+ All Categories
Home > Documents > Protein 4.1R regulates interphase microtubule organization ... · Microtubule organization is...

Protein 4.1R regulates interphase microtubule organization ... · Microtubule organization is...

Date post: 04-Jul-2020
Category:
Upload: others
View: 2 times
Download: 0 times
Share this document with a friend
10
Introduction Microtubule organization is essential for directional intracellular transport, the modulation of cell morphology and locomotion, and the formation of the spindle apparatus during cell division. In animal cells, the centrosome is the major microtubule-organizing center that, during interphase, originates a microtubule radial array. In fibroblasts, microtubules appear to project radially from a single spot, the microtubule-organizing center, implying that they remain tightly associated with the centrosome. By contrast, in neurons and polarized epithelial cells, many microtubules are released from centrosomes and become reorganized into nonradial arrays that project into neurites or away from the apical face of the cell (Dammermann et al., 2003; Doxsey, 2001). Centrosome-nucleated microtubules are polarized with their rapidly growing (plus) ends in the cytoplasm and their slowly growing (or minus) ends anchored at the centrosome. Cytoplasmic dynein is the predominant minus-end-directed microtubule motor in eukaryotic cells and usually conducts cargo in association with dynactin, a 20S complex consisting of at least nine polypeptides that appear to play an essential role in linking cargo to the dynein motor (Paschal et al., 1993). The dynein-dynactin complex has been shown to be a major contributor to microtubule organization and centrosome integrity (Quintyne et al., 1999). Red-blood-cell protein 4.1 (4.1R or 4.1R 80 ) was originally identified as an 80-kDa multifunctional protein of the membrane skeleton of human erythrocytes. In these cells, protein 4.1R stabilizes the spectrin-actin network and mediates the attachment of the underlying cytoskeleton to the overlaying lipid bilayer through interactions with integral membrane proteins (Conboy, 1993). In nonerythroid cells, multiple isoforms of 4.1R are expressed as a result of extensive alternative splicing of the 4.1R-encoding pre-mRNA (Conboy, 1999; Tang et al., 1990). This event is cell and tissue specific, and also dependent on the growth and differentiation stages of the cell (Baklouti et al., 1997; Conboy et al., 1991; Chasis et al., 1993; Hou and Conboy, 2001; Schischmanoff et al., 1997). Immunological studies have detected 4.1R epitopes at different subcellular locations. Concomitantly, the association of 4.1R with proteins localized at different intracellular sites has been reported (Cohen et al., 1998; De Cárcer et al., 1995; Hou et al., 2000; Kontrogianni-Konstantopoulos et al., 2001; Kontrogianni- Konstantopoulos et al., 2000; Krauss et al., 1997; Lallena and Correas, 1997; Lallena et al., 1998; Mattagajasingh et al., 6197 In human red blood cells, protein 4.1 (4.1R) stabilizes the spectrin-actin network and anchors it to the plasma membrane. To contribute to the characterization of functional roles of 4.1R in nonerythroid cells, we analysed the effect of ectopic expression of 4.1R isoforms on interphase microtubules in fibroblastic cells. We found that specific 4.1R isoforms disturbed the microtubule architecture but not the actin cytoskeleton. Biochemical sedimentation and/or confocal microscopy analyses showed that the pericentriolar components γ-tubulin and pericentrin remained at centrosomes, whereas the distributions of proteins p150 Glued and the dynein intermediate chain were altered. Remarkably, 4.1R was displaced from the centrosome. In microtubule depolymerizing-repolymerizing assays, 4.1R-transfected cells showed an ability to depolymerize and nucleate microtubules that was similar to that of untransfected cells; however, microtubules became disorganized soon after regrowth. In microtubule-depolymerized transfected cells and during the initial steps of microtubule regrowth, centrosomal 4.1R localized with γ-tubulin but did not when microtubules became disorganized. To learn more about centrosomal 4.1R function, isolated centrosomes were examined by confocal microscopy, western blot and in vitro microtubule aster-assembly assays. The experiments showed that 4.1R was present in isolated centrosome preparations, that it remained in the center of in-vitro-assembled microtubule asters and that more asters were assembled by the addition of protein 4.1R fused to glutathione-S-transferase. Together, these results indicate that 4.1R plays a key role at the centrosome, contributing to the maintenance of a radial microtubule organization. Key words: Protein 4.1R, Centrosome, Microtubules Summary Protein 4.1R regulates interphase microtubule organization at the centrosome Carmen M. Pérez-Ferreiro 1 , Isabelle Vernos 2 and Isabel Correas 1, * 1 Departamento de Biología Molecular, Centro de Biología Molecular ‘Severo Ochoa’ (UAM/CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain 2 European Molecular Biology Laboratory (EMBL), Cell Biology and Cell Biophysics Programme, 69117 Heidelberg, Germany *Author for correspondence (e-mail: [email protected]) Accepted 15 September 2004 Journal of Cell Science 117, 6197-6206 Published by The Company of Biologists 2004 doi:10.1242/jcs.01544 Research Article
Transcript
Page 1: Protein 4.1R regulates interphase microtubule organization ... · Microtubule organization is essential for directional intracellular transport, the modulation of cell morphology

IntroductionMicrotubule organization is essential for directionalintracellular transport, the modulation of cell morphology andlocomotion, and the formation of the spindle apparatus duringcell division. In animal cells, the centrosome is the majormicrotubule-organizing center that, during interphase,originates a microtubule radial array. In fibroblasts,microtubules appear to project radially from a single spot, themicrotubule-organizing center, implying that they remaintightly associated with the centrosome. By contrast, in neuronsand polarized epithelial cells, many microtubules are releasedfrom centrosomes and become reorganized into nonradialarrays that project into neurites or away from the apical faceof the cell (Dammermann et al., 2003; Doxsey, 2001).Centrosome-nucleated microtubules are polarized with theirrapidly growing (plus) ends in the cytoplasm and their slowlygrowing (or minus) ends anchored at the centrosome.Cytoplasmic dynein is the predominant minus-end-directedmicrotubule motor in eukaryotic cells and usually conductscargo in association with dynactin, a 20S complex consistingof at least nine polypeptides that appear to play an essentialrole in linking cargo to the dynein motor (Paschal et al., 1993).The dynein-dynactin complex has been shown to be a major

contributor to microtubule organization and centrosomeintegrity (Quintyne et al., 1999).

Red-blood-cell protein 4.1 (4.1R or 4.1R80) was originallyidentified as an 80-kDa multifunctional protein of themembrane skeleton of human erythrocytes. In these cells,protein 4.1R stabilizes the spectrin-actin network and mediatesthe attachment of the underlying cytoskeleton to the overlayinglipid bilayer through interactions with integral membraneproteins (Conboy, 1993).

In nonerythroid cells, multiple isoforms of 4.1R areexpressed as a result of extensive alternative splicing of the4.1R-encoding pre-mRNA (Conboy, 1999; Tang et al., 1990).This event is cell and tissue specific, and also dependent on thegrowth and differentiation stages of the cell (Baklouti et al.,1997; Conboy et al., 1991; Chasis et al., 1993; Hou andConboy, 2001; Schischmanoff et al., 1997). Immunologicalstudies have detected 4.1R epitopes at different subcellularlocations. Concomitantly, the association of 4.1R with proteinslocalized at different intracellular sites has been reported(Cohen et al., 1998; De Cárcer et al., 1995; Hou et al., 2000;Kontrogianni-Konstantopoulos et al., 2001; Kontrogianni-Konstantopoulos et al., 2000; Krauss et al., 1997; Lallena andCorreas, 1997; Lallena et al., 1998; Mattagajasingh et al.,

6197

In human red blood cells, protein 4.1 (4.1R) stabilizes thespectrin-actin network and anchors it to the plasmamembrane. To contribute to the characterization offunctional roles of 4.1R in nonerythroid cells, we analysedthe effect of ectopic expression of 4.1R isoforms oninterphase microtubules in fibroblastic cells. We foundthat specific 4.1R isoforms disturbed the microtubulearchitecture but not the actin cytoskeleton. Biochemicalsedimentation and/or confocal microscopy analysesshowed that the pericentriolar components γ-tubulinand pericentrin remained at centrosomes, whereas thedistributions of proteins p150Glued and the dyneinintermediate chain were altered. Remarkably, 4.1Rwas displaced from the centrosome. In microtubuledepolymerizing-repolymerizing assays, 4.1R-transfectedcells showed an ability to depolymerize and nucleatemicrotubules that was similar to that of untransfected cells;however, microtubules became disorganized soon after

regrowth. In microtubule-depolymerized transfected cellsand during the initial steps of microtubule regrowth,centrosomal 4.1R localized with γ-tubulin but did not whenmicrotubules became disorganized.

To learn more about centrosomal 4.1R function, isolatedcentrosomes were examined by confocal microscopy,western blot and in vitro microtubule aster-assemblyassays. The experiments showed that 4.1R was present inisolated centrosome preparations, that it remained in thecenter of in-vitro-assembled microtubule asters and thatmore asters were assembled by the addition of protein 4.1Rfused to glutathione-S-transferase. Together, these resultsindicate that 4.1R plays a key role at the centrosome,contributing to the maintenance of a radial microtubuleorganization.

Key words: Protein 4.1R, Centrosome, Microtubules

Summary

Protein 4.1R regulates interphase microtubuleorganization at the centrosomeCarmen M. Pérez-Ferreiro1, Isabelle Vernos2 and Isabel Correas1,*1Departamento de Biología Molecular, Centro de Biología Molecular ‘Severo Ochoa’ (UAM/CSIC), Universidad Autónoma de Madrid, 28049Madrid, Spain2European Molecular Biology Laboratory (EMBL), Cell Biology and Cell Biophysics Programme, 69117 Heidelberg, Germany*Author for correspondence (e-mail: [email protected])

Accepted 15 September 2004Journal of Cell Science 117, 6197-6206 Published by The Company of Biologists 2004doi:10.1242/jcs.01544

Research Article

Page 2: Protein 4.1R regulates interphase microtubule organization ... · Microtubule organization is essential for directional intracellular transport, the modulation of cell morphology

6198

2000; Nunomura et al., 1997), thus suggesting that 4.1R mightbe involved in many processes in nucleated cells. A possiblerole for 4.1R in organizing the nuclear architecture is suggestedby interactions observed between 4.1R and nuclearcomponents of the splicing machinery (Lallena and Correas,1997; Lallena et al., 1998). More recently, 4.1R has beenshown to be essential for proper nuclear assembly (Krauss etal., 2002). A role has also been proposed for 4.1R in organizingmicrotubule architecture and the mitotic spindle poles as 4.1Rinteracts with interphase microtubules (Perez-Ferreiro et al.,2001), with a novel centrosomal protein termed CPAP (Hunget al., 2000), and with the nuclear mitotic apparatus protein(Mattagajasingh et al., 1999).

Proteins with key microtubule-anchoring functions arereported to provoke disorganization of the interphasemicrotubule array when ectopically expressed in COS-7 cells(Dammermann and Merdes, 2002; Quintyne et al., 1999).However, they might not affect interphase microtubuleorganization in other cell lines. Thus, dynamitin, a componentof the dynactin complex, was reported to have no effect oninterphase microtubules in HeLa cells, whereas, conversely,COS-7 cells overexpressing dynamitin contained microtubulesthat were less focused than normal (Quintyne et al., 1999). Wehave described the colocalization of ectopically expressed 4.1Rwith the microtubule network in human T cells, and thedisruption of microtubule architecture in COS-7 cells (Perez-Ferreiro et al., 2001). To gain insights into the role of 4.1R inthe organization of interphase microtubules, we have performedtransfection experiments using different 4.1R-encoding cDNAs,microtubule depolymerization-repolymerization assays,confocal microscopy and biochemical sedimentation analysesin COS-7 cells. We have found that interphase microtubules areselectively perturbed by specific exogenous 4.1R isoforms andthat microtubule disorganization is accompanied by altereddistributions of the dynein-dynactin complex and, moreinterestingly, of centrosomal 4.1R. Results of furtherexperiments indicate that 4.1R plays an essential role inregulating microtubule organization at the centrosome.

Materials and MethodsCell culture and transfectionHuman T-lymphoid Molt-4 and COS-7 cells were grown as describedpreviously (Perez-Ferreiro et al., 2001). Transfection experimentswere performed by electroporation using the Electro Cell Manipulator600 (BTX, San Diego, CA). Cells were processed 48 hours aftertransfection.

cDNA cloning and composite cDNA constructsThe 4.1R135∆4,5,16, 4.1R135∆16, 4.1R135∆16,19, 4.1R80, 4.1R80∆16,4.1R80∆5,16 and 4.1R60∆16,18 cDNAs were cloned from Molt-4 Tcells and tagged as described (Luque and Correas, 2000; Luque et al.,1999). The 4.1R60∆16,18-GFP cDNA was constructed as detailedelsewhere (Luque et al., 2003). The glutathione-S-transferase (GST),GST C-terminus (GST-Cter), GST-4.1R60∆16,18 and GST-4.1R80∆16proteins were prepared as described elsewhere (Perez-Ferreiro et al.,2001).

AntibodiesAnti-c-Myc mouse monoclonal antibody 9E10 was obtained from theAmerican Type Culture Collection. Anti-4.1R (10b) antibody was an

affinity-purified polyclonal antibody generated as describedpreviously (Correas et al., 1986). Anti-α-tubulin (DM1A), anti-γ-tubulin (GTU-88), anti-actin (AC-40) and anti-dynein (intermediatechain) mouse monoclonal antibodies were purchased from Sigma.Anti-tubulin antibody YL1/2 was a rat monoclonal antibodypurchased from Sera-Lab. Anti-p150Glued antibody was a mousemonoclonal antibody from BD Transduction Laboratories. Anti-pericentrin antibody was a rabbit polyclonal antibody from Babco.Anti-GST antibody was a rabbit polyclonal antibody from Sigma.Horseradish-peroxidase-labeled secondary antibodies were obtainedfrom Southern Biotechnology Associates. Fluorescence-labeledsecondary antibodies and Alexa-Fluor-594/phalloidin were obtainedfrom Molecular Probes.

Immunofluorescence and confocal microscopyCOS-7 cells were fixed with 4% formalin (37% formaldehydesolution, Sigma), permeabilized and blocked (De Cárcer et al., 1995).In some cases, cells were fixed with methanol-acetone (1:1 v/v) for10 minutes at –20°C. Cells were incubated with the appropriateantibodies and processed as described (De Cárcer et al., 1995).Controls with primary antibodies omitted were included in eachexperiment. In figures 1-4, at least 100 transfected cells were analysedfrom four independent experiments. Images were obtained using aZeiss epifluorescence microscope or a Bio-Rad Radiance 2000confocal laser microscope.

Western-blot analysisProtein samples were separated by sodium-dodecyl-sulfatepolyacrylamide-gel electrophoresis (SDS-PAGE) and transferred toImmobilon polyvinylidine difluoride (Millipore) in Tris-borate buffer,pH 8.2. Membranes were processed and developed as described (DeCárcer et al., 1995).

Microtubule regrowth assaysCOS-7 cells were transfected, seeded on coverslips and grown for 48hours as described above. Microtubules were depolymerized in 10 µMnocodazole (Sigma) in Dulbecco’s modified Eagle’s medium(DMEM) for 4 hours and then washed three times with DMEM andincubated in fresh medium to allow microtubule regrowth. Coverslipswere fixed at timed intervals and processed for immunofluorescenceas described above.

Sedimentation assaysCOS-7 cells were harvested, lysed and sedimented as described(Echeverri et al., 1996). Sedimentation standards, bovine serumalbumin (4.4S), catalase (11.3S) and thyroglobulin (19S), were run inparallel. Equal volumes of each fraction were analysed by SDS-PAGEfollowed by western blotting (De Cárcer et al., 1995).

Centrosome isolationHuman centrosomes were isolated from Molt-4 T cells as previouslydescribed (Moudjou and Bornens, 1998). These cells were chosenbecause the centrosome isolation protocol used by Modjou andBornens (Moudjou and Bornens, 1998) is optimized for human T cellsand gives a high yield. Briefly, nocodazole- and cytochalasin-D-treated Molt-4 T cells (~109) were used for centrosome isolation. Thecentrosomal suspension obtained was layered onto a discontinuoussucrose gradient using the procedure described previously (Moudjouand Bornens, 1998). For immunofluorescence analysis, thecentrosome-containing fractions were spun onto coverslips andprobed with anti-α-tubulin, anti-γ-tubulin or anti-4.1R antibodiesusing previously described protocols (Moudjou and Bornens, 1998).

Journal of Cell Science 117 (25)

Page 3: Protein 4.1R regulates interphase microtubule organization ... · Microtubule organization is essential for directional intracellular transport, the modulation of cell morphology

6199Centrosomal 4.1R and microtubules

Microtubule nucleation assaysThe microtubule-nucleation ability of isolated centrosomes wasanalysed according to the method of Mitchison and Kirchner(Mitchison and Kirschner, 1984). The centrosome-containingfractions were incubated in 80 mM PIPES [piperazine-N,N′-bis(2-ethanesulfonic acid)], pH 6.8, 1 mM MgCl2, 1 mM EGTA and 1 mMGTP, containing 3 mg ml–1 bovine brain tubulin (Molecular Probes)for 8 minutes at 37°C. Microtubules were fixed by addingglutaraldehyde (1%), sedimented onto coverslips and then subjectedto immunofluorescence analysis using anti-α-tubulin and anti-4.1Rantibodies. 12 randomly chosen fields were examined on eachcoverslip under the microscope and the number of asters formed wascounted. To test the effect of addition of GST-4.1R60∆16,18, theisolated centrosomes were preincubated with the recombinant proteinfor 45 minutes at 4°C. GST alone was used as a negative control.

Pull-down assaysGST and the recombinant proteins GST-4.1R60∆16,18 and GST-Cterwere prepared as indicated (Perez-Ferreiro et al., 2001). COS-7 cellextracts were prepared by washing cells twice with PBS and addinglysis buffer (25 mM Tris, pH 7.6, 200 mM NaCl, 1% Triton X-100)containing 10 µg ml–1 pepstatin, leupeptin and aprotinin, and 1 mMphenylmethylsulfonyl fluoride. Cells were scraped off the plate,transferred to Eppendorf tubes and incubated for 10 minutes on ice.The lysates were centrifuged at high speed in a minicentrifuge at 4°Cfor 10 minutes. The supernatants were recovered and incubated for 1hour at 4°C with the glutathione/Sepharose-4B column loaded withthe corresponding GST proteins. After extensive washes with lysisbuffer, the beads were resuspended in Laemmli buffer and boiled for5 minutes. Recovery of the GST fusion proteins was confirmed bySDS-PAGE and Coomassie staining. The proteins were transferred tonitrocellulose membranes according to standard procedures andvisualized by immunoblotting as described above.

ResultsPerturbation of microtubule organization provoked byspecific 4.1R isoforms exogenously expressed in COS-7cellsWe have previously shown that ectopic expression of protein4.1R80∆16 in COS-7 cells provoked interphase microtubuledisorganization (Perez-Ferreiro et al., 2001). To gain insightsinto the role of protein 4.1R in the organization of interphasemicrotubules, we first examined whether microtubuleperturbation is a general effect caused by ectopic expression of4.1R in COS-7 cells. We chose to assay cDNAs encodingdifferent 4.1R isoform types: isoforms translated from the mostupstream start codon, ATG-1 (approximate molecular weight135 kDa), and from the downstream start sites, ATG-2(approximate molecular weight 80 kDa) and ATG-3(approximate molecular weight 60 kDa) (Fig. 1A). The three4.1R isoform types present different subcellular distribution:ATG1-translated 4.1R isoforms predominantly appear at non-nuclear sites (Luque et al., 1999); ATG2-translated 4.1Risoforms have a variable subcellular distribution (Gascard etal., 1998; Luque and Correas, 2000); and ATG3-translated4.1R isoforms are predominantly located in the nucleus (Luqueand Correas, 2000). Representative images are shown in Fig.1B. More than 95% of the cells ectopically expressing theATG1-translated isoform 4.1R135∆4,5,16 predominantlyshowed organized microtubules radiating from the centrosometo the periphery (Fig. 1B, top, arrows). Ectopic expression ofthe ATG1-translated isoforms 4.1R135∆16 and 4.1R135∆16,19

was also tested, the results being similar to those for4.1R135∆4,5,16 (not shown). By contrast, disorganized,unfocused microtubules that no longer radiated from thecentrosome were observed in most cells (∼70%) expressing theATG3-translated 4.1R isoform 4.1R60∆16,18 (Fig. 1B, middle,arrows). An examination of the consequences of ectopicexpression of the ATG2-translated 4.1R isoforms 4.1R80,4.1R80∆16 and 4.1R80∆5,16 yielded heterogeneous resultswhereby some isoforms affected microtubule organization

Fig. 1. Effect of exogenous 4.1R expression on interphase tubulinand actin cytoskeletons in COS-7 cells. (A) Schematic representationof the exon map for the 4.1R protein (top) and the cDNA constructsused in the transfection experiments shown in B,C (bottom). Exonsare coded as follows: striped, alternative; white, constitutive; black,noncoding. The number of each exon is shown at the bottom of themost upstream scheme. Three translation-initiation sites at exons 2′(ATG-1), 4 (ATG-2) and 8 (ATG-3) are indicated, as is the stopcodon (TGA) at exon 21. (B) Conventional fluorescence micrographsshowing COS-7 cells transfected with the indicated 4.1R-encodingcDNAs and subjected to double-immunofluorescence analysis withanti-tag 9E10 (left), and anti-tubulin YL1/2 (right) antibodies.(C) Cells were transfected with 4.1R60∆16,18-encoding cDNA andtriple stained with anti-tag 9E10 (top) and anti-tubulin YL1/2(middle) antibodies, and with Alexa-Fluor-594/phalloidin (bottom).Projections of complete x-y optical section stacks were acquired byconfocal microscopy. Arrows indicate transfected cells. Bar, 20 µm.

Page 4: Protein 4.1R regulates interphase microtubule organization ... · Microtubule organization is essential for directional intracellular transport, the modulation of cell morphology

6200

more than others (40%, 33% and 45%, respectively). Thiseffect correlated well with the subcellular localization of theexpressed ATG2-translated 4.1R isoform (Fig. 1B, bottom),which, as previously described, is variable (Gascard et al.,1998; Luque and Correas, 2000).

These results indicate that: (1) perturbation of themicrotubule cytoskeleton is not a general effect provoked bythe ectopic expression of 4.1R in COS-7 cells; and (2) thisperturbation seems to correlate with nuclear localization ofthe expressed 4.1R isoforms, suggesting that 4.1R wouldneed to be compartmentalized to induce this effect. To learnmore about the basis of the microtubule disorganizationobserved, we used the 4.1R60∆16,18 protein in this study,because this was the 4.1R isoform that had the strongesteffect.

Actin cytoskeleton is not affected by ectopic expressionof protein 4.1R60∆16,18We next investigated whether another component of thecytoskeleton, actin, was also perturbed by the ectopicexpression of 4.1R60∆16,18. Untransfected cells possessedfocused microtubules radiating from the centrosome and actinwas detected in stress fibers and membrane ruffles (Fig. 1C).In the same image, cells expressing 4.1R60∆16,18 showedunfocused microtubules (Fig. 1C, tubulin, arrows) but noevidence of perturbation in actin distribution was observed(Fig. 1C, actin, arrows). These results were observed inapproximately 98% of the transfected cells and indicate thatectopic expression of 4.1R60∆16,18 perturbs microtubule butnot actin organization.

γ-Tubulin and pericentrin distributions are not affected byectopic expression of protein 4.1R60∆16,18We next sought to establish whether the distribution of twomain centrosomal components involved in microtubulenucleation, γ-tubulin and pericentrin, were altered in cells

presenting disorganized microtubules. In control cells,microtubules were well organized and γ-tubulin and pericentrinlocalized to a single focus or paired foci near the nucleus (Fig.2, asterisks). In the same image, cells expressing protein4.1R60∆16,18 (Fig. 2, left, arrows), had disorganizedmicrotubules (Fig. 2, middle, arrows), whereas the stainingpatterns for γ-tubulin and pericentrin (Fig. 2, right, arrows)were similar to those in control cells. These results wereobserved in ∼95% of the transfected cells.

Dynactin and dynein distributions are affected by ectopicexpression of protein 4.1R60∆16,18One possible explanation for the microtubule disorganizationobserved in cells ectopically expressing protein 4.1R60∆16,18might be that the localization of some essential component(s)for stabilizing microtubule organization at centrosomes couldbe affected. Protein p150Glued is one of the subunits of thedynactin complex that appears to perform a key microtubule-anchoring function (Quintyne et al., 1999). We thereforeinvestigated whether cells overexpressing protein4.1R60∆16,18 have a normal or altered p150Glued distribution.Untransfected cells presented a single focus or paired foci nearthe nucleus and cytoplasmic staining (Fig. 3A, p150Glued,asterisks). By contrast, cells expressing protein 4.1R60∆16,18showed either p150Glued distributed in several foci dispersedthroughout the cytoplasm and cytoplasmic staining (Fig. 3A,p150Glued, top, arrow) or no detectable p150Glued fluorescencestaining at the centrosome (Fig. 3A, middle, insets).

Dynein intermediate chain (IC) mediates the dynein-dynactin interaction through association with p150Glued and isrequired for interphase microtubule organization atcentrosomes (Vaughan and Vallee, 1995). We were thereforeinterested to establish whether cells ectopically expressingprotein 4.1R60∆16,18 have normal or altered dyneindistribution. Untransfected control cells presented a singlefocus or paired foci near the nucleus (Fig. 3A, dynein, bottom,asterisks) whereas cells expressing protein 4.1R60∆16,18

Journal of Cell Science 117 (25)

Fig. 2. Effect of exogenous 4.1Rexpression on the distribution of themicrotubule nucleating proteins γ-tubulin and pericentrin. Cells weretriple labeled to detect exogenous4.1R60∆16,18, tubulin and γ-tubulin(top) or exogenous 4.1R60∆16,18,tubulin and pericentrin (bottom).Arrows and asterisks indicatetransfected and untransfected cells,respectively. Images are projectionsof x-y optical section stacksacquired by confocal microscopy.Bar, 20 µm.

Page 5: Protein 4.1R regulates interphase microtubule organization ... · Microtubule organization is essential for directional intracellular transport, the modulation of cell morphology

6201Centrosomal 4.1R and microtubules

showed, as in the case of protein p150Glued, either several focidispersed throughout the cytoplasm (not shown) or nodetectable dynein fluorescence staining at the centrosome (Fig.

3A, bottom, insets). The effects observed for dynein andp150Glued distributions are representative of most transfectedcells (~70%).

Fig. 3. Effects of exogenous 4.1R expression on centrosomal proteins of the dynein-dynactin complex and on centrosomal protein 4.1R.(A) Projection of optical section stacks obtained by confocal microscopy of cells triple labeled to detect exogenous 4.1R60∆16,18 (blue),p150Glued or dynein (green) and α-tubulin or γ-tubulin (red). Arrows and asterisks indicate transfected and untransfected cells, respectively. Theinsets are enlargements showing the distribution of each protein at the centrosome in the transfected cells. Notice that the transfected cells havealtered distributions of p150Glued and dynein. (B) Detergent lysates of cells transfected with empty plasmid (Ct) or with 4.1R60∆16,18/GFP-encoding cDNA (Tr) were sedimented into 5-20% sucrose gradients as described (Echeverri et al., 1996). Fractions were analysed byimmunoblotting using antibodies to p150Glued (p150Glued), to dynein intermediate chain (dynein) and to the control proteins actin (actin) and α-tubulin (α-tubulin). Notice that, in fraction 1 isolated from transfected cells (F1, arrow), p150 and dynein intermediate chain proteins arepresent, whereas they are absent from fraction 1 in control cells. F1 from transfected cells (Tr) also contains endogenous 4.1R (Endogenous4.1R) and exogenous 4.1R (Exogenous 4.1R). The sucrose gradient, the fractions collected and the position of sedimentation of the standardproteins, bovine serum albumin (4.4S), catalase (11.3S) and thyroglobulin (19S) are all indicated at the top. (C) Effect on endogenouscentrosomal 4.1R. Cells were triple stained to detect endogenous and exogenous 4.1R (4.1R), tubulin (tubulin) and γ-tubulin (γ-tubulin). Theimages are projections of optical-section stacks acquired by confocal microscopy and insets are enlargements showing the distributions of eachprotein at the centrosome in a transfected cell. Notice that γ-tubulin is detected as a pair of foci, whereas centrosomal endogenous 4.1R isabsent. By contrast, untransfected cells contain both centrosomal 4.1R and γ-tubulin (asterisks). Bar, 20 µm.

Page 6: Protein 4.1R regulates interphase microtubule organization ... · Microtubule organization is essential for directional intracellular transport, the modulation of cell morphology

6202

Overexpression of 4.1R60∆16,18 had similar effects onmicrotubule organization to those caused by some dynactincomponents, so we set out to determine whether interphasecells had disrupted dynactin structure (Echeverri et al., 1996;Quintyne et al., 1999). To address this matter, we examinedwhether dynactin-dynein remained a single complex thatsedimented at ~20S, as described by others (Paschal et al.,1993). In agreement with previous results from control COS-7 cells (Echeverri et al., 1996), p150Glued and dynein migratedas an ~18-19S and an ~20S complex, respectively (Fig. 3B,Ct). In samples prepared from cells ectopically expressing

4.1R60∆16,18, we observed no majoralterations in dynactin or dyneinsedimentation behaviors. We did notice,however, that a small proportion of p150Glued

and dynein fractionated with sedimentationcoefficients higher than 19S or 20S,respectively, in fraction 1 (Fig. 3B, Tr). It isplausible that p150Glued and dynein detectedin fraction 1 represent that isolated from thesmall cell population ectopically expressing4.1R60∆16,18 (∼10% of total cells) and aretherefore representative of the alteredlocalization observed for these proteins inthe confocal images (Fig. 3A). Interestingly,fraction 1 also contained endogenous 4.1Rproteins of ∼135-140 kDa that were absentfrom fraction 1 isolated from control cells(Fig. 3B, bottom). This result indicates thatthe sedimentation behavior of endogenousprotein 4.1R, like p150Glued and dynein, wasalso affected by the ectopic expression of4.1R60∆16,18.

Distribution of endogenous centrosomal4.1R is also perturbed by ectopicexpression of protein 4.1R60∆16,18Protein 4.1R has been detected at thecentrosome in interphase cells (Krauss et al.,1997) and associated with the centrosomal

protein CPAP (Hung et al., 2000). It seemed plausible thatcentrosomal 4.1R distribution could be altered in cells showingdisorganized microtubules by ectopically expressing4.1R60∆16,18. To test this hypothesis, control cells and cellsoverexpressing protein 4.1R60∆16,18 were triple stained withantibodies GTU-88 (to identify γ-tubulin), YL1/2 (to detecttubulin) and 10b (to identify simultaneously the endogenouscentrosomal 4.1R and the exogenously expressed 4.1Risoform). Several confocal images were analysed and thesection in which the centrosome was identified with the anti-γ-tubulin antibodies was carefully compared with the staining

Journal of Cell Science 117 (25)

Fig. 4. Effects of exogenous 4.1R expression onmicrotubule nucleation and retention at thecentrosome. Cells were treated with nocodazoleto promote microtubule disassembly and thenwashed to allow microtubule regrowth. At theindicated times, untransfected (A) andtransfected (B) cells were fixed and triple labeledwith the following antibodies: YL1/2 to detectmicrotubules (tubulin); GTU-88 to detect γ-tubulin (γ-tubulin); and 10b to detect endogenousand exogenous 4.1R (4.1R). Notice that, in cellswith disorganized microtubules, γ-tubulinremains at the centrosome, whereas centrosomal4.1R does not. The images of untransfected (A)and transfected (B) cells were taken from thesame field. The images are projections of optical-section stacks acquired by confocal microscopyand insets are enlargements showing thedistribution of each protein at the centrosome.Bar, 20 µm.

Page 7: Protein 4.1R regulates interphase microtubule organization ... · Microtubule organization is essential for directional intracellular transport, the modulation of cell morphology

6203Centrosomal 4.1R and microtubules

obtained for 4.1R. Most untransfected control cells (>96%)showed a normal microtubule cytoskeleton as detected withanti-tubulin antibody, and a brightly stained centrosome asdetected with anti-γ-tubulin and anti-4.1R, 10b, antibodies(Fig. 3C, asterisks). Most cells ectopically expressing4.1R60∆16,18 (∼70%) contained disorganized interphasemicrotubules and a brightly stained centrosome as detectedwith anti-γ-tubulin antibody, but had no detectable (Fig. 3C,insets) or highly reduced (not shown) 4.1R fluorescencestaining at the centrosome. These results indicate thatexogenous 4.1R alters the microtubule architecture and alsoaffects endogenous centrosomal 4.1R distribution.

Effects of 4.1R60∆16,18 overexpression on microtubulenucleation and organization at centrosomesCentrosome functions such as microtubule nucleation and theorganization of a radial microtubule network were examinedand correlated with the distribution of centrosomal 4.1R inthese processes. Microtubules were depolymerized usingnocodazole and the subsequent pattern of microtubuleregrowth was determined (Fig. 4). After increasing intervals ofregrowth (0 minutes to 3 hours), cells were fixed and triplestained to detect tubulin, γ-tubulin and 4.1R, and confocalimages were analysed. Untransfected cells (Fig. 4A) initiallyhad depolymerized microtubules (Fig. 4A, tubulin, 0′). After 5minutes of regrowth, cells showed single microtubule asters(Fig. 4A, tubulin, 5′). By 30 minutes of regrowth, cells haddeveloped a robust, radial array (Fig. 4A, tubulin, 30′). Duringall these processes, endogenous centrosomal 4.1R colocalizedwith γ-tubulin (Fig. 4A, insets). The distribution of endogenousnuclear 4.1R was also detected. Transfected cells (Fig. 4B),like control cells, were able to depolymerize microtubules (Fig.4B, tubulin, 0′). At this point, endogenous centrosomal 4.1Rlocalized with γ-tubulin (Fig. 4B, 0′ and corresponding insets).

After 5 minutes of regrowth, transfected cells showed singlemicrotubule asters and endogenous centrosomal 4.1Rdistribution was unaltered (5′; compare 4.1R with γ-tubulin).By 30 minutes of regrowth, microtubules and endogenouscentrosomal 4.1R distributions differed notably from those ofcontrol cells. Thus, disorganized, unfocused microtubules weredetected (Fig. 4B, tubulin, 30′), accompanied by a lack ofendogenous centrosomal 4.1R fluorescence staining (Fig. 4B,4.1R, 30′) but with an unaltered γ-tubulin distribution (Fig. 4B,γ-tubulin, 30′). The distribution of exogenous nuclear4.1R60∆16,18 is also shown in Fig. 4B. Exogenous 4.1R didnot contribute to the observed centrosomal staining because theanti-tag antibody did not stain the centrosomes of transfectedcells (not shown; see also Fig. 1). The results of theseexperiments imply that transfected cells could depolymerizemicrotubules correctly and nucleate them properly from thecentrosome, but that the ability of the centrosome to retainnewly assembled microtubules was disturbed. Interestingly, thecorrect microtubule depolymerization and nucleation from thecentrosome correlated with the presence of 4.1R at thecentrosome, whereas the inability of the centrosome to anchormicrotubules and maintain a radial microtubule organizationcorrelated with an altered distribution of centrosomal 4.1R.

Endogenous protein 4.1R is present in isolatedcentrosome preparations and localizes within the centerof microtubule asters assembled in vitroAlthough protein 4.1R was recently localized to thecentrosome by immunofluorescence and electron microscopyof mammalian tissue culture lines (Krauss et al., 1997), thepresence of 4.1R was not detected in either biochemicalexperiments or immunofluorescence assays in isolatedcentrosome preparations (Hung et al., 2000). To learn moreabout centrosomal 4.1R, we isolated centrosomes on sucrose

Fig. 5. Endogenous 4.1R is present in isolatedcentrosomes. (A) Centrosomes isolated from Molt-4 Tcells were subjected to double immunofluorescence withanti-4.1R (4.1R) and anti-γ-tubulin (γ-tubulin) or anti-α-tubulin (α-tubulin) antibodies. The samples were analysedby epifluorescence microscopy. (B) Western-blot analysesof isolated centrosomes were carried out with anti-4.1R(4.1R), anti-γ-tubulin (γ-tubulin) and anti-actin (actin)antibodies. (C) In-vitro-assembled microtubule asterswere triple stained with anti-α-tubulin (α-tubulin), anti-4.1R (4.1R) and anti-γ-tubulin (γ-tubulin) antibodies. Theimages are projections of optical-section stacks acquiredby confocal microscopy. Bar, 10 µm.

Page 8: Protein 4.1R regulates interphase microtubule organization ... · Microtubule organization is essential for directional intracellular transport, the modulation of cell morphology

6204

density gradients following the procedure described by Modjouand Bornens (Moudjou and Bornens, 1998). Fig. 5A showsrepresentative images of isolated centrosomes double stainedwith anti-4.1R and anti-γ-tubulin or anti-α-tubulin antibodies.As shown in Fig. 5A, protein 4.1R was present in the isolatedcentrosome fractions. Western-blot analysis of isolatedcentrosomes showed 4.1R major bands of ~135 kDa and ~80kDa and a minor ~60 kDa band (Fig. 5B, 4.1R). γ-Tubulin waspresent in the isolated centrosome preparations whereas actinwas absent (Fig. 5B, γ-tubulin and actin). It is not certainwhether the centrosomal 4.1R band of ∼135 kDa correspondsto that abnormally sedimenting with dynein and p150Glued infraction 1 (Fig. 3B) and that is therefore undetectable at thecentrosome of cells exogenously expressing 4.1R60∆16,18(Fig. 3C).

The isolated centrosomes were tested for their ability tonucleate and assemble microtubule asters in vitro and thelocation of 4.1R within the asters was determined by confocalmicroscopy. Most centrosomes (>80%) had the ability toassemble in vitro microtubule asters and protein 4.1R, like γ-tubulin, was localized at the central part of the microtubuleaster (Fig. 5C).

Effect of recombinant GST-4.1R on microtubule asterformationWe next analysed the effect that addition of GST-4.1R60∆16,18(GST-4.1R60) has on microtubule aster formation.Recombinant GST-4.1R80∆16 protein (GST-4.1R80) was alsoused in the assay because 4.1R80∆16, unlike 4.1R60∆16,18,distributed predominantly to the cytoplasm of cells (Luque andCorreas, 2000). The centrosomes were preincubated in theabsence (control sample, Ct) or the presence of GST, GST-4.1R80 or GST-4.1R60 and were then assayed for asterformation by the addition of tubulin. Addition of GST had noapparent effect on microtubule aster formation compared withcontrol samples. By contrast, addition of GST-4.1R60 increasedthe number of total microtubule asters assembled in vitro (Fig.6). The recombinant protein GST-4.1R60 was incorporated at

the center of the asters and along the microtubules (Fig. 6,small panels).

Protein 4.1R associates with γ-tubulin and α-tubulinThe results obtained above suggested that protein 4.1R mightfunction as a microtubule-anchoring factor, establishing abridge between microtubules and the centrosome. Weperformed pull-down assays to determine whether 4.1Rassociates with the centrosomal and microtubule componentsγ- and α-tubulin, respectively. Glutathione-Sepharose beadscoupled with GST-4.1R60∆16,18, GST-Cter or GST were usedon COS-7 cell extracts. As shown in Fig. 7, GST-4.1R60∆16,18,but not GST-Cter or GST control beads, copelleted γ- and α-tubulin from the cell extracts, indicating that 4.1R has theability to associate with γ- and α-tubulin through a region otherthan its C-terminus.

DiscussionMany studies have attempted to determine the mechanisms andproteins involved in focusing microtubule minus ends into thespindle poles. However, there is less information about themechanisms and proteins regulating microtubule organizationat the centrosome in interphase cells. The present report yieldsnew insights into the function of 4.1R in nonerythroid cellsby showing that 4.1R plays a key role at the centrosome,contributing to the maintenance of a radial microtubuleorganization.

Although protein 4.1R was recently localized at thecentrosome by immunofluorescence and electron microscopyof mammalian tissue culture lines (Krauss et al., 1997), thepresence of 4.1R was not detected in either biochemicalexperiments or immunofluorescence assays in isolatedcentrosome preparations (Hung et al., 2000). Our analysesusing isolated centrosomes showed that 4.1R was present at thecentrosome, that 4.1R localized at the central part ofmicrotubule asters assembled in vitro and that addition of GST-4.1R60∆16,18 (GST-4.1R60) enhanced the number of asters

Journal of Cell Science 117 (25)

Fig. 6. Effect of recombinant protein GST-4.1R60∆16,18 on microtubule aster formation. Theisolated centrosomes were preincubated with buffer(Ct), GST (GST), GST-4.1R80∆16 (GST-4.1R80) orGST-4.1R60∆16,18 (GST-4.1R60) for 45 minutes at4°C and then tubulin was added to start in-vitromicrotubule-aster formation. The microtubule astersstained with anti-α-tubulin antibody are shown(red). A representative aster (boxed field in theGST-4.1R60 panel) double stained with anti-α-tubulin (red) and anti-GST (green) antibodies isshown in the enlargements. The recombinant GST-4.1R60 protein is incorporated into the center of theasters and along the microtubules (merge). Fourexperiments were performed in duplicate. Theresults are shown in the histogram as the mean ± thestandard deviation of the number of microtubuleasters assembled in vitro. The images areprojections of optical-section stacks acquired byconfocal microscopy. Bar, 10 µm.

Page 9: Protein 4.1R regulates interphase microtubule organization ... · Microtubule organization is essential for directional intracellular transport, the modulation of cell morphology

6205Centrosomal 4.1R and microtubules

formed, facilitating aster assembly. This effect might be due tothe ability of 4.1R to associate with α- and γ-tubulin, assuggested by the pull-down experiments (Fig. 7) and by thedata on the incorporation of GST-4.1R60 in the astersassembled in vitro (Fig. 6). Previous studies from our groupshowed a direct interaction between protein 4.1R and tubulin(Correas and Avila, 1988). More recently, in vitro bindingassays revealed that 22 amino acids within a conserved regionof the 4.1R sequence were essential for tubulin binding (Perez-Ferreiro et al., 2001). In this study, the immunodetection ofGST-4.1R60 along the microtubules of the asters assembled invitro supports the direct association between 4.1R and α,β-tubulin. The incorporation of GST-4.1R60 into the center of theasters might simply be explained by such an interaction,although an interaction with other centrosomal componentcould also account for it. The results on the distribution and onthe effect of GST-4.1R60 in the asters assembled in vitro andthose on the pull-down experiments suggested us thatcentrosomal 4.1R might be acting as a microtubule-anchoringprotein.

Proteins with key microtubule-anchoring functions arereported to provoke disorganization of the interphasemicrotubule array when ectopically expressed in COS-7 cells(Dammermann and Merdes, 2002; Quintyne et al., 1999). Thesignificance of dynein and dynactin for microtubule anchoringand their participation in the regulation and turnover of themicrotubule network are clearly important (Bornens, 2002;

Dammermann et al., 2003). Indeed, when components of thedynactin complex were overexpressed in COS-7 cells,interphase microtubules were not focused into radial arrays anddynactin subunits did not accumulate at microtubule minusends (Burkhardt et al., 1997; Clark and Meyer, 1999; Quintyneet al., 1999). Using the same type of experiments, our resultsshowed that specific 4.1R isoforms provoked similarperturbation of the interphase microtubule organization andthat, importantly, 4.1R was displaced from the centrosome.These results, in conjunction with the finding that endogenous4.1R proteins and a population of p150Glued and dynein proteinsfractionated on sucrose density gradients with alteredsedimentation coefficients (>20S), suggested that microtubuleorganization is profoundly influenced by the activity andsubcellular localization of protein 4.1R.

Ninein, centrin and PCM1 are microtubule-anchoringfactors that require dynein/dynactin motor function for theirrecruitment to the centrosome (Dammermann and Merdes,2002). Depletion of any of these proteins provoked loss ofcentrosomal microtubule organization (Dammermann andMerdes, 2002). As for PCM1 depletion, the absence ofcentrosomal 4.1R was accompanied by centrosomalmicrotubule disorganization, altered distributions of dyneinand p150Glued and unaltered distribution of γ-tubulin. Bycontrast, unaltered distribution of centrosomal 4.1R correlatedwell with a correct microtubule depolymerization, nucleationand radial organization. These results indicate that, duringinterphase, 4.1R is important for the anchoring of microtubulesat the centrosome and for the maintenance of radial arrays ofmicrotubules.

Most of the data presented in this work were produced usingisoform 4.1R60∆16,18. The 4.1R60∆16,18-encoding cDNA wasoriginally isolated from Molt-4 T cells (Luque and Correas,2000) and, interestingly, endogenous 4.1R proteins of ∼60 kDahave also been detected in many other cell types (Luque andCorreas, 2000; Anderson et al., 1988). Whether any of theseendogenous 4.1R proteins correspond to isoform 4.1R60∆16,18is not known.

The ATG3-translated isoform 4.1R60∆16,18 had thestrongest effect on microtubule organization and presented apredominant nuclear localization. By contrast, ATG1-translated 4.1R isoforms had no effect and presented non-nuclear distribution. ATG2-translated 4.1R isoforms provokedmicrotubule disorganization if they had a predominant nuclearexpression (Fig. 1B, bottom). From these results, it could besuggested that overexpression of 4.1R isoforms with nuclearlocalization could provoke the dramatic phenotype we showwith isoform 4.1R60∆16,18. The mechanism by whichexogenous expression of nuclear 4.1R might affect microtubuleorganization is not yet established. One possible explanationcould be that the overexpressed nuclear 4.1R isoform retainedcentrosomal 4.1R and/or a cytoplasmic component essentialfor maintaining a correct microtubule organization.

In summary, the functional analysis using isolatedcentrosomes allowed us to analyse the effect on microtubuleaster assembly of adding GST-4.1R, whereas the 4.1R-overexpression assays were a useful tool to examinecentrosomes lacking endogenous 4.1R. A plausibleinterpretation of our results is that centrosomal protein 4.1Rcould be an anchoring protein that establishes a bridge betweenthe microtubules and the centrosome, hence participating in the

Fig. 7. Protein 4.1R associates with γ- and α-tubulin. (A) Coomassie-stained gels showing the purified (P) GST fusion proteins used in thebinding assays and the eluates (E) from glutathione beads coupledwith the indicated GST fusion proteins and incubated with COS-7cell extracts. (B) Immunoblots of the eluates revealed with antibodiesagainst γ- and α-tubulin. Positions of molecular-weight markers areindicated on the left.

Page 10: Protein 4.1R regulates interphase microtubule organization ... · Microtubule organization is essential for directional intracellular transport, the modulation of cell morphology

6206

dynamic interrelationships between the centrosome and thecytoarchitecture.

We thank J. E. Domínguez for invaluable discussions, F. Wandoselland M. A. Alonso for general comments, and A. Gosálbez fortechnical assistance. This work was supported by grant numberBMC2002-00978 from the Ministerio de Ciencia y Tecnología, Spain.CMP-F was a postdoctoral fellow from the Ministerio de Ciencia yTecnología, Spain.

ReferencesAnderson, R. A., Correas, I., Mazzucco, C., Castle, J. D. and Marchesi, V.

T. (1988). Tissue-specific analogues of erythrocyte protein 4.1 retainfunctional domains. J. Cell. Biochem. 37, 269-284.

Baklouti, F., Huang, S. C., Vulliamy, T. J., Delaunay, J. and Benz, E. J., Jr(1997). Organization of the human protein 4.1 genomic locus: new insightsinto the tissue-specific alternative splicing of the pre-mRNA. Genomics 39,289-302.

Bornens, M. (2002). Centrosome composition and microtubule anchoringmechanisms. Curr. Opin. Cell Biol. 14, 25-34.

Burkhardt, J. K., Echeverri, C. J., Nilsson, T. and Vallee, R. B. (1997).Overexpression of the dynamitin (p50) subunit of the dynactin complexdisrupts dynein-dependent maintenance of membrane organelle distribution.J. Cell Biol. 139, 469-484.

Clark, I. B. and Meyer, D. I. (1999). Overexpression of normal and mutantArp1alpha (centractin) differentially affects microtubule organization duringmitosis and interphase. J. Cell Sci. 112, 3507-3518.

Cohen, A. R., Woods, D. F., Marfatia, S. M., Walther, Z., Chishti, A. H.,Anderson, J. M. and Wood, D. F. (1998). Human CASK/LIN-2 bindssyndecan-2 and protein 4.1 and localizes to the basolateral membrane ofepithelial cells. J. Cell Biol. 142, 129-138.

Conboy, J. (1993). Structure, function, and molecular genetics of erythroidmembrane skeletal protein 4.1 in normal and abnormal red blood cells.Semin. Hematol. 30, 58-73.

Conboy, J. (1999). The role of alternative pre-mRNA splicing in regulatingthe structure and function of skeletal protein 4.1. Proc. Soc. Exp. Biol. Med.220, 73-78.

Conboy, J. G., Chan, J. Y., Chasis, J. A., Kan, Y. W. and Mohandas, N.(1991). Tissue- and development-specific alternative RNA splicing regulatesexpression of multiple isoforms of erythroid membrane protein 4.1. J. Biol.Chem. 266, 8273-8280.

Correas, I. and Avila, J. (1988). Erythrocyte protein 4.1 associates withtubulin. Biochem. J. 255, 217-221.

Correas, I., Speicher, D. W. and Marchesi, V. T. (1986). Structure of thespectrin-actin binding site of erythrocyte protein 4.1. J. Biol. Chem. 261,13362-13366.

Chasis, J. A., Coulombel, L., Conboy, J., McGee, S., Andrews, K., Kan, Y.W. and Mohandas, N. (1993). Differentiation-associated switches inprotein 4.1 expression. Synthesis of multiple structural isoforms duringnormal human erythropoiesis. J. Clin. Invest. 91, 329-338.

Dammermann, A. and Merdes, A. (2002). Assembly of centrosomal proteinsand microtubule organization depends on PCM-1. J. Cell Biol. 159, 255-266.

Dammermann, A., Desai, A. and Oegema, K. (2003). The minus end insight. Curr. Biol. 13, R614-R624.

De Cárcer, G., Lallena, M. J. and Correas, I. (1995). Protein 4.1 is acomponent of the nuclear matrix of mammalian cells. Biochem. J. 312, 871-877.

Doxsey, S. (2001). Re-evaluating centrosome function. Nat. Rev. Mol. CellBiol. 2, 688-698.

Echeverri, C. J., Paschal, B. M., Vaughan, K. T. and Vallee, R. B. (1996).Molecular characterization of the 50-kD subunit of dynactin reveals functionfor the complex in chromosome alignment and spindle organization duringmitosis. J. Cell Biol. 132, 617-633.

Gascard, P., Lee, G., Coulombel, L., Auffray, I., Lum, M., Parra, M.,Conboy, J. G., Mohandas, N. and Chasis, J. A. (1998). Characterizationof multiple isoforms of protein 4.1R expressed during erythroid terminaldifferentiation. Blood 92, 4404-4414.

Hou, V. C. and Conboy, J. G. (2001). Regulation of alternative pre-mRNAsplicing during erythroid differentiation. Curr. Opin. Hematol. 8, 74-79.

Hou, C. L., Tang, C., Roffler, S. R. and Tang, T. K. (2000). Protein 4.1R

binding to eIF3-p44 suggests an interaction between the cytoskeletalnetwork and the translation apparatus. Blood 96, 747-753.

Hung, L. Y., Tang, C. J. and Tang, T. K. (2000). Protein 4.1 R-135 interactswith a novel centrosomal protein (CPAP) which is associated with the γ-tubulin complex. Mol. Cell. Biol. 20, 7813-7825.

Kontrogianni-Konstantopoulos, A., Huang, S. C. and Benz, E. J., Jr(2000). A nonerythroid isoform of protein 4.1R interacts with componentsof the contractile apparatus in skeletal myofibers. Mol. Biol. Cell 11, 3805-3817.

Kontrogianni-Konstantopoulos, A., Frye, C. S., Benz, E. J., Jr and Huang,S. C. (2001). The prototypical 4.1R-10-kDa domain and the 4.1g-10-kDaparalog mediate fodrin-actin complex formation. J. Biol. Chem. 276, 20679-20687.

Krauss, S. W., Chasis, J. A., Rogers, C., Mohandas, N., Krockmalnic, G.and Penman, S. (1997). Structural protein 4.1 is located in mammaliancentrosomes. Proc. Natl. Acad. Sci. USA 94, 7297-7302.

Krauss, S. W., Heald, R., Lee, G., Nunomura, W., Gimm, J. A., Mohandas,N. and Chasis, J. A. (2002). Two distinct domains of protein 4.1 criticalfor assembly of functional nuclei in vitro. J. Biol. Chem. 277, 44339-44346.

Lallena, M. J. and Correas, I. (1997). Transcription-dependent redistributionof nuclear protein 4.1 to SC35-enriched nuclear domains. J. Cell Sci. 110,239-247.

Lallena, M. J., Martínez, C., Valcárcel, J. and Correas, I. (1998).Functional association of nuclear protein 4.1 with pre-mRNA splicingfactors. J. Cell Sci. 111, 1963-1971.

Luque, C. M. and Correas, I. (2000). A constitutive region is responsible fornuclear targeting of 4.1R: modulation by alternative sequences results indifferential intracellular localization. J. Cell Sci. 113, 2485-2495.

Luque, C. M., Lallena, M. J., Pérez-Ferreiro, C. M., de Isidro, Y., deCárcer, G., Alonso, M. A. and Correas, I. (1999). The N-terminal 209-aadomain of high molecular-weight 4.1R isoforms abrogates 4.1R targeting tothe nucleus. Proc. Natl. Acad. Sci. USA 96, 14925-14930.

Luque, C. M., Perez-Ferreiro, C. M., Perez-Gonzalez, A., Englmeier, L.,Koffa, M. D. and Correas, I. (2003). An alternative domain containing aleucine-rich sequence regulates nuclear cytoplasmic localization of protein4.1R. J. Biol. Chem. 278, 2686-2691.

Mattagajasingh, S. N., Huang, S. C., Hartenstein, J. S., Snyder, M.,Marchesi, V. T. and Benz, E. J. (1999). A nonerythroid isoform of protein4.1R interacts with the nuclear mitotic apparatus (NuMA) protein. J. CellBiol. 145, 29-43.

Mattagajasingh, S. N., Huang, S. C., Hartenstein, J. S. and Benz, E. J., Jr(2000). Characterization of the interaction between protein 4.1R and ZO-2.A possible link between the tight junction and the actin cytoskeleton. J. Biol.Chem. 275, 30573-30585.

Mitchison, T. and Kirschner, M. (1984). Microtubule assembly nucleated byisolated centrosomes. Nature 312, 232-237.

Moudjou, M. and Bornens, M. (1998). Method of centrosome isolation fromcultured animal cells. In Cell Biology: A Laboratory Handbook (ed. J. E.Celis), pp. 111-119. London, UK: Academic Press.

Nunomura, W., Takakuwa, Y., Tokimitsu, R., Krauss, S. W., Kawashima,M. and Mohandas, N. (1997). Regulation of CD44-protein 4.1 interactionby Ca2+ and calmodulin. Implications for modulation of CD44-ankyrininteraction. J. Biol. Chem. 272, 30322-30328.

Paschal, B. M., Holzbaur, E. L., Pfister, K. K., Clark, S., Meyer, D. I. andVallee, R. B. (1993). Characterization of a 50-kDa polypeptide incytoplasmic dynein preparations reveals a complex with p150Glued and anovel actin. J. Biol. Chem. 268, 15318-15323.

Perez-Ferreiro, C. M., Luque, C. M. and Correas, I. (2001). 4.1R proteinsassociate with interphase microtubules in human T cells: a 4.1R constitutiveregion is involved in tubulin binding. J. Biol. Chem. 276, 44785-44791.

Quintyne, N. J., Gill, S. R., Eckley, D. M., Crego, C. L., Compton, D. A.and Schroer, T. A. (1999). Dynactin is required for microtubule anchoringat centrosomes. J. Cell Biol. 147, 321-334.

Schischmanoff, P. O., Yaswen, P., Parra, M. K., Lee, G., Chasis, J. A.,Mohandas, N. and Conboy, J. G. (1997). Cell shape-dependent regulationof protein 4.1 alternative pre-mRNA splicing in mammary epithelial cells.J. Biol. Chem. 272, 10254-10259.

Tang, T. K., Qin, Z., Leto, T., Marchesi, V. T. and Benz, E. J., Jr (1990).Heterogeneity of mRNA and protein products arising from the protein 4.1gene in erythroid and nonerythroid tissues. J. Cell Biol. 110, 617-624.

Vaughan, K. T. and Vallee, R. B. (1995). Cytoplasmic dynein binds dynactinthrough a direct interaction between the intermediate chains and p150Glued.J. Cell Biol. 131, 1507-1516.

Journal of Cell Science 117 (25)


Recommended