3.
PLoS Comput Biol ; 12(11): e1005152, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27812098

ABSTRACT

Intracellular transport is an essential function in eucaryotic cells, facilitated by motor proteins-proteins converting chemical energy into kinetic energy. It is understood that motor proteins work in teams enabling unidirectional and bidirectional transport of intracellular cargo over long distances. Disruptions of the underlying transport mechanisms, often caused by mutations that alter single motor characteristics, are known to cause neurodegenerative diseases. For example, phosphorylation of kinesin motor domain at the serine residue is implicated in Huntington's disease, with a recent study of phosphorylated and phosphomimetic serine residues indicating lowered single motor stalling forces. In this article we report the effects of mutations of this nature on transport properties of cargo carried by multiple wild-type and mutant motors. Results indicate that mutants with altered stall forces might determine the average velocity and run-length even when they are outnumbered by wild type motors in the ensemble. It is shown that mutants gain a competitive advantage and lead to an increase in the expected run-length when the load on the cargo is in the vicinity of the mutant's stalling force or a multiple of its stalling force. A separate contribution of this article is the development of a semi-analytic method to analyze transport of cargo by multiple motors of multiple types. The technique determines transition rates between various relative configurations of motors carrying the cargo using the transition rates between various absolute configurations. This enables a computation of biologically relevant quantities like average velocity and run-length without resorting to Monte Carlo simulations. It can also be used to introduce alterations of various single motor parameters to model a mutation and to deduce effects of such alterations on the transport of a common cargo by multiple motors. Our method is easily implementable and we provide a software package for general use.


Subject(s)
Microtubules/chemistry , Microtubules/physiology , Models, Biological , Models, Chemical , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/physiology , Binding Sites , Biological Transport, Active/physiology , Computer Simulation , Energy Transfer/physiology , Models, Statistical , Motion , Protein Binding
4.
Methods Cell Biol ; 131: 277-309, 2016.
Article in English | MEDLINE | ID: mdl-26794520

ABSTRACT

Proper neuronal function critically depends on efficient intracellular transport and disruption of transport leads to neurodegeneration. Molecular pathways that support or regulate neuronal transport are not fully understood. A greater understanding of these pathways will help reveal the pathological mechanisms underlying disease. Drosophila melanogaster is the premier model system for performing large-scale genetic functional screens. Here we describe methods to carry out primary and secondary genetic screens in Drosophila aimed at identifying novel gene products and pathways that impact neuronal intracellular transport. These screens are performed using whole animal or live cell imaging of intact neural tissue to ensure integrity of neurons and their cellular environment. The primary screen is used to identify gross defects in neuronal function indicative of a disruption in microtubule-based transport. The secondary screens, conducted in both motoneurons and dendritic arborization neurons, will confirm the function of candidate gene products in intracellular transport. Together, the methodologies described here will support labs interested in identifying and characterizing gene products that alter intracellular transport in Drosophila.


Subject(s)
Axonal Transport/genetics , Axons/metabolism , Drosophila melanogaster/metabolism , Dyneins/metabolism , Kinesins/metabolism , Microtubule-Associated Proteins/metabolism , Animals , Axonal Transport/physiology , Dendrites/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Dynactin Complex , Dyneins/genetics , Larva/metabolism , Microtubules/genetics , Microtubules/metabolism , Neurodegenerative Diseases/pathology , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , RNA Interference , RNA, Small Interfering/genetics
5.
J Cell Biol ; 189(1): 143-58, 2010 Apr 05.
Article in English | MEDLINE | ID: mdl-20368622

ABSTRACT

Spinocerebellar ataxia type 5 (SCA5) is an autosomal dominant neurodegenerative disorder caused by mutations in the SPBTN2 gene encoding beta-III-spectrin. To investigate the molecular basis of SCA5, we established a series of transgenic Drosophila models that express human beta-III-spectrin or fly beta-spectrin proteins containing SCA5 mutations. Expression of the SCA5 mutant spectrin in the eye causes a progressive neurodegenerative phenotype, and expression in larval neurons results in posterior paralysis, reduced synaptic terminal growth, and axonal transport deficits. These phenotypes are genetically enhanced by both dynein and dynactin loss-of-function mutations. In summary, we demonstrate that SCA5 mutant spectrin causes adult-onset neurodegeneration in the fly eye and disrupts fundamental intracellular transport processes that are likely to contribute to this progressive neurodegenerative disease.


Subject(s)
Axonal Transport/genetics , Drosophila/genetics , Mutation , Nerve Degeneration/genetics , Spectrin/genetics , Spinocerebellar Ataxias/genetics , Animals , Animals, Genetically Modified , Drosophila/metabolism , Female , Humans , Male , Nerve Degeneration/metabolism , Spectrin/metabolism , Spinocerebellar Ataxias/metabolism
6.
Mol Biol Cell ; 19(11): 4918-29, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18799620

ABSTRACT

The dynein light intermediate chain (LIC) is a subunit unique to the cytoplasmic form of dynein, but how it contributes to dynein function is not fully understood. Previous work has established that the LIC homodimer binds directly to the dynein heavy chain and may mediate the attachment of dynein to centrosomes and other cargoes. Here, we report our characterization of the LIC in Drosophila. Unlike vertebrates, in which two Lic genes encode multiple subunit isoforms, the Drosophila LIC is encoded by a single gene. We determined that the single LIC polypeptide is phosphorylated, and that different phosphoisoforms can assemble into the dynein motor complex. Our mutational analyses demonstrate that, similar to other dynein subunits, the Drosophila LIC is required for zygotic development, germline specification of the oocyte, and mitotic cell division. We show that RNA interference depletion of LIC in Drosophila S2 cells does not block the recruitment of a dynein complex to kinetochores, but it does delay inactivation of Mad2 signaling and mitotic progression. Our observations suggest the LIC contributes to a broad range of dynein functions.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Dyneins/metabolism , Protein Subunits/metabolism , Spindle Apparatus/metabolism , Amino Acid Sequence , Animals , Cell Line , DNA Transposable Elements , Drosophila Proteins/chemistry , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Dyneins/chemistry , Genes, Insect , Kinetochores/metabolism , Mitosis , Molecular Sequence Data , Mutagenesis, Insertional , Mutation/genetics , Oogenesis , Protein Subunits/chemistry , RNA Interference
7.
J Cell Sci ; 121(Pt 16): 2643-51, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18653542

ABSTRACT

Intracellular transport and processing of ligands is critical to the activation of signal transduction pathways that guide development. Star is an essential gene in Drosophila that has been implicated in the trafficking of ligands for epidermal growth factor (EGF) receptor signaling. The role of cytoplasmic motors in the endocytic and secretory pathways is well known, but the specific requirement of motors in EGF receptor transport has not been investigated. We identified Star in a screen designed to recover second-site modifiers of the dominant rough eye phenotype of the Glued mutation Gl(1). The Glued (Gl) locus encodes the p150 subunit of the dynactin complex, an activator of cytoplasmic dynein-driven motility. We show that alleles of Gl and dynein genetically interact with both Star and EGFR alleles. Similarly to mutations in Star, the Gl(1) mutation is capable of modifying the phenotypes of the EGFR mutation Ellipse. These genetic interactions suggest a model in which Star, dynactin and dynein cooperate in the trafficking of EGF ligands. In support of this model, overexpression of the cleaved, active Spitz ligand can partially bypass defective trafficking and suppress the genetic interactions. Our direct observations of live S2 cells show that export of Spitz-GFP from the endoplasmic reticulum, as well as the trafficking of Spitz-GFP vesicles, depends on both Star and dynein.


Subject(s)
Drosophila Proteins/metabolism , Dyneins/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Membrane Proteins/metabolism , Animals , Animals, Genetically Modified , Cells, Cultured , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Dyneins/genetics , Dyneins/physiology , Endoplasmic Reticulum/metabolism , Epidermal Growth Factor/genetics , Epistasis, Genetic , ErbB Receptors/physiology , Eye/anatomy & histology , Eye/metabolism , Green Fluorescent Proteins/genetics , Membrane Proteins/genetics , Membrane Proteins/physiology , Mutagenesis, Insertional/physiology , Phenotype , Protein Binding , Protein Transport , Retroelements/genetics , Signal Transduction/physiology
8.
PLoS Genet ; 4(2): e36, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18282112

ABSTRACT

The localization of specific mRNAs can establish local protein gradients that generate and control the development of cellular asymmetries. While all evidence underscores the importance of the cytoskeleton in the transport and localization of RNAs, we have limited knowledge of how these events are regulated. Using a visual screen for motile proteins in a collection of GFP protein trap lines, we identified the Drosophila IGF-II mRNA-binding protein (Imp), an ortholog of Xenopus Vg1 RNA binding protein and chicken zipcode-binding protein. In Drosophila, Imp is part of a large, RNase-sensitive complex that is enriched in two polarized cell types, the developing oocyte and the neuron. Using time-lapse confocal microscopy, we establish that both dynein and kinesin contribute to the transport of GFP-Imp particles, and that regulation of transport in egg chambers appears to differ from that in neurons. In Drosophila, loss-of-function Imp mutations are zygotic lethal, and mutants die late as pharate adults. Imp has a function in Drosophila oogenesis that is not essential, as well as functions that are essential during embryogenesis and later development. Germline clones of Imp mutations do not block maternal mRNA localization or oocyte development, but overexpression of a specific Imp isoform disrupts dorsal/ventral polarity. We report here that loss-of-function Imp mutations, as well as Imp overexpression, can alter synaptic terminal growth. Our data show that Imp is transported to the neuromuscular junction, where it may modulate the translation of mRNA targets. In oocytes, where Imp function is not essential, we implicate a specific Imp domain in the establishment of dorsoventral polarity.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Oogenesis/physiology , Presynaptic Terminals/metabolism , RNA-Binding Proteins/metabolism , Alternative Splicing , Animals , Animals, Genetically Modified , Base Sequence , Biological Transport, Active , Body Patterning , DNA Primers/genetics , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/genetics , Female , Gene Expression , Genes, Insect , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mutation , Oogenesis/genetics , RNA-Binding Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
9.
Mol Biol Cell ; 18(6): 2254-63, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17429069

ABSTRACT

In Drosophila, the asymmetric localization of specific mRNAs to discrete regions within the developing oocyte determines the embryonic axes. The microtubule motors dynein and kinesin are required for the proper localization of the determinant ribonucleoprotein (RNP) complexes, but the mechanisms that account for RNP transport to and within the oocyte are not well understood. In this work, we focus on the transport of RNA complexes containing bicoid (bcd), an anterior determinant. We show in live egg chambers that, within the nurse cell compartment, dynein actively transports green fluorescent protein-tagged Exuperantia, a cofactor required for bcd RNP localization. Surprisingly, the loss of kinesin I activity elevates RNP motility in nurse cells, whereas disruption of dynein activity inhibits RNP transport. Once RNPs are transferred through the ring canal to the oocyte, they no longer display rapid, linear movements, but they are distributed by cytoplasmic streaming and gradually disassemble. By contrast, bcd mRNA injected into oocytes assembles de novo into RNP particles that exhibit rapid, dynein-dependent transport. We speculate that after delivery to the oocyte, RNP complexes may disassemble and be remodeled with appropriate accessory factors to ensure proper localization.


Subject(s)
Drosophila melanogaster , Oocytes/metabolism , RNA, Messenger/metabolism , Ribonucleoproteins/metabolism , Animals , Biological Transport/physiology , Cytoskeleton/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Dyneins/metabolism , Egg Proteins/genetics , Egg Proteins/metabolism , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Kinesins/genetics , Kinesins/metabolism , Macromolecular Substances , Male , Microtubules/metabolism , Oocytes/cytology , Ovary/anatomy & histology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
10.
Mol Biol Cell ; 15(7): 3005-14, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15090621

ABSTRACT

Variations in subunit composition and modification have been proposed to regulate the multiple functions of cytoplasmic dynein. Here, we examine the role of the Drosophila ortholog of tctex-1, the 14-kDa dynein light chain. We show that the 14-kDa light chain is a bona fide component of Drosophila cytoplasmic dynein and use P element excision to generate flies that completely lack this dynein subunit. Remarkably, the null mutant is viable and the only observed defect is complete male sterility. During spermatid differentiation, the 14-kDa light chain is required for the localization of a nuclear "cap" of cytoplasmic dynein and for proper attachment between the sperm nucleus and flagellar basal body. Our results provide evidence that the function of the 14-kDa light chain in Drosophila is distinct from other dynein subunits and is not required for any essential functions in early development or in the adult organism.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/physiology , Dyneins/metabolism , Dyneins/physiology , Spermatids/ultrastructure , Spermatogenesis , Amino Acid Sequence , Animals , Base Sequence , Cytoplasmic Dyneins , DNA Mutational Analysis , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/ultrastructure , Dyneins/analysis , Dyneins/genetics , Fertility/genetics , Male , Molecular Sequence Data , Mutagenesis, Insertional , Spermatids/chemistry , Spermatogenesis/genetics , Testis/chemistry , Testis/metabolism , Testis/ultrastructure
11.
Mol Biol Cell ; 14(4): 1355-65, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12686593

ABSTRACT

Sequence comparisons and structural analyses show that the dynein heavy chain motor subunit is related to the AAA family of chaperone-like ATPases. The core structure of the dynein motor unit derives from the assembly of six AAA domains into a hexameric ring. In dynein, the first four AAA domains contain consensus nucleotide triphosphate-binding motifs, or P-loops. The recent structural models of dynein heavy chain have fostered the hypothesis that the energy derived from hydrolysis at P-loop 1 acts through adjacent P-loop domains to effect changes in the attachment state of the microtubule-binding domain. However, to date, the functional significance of the P-loop domains adjacent to the ATP hydrolytic site has not been demonstrated. Our results provide a mutational analysis of P-loop function within the first and third AAA domains of the Drosophila cytoplasmic dynein heavy chain. Here we report the first evidence that P-loop-3 function is essential for dynein function. Significantly, our results further show that P-loop-3 function is required for the ATP-induced release of the dynein complex from microtubules. Mutation of P-loop-3 blocks ATP-mediated release of dynein from microtubules, but does not appear to block ATP binding and hydrolysis at P-loop 1. Combined with the recent recognition that dynein belongs to the family of AAA ATPases, the observations support current models in which the multiple AAA domains of the dynein heavy chain interact to support the translocation of the dynein motor down the microtubule lattice.


Subject(s)
Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Dyneins/chemistry , Dyneins/metabolism , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Binding Sites , Cytoplasm/metabolism , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Dyneins/genetics , Female , Genes, Insect , Microtubules/metabolism , Molecular Motor Proteins/genetics , Mutagenesis, Site-Directed , Oogenesis , Protein Structure, Tertiary , Ultraviolet Rays , Vanadates
12.
Biochemistry ; 41(13): 4302-11, 2002 Apr 02.
Article in English | MEDLINE | ID: mdl-11914076

ABSTRACT

The interactions of three subunits of cytoplasmic dynein from Drosophila melanogaster, LC8, Tctex-1, and the N-terminal domain of IC74 (N-IC74, residues 1-289), were characterized in vitro by affinity methods, limited proteolysis, and circular dichroism spectroscopy. These subunits were chosen for study because they are presumed to promote the assembly of the complex and to be engaged in the controlled binding and release of cargo. Limited proteolysis and mass spectrometry of N-IC74 in the presence of LC8 and Tctex-1 localized binding of Tctex-1 to the vicinity of K104 and K105, and localized binding of LC8 to the region downstream of K130. Circular dichroism, fluorescence, sedimentation velocity, and proteolysis studies indicate that N-IC74 has limited secondary and tertiary structure at near physiological solution conditions. Upon addition of LC8, N-IC74 undergoes a significant conformational change from largely unfolded to a more ordered structure. This conformational change is reflected in increased global protection of N-IC74 from proteolytic digestion following the interaction, and in a significant change in the CD signal. A smaller but reproducible change in the CD spectra was observed upon Tctex-1 binding as well. The increased structure introduced into N-IC74 upon light chain binding suggests a mechanism by which LC8 and Tctex-1 may regulate the assembly of the dynein complex.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cytoplasm/metabolism , Drosophila Proteins/chemistry , Dyneins/chemistry , Microtubule Proteins/chemistry , Microtubule Proteins/metabolism , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Nuclear Proteins , Xenopus Proteins , Amino Acid Sequence , Animals , Circular Dichroism , Cloning, Molecular , Dose-Response Relationship, Drug , Drosophila Proteins/metabolism , Drosophila melanogaster/chemistry , Dyneins/metabolism , Electrophoresis, Polyacrylamide Gel , Endopeptidase K/pharmacology , Guanidine/chemistry , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Precipitin Tests , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Protein Subunits , Spectrometry, Fluorescence , Time Factors , t-Complex Genome Region
SELECTION OF CITATIONS
SEARCH DETAIL