1
|
Li Q, Chen X, Lin L, Zhang L, Wang L, Bao J, Zhang D. Transcriptomic Dynamics of Active and Inactive States of Rho GTPase MoRho3 in Magnaporthe oryzae. J Fungi (Basel) 2022; 8:jof8101060. [PMID: 36294629 PMCID: PMC9605073 DOI: 10.3390/jof8101060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022] Open
Abstract
The small Rho GTPase acts as a molecular switch in eukaryotic signal transduction, which plays a critical role in polar cell growth and vesicle trafficking. Previous studies demonstrated that constitutively active (CA) mutant strains, of MoRho3-CA were defective in appressorium formation. While dominant-negative (DN) mutant strains MoRho3-DN shows defects in polar growth. However, the molecular dynamics of MoRho3-mediated regulatory networks in the pathogenesis of Magnaporthe oryzae still needs to be uncovered. Here, we perform comparative transcriptomic profiling of MoRho3-CA and MoRho3-DN mutant strains using a high-throughput RNA sequencing approach. We find that genetic manipulation of MoRho3 significantly disrupts the expression of 28 homologs of Saccharomyces cerevisiae Rho3-interacting proteins, including EXO70, BNI1, and BNI2 in the MoRho3 CA, DN mutant strains. Functional enrichment analyses of up-regulated DEGs reveal a significant enrichment of genes associated with ribosome biogenesis in the MoRho3-CA mutant strain. Down-regulated DEGs in the MoRho3-CA mutant strains shows significant enrichment in starch/sucrose metabolism and the ABC transporter pathway. Moreover, analyses of down-regulated DEGs in the in MoRho3-DN reveals an over-representation of genes enriched in metabolic pathways. In addition, we observe a significant suppression in the expression levels of secreted proteins suppressed in both MoRho3-CA and DN mutant strains. Together, our results uncover expression dynamics mediated by two states of the small GTPase MoRho3, demonstrating its crucial roles in regulating the expression of ribosome biogenesis and secreted proteins.
Collapse
Affiliation(s)
- Qian Li
- Meishan Vocational Technical College, Ministerial and Provincial Joint Innovation Centre for Safety Production of Cross-Strait Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Correspondence: (Q.L.); (D.Z.)
| | - Xi Chen
- State Key Laboratory for Ecological Pest Control of Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Lianyu Lin
- State Key Laboratory for Ecological Pest Control of Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Lianhu Zhang
- State Key Laboratory for Ecological Pest Control of Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China
| | - Li Wang
- Meishan Vocational Technical College, Ministerial and Provincial Joint Innovation Centre for Safety Production of Cross-Strait Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jiandong Bao
- State Key Laboratory for Ecological Pest Control of Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Dongmei Zhang
- State Key Laboratory for Ecological Pest Control of Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Correspondence: (Q.L.); (D.Z.)
| |
Collapse
|
2
|
Logan CM, Menko AS. Microtubules: Evolving roles and critical cellular interactions. Exp Biol Med (Maywood) 2019; 244:1240-1254. [PMID: 31387376 DOI: 10.1177/1535370219867296] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Microtubules are cytoskeletal elements known as drivers of directed cell migration, vesicle and organelle trafficking, and mitosis. In this review, we discuss new research in the lens that has shed light into further roles for stable microtubules in the process of development and morphogenesis. In the lens, as well as other systems, distinct roles for characteristically dynamic microtubules and stabilized populations are coming to light. Understanding the mechanisms of microtubule stabilization and the associated microtubule post-translational modifications is an evolving field of study. Appropriate cellular homeostasis relies on not only one cytoskeletal element, but also rather an interaction between cytoskeletal proteins as well as other cellular regulators. Microtubules are key integrators with actin and intermediate filaments, as well as cell–cell junctional proteins and other cellular regulators including myosin and RhoGTPases to maintain this balance.Impact statementThe role of microtubules in cellular functioning is constantly expanding. In this review, we examine new and exciting fields of discovery for microtubule’s involvement in morphogenesis, highlight our evolving understanding of differential roles for stabilized versus dynamic subpopulations, and further understanding of microtubules as a cellular integrator.
Collapse
Affiliation(s)
- Caitlin M Logan
- Pathology Anatomy and Cell Biology Department, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - A Sue Menko
- Pathology Anatomy and Cell Biology Department, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
3
|
Song Y, Kang M, Morfini G, Brady ST. Fast axonal transport in isolated axoplasm from the squid giant axon. Methods Cell Biol 2015; 131:331-48. [PMID: 26794522 DOI: 10.1016/bs.mcb.2015.07.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The giant axon of the squid provides a unique cell biological model for analyzing the biochemistry and cell biology of the axon. These axons may exceed 500 μm in diameter and can be readily dissected. Once the surrounding small axons and connective tissue are removed, the axoplasm can be extruded as an intact cylinder of isolated cytoplasm. This isolated axoplasm is morphologically indistinguishable from the intact axon, but without permeability barriers. Fast axonal transport will continue for more than 4 h after extrusion and can be visualized in real time. By perfusing defined concentrations of proteins and/or reagents into the axoplasm, this preparation represents a powerful model for study of intracellular trafficking and its underlying molecular mechanisms.
Collapse
Affiliation(s)
- Yuyu Song
- Marine Biological Laboratory, Woods Hole, MA, USA; Yale School of Medicine, Department of Genetics and Howard Hughes Medical Institute, Boyer Center, New Haven, CT, USA
| | - Minsu Kang
- Marine Biological Laboratory, Woods Hole, MA, USA; Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Gerardo Morfini
- Marine Biological Laboratory, Woods Hole, MA, USA; Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Scott T Brady
- Marine Biological Laboratory, Woods Hole, MA, USA; Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
4
|
Abstract
Vigorous transport of cytoplasmic components along axons over substantial distances is crucial for the maintenance of neuron structure and function. The transport of mitochondria, which serves to distribute mitochondrial functions in a dynamic and non-uniform fashion, has attracted special interest in recent years following the discovery of functional connections among microtubules, motor proteins and mitochondria, and their influences on neurodegenerative diseases. Although the motor proteins that drive mitochondrial movement are now well characterized, the mechanisms by which anterograde and retrograde movement are coordinated with one another and with stationary axonal mitochondria are not yet understood. In this Commentary, we review why mitochondria move and how they move, focusing particularly on recent studies of transport regulation, which implicate control of motor activity by specific cell-signaling pathways, regulation of motor access to transport tracks and static microtubule-mitochondrion linkers. A detailed mechanism for modulating anterograde mitochondrial transport has been identified that involves Miro, a mitochondrial Ca(2+)-binding GTPase, which with associated proteins, can bind and control kinesin-1. Elements of the Miro complex also have important roles in mitochondrial fission-fusion dynamics, highlighting questions about the interdependence of biogenesis, transport, dynamics, maintenance and degradation.
Collapse
Affiliation(s)
- William M Saxton
- Department of Molecular Cell and Developmental Biology, University of California, Cruz, CA 95060, USA.
| | | |
Collapse
|
5
|
STAGSTED JAN. Journey beyond immunology. Regulation of receptor internalization by major histocompatibility complex class I (MHC-I) and effect of peptides derived from MHC-I. APMIS 2011. [DOI: 10.1111/j.1600-0463.1998.tb05657.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
6
|
Perkins GA, Ellisman MH. Mitochondrial configurations in peripheral nerve suggest differential ATP production. J Struct Biol 2010; 173:117-27. [PMID: 20600951 DOI: 10.1016/j.jsb.2010.06.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 06/21/2010] [Indexed: 12/25/2022]
Abstract
Physiological states of mitochondria often correlate with distinctive morphology. Electron microscopy and tomographic reconstruction were used to investigate the three-dimensional structure of axonal mitochondria and mitochondria in the surrounding Schwann cells of the peripheral nervous system (PNS), both in the vicinity of nodes of Ranvier and far from these nodes. Condensed mitochondria were found to be abundant in the axoplasm, but not in the Schwann cell. Uncharacteristic of the classical morphology of condensed mitochondria, the outer and inner boundary membranes are in close apposition and the crista junctions are narrow, consistent with their function as gates for the diffusion of macromolecules. There is also less cristae surface area and lower density of crista junctions in these mitochondria. The density of mitochondria was greater at the paranode-node-paranode (PNP) as was the crista junction opening, yet there were fewer cristae in these organelles compared to those in the internodal region. The greater density of condensed mitochondria in the PNS axoplasm and in particular at the PNP suggests a need for these organelles to operate at a high workload of ATP production.
Collapse
Affiliation(s)
- Guy A Perkins
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California, San Diego, La Jolla, CA 92093-0608, United States.
| | | |
Collapse
|
7
|
APP anterograde transport requires Rab3A GTPase activity for assembly of the transport vesicle. J Neurosci 2009; 29:14534-44. [PMID: 19923287 DOI: 10.1523/jneurosci.1546-09.2009] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The amyloid precursor protein (APP) is anterogradely transported by conventional kinesin in a distinct transport vesicle, but both the biochemical composition of such a vesicle and the specific kinesin-1 motor responsible for transport are poorly defined. APP may be sequentially cleaved by beta- and gamma-secretases leading to accumulation of beta-amyloid (Abeta) peptides in brains of Alzheimer's disease patients, whereas cleavage of APP by alpha-secretases prevents Abeta generation. Here, we demonstrate by time-lapse analysis and immunoisolations that APP is a cargo of a vesicle containing the kinesin heavy chain isoform kinesin-1C, the small GTPase Rab3A, and a specific subset of presynaptic protein components. Moreover, we report that assembly of kinesin-1C and APP in this vesicle type requires Rab3A GTPase activity. Finally, we show cleavage of APP in transport vesicles by alpha-secretase activity, likely mediated by ADAM10. Together, these data indicate that maturation of APP transport vesicles, including recruitment of conventional kinesin, requires Rab3 GTPase activity.
Collapse
|
8
|
LaPointe NE, Morfini G, Pigino G, Gaisina IN, Kozikowski AP, Binder LI, Brady ST. The amino terminus of tau inhibits kinesin-dependent axonal transport: implications for filament toxicity. J Neurosci Res 2009; 87:440-51. [PMID: 18798283 DOI: 10.1002/jnr.21850] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The neuropathology of Alzheimer's disease (AD) and other tauopathies is characterized by filamentous deposits of the microtubule-associated protein tau, but the relationship between tau polymerization and neurotoxicity is unknown. Here, we examined effects of filamentous tau on fast axonal transport (FAT) using isolated squid axoplasm. Monomeric and filamentous forms of recombinant human tau were perfused in axoplasm, and their effects on kinesin- and dynein-dependent FAT rates were evaluated by video microscopy. Although perfusion of monomeric tau at physiological concentrations showed no effect, tau filaments at the same concentrations selectively inhibited anterograde (kinesin-dependent) FAT, triggering the release of conventional kinesin from axoplasmic vesicles. Pharmacological experiments indicated that the effect of tau filaments on FAT is mediated by protein phosphatase 1 (PP1) and glycogen synthase kinase-3 (GSK-3) activities. Moreover, deletion analysis suggested that these effects depend on a conserved 18-amino-acid sequence at the amino terminus of tau. Interestingly, monomeric tau isoforms lacking the C-terminal half of the molecule (including the microtubule binding region) recapitulated the effects of full-length filamentous tau. Our results suggest that pathological tau aggregation contributes to neurodegeneration by altering a regulatory pathway for FAT.
Collapse
Affiliation(s)
- Nichole E LaPointe
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Toda H, Mochizuki H, Flores R, Josowitz R, Krasieva TB, Lamorte VJ, Suzuki E, Gindhart JG, Furukubo-Tokunaga K, Tomoda T. UNC-51/ATG1 kinase regulates axonal transport by mediating motor-cargo assembly. Genes Dev 2009; 22:3292-307. [PMID: 19056884 DOI: 10.1101/gad.1734608] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Axonal transport mediated by microtubule-dependent motors is vital for neuronal function and viability. Selective sets of cargoes, including macromolecules and organelles, are transported long range along axons to specific destinations. Despite intensive studies focusing on the motor machinery, the regulatory mechanisms that control motor-cargo assembly are not well understood. Here we show that UNC-51/ATG1 kinase regulates the interaction between synaptic vesicles and motor complexes during transport in Drosophila. UNC-51 binds UNC-76, a kinesin heavy chain (KHC) adaptor protein. Loss of unc-51 or unc-76 leads to severe axonal transport defects in which synaptic vesicles are segregated from the motor complexes and accumulate along axons. Genetic studies show that unc-51 and unc-76 functionally interact in vivo to regulate axonal transport. UNC-51 phosphorylates UNC-76 on Ser(143), and the phosphorylated UNC-76 binds Synaptotagmin-1, a synaptic vesicle protein, suggesting that motor-cargo interactions are regulated in a phosphorylation-dependent manner. In addition, defective axonal transport in unc-76 mutants is rescued by a phospho-mimetic UNC-76, but not a phospho-defective UNC-76, demonstrating the essential role of UNC-76 Ser(143) phosphorylation in axonal transport. Thus, our data provide insight into axonal transport regulation that depends on the phosphorylation of adaptor proteins.
Collapse
Affiliation(s)
- Hirofumi Toda
- Division of Neurosciences, Beckman Research Institute of the City of Hope, California 91010, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Johnson JE, Perkins GA, Giddabasappa A, Chaney S, Xiao W, White AD, Brown JM, Waggoner J, Ellisman MH, Fox DA. Spatiotemporal regulation of ATP and Ca2+ dynamics in vertebrate rod and cone ribbon synapses. Mol Vis 2007; 13:887-919. [PMID: 17653034 PMCID: PMC2774461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE In conventional neurons, Ca2+ enters presynaptic terminals during an action potential and its increased local concentration triggers transient exocytosis. In contrast, vertebrate photoreceptors are nonspiking neurons that maintain sustained depolarization and neurotransmitter release from ribbon synapses in darkness and produce light-dependent graded hyperpolarizing responses. Rods transmit single photon responses with high fidelity, whereas cones are less sensitive and exhibit faster response kinetics. These differences are likely due to variations in presynaptic Ca2+ dynamics. Metabolic coupling and cross-talk between mitochondria, endoplasmic reticulum (ER), plasma membrane Ca2+ ATPase (PMCA), and Na+-Ca2+ exchanger (NCX) coordinately control presynaptic ATP production and Ca2+ dynamics. The goal of our structural and functional studies was to determine the spatiotemporal regulation of ATP and Ca2+ dynamics in rod spherules and cone pedicles. METHODS Central retina tissue from C57BL/6 mice was used. Laser scanning confocal microscopy (LSCM) experiments were conducted on fixed-frozen vertical sections. Primary antibodies were selected for their tissue/cellular specificity and ability to recognize single, multiple or all splice variants of selected isoforms. Electron microscopy (EM) and 3-D electron tomography (ET) studies used our standard procedures on thin- and thick-sectioned retinas, respectively. Calibrated fluo-3-Ca2+ imaging experiments of dark- and light-adapted rod and cone terminals in retinal slices were conducted. RESULTS Confocal microscopy showed that mitochondria, ER, PMCA, and NCX1 exhibited distinct retinal lamination patterns and differential distribution in photoreceptor synapses. Antibodies for three distinct mitochondrial compartments differentially labeled retinal areas with high metabolic demand: rod and cone inner segments, previously undescribed cone juxtanuclear mitochondria and the two plexiform layers. Rod spherule membranes uniformly and intensely stained for PMCA, whereas the larger cone pedicles preferentially stained for NCX1 at their active zones and PMCA near their mitochondria. EM and ET revealed that mitochondria in rod spherules and cone pedicles differed markedly in their number, location, size, volume, and total cristae surface area, and cristae junction diameter. Rod spherules had one large ovoid mitochondrion located near its active zone, whereas cone pedicles averaged five medium-sized mitochondria clustered far from their active zones. Most spherules had one ribbon synapse, whereas pedicles contained numerous ribbon synapses. Fluo-3 imaging studies revealed that during darkness rod spherules maintained a lower [Ca2+] than cone pedicles, whereas during light adaptation pedicles rapidly lowered their [Ca2+] below that observed in spherules. CONCLUSIONS These findings indicate that ATP demand and mitochondrial ATP production are greater in cone pedicles than rod spherules. Rod spherules employ high affinity/low turnover PMCA and their mitochondrion to maintain a relatively low [Ca2+] in darkness, which increases their sensitivity and signal-to-noise ratio. In contrast, cone pedicles utilize low affinity/high turnover NCX to rapidly lower their high [Ca2+] during light adaptation, which increases their response kinetics. Spatiotemporal fluo-3-Ca2+ imaging results support our immunocytochemical results. The clustering of cone pedicle mitochondria likely provides increased protection from Ca2+ overload and permeability transition. In summary, these novel studies reveal that several integrated cellular and subcellular components interact to regulate ATP and Ca2+ dynamics in rod and cone synaptic terminals. These results should provide a greater understanding of in vivo photoreceptor synaptic terminal exocytosis/endocytosis, Ca2+ overload and therapies for retinal degenerations.
Collapse
Affiliation(s)
- Jerry E. Johnson
- Department of Natural Sciences, University of Houston-Downtown, Houston, TX
- College of Optometry, University of Houston, Houston, TX
| | - Guy A. Perkins
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA
| | - Anand Giddabasappa
- College of Optometry, University of Houston, Houston, TX
- Department of Biology and Biochemistry, University of Houston, Houston, TX
| | - Shawntay Chaney
- College of Optometry, University of Houston, Houston, TX
- Department of Biology and Biochemistry, University of Houston, Houston, TX
| | - Weimin Xiao
- College of Optometry, University of Houston, Houston, TX
| | - Andrew D. White
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA
| | - Joshua M. Brown
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA
| | - Jenna Waggoner
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA
| | - Mark H. Ellisman
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA
- Department of Neurosciences, University of California San Diego, La Jolla, CA
| | - Donald A. Fox
- College of Optometry, University of Houston, Houston, TX
- Department of Biology and Biochemistry, University of Houston, Houston, TX
- Department of Pharmacology and Pharmaceutical Sciences, University of Houston, Houston, TX
| |
Collapse
|
11
|
Morfini G, Pigino G, Opalach K, Serulle Y, Moreira JE, Sugimori M, Llinás RR, Brady ST. 1-Methyl-4-phenylpyridinium affects fast axonal transport by activation of caspase and protein kinase C. Proc Natl Acad Sci U S A 2007; 104:2442-7. [PMID: 17287338 PMCID: PMC1892945 DOI: 10.1073/pnas.0611231104] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Parkinson's disease (PD), a late-onset condition characterized by dysfunction and loss of dopaminergic neurons in the substantia nigra, has both sporadic and neurotoxic forms. Neurotoxins such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and its metabolite 1-methyl-4-phenylpyridinium (MPP+) induce PD symptoms and recapitulate major pathological hallmarks of PD in human and animal models. Both sporadic and MPP+-induced forms of PD proceed through a "dying-back" pattern of neuronal degeneration in affected neurons, characterized by early loss of synaptic terminals and axonopathy. However, axonal and synaptic-specific effects of MPP+ are poorly understood. Using isolated squid axoplasm, we show that MPP+ produces significant alterations in fast axonal transport (FAT) through activation of a caspase and a previously undescribed protein kinase C (PKCdelta) isoform. Specifically, MPP+ increased cytoplasmic dynein-dependent retrograde FAT and reduced kinesin-1-mediated anterograde FAT. Significantly, MPP+ effects were independent of both nuclear activities and ATP production. Consistent with its effects on FAT, MPP+ injection in presynaptic domains led to a dramatic reduction in the number of membranous profiles. Changes in availability of synaptic and neurotrophin-signaling components represent axonal and synaptic-specific effects of MPP+ that would produce a dying-back pathology. Our results identify a critical neuronal process affected by MPP+ and suggest that alterations in vesicle trafficking represent a primary event in PD pathogenesis. We propose that PD and other neurodegenerative diseases exhibiting dying-back neuropathology represent a previously undescribed category of neurological diseases characterized by dysfunction of vesicle transport and associated with the loss of synaptic function.
Collapse
Affiliation(s)
- G. Morfini
- *Department of Anatomy and Cell Biology, University of Illinois, Chicago, IL 60612
- Marine Biological Laboratory, Woods Hole, MA 02543
- To whom correspondence may be addressed. E-mail:
, , or
| | - G. Pigino
- *Department of Anatomy and Cell Biology, University of Illinois, Chicago, IL 60612
- Marine Biological Laboratory, Woods Hole, MA 02543
| | - K. Opalach
- Marine Biological Laboratory, Woods Hole, MA 02543
| | - Y. Serulle
- Marine Biological Laboratory, Woods Hole, MA 02543
- Department of Physiology and Neuroscience, New York University School of Medicine New York, NY 10016; and
| | - J. E. Moreira
- Marine Biological Laboratory, Woods Hole, MA 02543
- Department of Cell and Molecular Biology, Riberão Preto School of Medicine, University of São Paulo, SP 14049-900, Ribeirão Preto, Brazil
| | - M. Sugimori
- Marine Biological Laboratory, Woods Hole, MA 02543
- Department of Physiology and Neuroscience, New York University School of Medicine New York, NY 10016; and
| | - R. R. Llinás
- Marine Biological Laboratory, Woods Hole, MA 02543
- Department of Physiology and Neuroscience, New York University School of Medicine New York, NY 10016; and
- To whom correspondence may be addressed. E-mail:
, , or
| | - S. T. Brady
- *Department of Anatomy and Cell Biology, University of Illinois, Chicago, IL 60612
- Marine Biological Laboratory, Woods Hole, MA 02543
- To whom correspondence may be addressed. E-mail:
, , or
| |
Collapse
|
12
|
Abstract
Organelle transport is vital for the development and maintenance of axons, in which the distances between sites of organelle biogenesis, function, and recycling or degradation can be vast. Movement of mitochondria in axons can serve as a general model for how all organelles move: mitochondria are easy to identify, they move along both microtubule and actin tracks, they pause and change direction, and their transport is modulated in response to physiological signals. However, they can be distinguished from other axonal organelles by the complexity of their movement and their unique functions in aerobic metabolism, calcium homeostasis and cell death. Mitochondria are thus of special interest in relating defects in axonal transport to neuropathies and degenerative diseases of the nervous system. Studies of mitochondrial transport in axons are beginning to illuminate fundamental aspects of the distribution mechanism. They use motors of one or more kinesin families, along with cytoplasmic dynein, to translocate along microtubules, and bidirectional movement may be coordinated through interaction between dynein and kinesin-1. Translocation along actin filaments is probably driven by myosin V, but the protein(s) that mediate docking with actin filaments remain unknown. Signaling through the PI 3-kinase pathway has been implicated in regulation of mitochondrial movement and docking in the axon, and additional mitochondrial linker and regulatory proteins, such as Milton and Miro, have recently been described.
Collapse
Affiliation(s)
- Peter J Hollenbeck
- Department of Biological Sciences, Purdue University, 915 West State Street, West Lafayette, IN 47907, USA.
| | | |
Collapse
|
13
|
Abstract
An abundance of mitochondria has been the hallmark of synapses since their first ultrastructural description 50 years ago. Mitochondria have been shown to be essential for synaptic form and function in many systems, but until recently it has not been clear exactly what role(s) they play in neurotransmission. Now, evidence from the nervous system of Drosophila identifies the specific subcellular events that are most dependent upon nearby mitochondria.
Collapse
Affiliation(s)
- Peter J Hollenbeck
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907, USA.
| |
Collapse
|
14
|
Jockusch WJ, Praefcke GJK, McMahon HT, Lagnado L. Clathrin-dependent and clathrin-independent retrieval of synaptic vesicles in retinal bipolar cells. Neuron 2005; 46:869-78. [PMID: 15953416 DOI: 10.1016/j.neuron.2005.05.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2004] [Revised: 03/03/2005] [Accepted: 05/02/2005] [Indexed: 10/25/2022]
Abstract
Synaptic vesicles can be retrieved rapidly or slowly, but the molecular basis of these kinetic differences has not been defined. We now show that substantially different sets of molecules mediate fast and slow endocytosis in the synaptic terminal of retinal bipolar cells. Capacitance measurements of membrane retrieval were made in terminals in which peptides and protein domains were introduced to disrupt known interactions of clathrin, the AP2 adaptor complex, and amphiphysin. All these manipulations caused a selective inhibition of the slow phase of membrane retrieval (time constant approximately 10 s), leaving the fast phase (approximately 1 s) intact. Slow endocytosis after strong stimulation was therefore dependent on the formation of clathrin-coated membrane. Fast endocytosis occurring after weaker stimuli retrieves vesicle membrane in a clathrin-independent manner. All compensatory endocytosis required GTP hydrolysis, but only a subset of released vesicles were primed for fast, clathrin-independent endocytosis.
Collapse
Affiliation(s)
- Wolf J Jockusch
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
15
|
Guo X, Macleod GT, Wellington A, Hu F, Panchumarthi S, Schoenfield M, Marin L, Charlton MP, Atwood HL, Zinsmaier KE. The GTPase dMiro Is Required for Axonal Transport of Mitochondria to Drosophila Synapses. Neuron 2005; 47:379-93. [PMID: 16055062 DOI: 10.1016/j.neuron.2005.06.027] [Citation(s) in RCA: 459] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2004] [Revised: 04/21/2005] [Accepted: 06/21/2005] [Indexed: 01/22/2023]
Abstract
We have identified EMS-induced mutations in Drosophila Miro (dMiro), an atypical mitochondrial GTPase that is orthologous to human Miro (hMiro). Mutant dmiro animals exhibit defects in locomotion and die prematurely. Mitochondria in dmiro mutant muscles and neurons are abnormally distributed. Instead of being transported into axons and dendrites, mitochondria accumulate in parallel rows in neuronal somata. Mutant neuromuscular junctions (NMJs) lack presynaptic mitochondria, but neurotransmitter release and acute Ca2+ buffering is only impaired during prolonged stimulation. Neuronal, but not muscular, expression of dMiro in dmiro mutants restored viability, transport of mitochondria to NMJs, the structure of synaptic boutons, the organization of presynaptic microtubules, and the size of postsynaptic muscles. In addition, gain of dMiro function causes an abnormal accumulation of mitochondria in distal synaptic boutons of NMJs. Together, our findings suggest that dMiro is required for controlling anterograde transport of mitochondria and their proper distribution within nerve terminals.
Collapse
Affiliation(s)
- Xiufang Guo
- Arizona Research Laboratories, Division of Neurobiology, University of Arizona, Tucson, Arizona 85721, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
|
17
|
Abstract
Over the past decade, a remarkable number and diversity of molecular motors have been described in eukaryotic cells. In addition to the identification of novel forms of myosin and dynein, the kinesins have been defined as an entirely new family of molecular motors. There may be as many as 30 different genes in a single organism encoding members of the kinesin superfamily. Why is such diversity in molecular motors needed? The biochemical and functional diversity of the originally defined form of kinesin provides some insights into the roles of molecular motors in cellular dynamics.
Collapse
Affiliation(s)
- S T Brady
- Dept of Cell Biology and Neuroscience, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75235-9111, USA
| |
Collapse
|
18
|
Morfini G, Szebenyi G, Brown H, Pant HC, Pigino G, DeBoer S, Beffert U, Brady ST. A novel CDK5-dependent pathway for regulating GSK3 activity and kinesin-driven motility in neurons. EMBO J 2004; 23:2235-45. [PMID: 15152189 PMCID: PMC419914 DOI: 10.1038/sj.emboj.7600237] [Citation(s) in RCA: 221] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2003] [Accepted: 04/20/2004] [Indexed: 11/09/2022] Open
Abstract
Neuronal transmission of information requires polarized distribution of membrane proteins within axonal compartments. Membrane proteins are synthesized and packaged in membrane-bounded organelles (MBOs) in neuronal cell bodies and later transported to axons by microtubule-dependent motor proteins. Molecular mechanisms underlying targeted delivery of MBOs to discrete axonal subdomains (i.e. nodes of Ranvier or presynaptic terminals) are poorly understood, but regulatory pathways for microtubule motors may be an essential step. In this work, pharmacological, biochemical and in vivo experiments define a novel regulatory pathway for kinesin-driven motility in axons. This pathway involves enzymatic activities of cyclin-dependent kinase 5 (CDK5), protein phosphatase 1 (PP1) and glycogen synthase kinase-3 (GSK3). Inhibition of CDK5 activity in axons leads to activation of GSK3 by PP1, phosphorylation of kinesin light chains by GSK3 and detachment of kinesin from transported cargoes. We propose that regulating the activity and localization of components in this pathway allows nerve cells to target organelle delivery to specific subcellular compartments. Implications of these findings for pathogenesis of neurodegenerative diseases such as Alzheimer's disease are discussed.
Collapse
Affiliation(s)
- Gerardo Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
- Marine Biological Laboratory, Woods Hole, MA, USA
| | - Györgyi Szebenyi
- Department of Cell Biology and Center for Basic Neuroscience, UT Southwestern, Dallas, TX, USA
| | - Hannah Brown
- Marine Biological Laboratory, Woods Hole, MA, USA
| | - Harish C Pant
- Marine Biological Laboratory, Woods Hole, MA, USA
- Laboratory of Neurochemistry, NINDS, Bethesda, MD, USA
| | - Gustavo Pigino
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Scott DeBoer
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Uwe Beffert
- Department of Molecular Genetics, UT Southwestern, Dallas, TX, USA
| | - Scott T Brady
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
- Marine Biological Laboratory, Woods Hole, MA, USA
- Anatomy and Cell Biology M/C 512, 808 S Wood St, University of Illinois at Chicago, Chicago, IL 60612, USA. Tel.: +1 312 996 6791; Fax: +1 312 413 0354; E-mail:
| |
Collapse
|
19
|
Morfini G, Pigino G, Beffert U, Busciglio J, Brady ST. Fast axonal transport misregulation and Alzheimer's disease. Neuromolecular Med 2003; 2:89-99. [PMID: 12428805 DOI: 10.1385/nmm:2:2:089] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2002] [Accepted: 06/07/2002] [Indexed: 11/11/2022]
Abstract
Pathological alterations in the microtubule-associated protein (MAP) tau are well-established in a number of neurodegenerative disorders, including Alzheimer's Disease (AD), frontotemporal dementia (FTD), progressive supranuclear palsy (PSP), and others. Tau protein and in some cases, neurofilament subunits exhibit abnormal phosphorylation on specific serine and threonine residues in these diseases. A large body of biochemical, genetic, and cell biological evidence implicate two major serine-threonine protein kinases, glycogen synthase kinase 3 (GSK-3) and cyclin-dependent kinase 5 (CDK5) as major kinases responsible for both normal and pathological phosphorylation of tau protein in vivo. What remains unclear is whether tau phosphorylation and/or neurofibrillary tangle (NFT) formation are causal or secondary to initiation of neuronal pathology. In fact, many studies have indicated that tau misphosphorylation is not the causal event. Interestingly, some of these kinase and phosphatase activities have recently merged as key regulators of fast axonal transport (FAT). Specifically, CDK5 and GSK-3 have been recently shown to regulate kinesin-driven motility. Given the essential role of FAT in neuronal function, an alternate model for pathogenesis can be proposed. In this model, misregulation of FAT induced by an imbalance in specific kinase-phosphatase activities within neurons represents an early and critical step for the initiation of neuronal pathology. Such a model may explain many of the unique characteristics of late onset of neurological diseases such as AD.
Collapse
Affiliation(s)
- Gerardo Morfini
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas 75390-9039, USA
| | | | | | | | | |
Collapse
|
20
|
Gunawardena S, Goldstein LSB. Cargo-carrying motor vehicles on the neuronal highway: Transport pathways and neurodegenerative disease. ACTA ACUST UNITED AC 2003; 58:258-71. [PMID: 14704957 DOI: 10.1002/neu.10319] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Within axons vital cargoes must be transported over great distances along microtubule tracks to maintain neuronal viability. Essential to this system are the molecular motors, kinesin and dynein, which transport a variety of neuronal cargoes. Elucidating the transport pathways, the identity of the cargoes transported, and the regulation of motor-cargo complexes are areas of intense investigation. Evidence suggests that essential components, including signaling proteins, neuroprotective and repair molecules, and vesicular and cytoskeletal components are all transported. In addition newly emerging data indicate that defects in axonal transport pathways may contribute to the initiation or progression of chronic neuronal dysfunction. In this review we concentrate on microtubule-based motor proteins, their linkers, and cargoes and discuss how factors in the axonal transport pathway contribute to disease states. As additional cargo complexes and transport pathways are identified, an understanding of the role these pathways play in the development of human disease will hopefully lead to new diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Shermali Gunawardena
- Department of Cellular and Molecular Medicine, Howard Hughes Medical Institute, School of Medicine, University of California, San Diego, La Jolla, California 92093-0683, USA
| | | |
Collapse
|
21
|
Mateer SC, McDaniel AE, Nicolas V, Habermacher GM, Lin MJS, Cromer DA, King ME, Bloom GS. The mechanism for regulation of the F-actin binding activity of IQGAP1 by calcium/calmodulin. J Biol Chem 2002; 277:12324-33. [PMID: 11809768 DOI: 10.1074/jbc.m109535200] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
IQGAP1 colocalizes with actin filaments in the cell cortex and binds in vitro to F-actin and several signaling proteins, including calmodulin, Cdc42, Rac1, and beta-catenin. It is thought that the F-actin binding activity of IQGAP1 is regulated by its reversible association with these signaling molecules, but the mechanisms have remained obscure. Here we describe the regulatory mechanism for calmodulin. Purified adrenal IQGAP1 was found to consist of two distinct protein pools, one of which bound F-actin and lacked calmodulin, and the other of which did not bind F-actin but was tightly associated with calmodulin. Based on this finding we hypothesized that calmodulin negatively regulates binding of IQGAP1 to F-actin. This hypothesis was tested in vitro using recombinant wild type and mutated IQGAP1s and in live cells that transiently expressed IQGAP1-YFP. In vitro, the affinity of wild type IQGAP1 for F-actin decreased with increasing concentrations of calmodulin, and this effect was dramatically enhanced by Ca(2+) and required the IQ domains of IQGAP1. In addition, we found that calmodulin bound wild type IQGAP1 much more efficiently in the presence of Ca(2+) than EGTA, and all 8 IQ motifs in each IQGAP1 dimer could bind calmodulin simultaneously. In live cells, IQGAP1-YFP localized to the cell cortex, but elevation of intracellular Ca(2+) reversibly induced the fluorescent fusion protein to become diffusely distributed. Taken together, these results support a model in which a rise in free intracellular Ca(2+) promotes binding of calmodulin to IQGAP1, which in turn inhibits IQGAP1 from binding to cortical actin filaments.
Collapse
Affiliation(s)
- Scott C Mateer
- Departments of Biology and Cell Biology, University of Virginia, Charlottesville, Virginia 22903, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Morfini G, Szebenyi G, Elluru R, Ratner N, Brady ST. Glycogen synthase kinase 3 phosphorylates kinesin light chains and negatively regulates kinesin-based motility. EMBO J 2002; 21:281-93. [PMID: 11823421 PMCID: PMC125832 DOI: 10.1093/emboj/21.3.281] [Citation(s) in RCA: 310] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2001] [Revised: 11/26/2001] [Accepted: 11/26/2001] [Indexed: 12/20/2022] Open
Abstract
Membrane-bounded organelles (MBOs) are delivered to different domains in neurons by fast axonal transport. The importance of kinesin for fast antero grade transport is well established, but mechanisms for regulating kinesin-based motility are largely unknown. In this report, we provide biochemical and in vivo evidence that kinesin light chains (KLCs) interact with and are in vivo substrates for glycogen synthase kinase 3 (GSK3). Active GSK3 inhibited anterograde, but not retrograde, transport in squid axoplasm and reduced the amount of kinesin bound to MBOs. Kinesin microtubule binding and microtubule-stimulated ATPase activities were unaffected by GSK3 phosphorylation of KLCs. Active GSK3 was also localized preferentially to regions known to be sites of membrane delivery. These data suggest that GSK3 can regulate fast anterograde axonal transport and targeting of cargos to specific subcellular domains in neurons.
Collapse
Affiliation(s)
- Gerardo Morfini
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9039, Marine Biological Laboratory, Woods Hole, MA 02543 and Department of Cell Biology and Anatomy, University of Cincinnati, Cincinnati, OH, USA Present address: Department of Otolaryngology, Washington University School of Medicine, St Louis, MO, USA Corresponding author at: Department of Cell Biology, University of Texas, Southwestern Medical Center, Dallas, TX 75390-9039, USA e-mail:
| | - Györgyi Szebenyi
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9039, Marine Biological Laboratory, Woods Hole, MA 02543 and Department of Cell Biology and Anatomy, University of Cincinnati, Cincinnati, OH, USA Present address: Department of Otolaryngology, Washington University School of Medicine, St Louis, MO, USA Corresponding author at: Department of Cell Biology, University of Texas, Southwestern Medical Center, Dallas, TX 75390-9039, USA e-mail:
| | - Ravindhra Elluru
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9039, Marine Biological Laboratory, Woods Hole, MA 02543 and Department of Cell Biology and Anatomy, University of Cincinnati, Cincinnati, OH, USA Present address: Department of Otolaryngology, Washington University School of Medicine, St Louis, MO, USA Corresponding author at: Department of Cell Biology, University of Texas, Southwestern Medical Center, Dallas, TX 75390-9039, USA e-mail:
| | - Nancy Ratner
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9039, Marine Biological Laboratory, Woods Hole, MA 02543 and Department of Cell Biology and Anatomy, University of Cincinnati, Cincinnati, OH, USA Present address: Department of Otolaryngology, Washington University School of Medicine, St Louis, MO, USA Corresponding author at: Department of Cell Biology, University of Texas, Southwestern Medical Center, Dallas, TX 75390-9039, USA e-mail:
| | - Scott T. Brady
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9039, Marine Biological Laboratory, Woods Hole, MA 02543 and Department of Cell Biology and Anatomy, University of Cincinnati, Cincinnati, OH, USA Present address: Department of Otolaryngology, Washington University School of Medicine, St Louis, MO, USA Corresponding author at: Department of Cell Biology, University of Texas, Southwestern Medical Center, Dallas, TX 75390-9039, USA e-mail:
| |
Collapse
|
23
|
Reilein AR, Rogers SL, Tuma MC, Gelfand VI. Regulation of molecular motor proteins. INTERNATIONAL REVIEW OF CYTOLOGY 2001; 204:179-238. [PMID: 11243595 DOI: 10.1016/s0074-7696(01)04005-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Motor proteins in the kinesin, dynein, and myosin superfamilies are tightly regulated to perform multiple functions in the cell requiring force generation. Although motor proteins within families are diverse in sequence and structure, there are general mechanisms by which they are regulated. We first discuss the regulation of the subset of kinesin family members for which such information exists, and then address general mechanisms of kinesin family regulation. We review what is known about the regulation of axonemal and cytoplasmic dyneins. Recent work on cytoplasmic dynein has revealed the existence of multiple isoforms for each dynein chain, making the study of dynein regulation more complicated than previously realized. Finally, we discuss the regulation of myosins known to be involved in membrane trafficking. Myosins and kinesins may be evolutionarily related, and there are common themes of regulation between these two classes of motors.
Collapse
Affiliation(s)
- A R Reilein
- Department of Cell and Structural Biology, University of Illinois, Urbana-Champaign, Urbana 61801, USA
| | | | | | | |
Collapse
|
24
|
Tsai MY, Morfini G, Szebenyi G, Brady ST. Release of kinesin from vesicles by hsc70 and regulation of fast axonal transport. Mol Biol Cell 2000; 11:2161-73. [PMID: 10848636 PMCID: PMC14910 DOI: 10.1091/mbc.11.6.2161] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The nature of kinesin interactions with membrane-bound organelles and mechanisms for regulation of kinesin-based motility have both been surprisingly difficult to define. Most kinesin is recovered in supernatants with standard protocols for purification of motor proteins, but kinesin recovered on membrane-bound organelles is tightly bound. Partitioning of kinesin between vesicle and cytosolic fractions is highly sensitive to buffer composition. Addition of either N-ethylmaleimide or EDTA to homogenization buffers significantly increased the fraction of kinesin bound to organelles. Given that an antibody against kinesin light chain tandem repeats also releases kinesin from vesicles, these observations indicated that specific cytoplasmic factors may regulate kinesin release from membranes. Kinesin light tandem repeats contain DnaJ-like motifs, so the effects of hsp70 chaperones were evaluated. Hsc70 released kinesin from vesicles in an MgATP-dependent and N-ethylmaleimide-sensitive manner. Recombinant kinesin light chains inhibited kinesin release by hsc70 and stimulated the hsc70 ATPase. Hsc70 actions may provide a mechanism to regulate kinesin function by releasing kinesin from cargo in specific subcellular domains, thereby effecting delivery of axonally transported materials.
Collapse
Affiliation(s)
- M Y Tsai
- Department of Cell Biology and Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9039, USA
| | | | | | | |
Collapse
|
25
|
Kaether C, Skehel P, Dotti CG. Axonal membrane proteins are transported in distinct carriers: a two-color video microscopy study in cultured hippocampal neurons. Mol Biol Cell 2000; 11:1213-24. [PMID: 10749925 PMCID: PMC14842 DOI: 10.1091/mbc.11.4.1213] [Citation(s) in RCA: 225] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Neurons transport newly synthesized membrane proteins along axons by microtubule-mediated fast axonal transport. Membrane proteins destined for different axonal subdomains are thought to be transported in different transport carriers. To analyze this differential transport in living neurons, we tagged the amyloid precursor protein (APP) and synaptophysin (p38) with green fluorescent protein (GFP) variants. The resulting fusion proteins, APP-yellow fluorescent protein (YFP), p38-enhanced GFP, and p38-enhanced cyan fluorescent protein, were expressed in hippocampal neurons, and the cells were imaged by video microscopy. APP-YFP was transported in elongated tubules that moved extremely fast (on average 4.5 micrometer/s) and over long distances. In contrast, p38-enhanced GFP-transporting structures were more vesicular and moved four times slower (0.9 micrometer/s) and over shorter distances only. Two-color video microscopy showed that the two proteins were sorted to different carriers that moved with different characteristics along axons of doubly transfected neurons. Antisense treatment using oligonucleotides against the kinesin heavy chain slowed down the long, continuous movement of APP-YFP tubules and increased frequency of directional changes. These results demonstrate for the first time directly the sorting and transport of two axonal membrane proteins into different carriers. Moreover, the extremely fast-moving tubules represent a previously unidentified type of axonal carrier.
Collapse
Affiliation(s)
- C Kaether
- European Molecular Biology Laboratory, Cell Biology Program, 69012 Heidelberg, Germany
| | | | | |
Collapse
|
26
|
Goldstein LS, Philp AV. The road less traveled: emerging principles of kinesin motor utilization. Annu Rev Cell Dev Biol 1999; 15:141-83. [PMID: 10611960 DOI: 10.1146/annurev.cellbio.15.1.141] [Citation(s) in RCA: 193] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Proteins of the kinesin superfamily utilize a conserved catalytic motor domain to generate movements in a wide variety of cellular processes. In this review, we discuss the rapid expansion in our understanding of how eukaryotic cells take advantage of these proteins to generate force and movement in diverse functional contexts. We summarize several recent examples revealing that the simplest view of a kinesin motor protein binding to and translocating a cargo along a microtubule track is inadequate. In fact, this paradigm captures only a small subset of the many ways in which cells harness force production of the generation of intracellular movements and functions. We also highlight several situations where the catalytic kinesin motor domain may not be used to generate movement, but instead may be used in other biochemical and functional contexts. Finally, we review some recent ideas about kinesin motor regulation, redundancy, and cargo attachment strategies.
Collapse
Affiliation(s)
- L S Goldstein
- Howard Hughes Medical Institute, Department of Pharmacology, University of California San Diego, La Jolla 92093-0683, USA.
| | | |
Collapse
|
27
|
Abstract
Research over the past 18 months has revealed that many membranous organelles move along both actin filaments and microtubules. It is highly likely that the activity of the microtubule motors, myosins and static linker proteins present on any organelle are co-ordinately regulated and that this control is linked to the processes of membrane traffic itself.
Collapse
Affiliation(s)
- V J Allan
- School of Biological Sciences, University of Manchester, 2.205 Stopford Building, Oxford Road, Manchester, M13 9PT, UK.
| | | |
Collapse
|
28
|
A role for cyclin-dependent kinase(s) in the modulation of fast anterograde axonal transport: effects defined by olomoucine and the APC tumor suppressor protein. J Neurosci 1998. [PMID: 9742142 DOI: 10.1523/jneurosci.18-19-07717.1998] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Proteins that interact with both cytoskeletal and membrane components are candidates to modulate membrane trafficking. The tumor suppressor proteins neurofibromin (NF1) and adenomatous polyposis coli (APC) both bind to microtubules and interact with membrane-associated proteins. The effects of recombinant NF1 and APC fragments on vesicle motility were evaluated by measuring fast axonal transport along microtubules in axoplasm from squid giant axons. APC4 (amino acids 1034-2844) reduced only anterograde movements, whereas APC2 (aa 1034-2130) or APC3 (aa 2130-2844) reduced both anterograde and retrograde transport. NF1 had no effect on organelle movement in either direction. Because APC contains multiple cyclin-dependent kinase (CDK) consensus phosphorylation motifs, the kinase inhibitor olomoucine was examined. At concentrations in which olomoucine is specific for cyclin-dependent kinases (5 microM), it reduced only anterograde transport, whereas anterograde and retrograde movement were both affected at concentrations at which other kinases are inhibited as well (50 microM). Both anterograde and retrograde transport also were inhibited by histone H1 and KSPXK peptides, substrates for proline-directed kinases, including CDKs. Our data suggest that CDK-like axonal kinases modulate fast anterograde transport and that other axonal kinases may be involved in modulating retrograde transport. The specific effect of APC4 on anterograde transport suggests a model in which the binding of APC to microtubules may limit the activity of axonal CDK kinase or kinases in restricted domains, thereby affecting organelle transport.
Collapse
|
29
|
Fullerton AT, Bau MY, Conrad PA, Bloom GS. In vitro reconstitution of microtubule plus end-directed, GTPgammaS-sensitive motility of Golgi membranes. Mol Biol Cell 1998; 9:2699-714. [PMID: 9763438 PMCID: PMC25545 DOI: 10.1091/mbc.9.10.2699] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/1997] [Accepted: 07/14/1998] [Indexed: 11/11/2022] Open
Abstract
Purified Golgi membranes were mixed with cytosol and microtubules (MTs) and observed by video enhanced light microscopy. Initially, the membranes appeared as vesicles that moved along MTs. As time progressed, vesicles formed aggregates from which membrane tubules emerged, traveled along MTs, and eventually generated extensive reticular networks. Membrane motility required ATP, occurred mainly toward MT plus ends, and was inhibited almost completely by the H1 monoclonal antibody to kinesin heavy chain, 5'-adenylylimidodiphosphate, and 100 microM but not 20 microM vanadate. Motility was also blocked by GTPgammaS or A1F4- but was insensitive to A1C13, NaF, staurosporin, or okadaic acid. The targets for GTPgammaS and A1F4- were evidently of cytosolic origin, did not include kinesin or MTs, and were insensitive to several probes for trimeric G proteins. Transport of Golgi membranes along MTs mediated by a kinesin has thus been reconstituted in vitro. The motility is regulated by one or more cytosolic GTPases but not by protein kinases or phosphatases that are inhibited by staurosporin or okadaic acid, respectively. The pertinent GTPases are likely to be small G proteins or possibly dynamin. The in vitro motility may correspond to Golgi-to-ER or Golgi-to-cell surface transport in vivo.
Collapse
Affiliation(s)
- A T Fullerton
- Department of Cell Biology and Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | | | | | | |
Collapse
|
30
|
Ratner N, Bloom GS, Brady ST. A role for cyclin-dependent kinase(s) in the modulation of fast anterograde axonal transport: effects defined by olomoucine and the APC tumor suppressor protein. J Neurosci 1998; 18:7717-26. [PMID: 9742142 PMCID: PMC6793030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Proteins that interact with both cytoskeletal and membrane components are candidates to modulate membrane trafficking. The tumor suppressor proteins neurofibromin (NF1) and adenomatous polyposis coli (APC) both bind to microtubules and interact with membrane-associated proteins. The effects of recombinant NF1 and APC fragments on vesicle motility were evaluated by measuring fast axonal transport along microtubules in axoplasm from squid giant axons. APC4 (amino acids 1034-2844) reduced only anterograde movements, whereas APC2 (aa 1034-2130) or APC3 (aa 2130-2844) reduced both anterograde and retrograde transport. NF1 had no effect on organelle movement in either direction. Because APC contains multiple cyclin-dependent kinase (CDK) consensus phosphorylation motifs, the kinase inhibitor olomoucine was examined. At concentrations in which olomoucine is specific for cyclin-dependent kinases (5 microM), it reduced only anterograde transport, whereas anterograde and retrograde movement were both affected at concentrations at which other kinases are inhibited as well (50 microM). Both anterograde and retrograde transport also were inhibited by histone H1 and KSPXK peptides, substrates for proline-directed kinases, including CDKs. Our data suggest that CDK-like axonal kinases modulate fast anterograde transport and that other axonal kinases may be involved in modulating retrograde transport. The specific effect of APC4 on anterograde transport suggests a model in which the binding of APC to microtubules may limit the activity of axonal CDK kinase or kinases in restricted domains, thereby affecting organelle transport.
Collapse
Affiliation(s)
- N Ratner
- Department of Cell Biology, Neurobiology, and Anatomy, University of Cincinnati School of Medicine, Cincinnati, Ohio 45267-0521, USA
| | | | | |
Collapse
|
31
|
Affiliation(s)
- J Lane
- School of Biological Sciences, University of Manchester, 2.205 Stopford Building, Oxford Road, Manchester, M13 9PT, UK.
| | | |
Collapse
|
32
|
Marlowe KJ, Farshori P, Torgerson RR, Anderson KL, Miller LJ, McNiven MA. Changes in kinesin distribution and phosphorylation occur during regulated secretion in pancreatic acinar cells. Eur J Cell Biol 1998; 75:140-52. [PMID: 9548371 DOI: 10.1016/s0171-9335(98)80056-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In secretory cells, microtubule- (Mt-) based motor enzymes are thought to support transport of secretory vesicles to the cell surface for subsequent release. At present, the role of Mts and kinesin in secretory vesicle transport in exocrine epithelial cells has not been defined. Furthermore, it is unclear whether an agonist-induced secretory event modifies kinesin function and distribution, thus altering vesicle transport. To this end, we utilized isolated rat pancreatic acini and cultured rat pancreatic acinar cells to examine the role of Mts and kinesin in regulated secretion. Exposure of cells to cytoskeletal antagonistic drugs demonstrated that the observed movements of apically clustered zymogen granules (ZGs) are supported by Mts, but not actin. Morphological studies of Mt organization in polarized acini show that Mt plus ends extend outward from the apical membrane toward the cell center. Immunofluorescence microscopy in both cell models revealed a clear association of kinesin with apical ZGs, while quantitative immunoblot analysis of pancreatic subcellular fractions confirmed kinesin enrichment on ZG membranes. In addition, microinjection of kinesin antibodies into cultured acinar cells inhibited ZG movements. Indirect immunofluorescence staining of isolated cells and quantitative Western blotting of isolated ZGs revealed that kinesin association with granule membranes increased up to 3-fold in response to a secretory stimulus. Autoradiographic studies of 32P-labeled acini showed up to a 6-fold increase in kinesin heavy chain (KHC) phosphorylation during stimulated secretion. These studies provide the first direct evidence that Mts and kinesin support ZG movements and that physiological agonists induce a marked phosphorylation of KHC while increasing the association of kinesin with ZG membranes. These changes during agonist stimulation suggest that the participation of kinesin in zymogen secretion is regulated.
Collapse
Affiliation(s)
- K J Marlowe
- Center for Basic Research in Digestive Diseases, Mayo Clinic and Foundation, Rochester, MN 55905, USA
| | | | | | | | | | | |
Collapse
|
33
|
Malchiodi-Albedi F, Petrucci TC, Picconi B, Iosi F, Falchi M. Protein phosphatase inhibitors induce modification of synapse structure and tau hyperphosphorylation in cultured rat hippocampal neurons. J Neurosci Res 1997. [DOI: 10.1002/(sici)1097-4547(19970601)48:5<425::aid-jnr4>3.0.co;2-g] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
34
|
Tang HZ, Hammerschlag R. Effects of exogenous triiodothyronine on fast axonal transport during tadpole metamorphosis. Neurochem Res 1996; 21:489-94. [PMID: 8734443 DOI: 10.1007/bf02527714] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Bullfrog tadpoles at metamorphic stages V, X and XVIII were immersed in 25 nM triiodothyronine (T3) to assess whether the 4-5 fold increase in fast axonal transport (FAxT) previously observed during this span of spontaneous metamorphosis (1) could be mimicked by precocious application of thyroid hormone. The trend initially observed was for T3 to stimulated [35S]methionine incorporation into lumbar DRG and inhibit incorporation in tail DRG. Both effects, however, appeared to be exerted primarily on satellite cells rather than neurons since most of the T3-induced changes in DRG were of a similar magnitude to those in the respective nerve trunks. Findings consistent with this observation resulted from use of the retrogradely transported lectin, ricin120, to determine the proportion of DRG incorporation occurring in neurons. When incorporation of [35S]methionine in lumbar DRG neurons was examined, T3 had no stimulatory effect at any of the metamorphic stages examined. When FAxT was assessed as [35S]protein accumulating proximal to a nerve trunk ligature, and expressed as a percentage of newly-synthesized protein in lumbar DRG neurons, no stimulatory effect of T3 was detected. The question remains whether the changes in FAxT in peripheral neurons observed during spontaneous metamorphosis may be induced by circulating hormones other than T3 or are secondary to changes in the target tissues.
Collapse
Affiliation(s)
- H Z Tang
- Division of Neurosciences, Beckman Research Institute of the City of Hope, Duarte, California 91010, USA
| | | |
Collapse
|
35
|
Abstract
The fact that multiple microtubule-based motors exist in brain inevitably raises questions about their function. Transcripts for at least seven kinesin superfamily genes and even more dynein heavy chain genes have been detected in brain cDNA libraries. The challenge now is to match their gene products to specific functions in cells of the nervous system. Recent studies have attempted to establish a function for each microtubule motor by using recombinant protein and immunochemical approaches.
Collapse
Affiliation(s)
- S T Brady
- Department of Cell Biology and Neuroscience, University of Texas Southwestern Medical Center, Dallas 75235-9111, USA.
| | | |
Collapse
|
36
|
Tang HZ, Hammerschlag R, Chan H, Smith RS. Changes in fast axonal transport in sensory neurons during tadpole metamorphosis. Int J Dev Neurosci 1995; 13:473-81. [PMID: 7484218 DOI: 10.1016/0736-5748(95)00007-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Fast axonal transport of radiolabeled protein was examined in lumbar and tail dorsal root ganglion (DRG) neurons at progressive stages of bullfrog tadpole metamorphosis. Accumulation of [35S]methionine-labeled protein proximal to a lumbar peripheral nerve ligature (at a fixed distance from the DRG) increased as tadpoles advanced from premetamorphosis through prometamorphosis to metamorphic climax. The rate of increase was steeper when expressed as a percentage of protein synthesized in the neurons of origin than when expressed as a percentage of total DRG protein synthesis. Further, the increase was not secondary to a rise in protein synthesis. In contrast, fast axonal transport decreased in DRG neurons of the tail at the onset of metamorphic climax, when tail resorption is initiated. The stage-related increase in protein transport in lumbar nerves is due, at least in part, to an increased rate of transport. As determined from optically detected anterograde organelles in individual lumbar nerve axons, an approximate doubling of the fast transport rate occurred between the premetamorphic stage and metamorphic climax. In addition, the rates of organelle transport in lumbar axons of adult bullfrogs were significantly greater than in corresponding axons of tadpoles at metamorphic climax, further suggesting that organelle velocity is a developmentally regulated parameter of fast axonal transport.
Collapse
Affiliation(s)
- H Z Tang
- Division of Neurosciences, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | | | | | | |
Collapse
|
37
|
Gross SD, Hoffman DP, Fisette PL, Baas P, Anderson RA. A phosphatidylinositol 4,5-bisphosphate-sensitive casein kinase I alpha associates with synaptic vesicles and phosphorylates a subset of vesicle proteins. J Cell Biol 1995; 130:711-24. [PMID: 7622570 PMCID: PMC2120523 DOI: 10.1083/jcb.130.3.711] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
In interphase cells, alpha-casein kinase I (alpha-CKI) is found associated with cytosolic vesicular structures, the centrosome, and within the nucleus. To identify the specific vesicular structures with which alpha-CKI is associated, established cell lines and primary rat neurons were immunofluorescently labeled with an antibody raised to alpha-CKI. In nonneuronal cells, alpha-CKI colocalizes with vesicular structures which align with microtubules and are partially coincident with both Golgi and endoplasmic reticulum markers. In neurons, alpha-CKI colocalizes with synaptic vesicle markers. When synaptic vesicles were purified from rat brain, they were highly enriched in a CKI, based on activity and immunoreactivity. The synaptic vesicle-associated CKI is an extrinsic kinase and was eluted from synaptic vesicles and purified. This purified CKI has properties most similar to alpha-CKI. When the activities of casein kinase I or II were specifically inhibited on isolated synaptic vesicles, CKI was shown to phosphorylate a specific subset of vesicle proteins, one of which was identified as the synaptic vesicle-specific protein SV2. As with alpha-CKI, the synaptic vesicle CKI is inhibited by phosphatidylinositol 4,5-bisphosphate (PIP2). However, synthesis of PIP2 was detected only in plasma membrane-containing fractions. Therefore, PIP2 may spatially regulate CKI. Since PIP2 synthesis is required for secretion, this inhibition of CKI may be important for the regulation of secretion.
Collapse
Affiliation(s)
- S D Gross
- Department of Pharmacology, University of Wisconsin Medical School, Madison 53706, USA
| | | | | | | | | |
Collapse
|
38
|
Allan V. Protein phosphatase 1 regulates the cytoplasmic dynein-driven formation of endoplasmic reticulum networks in vitro. J Cell Biol 1995; 128:879-91. [PMID: 7876311 PMCID: PMC2120396 DOI: 10.1083/jcb.128.5.879] [Citation(s) in RCA: 97] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Interphase Xenopus egg extracts form extensive tubular membrane networks in vitro. These networks are identified here as endoplasmic reticulum by the presence of ER resident proteins, as shown by immunofluorescence, and by the presence of single ribosomes and polysomes, as shown by electron microscopy. The effect of phosphorylation on ER movement in interphase was tested using the phosphatase inhibitor, okadaic acid. Okadaic acid treatment resulted in an increase of up to 27-fold in the number of ER tubules moving and in the extent of ER networks formed compared to control extracts. This activation was blocked by the broad-specificity kinase inhibitor 6-dimethylaminopurine. Okadaic acid had no effect, however, on the direction of ER tubule movement, which occurred towards the minus end of microtubules, and was sensitive to low concentrations of vanadate. Inhibition of phosphatases also had no effect on the speed or duration of ER tubule extensions, and did not stimulate the activity of soluble cytoplasmic dynein. The sensitivity of ER movement to okadaic acid closely matched that of protein phosphatase 1. Although the amount of ER motility was greatly increased by inhibiting protein phosphatase 1 (PP1), the amount of cytoplasmic dynein associated with the membrane was not altered. The data support a model in which phosphorylation regulates ER movement by controlling the activity of cytoplasmic dynein bound to the ER membrane.
Collapse
Affiliation(s)
- V Allan
- Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| |
Collapse
|
39
|
Smith RS, Snyder RE. Anterograde to retrograde reversal of fast axonal transport within cold blocked and rewarmed intact axons. Brain Res 1995; 672:205-13. [PMID: 7749742 DOI: 10.1016/0006-8993(94)01341-e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The possibility that anterograde to retrograde reversal of axonal transport might take place in mid axon at a site distant from any nerve termination was investigated in sciatic nerve preparations from Xenopus laevis. The nerve, containing a pulse of anterogradely transported protein labeled with [35S]methionine, was kept in a two-compartment temperature controlled chamber. One compartment containing the proximal nerve was maintained at room temperature throughout the duration of an experiment while the second compartment containing the distal nerve, and separated from the first by a thermal barrier, was initially cooled to 3-4 degrees C and later warmed to room temperature. Transport of labeled proteins in the nerve was detected with a position-sensitive detector of ionizing radiation. With the distal portion of the nerve cold, the pulse of labeled protein transported up to the thermal barrier and stopped. When the distal part of the nerve was warmed to room temperature, retrograde and anterograde pulses of label propagated away from the thermal barrier with no time delay. The retrograde pulse could be collected on the distal side of a proximally placed tie and could be eliminated by treatment of the proximal nerve with vinblastine or dinitrophenol. Functional and structural evidence indicated that the cold block and thermal barrier were not destructive to the axons. Electron microscopy showed that the numerical density of axonal microtubules distal to the cold block was decreased about seven fold during the cold treatment and that this decrease could be prevented by 10 mumol/l taxol.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- R S Smith
- Department of Anatomy, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
40
|
Abstract
Molecular cloning studies have provided valuable structural information about the different subunits of cytoplasmic dynein and their relationships to their axonemal dynein counterparts. Recent unexpected findings regarding the role of this enzyme in mitosis have emerged from mutational analyses and microinjection experiments, while studies of organelle transport in vivo have revealed clues to mechanisms for physiological regulation of dynein activity.
Collapse
Affiliation(s)
- T A Schroer
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218
| |
Collapse
|
41
|
Abstract
Great advances in the field of axonal transport have been made in the past year, including the identification of new molecular motors associated with microtubules and actin. In addition, studies on the mechanisms of bidirectional fast axonal transport have clarified new aspects of this process, such as the isolation of a kinesin-binding protein, kinectin, and the finding that phosphorylation regulates kinesin's dissociation from membranous organelles. New approaches to studying slow transport of cytoskeletal proteins have provided further evidence that the axonal cytoskeleton in mammalian systems is largely stationary, although a dynamic exchange occurs between polymers and a small pool of moving subunits.
Collapse
Affiliation(s)
- N Hirokawa
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Tokyo, Japan
| |
Collapse
|
42
|
Tixier-Vidal A, Barret A, Picart R, Mayau V, Vogt D, Wiedenmann B, Goud B. The small GTP-binding protein, Rab6p, is associated with both Golgi and post-Golgi synaptophysin-containing membranes during synaptogenesis of hypothalamic neurons in culture. J Cell Sci 1993; 105 ( Pt 4):935-47. [PMID: 8227215 DOI: 10.1242/jcs.105.4.935] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have recently localized a small GTP-binding protein (Rab6p) thought to be involved in vesicular membrane transport, to the medial and trans-cisternae of the Golgi apparatus in NRK (normal rat kidney) cells. Here, we have localized and quantified Rab6p during the development in culture of embryonic neurons, up to synapse formation, and compared its subcellular distribution and level of expression to that of synaptophysin, a major integral membrane protein of small synaptic vesicles. Using immunocytochemistry (laser scanning confocal microscopy, immunoelectron microscopy), fractionation and immunoisolation methods, we show that during the early phase of synaptogenesis, Rab6p is associated with synaptophysin-containing membranes of a trans-Golgi subcompartment, post-Golgi vesicles and small synaptic vesicles or their precursors. Concomitantly, Rab6p undergoes translocation from cytosol to membranes and its level of expression increases. However, at late stages, the association of Rab6p to small synaptic vesicles sharply decreases and its level of expression plateaus. These findings suggest a role for Rab6p in the post-Golgi transport of synaptophysin, at an early step of the biogenesis of small synaptic vesicles.
Collapse
Affiliation(s)
- A Tixier-Vidal
- Groupe de Neuroendocrinologie Cellulaire et Moléculaire, Collège de France (URA CNRS 1115), Paris
| | | | | | | | | | | | | |
Collapse
|