5
CYTOSKELETON Vasohibins encode tubulin detyrosinating activity Joppe Nieuwenhuis, 1 Athanassios Adamopoulos, 1 Onno B. Bleijerveld, 1 Abdelghani Mazouzi, 1 Elmer Stickel, 1 Patrick Celie, 1 Maarten Altelaar, 1,2 Puck Knipscheer, 3,4 Anastassis Perrakis, 1 Vincent A. Blomen, 1 * Thijn R. Brummelkamp 1,4,5 * Tubulin is subjected to a number of posttranslational modifications to generate heterogeneous microtubules. The modifications include removal and ligation of the C-terminal tyrosine of -tubulin. The enzymes responsible for detyrosination, an activity first observed 40 years ago, have remained elusive. We applied a genetic screen in haploid human cells to find regulators of tubulin detyrosination. We identified SVBP, a peptide that regulates the abundance of vasohibins (VASH1 and VASH2). Vasohibins, but not SVBP alone, increased detyrosination of -tubulin, and purified vasohibins removed the C-terminal tyrosine of -tubulin. We found that vasohibins play a cell typedependent role in detyrosination, although cells also contain an additional detyrosinating activity. Thus, vasohibins, hitherto studied as secreted angiogenesis regulators, constitute a long-sought missing link in the tubulin tyrosination cycle. M icrotubules are crucial constituents of the eukaryotic cytoskeleton, a dynamic struc- ture important for cell shape and intracel- lular transport, composed of polymerized - and b-tubulin heterodimers. Exten- sive enzymatic alterations create heterogeneous microtubules decorated with a variety of post- translational modifications including acetylation, (poly)glutamylation, (poly)glycylation, and poly- amination (1). Most -tubulin isoforms encode a tyrosine at their C terminus that can be proteo- lytically removed and religated. The incorporation of tyrosine, the first described posttranslational modification of tubulin, is carried out by the tu- bulin tyrosine ligase (TTL), which reverses the detyrosinated -tubulin state to the translated form (2). However, the activity of the detyrosin- ating enzyme (3), which initiates the tyrosination cycle, remains unclear. Tubulin detyrosination has been implicated in cardiac cell function (4), cell migration (5), mitosis (6), and trafficking in neurons (7). We applied a genetic approach in haploid human cells (8, 9) to identify tubulin-detyrosinating enzymes. De- tyrosinated -tubulin could be detected in wild- type Hap1 cells, and this signal was increased in TTL-deficient Hap1 cells and in cells treated with the microtubule-stabilizing agent paclitaxel (Fig. 1A), indicating that the tyrosination cycle is active in Hap1 cells. Next, mutagenized Hap1 cells were stained with antibodies recognizing the detyrosinated form of -tubulin after paclitaxel treatment, and cells displaying the highest and lowest 1% of detyrosinated -tubulin levels were isolated by fluorescence-activated cell sorting (FACS). Gene-trap insertion sites were mapped to identify genes enriched for mutations in cells exhibiting either high or low levels of -tubulin detyrosination (Fig. 1B) (9). TTL was identified as the strongest negative regulator of -tubulin de- tyrosination (647 independent gene-trap inser- tion events mapped in the locus in the highcell population versus 11 mutations in the lowpopulation; Fig. 1C). In addition, we identified both subunits (KATNA1 and KATNB1) of the microtubule-severing protein complex katanin as negative regulators, and CAMSAP2 and MAP4 as positive regulators, in agreement with previ- ous studies (1012). Among the genes that were enriched by at least a factor of 4 for mutations in the lowchannel, SVBP (small vasohibin bind- ing protein) was identified as the most signifi- cant hit (P = 4 × 10 -10 ). Using an antibody from a different supplier to enrich for cells with high and low levels of tubulin detyrosination, muta- tions in SVBP were similarly enriched in the pop- ulation displaying lowdetyrosination (n = 62 independent mutations), whereas no mutations in this locus could be identified in the population displaying highdetyrosination levels (fig. S1A). Neither TTL nor SVBP scored as regulators in 10 unrelated genetic screens examining diverse protein phenotypes (fig. S1B), which suggests that SVBP has a specific function in -tubulin detyrosination. The interaction of SVBP, encoding a short peptide (66 amino acids), with vasohibins (13) further suggested a possible function of SVBP in tubulin detyrosination. Although vasohibins have a predicted transglutaminase-like protease fold (14), enzymatic activity has not been demonstra- ted and substrates have not been proposed. They are found in the cytosol but are considered to act in the extracellular milieu after secretion through a noncanonical pathway (13, 15, 16). Mammalian cells contain two vasohibin paralogs, VASH1 and VASH2 (fig. S1, C and D), that may act re- dundantly. To assess the function of SVBP and vasohibins in detyrosination, we expressed SVBP, VASH1, and VASH2 in HeLa cells, a cell line with minimal levels of detyrosinated -tubulin (17) (Fig. 1D). Whereas SVBP did not increase de- tyrosinated -tubulin, expression of VASH1 or VASH2 modestly increased detyrosinated - tubulin. Coexpression of SVBP with vasohibins increased the abundance (13) and solubility (fig. S2) of vasohibins and further increased detyrosin- ation of -tubulin. Thus, SVBP together with vasohibins can increase -tubulin detyrosination. To determine whether endogenous vasohibins affected detyrosination of -tubulin, we gener- ated single- and double-knockout cell lines (fig. S3). Loss of VASH1 or VASH2 led to a modest decrease in the amount of detyrosinated tubulin in Hap1 cells, and their combined loss led to a further decrease (but not absence) of detyrosin- ated tubulin (Fig. 2A). The presence of detyro- sinated tubulin in vasohibin-deficient cells could not be attributed to expression of TUBA4A (fig. S4), an isoform lacking the C-terminal tyrosine. To investigate the role of the vasohibins in other cell types, we generated both VASH1-deficient and VASH1/VASH2-deficient cell lines in human embryonic kidney (HEK) 293T cells (fig. S5) as well as in the melanoma-derived cell line CHL-1 (fig. S6). In HEK293T cells, a minimal decrease in detyrosinated -tubulin was observed in VASH1 mutant cells, but a substantial decrease was ob- served in double-knockout cells (Fig. 2B). In CHL-1 cells, however, the double-knockout cells displayed undetectable levels of detyrosinated -tubulin (Fig. 2B) and a modest increase in the amount of tyrosinated tubulin (fig. S7A). Thus, vasohibins are important for -tubulin detyrosination and cells can also contain vasohibin-independent de- tyrosinating activity. Having identified vasohibin-dependent and -independent activities, we next sought to deter- mine whether these could affect the polymer- ized microtubule population. Paclitaxel stabilizes microtubules and thereby depletes the amount of free /b-tubulin dimers that are the substrate for TTL (18). Treatment of Hap1, HEK293T, and CHL-1 cells with paclitaxel led to a robust in- crease in -tubulin detyrosination. In Hap1 and HEK293T cells deficient for VASH1 and VASH2, a similar response was observed; this was also the case when translation was inhibited by cy- cloheximide in HEK293T cells (Fig. 2C and fig. S8). These findings suggest that the vasohibin- independent activity affects paclitaxel-stabilized microtubules. Comparative immunoblot analysis showed that vasohibins mediate at least 97% of -tubulin detyrosination in CHL-1 cells (fig. S9). RESEARCH Nieuwenhuis et al., Science 358, 14531456 (2017) 15 December 2017 1 of 4 1 Division of Biochemistry, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands. 2 Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, 3584 CH Utrecht, Netherlands. 3 Hubrecht InstituteKNAW, University Medical Center Utrecht, 3584 CT Utrecht, Netherlands. 4 CGC.nl, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands. 5 CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria. *Corresponding author. Email: vincent.blomen@scenicbiotech. com (V.A.B.); [email protected] (T.R.B.) on June 25, 2020 http://science.sciencemag.org/ Downloaded from

CYTOSKELETON Vasohibins encode tubulin detyrosinating …CYTOSKELETON Vasohibins encode tubulin detyrosinating activity Joppe Nieuwenhuis, 1Athanassios Adamopoulos, Onno B. Bleijerveld,1

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: CYTOSKELETON Vasohibins encode tubulin detyrosinating …CYTOSKELETON Vasohibins encode tubulin detyrosinating activity Joppe Nieuwenhuis, 1Athanassios Adamopoulos, Onno B. Bleijerveld,1

CYTOSKELETON

Vasohibins encode tubulindetyrosinating activity

Joppe Nieuwenhuis,1 Athanassios Adamopoulos,1 Onno B. Bleijerveld,1

Abdelghani Mazouzi,1 Elmer Stickel,1 Patrick Celie,1 Maarten Altelaar,1,2

Puck Knipscheer,3,4 Anastassis Perrakis,1 Vincent A. Blomen,1* Thijn R. Brummelkamp1,4,5*

Tubulin is subjected to a number of posttranslational modifications to generateheterogeneous microtubules. The modifications include removal and ligationof the C-terminal tyrosine of ⍺-tubulin. The enzymes responsible for detyrosination,an activity first observed 40 years ago, have remained elusive. We applied agenetic screen in haploid human cells to find regulators of tubulin detyrosination.We identified SVBP, a peptide that regulates the abundance of vasohibins (VASH1 andVASH2). Vasohibins, but not SVBP alone, increased detyrosination of ⍺-tubulin, andpurified vasohibins removed the C-terminal tyrosine of ⍺-tubulin. We found thatvasohibins play a cell type–dependent role in detyrosination, although cells alsocontain an additional detyrosinating activity. Thus, vasohibins, hitherto studied assecreted angiogenesis regulators, constitute a long-sought missing link in the tubulintyrosination cycle.

Microtubules are crucial constituents of theeukaryotic cytoskeleton, a dynamic struc-ture important for cell shape and intracel-lular transport, composed of polymerized⍺- and b-tubulin heterodimers. Exten-

sive enzymatic alterations create heterogeneousmicrotubules decorated with a variety of post-translational modifications including acetylation,(poly)glutamylation, (poly)glycylation, and poly-amination (1). Most ⍺-tubulin isoforms encode atyrosine at their C terminus that can be proteo-lytically removed and religated. The incorporationof tyrosine, the first described posttranslationalmodification of tubulin, is carried out by the tu-bulin tyrosine ligase (TTL), which reverses thedetyrosinated ⍺-tubulin state to the translatedform (2). However, the activity of the detyrosin-ating enzyme (3), which initiates the tyrosinationcycle, remains unclear.Tubulin detyrosination has been implicated in

cardiac cell function (4), cell migration (5), mitosis(6), and trafficking in neurons (7). We applied agenetic approach in haploid human cells (8, 9)to identify tubulin-detyrosinating enzymes. De-tyrosinated ⍺-tubulin could be detected in wild-type Hap1 cells, and this signal was increasedin TTL-deficient Hap1 cells and in cells treatedwith the microtubule-stabilizing agent paclitaxel

(Fig. 1A), indicating that the tyrosination cycle isactive in Hap1 cells. Next, mutagenized Hap1 cellswere stained with antibodies recognizing thedetyrosinated form of ⍺-tubulin after paclitaxeltreatment, and cells displaying the highest andlowest 1% of detyrosinated ⍺-tubulin levels wereisolated by fluorescence-activated cell sorting(FACS). Gene-trap insertion sites were mappedto identify genes enriched for mutations in cellsexhibiting either high or low levels of ⍺-tubulindetyrosination (Fig. 1B) (9). TTLwas identified asthe strongest negative regulator of ⍺-tubulin de-tyrosination (647 independent gene-trap inser-tion events mapped in the locus in the “high”cell population versus 11 mutations in the “low”population; Fig. 1C). In addition, we identifiedboth subunits (KATNA1 and KATNB1) of themicrotubule-severing protein complex kataninas negative regulators, and CAMSAP2 andMAP4as positive regulators, in agreement with previ-ous studies (10–12). Among the genes that wereenriched by at least a factor of 4 for mutations inthe “low” channel, SVBP (small vasohibin bind-ing protein) was identified as the most signifi-cant hit (P = 4 × 10−10). Using an antibody froma different supplier to enrich for cells with highand low levels of tubulin detyrosination, muta-tions in SVBP were similarly enriched in the pop-ulation displaying “low” detyrosination (n = 62independent mutations), whereas no mutationsin this locus could be identified in the populationdisplaying “high” detyrosination levels (fig. S1A).Neither TTL nor SVBP scored as regulators in10 unrelated genetic screens examining diverseprotein phenotypes (fig. S1B), which suggeststhat SVBP has a specific function in ⍺-tubulindetyrosination.The interaction of SVBP, encoding a short

peptide (66 amino acids), with vasohibins (13)

further suggested a possible function of SVBP intubulin detyrosination. Although vasohibins havea predicted transglutaminase-like protease fold(14), enzymatic activity has not been demonstra-ted and substrates have not been proposed. Theyare found in the cytosol but are considered to actin the extracellular milieu after secretion througha noncanonical pathway (13, 15, 16). Mammaliancells contain two vasohibin paralogs, VASH1and VASH2 (fig. S1, C and D), that may act re-dundantly. To assess the function of SVBP andvasohibins in detyrosination, we expressed SVBP,VASH1, and VASH2 in HeLa cells, a cell line withminimal levels of detyrosinated ⍺-tubulin (17)(Fig. 1D). Whereas SVBP did not increase de-tyrosinated ⍺-tubulin, expression of VASH1 orVASH2 modestly increased detyrosinated ⍺-tubulin. Coexpression of SVBP with vasohibinsincreased the abundance (13) and solubility (fig.S2) of vasohibins and further increased detyrosin-ation of ⍺-tubulin. Thus, SVBP together withvasohibins can increase ⍺-tubulin detyrosination.To determine whether endogenous vasohibins

affected detyrosination of ⍺-tubulin, we gener-ated single- and double-knockout cell lines (fig.S3). Loss of VASH1 or VASH2 led to a modestdecrease in the amount of detyrosinated tubulinin Hap1 cells, and their combined loss led to afurther decrease (but not absence) of detyrosin-ated tubulin (Fig. 2A). The presence of detyro-sinated tubulin in vasohibin-deficient cells couldnot be attributed to expression of TUBA4A (fig.S4), an isoform lacking the C-terminal tyrosine.To investigate the role of the vasohibins in othercell types, we generated both VASH1-deficientand VASH1/VASH2-deficient cell lines in humanembryonic kidney (HEK) 293T cells (fig. S5) aswell as in the melanoma-derived cell line CHL-1(fig. S6). In HEK293T cells, a minimal decreasein detyrosinated ⍺-tubulin was observed in VASH1mutant cells, but a substantial decrease was ob-served in double-knockout cells (Fig. 2B). In CHL-1cells, however, the double-knockout cells displayedundetectable levels of detyrosinated ⍺-tubulin(Fig. 2B) and a modest increase in the amount oftyrosinated tubulin (fig. S7A). Thus, vasohibinsare important for ⍺-tubulin detyrosination andcells can also contain vasohibin-independent de-tyrosinating activity.Having identified vasohibin-dependent and

-independent activities, we next sought to deter-mine whether these could affect the polymer-izedmicrotubule population. Paclitaxel stabilizesmicrotubules and thereby depletes the amountof free ⍺/b-tubulin dimers that are the substratefor TTL (18). Treatment of Hap1, HEK293T, andCHL-1 cells with paclitaxel led to a robust in-crease in ⍺-tubulin detyrosination. In Hap1 andHEK293T cells deficient for VASH1 and VASH2,a similar response was observed; this was alsothe case when translation was inhibited by cy-cloheximide in HEK293T cells (Fig. 2C and fig.S8). These findings suggest that the vasohibin-independent activity affects paclitaxel-stabilizedmicrotubules. Comparative immunoblot analysisshowed that vasohibins mediate at least 97% of⍺-tubulin detyrosination in CHL-1 cells (fig. S9).

RESEARCH

Nieuwenhuis et al., Science 358, 1453–1456 (2017) 15 December 2017 1 of 4

1Division of Biochemistry, Netherlands Cancer Institute,Plesmanlaan 121, 1066 CX Amsterdam, Netherlands.2Biomolecular Mass Spectrometry and Proteomics,Utrecht Institute for Pharmaceutical Sciences, Universityof Utrecht, 3584 CH Utrecht, Netherlands. 3HubrechtInstitute–KNAW, University Medical Center Utrecht,3584 CT Utrecht, Netherlands. 4CGC.nl, Plesmanlaan 121,1066 CX Amsterdam, Netherlands. 5CeMM ResearchCenter for Molecular Medicine of the Austrian Academy ofSciences, 1090 Vienna, Austria.*Corresponding author. Email: [email protected] (V.A.B.); [email protected] (T.R.B.)

on June 25, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 2: CYTOSKELETON Vasohibins encode tubulin detyrosinating …CYTOSKELETON Vasohibins encode tubulin detyrosinating activity Joppe Nieuwenhuis, 1Athanassios Adamopoulos, Onno B. Bleijerveld,1

Nieuwenhuis et al., Science 358, 1453–1456 (2017) 15 December 2017 2 of 4

α-Tubulin

Y-Tubulin

deY-Tubulinoverexposed

TTL

WT ΔTTLpaclitaxel

Hap1

+ +

α-Tubulin

Flag

eIF4G

deY-Tubulin

SVBP-FlagVASH1-FlagVASH2-Flag

++

++

+

+ +pcDNA-3.1(-) +

paclitaxel Antibody staining

FACS

De-crosslink and identifymutations by deep sequencing

mutagenizedHap1 cells

Cel

l num

ber

high

deY-Tubulin

deY-Tubulin

HeLa

overexposed

1 2 3 4

deY-Tubulin AB3201

Log10 Total No. of Insertions

Log2

Mut

atio

n In

dex

−6

−4

−2

0

2

4

6

KATNA1KATNB1

TTL

SVBP

MAP4

CAMSAP2

low

Low High

* *α-TubulindeY-Tubulin 0.75 1.001.000.46

α-TubulindeY-Tubulin * 0.55 1.00 0.97

50

50

50

50

50

250

50

37

37

15

Fig. 1. Identification of genetic regulators of tubulin detyrosination inhaploid human cells. (A) Wild-type Hap1 cells and Hap1 cells deficient for TTLwere treated with paclitaxel and subjected to immunoblot analysis usingantibodies directed against detyrosinated (deY) and tyrosinated (Y) tubulin andTTL.Total amounts of tubulin were used as a loading control.The relative ratios ofdetyrosinated versus total ⍺-tubulin levels are indicated. Asterisk indicates thatthe detyrosinated tubulin signal is not quantifiable. (B) Schematic overview of the

haploid genetic screen usingantibodies to detect detyrosinated tubulin. (C) Resultof the genetic screen for regulators of ⍺-tubulin detyrosination.The relativemutation frequency in the “high” versus the “low” cell population (referred to asmutation index) was plotted against the total amount of insertions mapped pergene. Positive regulators are labeled in yellow, negative regulators inblue. (D)HeLacells transfected with vectors directing the expression of FLAG-tagged SVBP,VASH1, VASH2, or combinations thereof, were subjected to immunoblot analysis.

ΔVASH2WT ΔVASH1

++ + + + +

paclitaxel

Hap1

WT ΔVASH1-2+ +

α-Tubulin

eIF4G

deY-Tubulin

WT+ +

CHL-1

HEK293T

HEK293T

WT+ +

WT

ΔVASH1-2

DAPI deY-Tubulin α-Tubulin Merge

WT

untreated

CHL-1

Clone# 1 21 2ΔVASH1 ΔVASH1-2WT WT

1 2

ΔVASH1-2

ΔVASH1-2 ΔVASH1-2 paclitaxel

Clone# 1 1 2 2 3 3 1 1 2 2 3 3

1.00 0.45 0.86 0.26 0.51 0.28 0.32 0.11 0.44 0.10 0.17 *

α-TubulindeY-Tubulin 1.00 0.80 0.81 0.75 0.16 0.33

α-TubulindeY-Tubulin

α-TubulindeY-Tubulin 1.00 * *0.40 *1.00 0.89 * 0.14 1.00 0.11 0.63

deY-Tubulin

α-Tubulin

eIF4G

Flag

pMX-e

mpt

y

pMX-V

ASH1-Flag

Hap1

+ +1 1

0.12 1.00

50

50

50

50

50

50

50

50

50

50

250 eIF4G

deY-Tubulin

α-Tubulin

CHL-1

Clone# 1 21 2ΔVASH1 ΔVASH1-2WT WT

1 2

1.000.84 0.13 0.26 * *

50

50

250

250250250

250

Fig. 2. VASH1 and VASH2 control tubulin detyrosination and affect thedetyrosination status of polymerized microtubules. (A) IndependentHap1 cell lines deficient for VASH1 or VASH1 and VASH2 were generated andsubjected to immunoblot analysis. Relative ratios of detyrosinated versustotal ⍺-tubulin levels are indicated. (B) Independent HEK293T and CHL-1 celllines deficient for VASH1 and/or VASH2 were generated and subjectedto immunoblot analysis. (C) Wild-type cells and cell lines deficient for VASH1

and VASH2 were treated with paclitaxel and subjected to immunoblot assayas in (A). (D) Wild-type CHL-1 cells and CHL-1 cells deficient for VASH1and VASH2 were treated with paclitaxel and stained with antibodies to detectdetyrosinated ⍺-tubulin (green) and ⍺-tubulin (red). Blue indicates 4´,6-diamidino-2-phenylindole (DAPI) nuclear counterstain. Paclitaxel treatmentled to a factor of 1.89 increase in the detyrosination/total tubulin ratio of wild-type cells. Scale bars, 25 mm.

RESEARCH | REPORTon June 25, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 3: CYTOSKELETON Vasohibins encode tubulin detyrosinating …CYTOSKELETON Vasohibins encode tubulin detyrosinating activity Joppe Nieuwenhuis, 1Athanassios Adamopoulos, Onno B. Bleijerveld,1

Nieuwenhuis et al., Science 358, 1453–1456 (2017) 15 December 2017 3 of 4

α-Tubulin

Flag

eIF4G

deY-Tubulin

SVBP-FlagVASH1-Flag

VASH2-Flag

pcDNA-3.1(-)

VASH1-C169A-Flag

VASH2-C93A-Flag

HeLa

+ ++

++

+ ++

+

KDYEEVGVDSVEGEGEEEGEE2+

DetyrosinatedKDYEEVGVDSVEGEGEEEGEEY2+

Tyrosinated

29 30 31 32 33 34Time (min)

4.01E8

0

VASH1

Abu

ndan

ce

2.27E6

033

Abu

ndan

ce

29 30 31 32 33 34Time (min)

6.56E8

0

VASH1 C169A 4.48E7

030

Lys-N digestion productTUBA1A/B

eGFP -VEGEGEEEGEEX S

F

Y

deY-Tubulin

GFP

eIF4G

deY-Tubulin

GFPYG

GY

eIF4G

++ +

++

+

++ +

++

+

# Assigned spectra C169A VASH1 (inactive)

WT VASH1 (active)

39

9

1

1

41

1

AALE KDYEEVGVDSVEGEGEEEGEEY

AALE KDYEEVGVDSVEGEGEEEGEE

AALE KDYEEVGVDSVEGEGEEEGE

++ +

++

+

A

HEK293T

VASH1-FlagVASH1-C169A-Flag

VASH2-FlagSVBP-Flag

0.751.00 * * *α-TubulindeY-Tubulin

50

50

250

37

15

time (min) 0 1 2 4 8

deY-Tubulin

α-Tubulin

deY-Tubulin

α-Tubulin

+VASH1:SVBP

HeLa Tubulin dimersSVBP

VASH1VASH1-C169A

++

+

+

+

Y-Tubulin

deY-Tubulin

α-Tubulin50

50

50

1.000.63 * * *α-TubulindeY-Tubulin

+

SVBP-HisVASH1-Tag

VASH1-C169A-Tag

+ + ++

+

VASH1

SVBP

+

10

40

30

20

reaction time (min)de

Y-Tu

bulin

(A

U)

slope: 0.26±0.04

P=0.013

slope: 0.10±0.02

0 2 4 6 80.0

0.5

1.0

1.5

2.0

2.5

coomassie

input tubulin:

+GTP

-GTP

+ GTPGTP-

Fig. 3. The catalytic activity of vasohibins specifically removesthe tyrosine residue of a-tubulin. (A) HeLa cells were transfectedwith the indicated plasmids and subjected to immunoblot analysis.(B) Coomassie staining of a gel loaded with the purified products ofSVBP,VASH1, SVBP-VASH1, and SVBP-VASH1-C169A expressed ininsect cells. (C) In vitro detyrosination assay, using recombinant SVBP,VASH1,VASH1-SVBP, and catalytic inactiveVASH1-SVBP,with purified HeLa ⍺/b-tubulinas substrate.Tubulin tyrosination and detyrosination levels were determinedusing immunoblot analysis. (D) Purified VASH1-SVBP was incubated with invitro generated microtubules from HeLa cells, and immunoblot signals werequantified to establish the detyrosination rate relative to nontreated⍺/b-tubulin (n = 3). (E) NanoLC-MS/MS analysis of HeLa tubulin incubatedwith catalytic active or inactive VASH1-SVBP complexes. Extracted ion

chromatograms of the detyrosinated and tyrosinated peptides are showntogether with the number of assigned spectra of the respective peptides.(F) HEK293Tcells were cotransfected with vectors encoding the expressionof enhanced GFP molecules with C-terminal extensions corresponding tothe C terminus TUBA1A/B with the indicated modifications as well as SVBPand VASH1, VASH2, or catalytically inactive VASH1. Transfected cells weresubjected to immunoblot analysis. Amino acid abbreviations: A, Ala; D, Asp;E, Glu; F, Phe; G, Gly; K, Lys; L, Leu; S, Ser; V, Val; Y, Tyr.

RESEARCH | REPORTon June 25, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 4: CYTOSKELETON Vasohibins encode tubulin detyrosinating …CYTOSKELETON Vasohibins encode tubulin detyrosinating activity Joppe Nieuwenhuis, 1Athanassios Adamopoulos, Onno B. Bleijerveld,1

Paclitaxel treatment also increased detyrosina-tion in these cells, which suggests that vasohibinsalso affect the detyrosination status of poly-merized microtubules. To address this further,we stained CHL-1 cells using antibodies directedagainst ⍺-tubulin and tyrosinated and detyrosi-nated tubulin (Fig. 2D and fig. S7B). The signalfor detyrosinated tubulin was absent in vasohibin-deficient cells, but it colocalized with micro-tubules in both interphase and mitotic wild-typecells (Fig. 2D and fig. S10). Thus, vasohibins af-fect the detyrosination state of polymerizedmicrotubules, although their activity appearednot to be absolutely needed for chromosomecongression (6).To directly test whether vasohibins act as

transglutaminase peptidases toward tyrosinated⍺-tubulin, we designed VASH1 and VASH2 mu-tants that affected their respective predicted cat-alytic sites, Cys169 and Cys93 (14). Coexpressionof VASH1-Cys169Ala and VASH2-Cys93Ala withSVBP showed that these cysteines were essentialfor vasohibin-dependent induction of detyrosin-ated ⍺-tubulin (Fig. 3A).To study whether vasohibins can produce

detyrosinated ⍺-tubulin in vitro, we coexpressedthe VASH1-SVBP complex in insect cells (Fig.3B) and purified a stable and soluble com-plex. Small-angle x-ray scattering (SAXS) cou-pled to size exclusion chromatography revealeda well-folded, structurally robust, elongated com-plex with 1:1 stoichiometry (fig. S11). PurifiedVASH1-SVBP reduced the tyrosinated form oftubulin while increasing the detyrosinated formas examined by specific antibodies, with anapparent Michaelis constant KM of ~700 nMon ⍺/b-tubulin (fig. S12). VASH1 alone wasexpressed in small amounts, again suggestingthat SVBP is needed for folding, and thus sol-ubility, of VASH1.Relative to nontreated ⍺/b-tubulin, the in vitro

detyrosination rate of VASH1-SVBP was higherusing guanosine triphosphate (GTP)–inducedpolymerized stabilized microtubules as a sub-strate by a factor of ~2.5 (P = 0.013) (Fig. 3D).Whereas immunoblot analysis suggested that de-tyrosinated ⍺-tubulin is generated by vasohibins,it is a possibility that other reaction productscould also be generated, including the degluta-minated D2- or D3-forms of ⍺-tubulin (19, 20).Nano–liquid chromatography combined with

tandem mass spectrometry (nanoLC-MS/MS)indicated thatVASH1-SVBPdetyrosinated⍺-tubulinwithout affecting the adjacent glutamic acidresidues (Fig. 3E). Thus, VASH1 acts as a pepti-dase to catalyze removal of the C-terminal tyro-sine of ⍺-tubulin.We next designed experiments to study the spe-

cificity of vasohibins. Tubulin isoforms encode dif-ferent C-terminal tails. These tails were attachedto theC terminus of green fluorescent protein (GFP)and coexpressed with VASH1, VASH1-Cys169Ala,and VASH2. All isoforms containing a tyrosine attheir C terminus could be detyrosinated; TUBA8A,which encodes for a C-terminal phenylalanine,could also be modified by vasohibins (fig. S13).To further determine the substrate specificity,we generated mutants in the TUBA1A/B mini-mal substrate. Only variants with a C-terminaltyrosine or phenylalanine were processed byvasohibins (Fig. 3F), suggesting a requirementfor an aromatic ring at the C-terminal position.Extension of the C terminus with a glycine pre-vented enzymatic conversion by vasohibins, sug-gesting that the terminal free carboxyl group isrequired and that vasohibins do not cleave inter-nally. These experiments start to provide a ratio-nale for the specific proteolysis of the C terminusof ⍺-tubulin.Previously, vasohibins have been studied as

secreted molecules affecting angiogenesis, al-though the mechanism of secretion remainsunclear (21–23). The enzymatic activity describedhere addresses long-standing questions aboutthe nature of molecules that are able to start thedetyrosination-tyrosination cycle. Additional stud-ies are required to address whether certain iso-forms or modified versions of vasohibins functionspecifically inside the cell to detyrosinate tubulin.Although detyrosination was envisioned as asimple reaction carried out by a carboxypepti-dase, the identification of VASH1, VASH2, theregulating peptide SVBP, and a yet-unidentifiedactivity extend our view on the complexity of thisprocess.

REFERENCES AND NOTES

1. S. Gadadhar, S. Bodakuntla, K. Natarajan, C. Janke, J. Cell Sci.130, 1347–1353 (2017).

2. H. Murofushi, J. Biochem. 87, 979–984 (1980).3. M. E. Hallak, J. A. Rodriguez, H. S. Barra, R. Caputto, FEBS Lett.

73, 147–150 (1977).4. P. Robison et al., Science 352, aaf0659 (2016).

5. J. R. Yoon et al., Breast Cancer Res. Treat. 129, 691–701(2011).

6. M. Barisic et al., Science 348, 799–803 (2015).7. C. P. Garnham, A. Roll-Mecak, Cytoskeleton 69, 442–463

(2012).8. J. E. Carette et al., Science 326, 1231–1235 (2009).9. M. Brockmann et al., Nature 546, 307–311 (2017).10. D. Zhang et al., Nat. Cell Biol. 13, 361–370 (2011).11. K. Jiang et al., Dev. Cell 28, 295–309 (2014).12. J. T. Fassett et al., Am. J. Physiol. Heart Circ. Physiol. 304,

H749–H758 (2013).13. Y. Suzuki et al., J. Cell Sci. 123, 3094–3101 (2010).14. L. Sanchez-Pulido, C. P. Ponting, Bioinformatics 32, 1441–1445

(2016).15. M. Saito, Y. Suzuki, S. Yano, T. Miyazaki, Y. Sato, J. Biochem.

160, 227–232 (2016).16. X. Xue et al., Oncogene 32, 1724–1734 (2013).

17. J. C. Bulinski, J. E. Richards, G. Piperno, J. Cell Biol. 106,1213–1220 (1988).

18. A. E. Prota et al., J. Cell Biol. 200, 259–270 (2013).

19. L. Paturle-Lafanechère et al., Biochemistry 30, 10523–10528(1991).

20. C. Aillaud et al., Mol. Biol. Cell 27, 640–653 (2016).

21. K. Yoshinaga et al., Cancer Sci. 102, 446–451 (2011).

22. H. Kimura et al., Blood 113, 4810–4818 (2009).

23. J. Kern, M. Steurer, G. Gastl, E. Gunsilius, G. Untergasser,BMC Cancer 9, 284 (2009).

ACKNOWLEDGMENTS

We thank R. Medema, J. van den Berg, H. Janssen, andmembers of the Brummelkamp laboratory for discussions;A. Fish for advice in performing biochemical assays;M. Stadnik-Spiewak for assisting in protein expressionexperiments; D. Peeper for CHL-1 cells; and T. Sixmafor carefully reading the manuscript. Supported byNWO Vici Grant 016.Vici.170.033, KWF grant NKI2015-7609,the Cancer Genomics Center (CGC.nl), and the AmmodoKNAW Award 2015 for Biomedical Sciences (T.R.B.);the Netherlands Organization for Scientific Research (NWO)as part of the National Roadmap Large-scale ResearchFacilities of the Netherlands, Proteins@Work project number184.032.201 (O.B.B. and M.A.); and VIDI grant 723.012.102(M.A.). The HAP1 cell line is available from T.R.B. undera material transfer agreement with the NetherlandsCancer Institute. Sequencing data have been depositedin the NCBI Sequence Read Archive (www.ncbi.nlm.nih.gov.sra)under accession number SRP119153. Processed screenresults are accessible in an interactive database(https://phenosaurus.nki.nl/).

SUPPLEMENTARY MATERIALS

www.sciencemag.org/content/358/6369/1453/suppl/DC1Materials and MethodsFigs. S1 to S13Table S1References (24–34)

3 August 2017; accepted 3 November 2017Published online 16 November 201710.1126/science.aao5676

Nieuwenhuis et al., Science 358, 1453–1456 (2017) 15 December 2017 4 of 4

RESEARCH | REPORTon June 25, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 5: CYTOSKELETON Vasohibins encode tubulin detyrosinating …CYTOSKELETON Vasohibins encode tubulin detyrosinating activity Joppe Nieuwenhuis, 1Athanassios Adamopoulos, Onno B. Bleijerveld,1

Vasohibins encode tubulin detyrosinating activity

Maarten Altelaar, Puck Knipscheer, Anastassis Perrakis, Vincent A. Blomen and Thijn R. BrummelkampJoppe Nieuwenhuis, Athanassios Adamopoulos, Onno B. Bleijerveld, Abdelghani Mazouzi, Elmer Stickel, Patrick Celie,

originally published online November 16, 2017DOI: 10.1126/science.aao5676 (6369), 1453-1456.358Science 

, this issue p. 1448, p. 1453; see also p. 1381Science-tubulin.α also showed that vasohibins can remove the C-terminal tyrosine of et al.strategy, Nieuwenhuis

development. The authors also developed an inhibitor targeting this family of enzymes. Using a completely different essential for their activity. Vasohibin/SVBP complexes were involved in neuron polarization and brain cortexidentified vasohibins as enzymes that perform the TCP function and found that their small interacting partner SBVP was

et al.detyrosination have remained elusive for 40 years (see the Perspective by Akhmanova and Maiato). Aillaud their multiple cellular functions and are altered in disease. Tubulin carboxypeptidases (TCPs) responsible for

-tubulin detyrosination/tyrosination cycle create landmarks on microtubules that are essential forαEnzymes of the Tubulin carboxypeptidase identity revealed

ARTICLE TOOLS http://science.sciencemag.org/content/358/6369/1453

MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2017/11/15/science.aao5676.DC1

CONTENTRELATED

http://science.sciencemag.org/content/sci/358/6369/1381.fullhttp://science.sciencemag.org/content/sci/358/6369/1448.full

REFERENCES

http://science.sciencemag.org/content/358/6369/1453#BIBLThis article cites 34 articles, 9 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

is a registered trademark of AAAS.ScienceScience, 1200 New York Avenue NW, Washington, DC 20005. The title (print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement ofScience

Science. No claim to original U.S. Government WorksCopyright © 2017 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of

on June 25, 2020

http://science.sciencemag.org/

Dow

nloaded from