Skip to main content
Log in

A microtubule-based, dynein-dependent force induces local cell protrusions: Implications for neurite initiation

  • Research Article
  • Published:
Brain Cell Biology

Abstract

A key event in neurite initiation is the accumulation of microtubule bundles at the neuron periphery. We hypothesized that such bundled microtubules may generate a force at the plasma membrane that facilitates neurite initiation. To test this idea we observed the behavior of microtubule bundles that were induced by the microtubule-associated protein MAP2c. Endogenous MAP2c contributes to neurite initiation in primary neurons, and exogeneous MAP2c is sufficient to induce neurites in Neuro-2a cells. We performed nocodazol washout experiments in primary neurons, Neuro-2a cells and COS-7 cells to investigate the underlying mechanism. During nocodazol washout, small microtubule bundles formed rapidly in the cytoplasm and immediately began to move toward the cell periphery in a unidirectional manner. In neurons and Neuro-2a cells, neurite-like processes extended within minutes and concurrently accumulated bundles of repolymerized microtubules. Speckle microscopy in COS-7 cells indicated that bundle movement was due to transport, not treadmilling. At the periphery bundles remained under a unidirectional force and induced local cell protrusions that were further enhanced by suppression of Rho kinase activity. Surprisingly, this bundle motility was independent of classical actin- or microtubule-based tracks. It was, however, reversed by function-blocking antibodies against dynein. Suppression of dynein expression in primary neurons by RNA interference severely inhibited the generation of new neurites, but not the elongation of existing neurites formed prior to dynein knockdown. Together, these cell biological data suggest that neuronal microtubule-associated proteins induce microtubule bundles that are pushed outward by dynein and locally override inward contraction to initiate neurite-like cell protrusions. A similar force-generating mechanism might participate in spontaneous initiation of neurites in developing neurons.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Similar content being viewed by others

References

  • Abal, M., Piel, M., Bouckson-Castaing, V., Mogensen, M., Sibarita, J. B., and Bornens, M. (2002). Microtubule release from the centrosome in migrating cells. J. Cell Biol. 159, 731–737.

    Article  PubMed  CAS  Google Scholar 

  • Adames, N. R. and Cooper, J. A. (2000). Microtubule interactions with the cell cortex causing nuclear movements in Saccharomyces cerevisiae. J. Cell Biol. 149, 863–874.

    Article  PubMed  CAS  Google Scholar 

  • Ahmad, F. J., Hughey, J., Wittmann, T., Hyman, A., Greaser, M., and Baas, P. W. (2000). Motor proteins regulate force interactions between microtubules and microfilaments in the axon. Nat. Cell Biol. 2, 276–280.

    Article  PubMed  CAS  Google Scholar 

  • Ahmad, F. J., Yu, W., McNally, F. J., and Baas, P. W. (1999). An essential role for katanin in severing microtubules in the neuron. J. Cell Biol. 145, 305–315.

    Article  PubMed  CAS  Google Scholar 

  • Baas, P. W. and Ahmad, F. J. (2001). Force generation by cytoskeletal motor proteins as a regulator of axonal elongation and retraction. Trends Cell Biol. 11, 244–249.

    Article  PubMed  CAS  Google Scholar 

  • Baas, P. W., Black, M. M., and Banker, G. A. (1989). Changes in microtubule polarity orientation during the development of hippocampal neurons in culture. J. Cell Biol. 109, 3085–3094.

    Article  PubMed  CAS  Google Scholar 

  • Brummelkamp, T. R., Bernards, R., and Agami, R. (2002). A system for stable expression of short interfering RNAs in mammalian cells. Science 296, 550–553.

    Article  PubMed  CAS  Google Scholar 

  • Calabrese, B. and Halpain, S. (2005). Essential role for the PKC target MARCKS in maintaining dendritic spine morphology. Neuron 48, 77–90.

    Article  PubMed  CAS  Google Scholar 

  • Dai, J. and Sheetz, M. P. (1995). Mechanical properties of neuronal growth cone membranes studied by tether formation with laser optical tweezers. Biophys. J. 68, 988–996.

    PubMed  CAS  Google Scholar 

  • Dehmelt, L., Smart, F. M., Ozer, R. S., and Halpain, S. (2003). The role of microtubule-associated protein 2c in the reorganization of microtubules and lamellipodia during neurite initiation. J. Neurosci. 23, 9479–9490.

    PubMed  CAS  Google Scholar 

  • Dujardin, D. L., Barnhart, L. E., Stehman, S. A., Gomes, E. R., Gundersen, G. G., and Vallee, R. B. (2003). A role for cytoplasmic dynein and LIS1 in directed cell movement. J. Cell Biol. 163, 1205–1211.

    Article  PubMed  CAS  Google Scholar 

  • Dujardin, D. L. and Vallee, R. B. (2002). Dynein at the cortex. Curr. Opin. Cell Biol. 14, 44–49.

    Article  PubMed  CAS  Google Scholar 

  • Edson, K., Weisshaar, B., and Matus, A. (1993). Actin depolymerisation induces process formation on MAP2-transfected non-neuronal cells. Development 117, 689–700.

    PubMed  CAS  Google Scholar 

  • Fan, J. and Amos, L. A. (2001). Antibodies to cytoplasmic dynein heavy chain map the surface and inhibit motility. J. Mol. Biol. 307, 1317–1327.

    Article  PubMed  CAS  Google Scholar 

  • Fink, G. and Steinberg, G. (2006). Dynein-dependent motility of microtubules and nucleation sites supports polarization of the tubulin array in the fungus Ustilago maydis. Mol. Biol. Cell. 17, 3242–3253.

    Article  PubMed  CAS  Google Scholar 

  • Habermann, A., Schroer, T. A., Griffiths, G., and Burkhardt, J. K. (2001). Immunolocalization of cytoplasmic dynein and dynactin subunits in cultured macrophages: enrichment on early endocytic organelles. J. Cell Sci. 114, 229–240.

    PubMed  CAS  Google Scholar 

  • Harada, A., Teng, J., Takei, Y., Oguchi, K., and Hirokawa, N. (2002). MAP2 is required for dendrite elongation, PKA anchoring in dendrites, and proper PKA signal transduction. J. Cell Biol. 158, 541–549.

    Article  PubMed  CAS  Google Scholar 

  • Hasaka, T. P., Myers, K. A., and Baas, P. W. (2004). Role of actin filaments in the axonal transport of microtubules. J. Neurosci. 24, 11291–11301.

    Article  PubMed  CAS  Google Scholar 

  • He, Y., Francis, F., Myers, K. A., Yu, W., Black, M. M., and Baas, P. W. (2005). Role of cytoplasmic dynein in the axonal transport of microtubules and neurofilaments. J. Cell Biol. 168, 697–703.

    Article  PubMed  CAS  Google Scholar 

  • Heil-Chapdelaine, R. A., Tran, N. K., and Cooper, J. A. (2000). Dynein-dependent movements of the mitotic spindle in Saccharomyces cerevisiae Do not require filamentous actin. Mol. Biol. Cell 11, 863–872.

    PubMed  CAS  Google Scholar 

  • Helfand, B. T., Mikami, A., Vallee, R. B., and Goldman, R. D. (2002). A requirement for cytoplasmic dynein and dynactin in intermediate filament network assembly and organization. J. Cell Biol. 157, 795–806.

    Article  PubMed  CAS  Google Scholar 

  • Homma, N., Takei, Y., Tanaka, Y., Nakata, T., Terada, S., Kikkawa, M., Noda, Y., and Hirokawa, N. (2003). Kinesin superfamily protein 2A (KIF2A) functions in suppression of collateral branch extension. Cell 114, 229–239.

    Article  PubMed  CAS  Google Scholar 

  • Keating, T. J., Peloquin, J. G., Rodionov, V. I., Momcilovic, D., and Borisy, G. G. (1997). Microtubule release from the centrosome. Proc. Natl. Acad. Sci. U.S.A. 94, 5078–5083.

    Article  PubMed  CAS  Google Scholar 

  • Krylyshkina, O., Kaverina, I., Kranewitter, W., Steffen, W., Alonso, M. C., Cross, R. A., and Small, J. V. (2002). Modulation of substrate adhesion dynamics via microtubule targeting requires kinesin-1. J. Cell Biol. 156, 349–359.

    Article  PubMed  CAS  Google Scholar 

  • Letourneau, P. C., Shattuck, T. A., and Ressler, A. H. (1987). “Pull” and “push” in neurite elongation: observations on the effects of different concentrations of cytochalasin B and taxol. Cell Motil Cytoskeleton. 8, 193–209.

    Article  PubMed  CAS  Google Scholar 

  • Liu, Z., Steward, R., and Luo, L. (2000). Drosophila Lis1 is required for neuroblast proliferation, dendritic elaboration and axonal transport. Nat. Cell Biol. 2, 776–783.

    Article  PubMed  CAS  Google Scholar 

  • Meijering, E., Jacob, M., Sarria, J. C., Steiner, P., Hirling, H., and Unser, M. (2004). Design and validation of a tool for neurite tracing and analysis in fluorescence microscopy images. Cytometry A 58, 167–176.

    Article  PubMed  CAS  Google Scholar 

  • Menezes, J. R. and Luskin, M. B. (1994). Expression of neuron-specific tubulin defines a novel population in the proliferative layers of the developing telencephalon. J. Neurosci. 14, 5399–5416.

    PubMed  CAS  Google Scholar 

  • Mitchison, T. J. (1989). Polewards microtubule flux in the mitotic spindle: evidence from photoactivation of fluorescence. J. Cell Biol. 109, 637–652.

    Article  PubMed  CAS  Google Scholar 

  • Nilsson, H., Steffen, W., and Palazzo, R. E. (2001). In vitro reconstitution of fish melanophore pigment aggregation. Cell Motil. Cytoskeleton 48, 1–10.

    Article  PubMed  CAS  Google Scholar 

  • Ozer, R. S. and Halpain, S. (2000). Phosphorylation-dependent localization of microtubule-associated protein MAP2c to the actin cytoskeleton. Mol. Biol. Cell 11, 3573–3587.

    PubMed  CAS  Google Scholar 

  • Paschal, B. M., Shpetner, H. S., and Vallee, R. B. (1987). MAP 1C is a microtubule-activated ATPase which translocates microtubule in vitro and has dynein-like properties. J. Cell Biol. 105, 1273–1282.

    Article  PubMed  CAS  Google Scholar 

  • Paschal, B. M. and Vallee, R. B. (1987). Retrograde transport by the microtubule-associated protein MAP 1C. Nature 330, 181–183.

    Article  PubMed  CAS  Google Scholar 

  • Riederer, B. M., Pellier, V., Antonsson, B., Di Paolo, G., Stimpson, S. A., Lutjens, R., Catsicas, S., and Grenningloh, G. (1997). Regulation of microtubule dynamics by the neuronal growth-associated protein SCG10. Proc. Natl. Acad. Sci. U.S.A. 94, 741–745.

    Article  PubMed  CAS  Google Scholar 

  • Rogers, G. C., Rogers, S. L., Schwimmer, T. A., Ems-McClung, S. C., Walczak, C. E., Vale, R. D., Scholey, J. M., and Sharp, D. J. (2004). Two mitotic kinesins cooperate to drive sister chromatid separation during anaphase. Nature 427, 364–370.

    Article  PubMed  CAS  Google Scholar 

  • Rogers, G. C., Rogers, S. L., and Sharp, D. J. (2005). Spindle microtubules in flux. J. Cell Sci. 118, 1105–1116.

    Article  PubMed  CAS  Google Scholar 

  • Rolls, M. M., Stein, P. A., Taylor, S. S., Ha, E., McKeon, F., and Rapoport, T. A. (1999). A visual screen of a GFP-fusion library identifies a new type of nuclear envelope membrane protein. J. Cell. Biol. 146:29–44.

    PubMed  CAS  Google Scholar 

  • Rotsch, C. and Radmacher, M. (2000). Drug-induced changes of cytoskeletal structure and mechanics in fibroblasts: an atomic force microscopy study. Biophys. J. 78, 520–535.

    Article  PubMed  CAS  Google Scholar 

  • Salmon, W. C., Adams, M. C. and Waterman-Storer, C. M. (2002). Dual-wavelength fluorescent speckle microscopy reveals coupling of microtubule and actin movements in migrating cells. J. Cell Biol. 158, 31–37.

    Article  PubMed  CAS  Google Scholar 

  • Shiraishi, Y., Mizutani, A., Yuasa, S., Mikoshiba, K., and Furuichi, T. (2003). Glutamate-induced declustering of post-synaptic adaptor protein Cupidin (Homer 2/vesl-2) in cultured cerebellar granule cells. J. Neurochem. 87, 364–376.

    Article  PubMed  CAS  Google Scholar 

  • Steffen, W., Karki, S., Vaughan, K. T., Vallee, R. B., Holzbaur, E. L., Weiss, D. G., and Kuznetsov, S. A. (1997). The involvement of the intermediate chain of cytoplasmic dynein in binding the motor complex to membranous organelles of Xenopus oocytes. Mol. Biol. Cell 8, 2077–2088.

    PubMed  CAS  Google Scholar 

  • Takemura, R., Okabe, S., Umeyama, T., and Hirokawa, N. (1995). Polarity orientation and assembly process of microtubule bundles in nocodazole-treated, MAP2c-transfected COS cells. Mol. Biol. Cell 6, 981–996.

    PubMed  CAS  Google Scholar 

  • Teng, J., Takei, Y., Harada, A., Nakata, T., Chen, J., and Hirokawa, N. (2001). Synergistic effects of MAP2 and MAP1B knockout in neuronal migration, dendritic outgrowth, and microtubule organization. J. Cell Biol. 155, 65–76.

    Article  PubMed  CAS  Google Scholar 

  • Valetti, C., Wetzel, D. M., Schrader, M., Hasbani, M. J., Gill, S. R., Kreis, T. E., and Schroer, T. A. (1999). Role of dynactin in endocytic traffic: effects of dynamitin overexpression and colocalization with CLIP-170. Mol. Biol. Cell 10, 4107–4120.

    PubMed  CAS  Google Scholar 

  • Waterman-Storer, C. M., Desai, A., Bulinski, J. C., and Salmon, E. D. (1998). Fluorescent speckle microscopy, a method to visualize the dynamics of protein assemblies in living cells. Curr. Biol. 8, 1227–1230.

    Article  PubMed  CAS  Google Scholar 

  • Wu, G., Fang, Y., Lu, Z. H. and Ledeen, R. W. (1998). Induction of axon-like and dendrite-like processes in neuroblastoma cells. J. Neurocytol. 27, 1–14.

    Article  PubMed  CAS  Google Scholar 

  • Zhang, X. F., Schaefer, A. W., Burnette, D. T., Schoonderwoert, V. T., and Forscher, P. (2003). Rho-dependent contractile responses in the neuronal growth cone are independent of classical peripheral retrograde actin flow. Neuron 40, 931–944.

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to S. Halpain.

Electronic supplementary material

Fig. S1.

Fluorescent speckle microscopy: Treadmilling cannot account for microtubule bundle movements. GFP-MAP2c transfected, nocodazole treated COS-7 cells were injected with low concentrations of rhodamine-X-labeled tubulin before drug washout to allow formation of speckle-like fiduciary marks within microtubule bundles. (A) and (B) After drug washout, imaging revealed slow directional flow of speckles within peripheral microtubule bundles towards the cell center (indicated by yellow arrows. See Movie S6). Kymograph analysis was performed in the area marked with the red bar to analyze speckle movements within a stationary microtubule bundle. (C) Likewise, kymograph analysis was performed to measure translocation of moving bundles within a different cell. (D). Identically scaled kymographs derived from panels B) and C) for measuring speckle and bundle movements; green lines are overlaid on individual events to indicate relative velocity. Speckle movements were approximately 50-fold slower than bundle movements (error bars represent standard error of the mean).

Fig. S2.

Rho kinase-dependent contractility inhibits microtubule bundle induced cell protrusion. Similar to Fig 3A, GFP-MAP2c transfected COS-7 cells were treated with nocodazole and imaged after drug washout. 45min following washout, the ROCK inhibitor Y27632 (60 μM) was added. DIC (A) and epi-fluorescence (B) sequences are shown before (−10:00 to 0:00) and after (0:00 to 20:00) addition of Y27632. In GFP-MAP2c transfected cells, more protrusions were formed after Y27632 application (arrowheads in A 20:00 and arrows in B 10:00 and 20:00) compared to the time period before drug addition. Time points represent minutes and seconds.

Fig. S3.

Anti-dynein antibodies m74-2 and m74-1 inhibit dynein-dependent retrograde transport of transferrin in COS-7 cells. COS-7 cells were injected with control antibodies (mouse IgG; n = 26), anti-dynein antibody m74-2 (n = 11) or m74-1 (n = 9, data not shown) supplemented with Alexa-488-Dextran to identify injected cells. After serum starving for 1 h, Texas Red labeled transferrin was applied for 10min, followed by rinsing in PBS (2min) and fixation. Nuclei were stained using DAPI and cells were scored for retrograde transport of internalized transferrin. Whereas perinuclear accumulation of transferrin was detectable in 81% of control cells injected with isotype control antibody, this dynein-dependent process was completely blocked in cells injected with either antibody m74-1 or m74-2.

Fig. S4.

Specificity of dynein function blocking antibodies. (A) COS-7 or (B) Neuro-2a cell extracts were probed with antibodies m74-2 and m74-1 targeted against the dynein intermediate chain in TBS+500 mM NaCl. Both m74-2 and m74-1 recognize a single predominant band in the tested cell types.

Fig. S5.

Estimation of the number of microtubules in MAP2c induced microtubule bundles. (A) GFP-MAP2c transfected COS7 cells or control cells were treated with nocodazole overnight and injected with comparable levels of rhodamine-labeled tubulin. Shortly after washout, microtubules and microtubule bundles were imaged using time-lapse microscopy (see Movie S5). The first two panels show equally scaled frames optimized to depict the smallest detectable microtubule structures. The third panel is identical to panel 2, except that it is displayed at a wider intensity scale to demonstrate that also the thickest bundles were not overexposed in the original acquisition and thus suited for quantitative analysis. Individual frames of these recordings were analyzed by measuring integrated, background corrected intensity profiles along line segments placed across microtubule structures (see small inset in middle panel). Although we do not take into account potential non-linear effects, such as fluorescence quenching, such an analysis can provide a reasonable estimate of microtubule number within bundles. (B) Frequency histograms were used to visualize the distribution of intensity measurements. Peripheral microtubules from control cells all measured within a very similar range of intensities (∼20 arbitrary units). As judged by their dynamic behavior, the peripheral array of microtubule structures in control cells was mostly composed of individual microtubules (in control cells, we avoided measuring regions in which microtubules overlapped with each other). Therefore, we conclude that this intensity range is typical for single microtubules. In GFP-MAP2c transfected cells, we detected a significant number of single microtubules, although these represent only 2% of the total population, as the vast majority of microtubules are contained in bundles, which range from small-sized bundles composed of ∼2–5 microtubules to thicker bundles of up to ∼50 microtubules.

Fig. S6.

Microtubule bundles accumulate at the cell periphery in the absence of microtubule tracks. After nocodazole washout, GFP-MAP2c transfected cells were stained for total tubulin. Untransfected cells displayed radial microtubule networks, which were similar to those seen in control cells not treated with nocodazole. In contrast, transfected cells (*) concentrated nearly all microtubules in bundles at the cell periphery. In transfected cells, no underlying microtubule network was observed that might have facilitated unidirectional microtubule bundle movements.

Fig. S7.

Microtubule-dependent induction of local cell protrusions. (A) Primary hippocampal neurons were co-transfected with vectors coding for GFP-tubulin and mRFP-actin to visualize microtubule and actin reorganization. Five days after plating, neurons were treated with 10 μM nocodazole overnight and imaged during drug washout. During a timeperiod of 2–10 minutes after washout, bundled microtubules accumulate at the cell periphery and induce a local cell protrusion (cyan arrows). Concurrently, a nascent actin rich growth cone forms at the tip of the newly formed neurite (yellow arrows). (B) Primary hippocampal neurons were transfected with GFP-MAP2c treated with 10μM nocodazole one day after plating. The left panel shows the complete neuron shortly after drug washout. The red box indicates a region, which is shown magnified in the middle panel. During the early phases of drug washout, rapid movements of microtubules were observed (orange arrows, see also Movie S11). After washout, cells were stained for the neuronal marker TUJ1 (right panels). (C) After peripheral accumulation, bundles frequently protruded the cell membrane (green arrows, see also Movie S12), suggesting that MAP2c-induced microtubule bundles produce an anterograde force in primary neurons.

Fig. S8.

Validation of dynein heavy chain depletion. Primary hippocampal neurons were transfected with vectors coding for GFP together with control or dynein heavy chain targeted shRNA and stained for dynein heavy chain 4 or 11 days after plating. (A) Representative examples of cells transfected with control or dynein targeted shRNA (11 DIV). (B) Transfection efficiency was below 30%. Thus we measured dynein depletion on a per cell basis by quantification of maximal perinuclear dynein heavy chain immunofluorescence. Dynein levels were strongly reduced in the time interval between 4-11 days post-plating (n ≥ 20; *** = p < 0.0001; Student's T-test, two-tailed; error bars represent standard error of the mean).

Mechanisms in Neurite Initiation: Dynein-Dependent Induction of Cell Protrusions

Movie S1: Dynamics of Microtubule Bundle Formation in Neuro-2a cells.

GFP-MAP2c transfected Neuro-2a cells were treated with nocodazole overnight and GFP-MAP2c fluorescence was imaged during drug washout. Shortly after nocodazole washout, microtubules repolymerize and quickly form radially oriented microtubule bundles. Time signature represents hours:minutes.

Movie S2: Microtubule Bundle Translocation in Neuro-2a cells.

GFP-MAP2c transfected Neuro-2a cells were treated and imaged as in Movie S1. As shown in this example, we observed microtubule bundle translocation within the cells cytoplasm in some cases. Time signature represents hours:minutes.

Movie S3 and Movie S4: Microtubule Bundle Translocation in COS-7 cells. GFP-MAP2c transfected COS-7 cells were treated with nocodazole overnight and GFP-MAP2c fluorescence was imaged during drug washout. Two examples are shown to exemplify variability in bundle size. However, bundle translocation behavior was similar across all observed cells. Time signature represents hours:minutes.

Movie S4

Movie S5: Microtubule Bundle Translocation in COS-7 cells.

GFP-MAP2c transfected or control COS-7 cells were treated with nocodazole overnight and injected with labeled tubulin. Microtubules were imaged shortly after drug washout in both control and GFP-MAP2c transfected cells. Both panels were scaled identical for comparison. Single microtubules in control cells displayed typical dynamic behavior; single and bundled microtubules in GFP-MAP2c transfected cells were transported rapidly in the absence of microtubule tracks. Time signature represents minutes:seconds.

Movie S6: Speckle Analysis of Microtubule Treadmilling in Peripheral Microtubule Bundles.

GFP-MAP2c transfected COS-7 cells were treated with nocodazole overnight, injected with low amounts of Rhodamine-X-labeled tubulin. One hour after injection, nocodazole was washed out; microtubules were allowed to re-grow and accumulate at the cell periphery. Fluorescent speckles of Rhodamine-X-tubulin were imaged using confocal microscopy. Speckles revealed slow directional treadmilling towards the cell center. Time signature represents minutes:seconds.

Movie S7 and Movie S8: Simultaneous Imaging of Speckle and Bundle Translocation in Nascent Microtubule Bundles.

GFP-MAP2c transfected COS-7 cells were treated with nocodazole and injected with Rhodamine-X-labeled tubulin. GFP-MAP2c (shown in green) and Rhodamine-X-tubulin (shown in red) were imaged in short succession (with a delay between the individual channels of less than 1 second). Two examples are shown. Movie S8 shows a smaller thicker bundle, whereas Movie S7 shows a longer, single microtubule or thin bundle. Both microtubule bundles and speckles moved simultaneously at similar speeds. Time signature represents minutes:seconds.

Movie S8

Movie S9: Microtubule Bundles Induce Membrane Protrusions upon Cytochalasin D Treatment. GFP-MAP2c transfected COS-7 cells or untransfected control cells were treated with nocodazole overnight. After drug washout, microtubules were allowed to re-grow and accumulate at the cell periphery. 4μM cytochalasin D was added while cells were imaged using Nomarski DIC optics. In GFP-MAP2c transfected cells, the plasma membrane was deflected at cell margins shortly after cytochalasin D addition. Time signature represents hours:minutes.

Movie S10: Microtubule Bundle Translocation in Latrunculin A treated COS-7 cells.

GFP-MAP2c transfected COS-7 cells were treated with nocodazole overnight. GFP-MAP2c fluorescence was imaged while cells were first treated with 10μM latrunculin A for 20min to disrupt actin filaments, followed by nocodazole washout in the continued presence of 10μM latrunculin A. Robust bundle movements were observed under these conditions. Time signature represents hours:minutes.

Movie S11: Microtubule Bundle Translocation in Primary Hippocampal Neurons.

GFP-MAP2c transfected primary hippocampal neurons were treated with nocodazole overnight and GFP-MAP2c fluorescence was imaged during drug washout. Shortly after nocodazole washout, microtubules repolymerize and start to move unidirectionally. Time signature represents minutes:seconds.

Movie S12: Microtubule Bundles Induce Local Neurite-Like Protrusions in Primary Hippocampal Neurons. Primary hippocampal neurons were treated with nocodazole overnight and GFP-MAP2c fluorescence was imaged during drug washout. Microtubule bundles accumulate at the neurons periphery and frequently induce local neurite-like protrusions. Time signature represents minutes:seconds.

Movie S13: Model for Peripheral Microtubule Bundle Accumulation by Unidirectional Transport. Bundles were drawn in Photoshop with random orientation. (+) denotes microtubule plus-ends. Unidirectional transport is sufficient to accumulate microtubule bundles in the cell periphery in a polarized array with microtubule plus-ends facing outward.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Dehmelt, L., Nalbant, P., Steffen, W. et al. A microtubule-based, dynein-dependent force induces local cell protrusions: Implications for neurite initiation. Brain Cell Bio 35, 39–56 (2006). https://doi.org/10.1007/s11068-006-9001-0

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11068-006-9001-0

Keywords

Navigation