mammalian erythroblast enucleation requires pi3k-dependent ...mammalian erythroblast enucleation...

10
Mammalian erythroblast enucleation requires PI3K-dependent cell polarization Junxia Wang 1, *, Tzutzuy Ramirez 1, *, Peng Ji 2,§ , Senthil Raja Jayapal 2,3 , Harvey F. Lodish 2 and Maki Murata-Hori 1,4,` 1 Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, 117604, Singapore 2 Whitehead Institute for Biological Research, Department of Biology, Massachusetts Institute of Technology, 9 Cambridge Center, Floor 6, Cambridge, MA 02142, USA 3 Stem Cell Group 4, Genome Institute of Singapore, #08-01, Genome, 60 Biopolis Street, Singapore 138672 4 Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543 *These authors contributed equally to this work § Present address: Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Ward 3-140, Chicago, IL 60611, USA ` Author for correspondence ([email protected]) Accepted 11 August 2011 Journal of Cell Science 125, 340–349 ß 2012. Published by The Company of Biologists Ltd doi: 10.1242/jcs.088286 Summary Enucleation, the final step in terminal differentiation of mammalian red blood cells, is an essential process in which the nucleus surrounded by the plasma membrane is budded off from the erythroblast to form a reticulocyte. Most molecular events in enucleation remain unclear. Here we show that enucleation requires establishment of cell polarization that is regulated by the microtubule-dependent local activation of phosphoinositide 3-kinase (PI3K). When the nucleus becomes displaced to one side of the cell, actin becomes restricted to the other side, where dynamic cytoplasmic contractions generate pressure that pushes the viscoelastic nucleus through a narrow constriction in the cell surface, forming a bud. The PI3K products PtdIns(3,4)P 2 and PtdIns(3,4,5)P 3 are highly localized at the cytoplasmic side of the plasma membrane. PI3K inhibition caused impaired cell polarization, leading to a severe delay in enucleation. Depolymerization of microtubules reduced PI3K activity, resulting in impaired cell polarization and enucleation. We propose that enucleation is regulated by microtubules and PI3K signaling in a manner mechanistically similar to directed cell locomotion. Key words: Erythroblast enucleation, PI3-kinase, Microtubules, Cell polarization Introduction In the last step of erythropoiesis, mammalian erythroblasts undergo enucleation, a process that is crucial for the formation of mature functional red blood cells. During enucleation, the erythroblast extrudes its nucleus tightly apposed to the plasma membrane, forming a reticulocyte (Ihle and Gilliland, 2007; Koury et al., 2002; Richmond et al., 2005). Pioneering studies using electron microscopy revealed that at the earliest stage of enucleation the erythroblast nucleus becomes located close to the cell membrane away from the center of the cell (Simpson and Kling, 1967; Skutelsky and Danon, 1967) and that a cytokinetic- like furrow is formed in the region between the extruded nucleus and the incipient reticulocyte (Koury et al., 1989; Skutelsky and Danon, 1967). Actin filaments (F-actin) accumulate in the cytokinetic-like furrow (Ji et al., 2008; Koury et al., 1989) and disruption of F-actin (Ji et al., 2008; Koury et al., 1989; Yoshida et al., 2005) or depletion of mDia2, a regulator of actin polymerization (Ji et al., 2008), blocked enucleation, suggesting that actin-based forces drive nuclear extrusion. However, many questions remain unanswered concerning the process of erythroblast enucleation. In particular, little is known how an asymmetry is established within the erythroblast (i.e. how the nucleus becomes localized to one side of the cell and the cytoplasm to the other), although this polarized state appears to be important for enucleation. Moreover, the detailed organization of actin and microtubules in polarized erythroblasts is unknown. Phosphoinositide 3-kinase (PI3K) is well known as a central regulator of chemotaxis. In migrating Dictyostelium discoideum, neutrophils and fibroblasts, the PI3K products PtdIns(3,4)P 3 and PtdIns(3,4,5)P 3 accumulate locally at the leading edge of the surface membrane and control cell polarization (Haugh et al., 2000; Parent et al., 1998; Servant et al., 2000). Although involvement of PI3K in the early stages of Epo (erythropoietin)- regulated differentiation of erythroid progenitors has been established (Ghaffari et al., 2006; Zhao et al., 2006), little is known about its role in the much later steps of enucleation. We investigated how erythroblasts establish cell polarization and whether this polarization plays a role in expelling the nucleus from the cell. We used a powerful combination of an in vitro cell culture system that mimics normal terminal erythroid proliferation, differentiation and enucleation (Ji et al., 2008), combined with several microscopic imaging techniques. Our results show that proper enucleation requires establishment and maintenance of cell polarization mediated by PI3K in a manner similar to that seen in migrating cells. Results Erythroblast enucleation is initiated through establishment of cell polarization, followed by dynamic cytoplasmic contractions We first wanted to determine when the terminal erythroblast becomes polarized and how the nucleus is extruded from the 340 Research Article Journal of Cell Science

Upload: others

Post on 21-Oct-2020

10 views

Category:

Documents


0 download

TRANSCRIPT

  • Mammalian erythroblast enucleation requiresPI3K-dependent cell polarization

    Junxia Wang1,*, Tzutzuy Ramirez1,*, Peng Ji2,§, Senthil Raja Jayapal2,3, Harvey F. Lodish2 andMaki Murata-Hori1,4,`

    1Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, 117604, Singapore2Whitehead Institute for Biological Research, Department of Biology, Massachusetts Institute of Technology, 9 Cambridge Center, Floor 6,Cambridge, MA 02142, USA3Stem Cell Group 4, Genome Institute of Singapore, #08-01, Genome, 60 Biopolis Street, Singapore 1386724Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543

    *These authors contributed equally to this work§Present address: Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Ward 3-140, Chicago, IL 60611, USA`Author for correspondence ([email protected])

    Accepted 11 August 2011Journal of Cell Science 125, 340–349� 2012. Published by The Company of Biologists Ltddoi: 10.1242/jcs.088286

    SummaryEnucleation, the final step in terminal differentiation of mammalian red blood cells, is an essential process in which the nucleussurrounded by the plasma membrane is budded off from the erythroblast to form a reticulocyte. Most molecular events in enucleationremain unclear. Here we show that enucleation requires establishment of cell polarization that is regulated by the microtubule-dependent

    local activation of phosphoinositide 3-kinase (PI3K). When the nucleus becomes displaced to one side of the cell, actin becomesrestricted to the other side, where dynamic cytoplasmic contractions generate pressure that pushes the viscoelastic nucleus through anarrow constriction in the cell surface, forming a bud. The PI3K products PtdIns(3,4)P2 and PtdIns(3,4,5)P3 are highly localized at the

    cytoplasmic side of the plasma membrane. PI3K inhibition caused impaired cell polarization, leading to a severe delay in enucleation.Depolymerization of microtubules reduced PI3K activity, resulting in impaired cell polarization and enucleation. We propose thatenucleation is regulated by microtubules and PI3K signaling in a manner mechanistically similar to directed cell locomotion.

    Key words: Erythroblast enucleation, PI3-kinase, Microtubules, Cell polarization

    IntroductionIn the last step of erythropoiesis, mammalian erythroblasts

    undergo enucleation, a process that is crucial for the formation of

    mature functional red blood cells. During enucleation, the

    erythroblast extrudes its nucleus tightly apposed to the plasma

    membrane, forming a reticulocyte (Ihle and Gilliland, 2007;

    Koury et al., 2002; Richmond et al., 2005). Pioneering studies

    using electron microscopy revealed that at the earliest stage of

    enucleation the erythroblast nucleus becomes located close to the

    cell membrane away from the center of the cell (Simpson and

    Kling, 1967; Skutelsky and Danon, 1967) and that a cytokinetic-

    like furrow is formed in the region between the extruded nucleus

    and the incipient reticulocyte (Koury et al., 1989; Skutelsky and

    Danon, 1967). Actin filaments (F-actin) accumulate in the

    cytokinetic-like furrow (Ji et al., 2008; Koury et al., 1989) and

    disruption of F-actin (Ji et al., 2008; Koury et al., 1989; Yoshida

    et al., 2005) or depletion of mDia2, a regulator of actin

    polymerization (Ji et al., 2008), blocked enucleation, suggesting

    that actin-based forces drive nuclear extrusion. However, many

    questions remain unanswered concerning the process of

    erythroblast enucleation. In particular, little is known how an

    asymmetry is established within the erythroblast (i.e. how the

    nucleus becomes localized to one side of the cell and the

    cytoplasm to the other), although this polarized state appears to

    be important for enucleation. Moreover, the detailed organization

    of actin and microtubules in polarized erythroblasts is unknown.

    Phosphoinositide 3-kinase (PI3K) is well known as a central

    regulator of chemotaxis. In migrating Dictyostelium discoideum,

    neutrophils and fibroblasts, the PI3K products PtdIns(3,4)P3 and

    PtdIns(3,4,5)P3 accumulate locally at the leading edge of the

    surface membrane and control cell polarization (Haugh et al.,

    2000; Parent et al., 1998; Servant et al., 2000). Although

    involvement of PI3K in the early stages of Epo (erythropoietin)-

    regulated differentiation of erythroid progenitors has been

    established (Ghaffari et al., 2006; Zhao et al., 2006), little is

    known about its role in the much later steps of enucleation.

    We investigated how erythroblasts establish cell polarization

    and whether this polarization plays a role in expelling the nucleus

    from the cell. We used a powerful combination of an in vitro

    cell culture system that mimics normal terminal erythroid

    proliferation, differentiation and enucleation (Ji et al., 2008),

    combined with several microscopic imaging techniques. Our

    results show that proper enucleation requires establishment and

    maintenance of cell polarization mediated by PI3K in a manner

    similar to that seen in migrating cells.

    ResultsErythroblast enucleation is initiated through

    establishment of cell polarization, followed by dynamic

    cytoplasmic contractions

    We first wanted to determine when the terminal erythroblast

    becomes polarized and how the nucleus is extruded from the

    340 Research Article

    Journ

    alof

    Cell

    Scie

    nce

    mailto:[email protected]

  • erythroblast. To this end, we conducted a detailed microscopic

    analysis of the enucleation process using an in vitro cell culture

    system employing mouse fetal liver erythroblasts. Enucleation

    begins ,35 hours after stimulation of erythroid progenitors (Jiet al., 2008). This system employs normal primary erythroid cells

    and thus does not have the obvious abnormalities associated with

    virus-infected and/or transformed cell lines. Moreover, the time-

    course of erythroid differentiation in this system has been well

    established (Ji et al., 2008), providing us in a time-dependent

    manner with erythroblasts at different stages of differentiation.

    We could follow cultured erythroblast cells after they underwent

    a final mitotic division (Fig. 1A, 24 minutes) and generated twolate erythroblasts in which the nuclei were located at the center of

    each cell (Fig. 1A, 44 minutes). At ,3 hours after each wasgenerated (210±8.64 minutes, n58), these late erythroblastsunderwent significant polarization, displacing the nucleus

    to one side of the cell (Fig. 1A, 115 and 153 minutes, arrows).

    Around 1 hour after cell polarization, enucleation initiated

    (74.7±8.64 minutes, n58) and dynamic and random contractionsoccurred at the side of the cytoplasm opposite to the nucleus

    (n516; Fig. 1B and supplementary material Movie 1). Strikingly,a part of the nucleus suddenly protruded from the cell, forming a

    bleb-like structure from a limited area of the cortex adjacent to the

    nucleus (3.35±0.64 mm diameter; Fig. 1B, arrows). Then the

    whole nucleus was quickly squeezed out (5.98±2.73 minutes), in

    the process undergoing extensive deformations into an hourglass-

    like shape (supplementary material Movie 1). Fixed late

    erythroblasts at different stages of enucleation (Fig. 1C) were

    morphologically similar to live cells (Fig. 1B).

    Before late erythroblasts became polarized, F-actin formed

    small particles and patch-like structures distributed throughout

    the cytoplasm (Fig. 1D, Non-polarized). After cell polarization,

    F-actin was mostly restricted to the side of the cytoplasm away

    from the nucleus, and little was localized at the side with the

    nucleus (Fig. 1D, Polarized). While the nucleus was being

    expelled, a fraction of F-actin concentrated at the neck region of

    the bleb (Fig. 1D, Extruding nucleus, F-actin, arrowheads) where

    it was previously described as the cytokinetic-like furrow or

    contractile actin ring (Ji et al., 2008; Koury et al., 1989;

    Skutelsky and Danon, 1967).

    Similar to F-actin, after late erythroblasts became polarized small

    particles of active myosin II (myosin II regulatory light chain

    phosphorylated at Ser19; pMRLC) (Matsumura et al., 1998) became

    restricted to the side of the cytoplasm away from the nucleus

    (Fig. 1D, pMRLC). A fraction of active myosin II also became

    localized to the neck region of the bleb (Fig. 1D, pMRLC, arrows).

    Although previous studies have shown that F-actin is required

    for enucleation (Ji et al., 2008; Koury et al., 1989; Repasky

    Fig. 1. Enucleation is initiated through

    establishment of cell polarization, followed by

    contractions of asymmetrically localized

    cytoplasmic actomysoin. (A) Time-lapse images

    of late erythroblasts undergoing cell polarization.

    White dots depict the nucleus. Time elapsed in

    minutes after completion of the final mitotic

    division of the erythroid progenitor cell is shown.

    (B) Time-lapse images of late erythroblasts

    undergoing enucleation. (C) Late erythroblasts

    were fixed and stained for DNA (blue). The

    populations of late erythroblasts were classified

    into non-polarized cells, polarized cells that show

    nuclear displacement, early phase of nuclear

    extrusion and late phase of nuclear extrusion.

    (D) Immunofluorescence of late erythroblasts at

    different stages of enucleation. Single confocal

    images of F-actin (red), active myosin II

    (pMRLC; green) and DNA (blue) are shown.

    Note that both F-actin and active myosin II

    become mostly restricted to the opposite side of

    the cytoplasm after cells become polarized. A

    small fraction of both F-actin and active myosin

    (arrows) is found accumulated in the neck region

    of the bleb-like protrusion. (E) DMSO,

    cytochalasin D or blebbistatin was applied to

    cells at 36 hours and then polarized late

    erythroblasts were identified for time-lapse

    recording. Scale bars: 5 mm.

    Mechanics of mammalian erythroblast enucleation 341

    Journ

    alof

    Cell

    Scie

    nce

  • and Eckert, 1981; Yoshida et al., 2005), it remained

    unknown whether enucleation is powered by contractions of

    asymmetrically localized actomyosin. To this end, we disrupted

    F-actin or inhibited myosin II function specifically in polarized

    late erythroblasts. We applied specific inhibitors of actin or

    myosin II to cells at 36 hours of culture, and polarized cells were

    then identified for time-lapse recording. In polarized cells treated

    with cytochalasin D, an agent that disrupts F-actin, dynamic

    contractions were quickly and completely suppressed and

    enucleation was blocked (Fig. 1E, Cytochalasin D).

    Similarly, treatment of polarized cells with blebbistatin, an

    inhibitor of myosin II activity (Straight et al., 2003), at a

    concentration (100 mM) that completely inhibits cytokinesis(Mukhina et al., 2007), caused the suppression of cytoplasmic

    contractions and a failure of enucleation (Fig. 1E, Blebbistatin,

    top). Strikingly, even in cells already extruding the nucleus,

    following blebbistatin treatment the nucleus regressed into

    the cell (Fig. 1E, Blebbistatin, bottom). These results suggest

    that nuclear extrusion is achieved by pressure generated by

    contractions of asymmetrically localized actomyosin and thus

    that the establishment of cell polarization is crucial for

    enucleation. Our observations also indicate that completion of

    enucleation probably involves actomyosin-driven constriction of

    the cortical actin ring at the neck of the bleb-like protrusion.

    PI3K activity is required for proper enucleation by

    regulating the establishment and maintenance of

    cell polarization

    How is cell polarization established and maintained during

    enucleation? We speculated that PI3K, which regulates cell

    polarization in migrating cells, might be involved in this process.

    To test this hypothesis, we first examined the localization of the

    PI3K products PtdIns(3,4)P2 and PtdIns(3,4,5)P3 by expressing

    the reporter (3)Akt(PH)–GFP (the trimerized PH domain of Akt

    tagged with green fluorescent protein) in late erythroblasts

    (Fig. 2A) (Luo et al., 2005).

    In non-polarized cells, (3)Akt(PH)–GFP was localized

    throughout the entire plasma membrane (n511; Fig. 2A, Non-polarized). Strikingly, when cells became polarized, (3)Akt(PH)–

    GFP became restricted in localization to the segment of plasma

    membrane facing the cytoplasm (n58; Fig. 2A, Polarized); thislocalization pattern remained unchanged during nuclear extrusion

    (n516; Fig. 2A, Extruding nucleus). Three-dimensional imagesof (3)Akt(PH)–GFP and mCherry, used as a cytoplasmic volume

    marker, showed that the asymmetric distribution of (3)Akt(PH)–

    GFP in polarized cells was not caused by volume effects in

    the cytoplasm because mCherry was uniformly distributed

    throughout the cell (Fig. 2B). In late erythroblasts,

    (3)Akt(PHR25C)–GFP, a mutant that is unable to bind to

    PtdIns(3,4)P2 or PtdIns(3,4,5)P3 (Franke et al., 1997), failed to

    localize to the plasma membrane and was diffusely distributed in

    the cytoplasm, indicating that (3)Akt(PH)–GFP is a specific

    reporter for PI3K activity (supplementary material Fig. S1).

    Next, we wanted to determine whether PI3K activity is

    required for cell polarization during enucleation. Because PI3K

    activity is involved in the early steps of Epo-triggered cell

    division and differentiation of erythroid progenitor cells (Ghaffari

    et al., 2006; Zhao et al., 2006), we needed to use a specific

    inhibitor of PI3K to block its activity only in late erythroblasts –

    more specifically during enucleation. To this end, we applied

    Fig. 2. (3)Akt(PH)–GFP is asymmetrically localized in

    polarized late erythroblasts. (A) Two-dimensional and

    (B) three-dimensional images of (3)Akt(PH)–GFP in late

    erythroblasts that are non-polarized, polarized and extruding

    its nucleus are shown. White dots depict the nucleus. The

    color bar indicates the intensity of GFP signal from the lowest

    (black) to highest (red). (C) Localization of (3)Akt(PH)–GFP

    in cells treated with DMSO or LY294002. Each graph shows

    the distribution of the GFP (green) and mCherry (red)

    fluorescence intensity (y axis) in the corresponding distance

    (x axis; double-headed arrows). Scale bars: 5 mm.

    Journal of Cell Science 125 (2)342

    Journ

    alof

    Cell

    Scie

    nce

  • LY294002, a well-established specific inhibitor of PI3K, to

    late erythroblasts expressing (3)Akt(PH)–GFP. Strikingly, after

    ,1 hour of incubation the membrane association of (3)Akt(PH)–GFP became strongly diminished in 24 cells out of 28 (24/28)

    treated with LY294002 but not in the DMSO-treated control cells

    (0/17) (Fig. 2C), indicating that LY294002 effectively inhibits

    PI3K activity in late erythroblasts.

    To determine whether PI3K inhibition affects the establishment

    of cell polarization, we applied LY294002 to cells at 31 hours in

    culture, where ,80% of late erythroblasts were not yet polarized(see supplementary material Fig. S2). After 1 hour of incubation,

    non-polarized cells were identified and monitored by time-lapse

    recording. The majority of control non-polarized cells underwent

    cell polarization within 3 hours (80%, n540), whereas 75% of thenon-polarized cells treated with LY294002 failed to become

    polarized during this time period (n536; Fig. 3A and Fig. 4). Thedistance between the cellular and nuclear centroids in polarized

    control cells was 1.59±0.14 mm, whereas the maximum distancebetween these centroids was 0.62±0.09 mm in LY294002-treatedcells that were not able to undergo cell polarization (Fig. 3B),

    suggesting that the nucleus remained localized in the cell center in

    LY294002-treated cells. F-actin was found localized throughout

    the cytoplasm in the LY294002-treated cells that remained non-

    polarized (n523; Fig. 3C).LY294002- and DMSO-treated (control) cells were also fixed in

    order to perform quantitative analysis on the progression of

    enucleation by counting the number of late erythroblasts at each

    stage of enucleation per total number of late erythroblasts and

    enucleated cells. We observed only a slight (although statistically

    significant) decrease in the percentage of polarized cells as well

    Fig. 3. PI3K activity is required for

    establishment of cell polarization during

    enucleation. (A) Time-lapse images of non-

    polarized cells treated with DMSO or

    LY294002. White dots depict the nucleus.

    (B) Quantification of data obtained with

    cells similar to those shown in A. The

    distance between the nuclear and cellular

    centroids in individual cells treated with

    DMSO (blue) or LY294002 (red) over time

    was plotted. Four representative cells from

    each treatment are shown. (C) Z-projections

    of F-actin in late erythroblasts treated with

    DMSO or LY294002. (D,E) Cells treated

    with DMSO or LY294002 from 31 to

    35 hours (D) or from 31 to 48 hours in

    culture (E) were fixed and stained for F-

    actin and DNA. We considered an

    enucleated nucleus as an enucleated cell.

    More than 100 cells were counted in each

    experiment. Three independent experiments

    for each treatment were performed; values

    are means ± s.e.m. (*P,0.05). Scale bars:

    5 mm.

    Mechanics of mammalian erythroblast enucleation 343

    Journ

    alof

    Cell

    Scie

    nce

  • as enucleated cells in cells treated with LY294002 from 31 to35 hours in culture, compared with control cells (polarized cells,17.56±1.84% vs 24.48±2.67%; enucleated cells, 7.54±0.68% vs

    14.24±1.80%; P,0.05, Fig. 3D). Similar results were obtainedwhen late erythroblasts were treated with another PI3K inhibitor,wortmannin (supplementary material Fig. S3), suggesting that the

    defects in enucleation caused by LY294002 were indeed due toinhibition of PI3K activity and that PI3K inhibition did notcompletely block cell polarization. When cells were treated with

    LY294002 from 31 to 48 hours in culture, the decrease inpolarization and enucleation was no longer statistically significant(Fig. 3E). These results suggest that PI3K inhibition caused a severedelay rather than arrest in the establishment of cell polarization.

    To determine whether PI3K inhibition affects nuclearextrusion, LY294002 was applied to cells at 35 hours in cultureand, after 1 hour of incubation, polarized cells were identified

    and monitored by time-lapse video microscopy. Strikingly, 76%of polarized cells treated with LY294002 exhibited a wide rangeof delay and/or arrest in nuclear extrusion (35/46), whereas in all

    control cells enucleation was completed within 25 minutes (46/46; Fig. 5A). Some cells completed nuclear extrusion after adelay (11/46), whereas others moved out of the field of view

    before the extrusion was completed or did not complete nuclearextrusion during time-lapse imaging (24/46, Fig. 4 andsupplementary material Fig. S4).

    Cells that showed a delay in nuclear extrusion typically exhibited

    one or both of the following phenotypes: (a) Nuclear extrusion wasinitiated but the nucleus regressed into the cell before the extrusionwas completed; some cells repeated this ‘trial and error’ of nuclear

    extrusion several times. (b) Nuclear extrusion was initiated butfailed to complete, with formation of an hourglass-shaped nucleus(Fig. 5A, supplementary material Movies 2 and 3).

    In control cells, nuclear extrusion was completed, with adistance between the cellular and nuclear centroids of6.11±0.40 mm (Fig. 5B, DMSO), whereas in LY294002-treated

    cells that showed a delay in nuclear extrusion the distance

    between these centroids was 1.59±0.32 mm and remainedrelatively unchanged until extrusion was completed (Fig. 5B,

    LY294002). Quantitative analysis revealed no significant

    decrease in the percentage of enucleated cells among cells

    treated with LY294002 from 35 to 39 hours in culture compared

    with control cells (Fig. 5C), suggesting that PI3K inhibition

    caused a delay rather than a complete arrest in nuclear extrusion.

    These results suggest that PI3K activity is important to maintain

    cell polarization. Taken together, our observations suggest that

    PI3K activity is required for proper enucleation by regulating the

    establishment and maintenance of cell polarization.

    Microtubule-dependent local activation of PI3K is required

    for proper cell polarization during enucleation

    Next, we wanted to determine how PI3K activity is regulated

    during enucleation. In both migrating neutrophils (Xu et al.,

    2005) and macrophages during phagocytosis (Khandani et al.,

    2007), localized PI3K activity is dependent on microtubules.

    Thus, we speculated that microtubules might regulate PI3K

    activity during enucleation. Before testing this hypothesis, it

    was important to analyze the microtubule organization during

    enucleation because this had not been determined, especially in

    polarized late erythroblasts.

    Immunofluorescence showed that, as previously described,

    before cells became polarized microtubule arrays were radially

    symmetric, forming a cage-like structure surrounding the

    nucleus (Fig. 6A) (Koury et al., 1989). Concomitant with cell

    polarization, microtubules formed an asymmetric array oriented

    toward the cortex on the side of the cytoplasm opposite to the

    nucleus (Fig. 6A, Polarized and Extruding nucleus). We also

    found that, during enucleation, one or two foci of GFP–c-tubulinwere localized together in the cytoplasm in close proximity to the

    nucleus at the side of the cytoplasm (n56; Fig. 6B).

    Fig. 4. Summary of the defects in nuclear displacement and extrusion in cells treated with LY294002 or nocodazole. Live cells were treated with 50 mMLY294002 or 10 mM nocodazole in culture and the percentage of cells showing defects in enucleation was calculated. Control cells were treated with DMSO.Figures in parentheses indicate the number of cells showing the phenotype out of the total number of cells examined.

    Journal of Cell Science 125 (2)344

    Journ

    alof

    Cell

    Scie

    nce

  • Similar localization patterns of endogenous c-tubulin (n521)and pericentrin (n511) were observed in late erythroblasts(supplementary material Fig. S5A,B). Moreover, when late

    erythroblasts were stained for EB1, a protein that binds to plus-

    ends of growing microtubules (Mimori-Kiyosue et al., 2000),

    typical comet-like structures were observed regardless of the stageof enucleation (Fig. 6C, arrows), suggesting that microtubules

    are dynamic during enucleation. These results suggest thatmicrotubule organization is dramatically rearranged into anasymmetric array with the centrosome when late erythroblasts

    become polarized.

    To determine whether microtubules regulate PI3K activityduring enucleation, late erythroblasts expressing (3)Akt

    (PH)–GFP were treated with nocodazole, a microtubule-depolymerization agent. Strikingly, in nocodazole-treated cellswhere microtubules were severely disrupted (3)Akt(PH)–GFP

    failed to associate with the plasma membrane and insteadwas diffusely localized in the cytoplasm (11/15). By contrast,(3)Akt(PH)–GFP was associated with the membrane in controlcells (12/12) (Fig. 6D,E), suggesting that microtubules are

    important for maintaining PI3K activity.

    Nocodazole treatment caused defects in enucleation similar to

    those seen in cells treated with LY294002. Live-cell analysisshowed that when nocodazole was applied to cells at 32 hours inculture only 20% of non-polarized cells were able to undergopolarization within 3 hours (n520; Fig. 6F, Fig. 4 andsupplementary material Fig. S5C). F-actin was localizedthroughout the cytoplasm in nocodazole-treated cells thatremained non-polarized (n520; supplementary materialFig. S5D). When nocodazole was applied to cells at 36 hoursin culture, 63% of polarized cells exhibited a severe delay innuclear extrusion (n535; Fig. 6G, Fig. 4, supplementary materialFig. S4, Fig. S5E and Movie 4), with phenotypes similar to thoseseen in LY294002-treated cells (Fig. 5A).

    Quantitative analysis revealed only a slight decrease in the

    percentage of polarized and enucleated cells among cells treatedwith nocodazole from 32 to 36 hours in culture, compared withcontrol cells (supplementary material Fig. S5F). It is noteworthy

    that treatment of erythroblasts with nocodazole from 32 to48 hours in culture resulted in severe defects in nuclear andcellular morphology (data not shown). The decrease was notsignificant when cells were treated with nocodazole from 36 to

    40 hours in culture (supplementary material Fig. S5G). Theseresults suggest that microtubules are required for properenucleation by regulating the establishment and maintenance of

    cell polarization. As detailed below, in large measure thisresolves past controversies about whether or not microtubulesplay an essential role in enucleation (Keerthivasan et al., 2010;

    Koury et al., 1989; Sonoda et al., 1998). Unlike the crucial role ofmicrotubules in the regulation of PI3K activity, PI3K inhibitioncaused no drastic defects in microtubule organization in late

    erythroblasts (Fig. 6H). Taken together, our observations suggestthat microtubule-dependent local activation of PI3K is requiredfor proper enucleation.

    PI3K regulates cell polarization through promoting themovement of the nucleus but not the MTOCduring enucleation

    In migrating fibroblasts, reorientation of the microtubule-organizing center (MTOC) is the initial polarization event and

    occurs by the movement of the nucleus but not the MTOC(Gomes et al., 2005; Schmoranzer et al., 2009). Our live-cellanalyses showed that when late erythroblasts became polarized

    the nucleus moved to one side of the cell (Fig. 3A,B), suggestingthat cell polarization might be initiated in a way similar to thatseen in migrating cells and that PI3K might regulate this process.

    Fig. 5. PI3K activity is required for maintenance of cell polarization

    during enucleation. (A) Time-lapse images of polarized cells treated with

    DMSO or LY294002. Time 0 indicates the onset of nuclear extrusion. White

    dots depict the nucleus. (B) Quantification of data obtained with cells similar to

    those shown in A. The distance between the nuclear and cellular centroids in

    individual cells treated with DMSO (blue) or LY294002 (red) over time after the

    onset of nuclear extrusion (time 0) was plotted. Representative cells for each

    treatment (five for DMSO and seven for LY294002) are shown. (C) Cells treated

    with DMSO or LY294002 from 35 to 39 hours in culture were fixed and stained

    for F-actin and DNA. Three independent experiments for each treatment were

    performed; values are means ± s.e.m. (*P,0.05). Scale bar: 5 mm.

    Mechanics of mammalian erythroblast enucleation 345

    Journ

    alof

    Cell

    Scie

    nce

  • To test this, cells treated with LY294002 from 31 to 35 hours in

    culture were fixed and stained for the nucleus and c-tubulin (seesupplementary material Fig. S6). Then, we measured the distance

    between the cellular centroid and the MTOC or the nuclear

    centroid (Fig. 7).

    In control cells (Fig. 7, DMSO), the average distance between

    the MTOC and the cellular centroid in non-polarized cells

    (1.75±0.06 mm) was similar to that in polarized cells(1.71±0.08 mm), suggesting that the position of the MTOCremains unchanged during cell polarization. By contrast, as

    observed in live-cell analysis (Fig. 3A,B), the distance between

    the nuclear and cellular centroids was significantly increased

    when the cells became polarized (Fig. 7, DMSO). These results

    indicate that during the early stages of enucleation, MTOC

    reorientation occurs by the movement of the nucleus but not the

    MTOC.

    In non-polarized cells, LY294002 treatment had no effect on

    the distances between the MTOC or the nuclear centroid and the

    cellular centroid (Fig. 7, Non-polarized, LY294002). In the few

    LY294002-treated cells that managed to become polarized, the

    Fig. 6. Enucleation requires microtubule-dependent local activation of PI3K. (A) Microtubule organization in cells prior to and during enucleation. Z-

    projections of microtubules (green) and DNA (blue) are shown. (B) Time-lapse fluorescence images of cells expressing GFP–c tubulin (GFP-cTub) during

    enucleation. Asterisks indicate the position of the nucleus. Dashed lines outline the nucleus and the cell edge. (C) Z-projections of EB1 (red) and DNA (blue) are

    shown. Typical comet-like structures of EB1 are detected (arrows). (D) Localization of (3)Akt(PH)–GFP in live cells treated with DMSO or nocodazole. Each

    graph shows the distribution of the GFP (green) and mCherry (red) fluorescence intensity (y axis) in the corresponding distance (x axis; double-headed arrows).

    (E) Microtubule organization (red) and localization of (3)Akt(PH)–GFP (green) in fixed cells treated with DMSO (top) or nocodazole (bottom). Z-projections of

    microtubules (red) are shown. (F) Time-lapse images of non-polarized cells treated with DMSO or nocodazole. Dashed lines depict the nucleus. (G) Time-lapse

    images of polarized cells treated with DMSO or nocodazole. Time 0 indicates the onset of nuclear extrusion. Dashed lines depict the nucleus. (H) Microtubule

    organization in late erythroblasts treated with DMSO (top) or LY294002 (bottom). Z-projections of microtubules (green) are shown. Scale bars: 5 mm.

    Journal of Cell Science 125 (2)346

    Journ

    alof

    Cell

    Scie

    nce

  • distance between the nuclear and the cellular centroids wasincreased, whereas the position of the MTOC remained

    unchanged, similar to that seen in control DMSO-treated cells(Fig. 7, Polarized, LY294002). These results suggest that PI3Kinhibition causes a delay in nuclear repositioning. Taken

    together, our observations suggest that PI3K regulates cellpolarization through promoting the movement of the nucleusbut not the MTOC during enucleation.

    DiscussionOne of the long-standing questions in erythropoiesis is how theerythroblast extrudes its nucleus to form the reticulocyte that laterbecomes the mature red blood cell. In particular, little is known

    about how erythroblasts establish and maintain a polarized stateduring enucleation. The live-cell analyses we performed in thisstudy have provided a deeper understanding of the mechanisms

    underlying the regulation of cell polarization and its importancein enucleation.

    Early work suggested a mechanism of enucleation similar tothat used in cytokinesis of mitotic cells, where local cortical

    contractions occur between the two daughter cells (Skutelsky andDanon, 1967; Koury et al., 1989). By contrast, our observationsdemonstrate that nuclear extrusion is driven by contractile forces

    generated by asymmetrically distributed cytoplasmic actomyosinin late erythroblasts. This finding reiterates the importance ofestablishment and maintenance of cell polarization during

    enucleation.

    Although previous studies revealed that PI3K was required forearly Epo-dependent stages of erythropoiesis (Ghaffari et al.,2006; Zhao et al., 2006), here we demonstrated a second and

    novel role for PI3K in the establishment and maintenance of cellpolarization during enucleation. Moreover, our results alsoshowed that PI3K promotes cell polarization by repositioning

    of the nucleus during enucleation. In migrating fibroblasts, cellpolarization is established by the movement of the nucleusinstead of the MTOC (Gomes et al., 2005). Our results indicate

    that this is also the case in late erythroblasts during enucleation.A possible mechanism that regulates the repositioning of thenucleus by PI3K is discussed below.

    The roles of microtubules in enucleation have beencontroversial (Keerthivasan et al., 2010; Koury et al., 1989;

    Sonoda et al., 1998). We observed different effects ofmicrotubule disruption agents on enucleation when cells weretreated at different stages of erythropoiesis and/or for different

    time periods of incubation. Imaging and quantitative analysis oflive and fixed cells revealed that disruption of microtubulescaused a severe delay in enucleation, but did not affect the finalextent of enucleation. This indicates that microtubules are

    required for, but not essential to, enucleation. Currently it isunknown how microtubules regulate PI3K activation duringenucleation. Interestingly, biochemical analyses showed that the

    regulatory subunits of PI3K can bind to tubulin (Inukai et al.,2000; Kapeller et al., 1995), but we do not know whether thisoccurs in late erythroblasts or whether this affects activation or

    localization of PI3K.

    Interestingly, our results show that cell polarization is initiatedthrough repositioning of the nucleus but not MTOC during

    enucleation, similar to that seen in migrating fibroblasts (Gomeset al., 2005; Schmoranzer et al., 2009). Nuclear repositioningoccurs concomitantly with the rearrangement of the microtubule

    array that leads to asymmetric distribution of actomyosin andthus the generation of asymmetric force (see below). This forcemost probably drives the repositioning of the nucleus, somewhatsimilar to that seen in migrating cells where nuclear movement

    needs actin flow (Gomes et al., 2005).

    In comparison with migrating cells, in late erythroblasts the

    nucleus moves only a short distance (,1 mm) for the cell tobecome polarized. Then why do late erythroblasts need a longtime (2–3 hours) to establish polarization? This is probablybecause either rearrangement of the microtubule array or stable

    association of the nucleus with the plasma membrane, or both,might be a relatively slow event.

    Our proposed model for erythroblast enucleation issummarized in Fig. 8. The reorientation from a radialmicrotubule array to an asymmetric array somehow results inasymmetric activation of PI3K. Asymmetric accumulation of the

    PI3K products in the plasma membrane then induce asymmetricactin assembly and disassembly by stimulating their effectors,such as a-actinin (Franke et al., 1997), profilin (Lassing andLindberg, 1985) or gelsolin (Janmey et al., 1987), as well as Rac1activity (Ji et al., 2008) probably through activation of itsupstream regulator (Han et al., 1998; Kunisaki et al., 2006). This

    asymmetric actin assembly and disassembly and actomyosincontractions promote nuclear displacement as well as nuclearextrusion.

    In cells defective in this microtubule-regulated PI3K-dependent polarization mechanism, enucleation could beachieved by random cytoplasmic contractions alone but in a

    highly ineffective and slow manner. It is likely that such defectsin enucleation cause some proteins to be wrongly sorted to thereticulocytes or the extruded nucleus, leading to the formation of

    abnormal reticulocytes.

    Our results suggest that enucleation is carried out in a waymechanistically similar to directed cell migration. Some

    migrating cells form a polarized bleb-like protrusion in order tomove forward (Charras and Paluch, 2008; Haston and Shields,1984; Paluch et al., 2006), and the difference between the role of

    the bleb-like protrusion in cell migration and enucleationis probably influenced by differences in their cytoskeletalorganization. The unexpected findings in the present study thus

    Fig. 7. PI3K regulates cell polarization through promoting the movement

    of the nucleus but not the MTOC. Average distance between the centroids

    of the nucleus (red) or the MTOC (blue) and the cell in late erythroblasts

    treated with DMSO or LY294002. *P,0.05.

    Mechanics of mammalian erythroblast enucleation 347

    Journ

    alof

    Cell

    Scie

    nce

  • begin to elucidate the regulatory and mechanistic mechanisms

    underlying mammalian erythroblast enucleation.

    Materials and MethodsCell culture and infection

    Pregnant C57 BL/6J mice were purchased from the Genome Institute of Singaporeand the Center for Animal Resources (National University of Singapore). Thepurification and culturing of mouse fetal liver erythroblast precursors (CFU-e;TER119-negative cells) were performed as previously described (Ji et al., 2008;Zhang et al., 2003). For retroviral gene transfer experiments, we used the murinestem cell retroviral vector system (MSCV; Clontech, BD Biosciences). Infection ofprimary cells with mouse-specific retroviruses encoding fusion proteins of(3)Akt(PH)–GFP (Luo et al., 2005), (3)Akt(PHR25C)–GFP, mCherry, RFP orGFP–c-tubulin was performed as previously described (Ji et al., 2008). All animalexperiments were performed according to the relevant regulatory standards.

    Microscopy and image processing

    For phase-contrast and bright-field time-lapse live-cell imaging, purified TER119-negative cells grown on fibronectin-coated glass chamber dishes (McKenna andWang, 1989) were maintained at 37 C̊ in a custom-made incubator built on top ofan Axiovert 135 or Axiovert 200M inverted microscope (Carl Zeiss) and viewedwith a 1006, Plan Apo NA 1.25 or 1006, NA 1.30, Plan-NEOFLUAR lens.Images were acquired with a video camera (Mintron) or a cooled charge-coupleddevice camera (CoolSNAPHQ, Roper Scientific) and processed with MetaView(Universal Imaging) or Image-Pro Plus software. Images of cells expressing(3)Akt(PH)–GFP, cells expressing (3)Akt(PH)–GFP and mCherry or RFP, cellsexpressing (3)Akt(PHR25C)–GFP and cells expressing GFP–c-tubulin wereacquired using an Axiovert 200M inverted microscope (Carl Zeiss) equippedwith 1006, NA 1.4 Plan-Apochromat lens, a spinning disk confocal (YokogawaCSU-21) scan head and Hamamatsu Orca-ER cooled CCD camera. Bright fieldimages (Fig. 1B,D) were uniformly filtered to reduce noise using ImageJ (NIH,Bethesda, MD).

    For imaging of F-actin, stacks of images acquired using a LSM 510 Metaconfocal microscope system (1006, NA 1.4 Plan-Apochromat lens) werecollected at a vertical interval of 0.75 mm and processed with IMARIS(Bitplane, Scientific Solutions).

    For imaging of microtubules, Z-stacks of wide-field images were collected atintervals of 0.3 mm, were deconvoluted (for Fig. 3A) using AutoQuant (MediaCybernetics) and processed with a pattern recognition program that detects linearstructures as described previously (Wang, 2003). For imaging of EB1 andpericentrin, stacks of wide-field images were collected at vertical intervals of0.3 mm and projected with a maximum-intensity algorithm using ImageJ. For c-tubulin, single wide-field images were collected.

    Immunofluorescence

    Immunofluorescence staining was performed according to the methods previouslydescribed (Wheatley and Wang, 1998). We used primary antibodies against rabbitSer19-phosphorylated myosin light chain II (1:50, Cell Signaling), mouse anti-b-tubulin (1:1000, AbCam), mouse anti-EB1 (1:50, BD Biosciences, Pharmingen),mouse anti-c-tubulin (1:1000, Sigma); rabbit anti-pericentrin (1:700, Abcam).Secondary antibodies Alexa-Fluor-488-conjugated goat anti-rabbit IgG, Alexa-Fluor-488-conjugated goat anti-mouse IgG and Alexa-Fluor-546-conjugated goatanti-mouse IgG (Molecular Probes), all diluted 1:100. To observe F-actin,Rhodamine–phalloidin (Molecular Probes) was used at 1:300 dilution. Hoechst33258 (1 mg/ml) was used to locate the nucleus.

    Drug treatment

    Cytochalasin D (Sigma), blebbistatin (Toronto Research Chemicals), nocodazole(Sigma), LY294002 (Invitrogen) and wortmannin (Sigma) were dissolved inDMSO to make stock solutions of 25 mM, 100 mM, 3.3 mM, 50 mM and 1 mM,respectively, and applied to a culture dish at final concentration of 2 mM, 100 mM,10 mM, 50 mM and 1 mM, respectively. Note that .10 mM nocodazole wasrequired to completely depolymerize microtubules in late erythroblasts.

    Data analysis

    To measure the size of the nuclear area, the area of the ellipse corresponding to theshape of the nucleus was calculated using ImageJ. To track the distance betweenthe centroids of the cell and the centroids of the nucleus or the MTOC, eachperimeter was drawn and the centroid corresponding to the nucleus and the cellwas calculated using ImageJ. The distance between these centroids was calculated.

    AcknowledgementsWe thank TLL Microscopy and Animal Care facilities and ShazminaRafee for their technical assistance, Koichi Okumura (CancerScience Institute of Singapore) for helpful suggestions, and G.Wright and J. Brocher for advice on image processing. We thank allmembers of the Murata-Hori laboratory for discussions and/orcritical reading of the manuscript.

    FundingThis study was supported by intramural funds from the Temasek LifeSciences Laboratory to M.M.-H. and by the National Institutes ofHealth [grant number P01 HL 32262] to H.F.L. Deposited in PMCfor release after 12 months.

    Supplementary material available online at

    http://jcs.biologists.org/lookup/suppl/doi:10.1242/jcs.088286/-/DC1

    ReferencesCharras, G. and Paluch, E. (2008). Blebs lead the way: how to migrate without

    lamellipodia. Nat. Rev. Mol. Cell Biol. 9, 730-736.

    Franke, T. F., Kaplan, D. R., Cantley, L. C. and Toker, A. (1997). Direct regulation

    of the Akt proto-oncogene product by phosphatidylinositol-3,4-bisphosphate. Science275, 665-668.

    Ghaffari, S., Kitidis, C., Zhao, W., Marinkovic, D., Fleming, M. D., Luo, B.,

    Marszalek, J. and Lodish, H. F. (2006). AKT induces erythroid-cell maturation ofJAK2-deficient fetal liver progenitor cells and is required for Epo regulation of

    erythroid-cell differentiation. Blood 107, 1888-1891.

    Gomes, E. R., Jani, S. and Gundersen, G. G. (2005). Nuclear movement regulated byCdc42, MRCK, myosin, and actin flow establishes MTOC polarization in migrating

    cells. Cell 121, 451-463.

    Han, J., Luby-Phelps, K., Das, B., Shu, X., Xia, Y., Mosteller, R. D., Krishna, U. M.,

    Falck, J. R., White, M. A. and Broek, D. (1998). Role of substrates and products of

    PI 3-kinase in regulating activation of Rac-related guanosine triphosphatases by Vav.Science 279, 558-560.

    Haston, W. S. and Shields, J. M. (1984). Contraction waves in lymphocyte locomotion.

    J. Cell Sci. 68, 227-241.

    Haugh, J. M., Codazzi, F., Teruel, M. and Meyer, T. (2000). Spatial sensing in

    fibroblasts mediated by 39 phosphoinositides. J. Cell Biol. 151, 1269-1280.Ihle, J. N. and Gilliland, D. G. (2007). Jak2: normal function and role in hematopoietic

    disorders. Curr. Opin. Genet. Dev. 17, 8-14.

    Inukai, K., Funaki, M., Nawano, M., Katagiri, H., Ogihara, T., Anai, M., Onishi, Y.,

    Sakoda, H., Ono, H., Fukushima, Y. et al. (2000). The N-terminal 34 residues of the

    55 kDa regulatory subunits of phosphoinositide 3-kinase interact with tubulin.

    Biochem. J. 346, 483-489.

    Janmey, P. A., Iida, K., Yin, H. L. and Stossel, T. P. (1987). Polyphosphoinositide

    micelles and polyphosphoinositide-containing vesicles dissociate endogenous gelso-lin-actin complexes and promote actin assembly from the fast-growing end of actin

    filaments blocked by gelsolin. J. Biol. Chem. 262, 12228-12236.

    Ji, P., Jayapal, S. R. and Lodish, H. F. (2008). Enucleation of cultured mouse fetalerythroblasts requires Rac GTPases and mDia2. Nat. Cell Biol. 10, 314-321.

    Kapeller, R., Toker, A., Cantley, L. C. and Carpenter, C. L. (1995). Phosphoinositide

    3-kinase binds constitutively to alpha/beta-tubulin and binds to gamma-tubulin inresponse to insulin. J. Biol. Chem. 270, 25985-25991.

    Keerthivasan, G., Small, S., Liu, H., Wickrema, A. and Crispino, J. D. (2010).

    Vesicle trafficking plays a novel role in erythroblast enucleation. Blood 116, 3331-3340.

    Fig. 8. Proposed model for PI3K-mediated regulation of erythroblast enucleation. The reorientation from a radial microtubule array to an asymmetric array

    results in localized activation of PI3K and thereby promotes asymmetric actin assembly. Enucleation is triggered and continued by a pressure generated by

    contractions of cytoplasmic actomyosin (large arrow) and is probably completed by contractions of concentrated actomyosin at the bleb neck (small arrows).

    Journal of Cell Science 125 (2)348

    Journ

    alof

    Cell

    Scie

    nce

    http://dx.doi.org/10.1038%2Fnrm2453http://dx.doi.org/10.1038%2Fnrm2453http://dx.doi.org/10.1126%2Fscience.275.5300.665http://dx.doi.org/10.1126%2Fscience.275.5300.665http://dx.doi.org/10.1126%2Fscience.275.5300.665http://dx.doi.org/10.1182%2Fblood-2005-06-2304http://dx.doi.org/10.1182%2Fblood-2005-06-2304http://dx.doi.org/10.1182%2Fblood-2005-06-2304http://dx.doi.org/10.1182%2Fblood-2005-06-2304http://dx.doi.org/10.1016%2Fj.cell.2005.02.022http://dx.doi.org/10.1016%2Fj.cell.2005.02.022http://dx.doi.org/10.1016%2Fj.cell.2005.02.022http://dx.doi.org/10.1126%2Fscience.279.5350.558http://dx.doi.org/10.1126%2Fscience.279.5350.558http://dx.doi.org/10.1126%2Fscience.279.5350.558http://dx.doi.org/10.1126%2Fscience.279.5350.558http://dx.doi.org/10.1083%2Fjcb.151.6.1269http://dx.doi.org/10.1083%2Fjcb.151.6.1269http://dx.doi.org/10.1016%2Fj.gde.2006.12.009http://dx.doi.org/10.1016%2Fj.gde.2006.12.009http://dx.doi.org/10.1042%2F0264-6021%3A3460483http://dx.doi.org/10.1042%2F0264-6021%3A3460483http://dx.doi.org/10.1042%2F0264-6021%3A3460483http://dx.doi.org/10.1042%2F0264-6021%3A3460483http://dx.doi.org/10.1038%2Fncb1693http://dx.doi.org/10.1038%2Fncb1693http://dx.doi.org/10.1074%2Fjbc.270.43.25985http://dx.doi.org/10.1074%2Fjbc.270.43.25985http://dx.doi.org/10.1074%2Fjbc.270.43.25985http://dx.doi.org/10.1182%2Fblood-2010-03-277426http://dx.doi.org/10.1182%2Fblood-2010-03-277426http://dx.doi.org/10.1182%2Fblood-2010-03-277426

  • Khandani, A., Eng, E., Jongstra-Bilen, J., Schreiber, A. D., Douda, D., Samavarchi-Tehrani, P. and Harrison, R. E. (2007). Microtubules regulate PI-3K activity andrecruitment to the phagocytic cup during Fcgamma receptor-mediated phagocytosis innonelicited macrophages. J. Leukoc. Biol. 82, 417-428.

    Koury, M. J., Sawyer, S. T. and Brandt, S. J. (2002). New insights into erythropoiesis.Curr. Opin. Hematol. 9, 93-100.

    Koury, S. T., Koury, M. J. and Bondurant, M. C. (1989). Cytoskeletal distributionand function during the maturation and enucleation of mammalian erythroblasts. J.Cell Biol. 109, 3005-3013.

    Kunisaki, Y., Nishikimi, A., Tanaka, Y., Takii, R., Noda, M., Inayoshi, A.,

    Watanabe, K., Sanematsu, F., Sasazuki, T., Sasaki, T. et al. (2006). DOCK2 is aRac activator that regulates motility and polarity during neutrophil chemotaxis. J. CellBiol. 174, 647-652.

    Lassing, I. and Lindberg, U. (1985). Specific interaction between phosphatidylinositol4,5-bisphosphate and profilactin. Nature 314, 472-474.

    Luo, J., Field, S. J., Lee, J. Y., Engelman, J. A. and Cantley, L. C. (2005). The p85regulatory subunit of phosphoinositide 3-kinase down-regulates IRS-1 signaling viathe formation of a sequestration complex. J. Cell Biol. 170, 455-464.

    Matsumura, F., Ono, S., Yamakita, Y., Totsukawa, G. and Yamashiro, S. (1998).Specific localization of serine 19 phosphorylated myosin II during cell locomotionand mitosis of cultured cells. J. Cell Biol. 140, 119-129.

    McKenna, N. M. and Wang, Y. L. (1989). Culturing cells on the microscope stage.Methods Cell Biol. 29, 195-205.

    Mimori-Kiyosue, Y., Shiina, N. and Tsukita, S. (2000). The dynamic behavior of theAPC-binding protein EB1 on the distal ends of microtubules. Curr. Biol. 10, 865-868.

    Mukhina, S., Wang, Y. L. and Murata-Hori, M. (2007). Alpha-actinin is required fortightly regulated remodeling of the actin cortical network during cytokinesis. Dev.Cell 13, 554-565.

    Paluch, E., Sykes, C., Prost, J. and Bornens, M. (2006). Dynamic modes of thecortical actomyosin gel during cell locomotion and division. Trends Cell Biol. 16, 5-10.

    Parent, C. A., Blacklock, B. J., Froehlich, W. M., Murphy, D. B. and Devreotes,P. N. (1998). G protein signaling events are activated at the leading edge ofchemotactic cells. Cell 95, 81-91.

    Repasky, E. A. and Eckert, B. S. (1981). The effect of cytochalasin B on theenucleation of erythroid cells in vitro. Cell Tissue Res. 221, 85-91.

    Richmond, T. D., Chohan, M. and Barber, D. L. (2005). Turning cells red: signal

    transduction mediated by erythropoietin. Trends Cell Biol. 15, 146-155.

    Schmoranzer, J., Fawcett, J. P., Segura, M., Tan, S., Vallee, R. B., Pawson, T. and

    Gundersen, G. G. (2009). Par3 and dynein associate to regulate local microtubule

    dynamics and centrosome orientation during migration. Curr. Biol. 19, 1065-1074.

    Servant, G., Weiner, O. D., Herzmark, P., Balla, T., Sedat, J. W. and Bourne, H. R.

    (2000). Polarization of chemoattractant receptor signaling during neutrophil

    chemotaxis. Science 287, 1037-1040.

    Simpson, C. F. and Kling, J. M. (1967). The mechanism of denucleation in circulating

    erythroblasts. J. Cell Biol. 35, 237-245.

    Skutelsky, E. and Danon, D. (1967). An electron microscopic study of nuclear

    elimination from the late erythroblast. J. Cell Biol. 33, 625-635.

    Sonoda, Y., Sasaki, K., Suda, M., Itano, C. and Iwatsuki, H. (1998). Effects of

    colchicine on the enucleation of erythroid cells and macrophages in the liver of mouse

    embryos: ultrastructural and three-dimensional studies. Anat. Rec. 251, 290-296.

    Straight, A. F., Cheung, A., Limouze, J., Chen, I., Westwood, N. J., Sellers, J. R. and

    Mitchison, T. J. (2003). Dissecting temporal and spatial control of cytokinesis with a

    myosin II Inhibitor. Science 299, 1743-1747.

    Wang, Y. L. (2003). Computational restoration of fluorescence images: noise reduction,

    deconvolution, and pattern recognition. Methods Cell Biol. 72, 337-348.

    Wheatley, S. P. and Wang, Y. L. (1998). Indirect immunofluorescence microscopy in

    cultured cells. Methods Cell Biol. 57, 313-332.

    Xu, J., Wang, F., Van Keymeulen, A., Rentel, M. and Bourne, H. R. (2005).

    Neutrophil microtubules suppress polarity and enhance directional migration. Proc.

    Natl. Acad. Sci. USA 102, 6884-6889.

    Yoshida, H., Kawane, K., Koike, M., Mori, Y., Uchiyama, Y. and Nagata, S. (2005).

    Phosphatidylserine-dependent engulfment by macrophages of nuclei from erythroid

    precursor cells. Nature 437, 754-758.

    Zhang, J., Socolovsky, M., Gross, A. W. and Lodish, H. F. (2003). Role of Ras

    signaling in erythroid differentiation of mouse fetal liver cells: functional analysis by

    a flow cytometry-based novel culture system. Blood 102, 3938-3946.

    Zhao, W., Kitidis, C., Fleming, M. D., Lodish, H. F. and Ghaffari, S. (2006).

    Erythropoietin stimulates phosphorylation and activation of GATA-1 via the PI3-

    kinase/AKT signaling pathway. Blood 107, 907-915.

    Mechanics of mammalian erythroblast enucleation 349

    Journ

    alof

    Cell

    Scie

    nce

    http://dx.doi.org/10.1189%2Fjlb.0706469http://dx.doi.org/10.1189%2Fjlb.0706469http://dx.doi.org/10.1189%2Fjlb.0706469http://dx.doi.org/10.1189%2Fjlb.0706469http://dx.doi.org/10.1097%2F00062752-200203000-00002http://dx.doi.org/10.1097%2F00062752-200203000-00002http://dx.doi.org/10.1083%2Fjcb.109.6.3005http://dx.doi.org/10.1083%2Fjcb.109.6.3005http://dx.doi.org/10.1083%2Fjcb.109.6.3005http://dx.doi.org/10.1083%2Fjcb.200602142http://dx.doi.org/10.1083%2Fjcb.200602142http://dx.doi.org/10.1083%2Fjcb.200602142http://dx.doi.org/10.1083%2Fjcb.200602142http://dx.doi.org/10.1038%2F314472a0http://dx.doi.org/10.1038%2F314472a0http://dx.doi.org/10.1083%2Fjcb.200503088http://dx.doi.org/10.1083%2Fjcb.200503088http://dx.doi.org/10.1083%2Fjcb.200503088http://dx.doi.org/10.1083%2Fjcb.140.1.119http://dx.doi.org/10.1083%2Fjcb.140.1.119http://dx.doi.org/10.1083%2Fjcb.140.1.119http://dx.doi.org/10.1016%2FS0091-679X%2808%2960195-8http://dx.doi.org/10.1016%2FS0091-679X%2808%2960195-8http://dx.doi.org/10.1016%2FS0960-9822%2800%2900600-Xhttp://dx.doi.org/10.1016%2FS0960-9822%2800%2900600-Xhttp://dx.doi.org/10.1016%2Fj.devcel.2007.08.003http://dx.doi.org/10.1016%2Fj.devcel.2007.08.003http://dx.doi.org/10.1016%2Fj.devcel.2007.08.003http://dx.doi.org/10.1016%2Fj.tcb.2005.11.003http://dx.doi.org/10.1016%2Fj.tcb.2005.11.003http://dx.doi.org/10.1016%2Fj.tcb.2005.11.003http://dx.doi.org/10.1016%2FS0092-8674%2800%2981784-5http://dx.doi.org/10.1016%2FS0092-8674%2800%2981784-5http://dx.doi.org/10.1016%2FS0092-8674%2800%2981784-5http://dx.doi.org/10.1007%2FBF00216572http://dx.doi.org/10.1007%2FBF00216572http://dx.doi.org/10.1016%2Fj.tcb.2005.01.007http://dx.doi.org/10.1016%2Fj.tcb.2005.01.007http://dx.doi.org/10.1016%2Fj.cub.2009.05.065http://dx.doi.org/10.1016%2Fj.cub.2009.05.065http://dx.doi.org/10.1016%2Fj.cub.2009.05.065http://dx.doi.org/10.1126%2Fscience.287.5455.1037http://dx.doi.org/10.1126%2Fscience.287.5455.1037http://dx.doi.org/10.1126%2Fscience.287.5455.1037http://dx.doi.org/10.1083%2Fjcb.35.1.237http://dx.doi.org/10.1083%2Fjcb.35.1.237http://dx.doi.org/10.1083%2Fjcb.33.3.625http://dx.doi.org/10.1083%2Fjcb.33.3.625http://dx.doi.org/10.1002%2F%28SICI%291097-0185%28199807%29251%3A3%3C290%3A%3AAID-AR3%3E3.0.CO%3B2-%23http://dx.doi.org/10.1002%2F%28SICI%291097-0185%28199807%29251%3A3%3C290%3A%3AAID-AR3%3E3.0.CO%3B2-%23http://dx.doi.org/10.1002%2F%28SICI%291097-0185%28199807%29251%3A3%3C290%3A%3AAID-AR3%3E3.0.CO%3B2-%23http://dx.doi.org/10.1126%2Fscience.1081412http://dx.doi.org/10.1126%2Fscience.1081412http://dx.doi.org/10.1126%2Fscience.1081412http://dx.doi.org/10.1016%2FS0091-679X%2803%2972016-0http://dx.doi.org/10.1016%2FS0091-679X%2803%2972016-0http://dx.doi.org/10.1016%2FS0091-679X%2808%2961588-5http://dx.doi.org/10.1016%2FS0091-679X%2808%2961588-5http://dx.doi.org/10.1073%2Fpnas.0502106102http://dx.doi.org/10.1073%2Fpnas.0502106102http://dx.doi.org/10.1073%2Fpnas.0502106102http://dx.doi.org/10.1038%2Fnature03964http://dx.doi.org/10.1038%2Fnature03964http://dx.doi.org/10.1038%2Fnature03964http://dx.doi.org/10.1182%2Fblood-2003-05-1479http://dx.doi.org/10.1182%2Fblood-2003-05-1479http://dx.doi.org/10.1182%2Fblood-2003-05-1479http://dx.doi.org/10.1182%2Fblood-2005-06-2516http://dx.doi.org/10.1182%2Fblood-2005-06-2516http://dx.doi.org/10.1182%2Fblood-2005-06-2516

    Fig 1Fig 2Fig 3Fig 4Fig 5Fig 6Fig 7Ref 1Ref 2Ref 3Ref 4Ref 5Ref 6Ref 7Ref 8Ref 9Ref 10Ref 11Ref 12Ref 13Fig 8Ref 14Ref 15Ref 16Ref 17Ref 18Ref 19Ref 20Ref 21Ref 22Ref 23Ref 24Ref 25Ref 26Ref 27Ref 28Ref 29Ref 30Ref 31Ref 32Ref 33Ref 34Ref 35Ref 36Ref 37Ref 38Ref 39