SlideShare a Scribd company logo
1 of 28
Download to read offline
and Mansoor Husain
MomenMansoor Tanwir, Taha Tabish, Abdul
Talat Afroze, Ge Yang, Amir Khoshbin,
Vascular Smooth Muscle Cells
Mediates Cell Cycle Progression in
ATPase-4 (PMCA4)2+Membrane Ca
Calcium Efflux Activity of Plasma
Gene Regulation:
doi: 10.1074/jbc.M113.533638 originally published online January 21, 2014
2014, 289:7221-7231.J. Biol. Chem.
10.1074/jbc.M113.533638Access the most updated version of this article at doi:
.JBC Affinity SitesFind articles, minireviews, Reflections and Classics on similar topics on the
Alerts:
When a correction for this article is posted•
When this article is cited•
to choose from all of JBC's e-mail alertsClick here
Supplemental material:
http://www.jbc.org/content/suppl/2014/01/21/M113.533638.DC1.html
http://www.jbc.org/content/289/10/7221.full.html#ref-list-1
This article cites 48 references, 30 of which can be accessed free at
atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/DownloadedfromatTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
Calcium Efflux Activity of Plasma Membrane Ca2؉
ATPase-4
(PMCA4) Mediates Cell Cycle Progression in Vascular Smooth
Muscle Cells*□S
Received for publication,November 11, 2013, and in revised form, January 10, 2014 Published, JBC Papers in Press,January 21, 2014, DOI 10.1074/jbc.M113.533638
Talat Afroze‡
, Ge Yang§
, Amir Khoshbin‡
, Mansoor Tanwir‡
, Taha Tabish‡
, Abdul Momen‡
, and Mansoor Husain‡§¶ʈ1
From the ‡
Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario
M5G 2C4, Canada, §
Institute of Medical Sciences, University of Toronto, Toronto, Ontario M5S 1A8, Canada, and ¶
Heart and Stroke
Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario M5G 1L7, and ʈ
Department of Medicine,
University of Toronto, Toronto, Ontario M5G 2C4, Canada
Background: PMCA4 actions in vascular smooth muscle are not understood.
Results: Alternative splicing of PMCA4 changes during vessel injury. Cell cycle arrest and downstream effectors of PMCA4
deletion are rescued by PMCA4a, PMCA4b, and PMCA4b unable to bind PDZ- proteins but not by inactive PMCA4.
Conclusion: Ca2ϩ
efflux activity of PMCA4 regulates G1 progression.
Significance: PMCA4 Ca2ϩ
efflux regulates cell cycle.
We explored the role played by plasma membrane calcium
ATPase-4 (PMCA4) and its alternative splice variants in the cell
cycle of vascular smooth muscle cells (VSMC). A novel variant
(PMCA4e) was discovered. Quantitative real-time-PCR-quanti-
fied PMCA4 splice variant proportions differed in specific
organs. The PMCA4a:4b ratio in uninjured carotid arteries
(ϳ1:1) was significantly reduced by wire denudation injury (to
ϳ1:3) by modulation of alternative splicing, as confirmed by
novel antibodies against PMCA4a/e and PMCA4b. Laser cap-
ture microdissection localized this shift to the media and adven-
titia. Primary carotid VSMC from PMCA4 knock-out (P4KO)
mice showed impaired [3
H]thymidine incorporation and G1
phase arrest as compared with wild type (P4WT). Electropora-
tion of expression constructs encoding PMCA4a, PMCA4b, and
a PMCA4b mutant lacking PDZ binding rescued this phenotype
of P4KO cells, whereas a mutant with only 10% of normal Ca2؉
efflux activity could not. Microarray of early G1-synchronized
VSMC showed 39-fold higher Rgs16 (NFAT (nuclear factor of
activated T-cells) target; MAPK inhibitor) and 69-fold higher
Decorin (G1 arrest marker) expression in P4KO versus P4WT.
Validation by Western blot also revealed decreased levels of
Cyclin D1 and NFATc3 in P4KO. Microarrays of P4KO VSMC
rescued by PMCA4a or PMCA4b expression showed reversal of
perturbed Rgs16, Decorin, and NFATc3 expression levels. How-
ever, PMCA4a rescue caused a 44-fold reduction in AP-2␤, a
known anti-proliferative transcription factor, whereas
PMCA4b rescue resulted in a 50-fold reduction in p15 (Cyclin
D1/Cdk4 inhibitor). We conclude that Ca2؉
efflux activity of
PMCA4 underlies G1 progression in VSMC and that PMCA4a
and PMCA4b differentially regulate specific downstream
mediators.
Progression through the cell cycle involves Ca2ϩ
transport-
ers including Ca2ϩ
channels and Ca2ϩ
efflux pumps (for review,
see Ref. 1), and sarco-endoplasmic reticulum Ca2ϩ
ATPase-2
(SERCA2) splice variants are differentially expressed at various
stages of the cell cycle (for review, see Ref. 2). However, whether
and how plasma membrane Ca2ϩ
ATPases (PMCAs),2
a family
of low capacity, high affinity plasma membrane-associated
Ca2ϩ
efflux pumps encoded by four genes (PMCA1–4) (for
review, see Refs. 3 and 4) also participated in this process was
not known.
Alternative splicing at the amino (N) and carboxyl (C) termi-
nal sites of PMCA4 pre-mRNA can produce x or z splice vari-
ants at site N and a, b, or d splice variants at site C, each leading
to the expression of a different protein (4–6). PMCA4 is
expressed later in development than PMCA1 but is expressed
more or less ubiquitously in adult mammalian organs (4).
PMCA gene transcription is controlled by c-Myb, leading to
changes in the free intracellular Ca2ϩ
concentration ([Ca2ϩ
]i)
during the cell cycle progression of vascular smooth muscle
cells (VSMC) (7–9).
Our initial observations in VSMC showed that inhibiting
c-Myb expression or function prevented cell cycle progression
and lowered [Ca2ϩ
]i (10). We found these changes to be medi-
ated by increased PMCA1 and PMCA4 expression. Indeed,
* This work was supported by operating grants (to M. H.) from the Canadian
Institutes of Health Research (MOP-64352) and the Heart and Stroke Foun-
dation of Ontario (T-5254 and NA6713).
□S
This article contains supplemental Tables S1–S8 and Figs. S1–S9. The nucle-
otide sequence(s) reported in this paper has been submitted to the Gen-
BankTM/EBI Data Bank with accession number(s) JF683384, EU493095, and
EU493094
PMCA4 microarray data have been placed in the PubMed microarray database
GEO under accession number GSE38320.
1
A Clinician-Scientist of the Canadian Institutes of Health Research (CL-
42617), currently a Career Investigator of the Heart and Stroke Foundation
of Ontario (CI-5503). To whom correspondence should be addressed:
TMDT-3-909, 200 Elizabeth St., Toronto, ON, Canada, M5G-2C4. Tel.: 416-
581-7489; E-mail: mansoor.husain@uhn.ca.
2
The abbreviations used are: PMCA, plasma membrane Ca2ϩ
ATPases; qRT,
quantitative real-time; VSMC, vascular smooth muscle cells; EGFP,
enhanced GFP; IHC, immunohistochemistry; NFAT, nuclear factor of acti-
vated T-cells; CREB, cAMP-response element-binding protein; PI, pro-
pidium iodide; NS, not significant; Rgs16, regulator of G-protein signaling-
16; Dcn, Decorin.
THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 289, NO. 10, pp. 7221–7231, March 7, 2014
© 2014 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A.
MARCH 7, 2014•VOLUME 289•NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 7221
atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
transient overexpression of PMCA1a in VSMC elevated the
Ca2ϩ
efflux rate, decreased the [Ca2ϩ
]i, and reduced the rate of
cell proliferation by over 2.5-fold (10). Whereas PMCA4b over-
expression in mice increased blood pressure and myogenic tone
(11), mice with functional knock-out of PMCA4 showed
impaired phasic contractions and apoptosis in portal vein
smooth muscle in vitro provided the mice were also heterozy-
gous for the PMCA1 gene deletion (12). Together, these find-
ings suggested that PMCA4 plays a role in VSMC biology.
We now present evidence that the relative proportions of
PMCA4a and PMCA4b splice variants are regulated after wire
denudation injury of carotid arteries in vivo. Quiescent VSMC
in vivo have roughly equal proportions of PMCA4a and
PMCA4b splice variants until arterial injury leads to a signifi-
cant decrease in the ratio of PMCA4a to PMCA4b. Mice lacking
functional PMCA4 (P4KO) show reduced remodeling after
injury in vivo, although primary VSMC isolated from such mice
manifest reduced cell proliferation and a G1 phase arrest that
can be rescued by either the PMCA4a or PMCA4b splice vari-
ant alone. A PMCA4b mutant lacking the carboxyl-terminal
PDZ binding domain also rescued the G1 phase arrest of P4KO
VSMC, whereas a PMCA4b mutant with only 10% of the Ca2ϩ
efflux activity of the wild-type pump could not. Microarray data
show that functional PMCA4 ablation caused strong increases
in Decorin (a G1 arrest marker) and regulator of G-protein sig-
naling-16 (Rgs16; mitogen-activated protein kinase (MAPK)
inhibitor, nuclear factor of activated T-cells (NFAT) target)
expression, although rescue with either PMCA4a or PMCA4b
restored normal Decorin and Rgs16 expression levels. Having
said this, PMCA4a and PMCA4b rescues did differ in their
global outcomes, with PMCA4a causing decreased expression
of the transcription factor AP-2␤ (an inhibitor of c-Myc,
AP-2␣, and cell cycle), whereas PMCA4b caused decreased
expression of p15 (Cdkn2b (cyclin-dependent kinase inhibitor
2b)), an inhibitor of Cyclin D1/Cdk4. Together, these findings
lead us to conclude that the Ca2ϩ
efflux activity of PMCA4
mediates G1 phase cell cycle progression in VSMC and that
PMCA4a and PMCA4b appear to work through different effec-
tor proteins to do so.
MATERIALS AND METHODS
Mice and Cell Culture—C57BL6/J mice were purchased
(Charles River Laboratories, Wilmington, MA). Frozen
PMCA4 knock-out hearts (lacking exons 2 and 3) were a gift
from Ludwig Neyses (Manchester, UK) (13, 14). Because dele-
tion of exons 2–3 disrupts the reading frame of the resulting
PMCA4 locus, these mice do not produce any PMCA4 protein
(supplemental Fig. S2, panel B). PMCA4 functional knock-out
mice distinct from the Neyses laboratory mice and lacking
exon-11 (encoding codons 440–520, including aspartate 466,
which is the catalytic phosphorylation site; GenBankTM
acces-
sion number NM_001167949.2) were a kind gift from Gary
Shull (12). These exon-11-deficient mice express a mutant
PMCA4 mRNA as a consequence of exon-10 splicing to
exon-12 (12), which preserves the reading frame and enables
these mice to produce functionless mutant PMCA4a and
PMCA4b proteins (devoid of a catalytic site). These mice are
referred to as P4KO mice, with their wild-type littermates
denoted as P4WT. Protocols for mouse tissue harvest and
carotid artery wire denudation injury have been described (15)
and were approved by the Animal Care Committee of Univer-
sity Health Network. Primary VSMC cultures were maintained
in Dulbecco’s modified Eagle’s medium (DMEM) containing
10% fetal calf serum (Wisent; Montreal, Quebec, Canada), 50
ng/ml rat recombinant PDGF-BB (Sigma), and 1% penicillin-
streptomycin in humidified air containing 5% CO2 at 37 °C.
Cloning of PMCA4 cDNA Variants—PMCA4a, PMCA4b,
and PMCA4e cDNA were cloned from total RNA extracted
from mouse bladder, a mouse VSMC cell line (MOVAS; ATCC
number CRL-2797), and mouse brain of C57 BL6/J mice,
respectively (NucleoSpin RNA II kit; Takara-Clontech, Moun-
tain View, CA) and PCR-cloned using mouse PMCA4-specific
primers (see supplement Table S1) targeting either the 5Ј or 3Ј
half of PMCA4 mRNA into pGemT-Easy vector (Promega).
Clones containing the 5Ј and 3Ј halves of PMCA4 were digested
with BamHI, gel-eluted, and ligated to generate full-length
cDNA clones for each splice variant and sequenced. These
cDNA sequences have been deposited in GenBankTM
under
accession numbers JF683384 (PMCA4a), EU493095
(PMCA4b), and EU493094 (PMCA4e).
Antibody Generation and Validation—Peptides (supple-
mental Fig. IB) specific for PMCA4b and PMCA4a/e carboxyl
termini were designed (Aves Labs; Tigard, OR) and used to
inject chickens. Polyclonal chicken antibodies with expected
specificity for PMCA4 splice variants were collected and affin-
ity-purified on columns containing the immobilized peptide
antigen.
Western Blotting—Protein extracts made from primary
VSMC and various mouse organs including uninjured and
injured carotid arteries were resolved on mini-PAGE gels
(Novex; Invitrogen), blotted, and hybridized to various anti-
bodies and visualized either by HRP-conjugated secondary
antibodies (ECL kit; PerkinElmer Life Sciences) or by near
infrared dye conjugated secondary antibodies using the Odys-
sey laser scanner (LiCor, Lincoln, NE).
qRT-PCR—Total RNA was extracted from various organs of
C57BL6/J mice with the PicoPure RNA extraction kit (Invitro-
gen). After DNase treatment and the addition of 1.0 pg of EGFP
RNA to each sample (reverse transcription efficiency control),
cDNA was synthesized, and qRT-PCR was carried out with
primers specific for EGFP (cDNA synthesis efficiency), ␤2-mi-
croglobulin (housekeeping gene), PMCA4a, PMCA4b, and
PMCA4e splice variants (see supplemental Table S1) using
SYBR GREEN master mix in an ABI HT-7900 (Invitrogen)
thermal cycler. qRT-PCR primers for PMCA4 splice variants
were first used to gel elute amplified products from each primer
pair, clone them into pGemT-Easy vector, and sequence them
to confirm the identity of the amplicon for each splice variant.
Next, the PMCA4a, PMCA4b, and PMCA4e primer pairs were
tested under various amplification conditions to measure
amplification efficiency. Amplification conditions were chosen
where the individual Ct values for each splice variant differed by
Ͻ5% from the mean Ct value for all three variants. This ensured
that amplification efficiency would not vary significantly
among the PCR reactions for the three PMCA4 splice variants.
The amplification conditions employed consisted of a hot start
PMCA4 Efflux Activity Regulates Cell Cycle in VSMC
7222 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289•NUMBER 10•MARCH 7, 2014
atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
of 94 °C for 10 min followed by 50 cycles of 94 °C for 15 s (dena-
turation), 69 °C for 30 s (annealing), and 72 °C for 30 s (exten-
sion). Gel eluted amplicons for EGFP, microglobulin, and the
three PMCA4 splice variants were diluted to generate copy
number standards containing 1 ϫ 106
, 1 ϫ 105
, 1 ϫ 104
, 1 ϫ 103
,
100, and 10 copies/␮l. The absolute copy number for each par-
ticular mRNA was calculated via the absolute quantification
method (Protocol 4304965; Applied Biosystems) that employs
Ct values of copy number standards to calculate absolute copy
numbers of each template in each sample. The absolute copy
numbers of PMCA4 splice variants were then normalized to the
copies of EGFP (cDNA synthesis efficiency control) and micro-
globulin (housekeeping gene control) cDNA in each sample.
Finally, normalized copy numbers of each PMCA4 splice vari-
ant in each sample were summed, and copy numbers of each
splice variant were divided by this sum to obtain the relative
proportion of each splice variant in each sample.
Immunohistochemistry (IHC), Immunofluorescent Staining,
and Morphometry—Mouse carotid artery cross-sections (para-
formaldehyde-fixed, paraffin-embedded) were used for immu-
nostaining with various antibodies and fluorophore-conju-
gated secondary antibodies (Jackson ImmunoResearch and
Invitrogen) and visualized by confocal imaging on the Olympus
Fluoview microscope using Fluoview software (Advanced Opti-
cal Microscopy Facility, University Health Network). Regions
of interest were drawn on the images, and fluorescence inten-
sity was quantified in the regions of interest using Fluoview
software. Quantification was done on at least two different
regions of interests of both uninjured and injured carotid artery
sections embedded in one paraffin section (three uninjured and
three injured arteries per section), immunostained together on
one slide, and imaged via z-stacks using the same laser intensity
for all sections. For carotid artery morphometry, sections from
P4KO and P4WT mice (day 9 post injury) were immunostained
with anti-PMCA4a/e antibody, and the area of the entire media
and entire remodeled adventitia was measured at 40ϫ resolu-
tion on the Olympus Fluoview microscope using Fluoview
software.
Laser Capture Microdissection—Mouse carotid arteries
(uninjured and injured) or mouse bladder fresh-frozen in OCT
(Optical Cutting Temperature Compound, Sakura Finetek,
Torrance, CA) were sectioned (10 ␮m thick) and mounted on
laser capture microdissection microscope slides. Sections were
lightly stained with Histogene (Invitrogen), fixed with 70% eth-
anol, and taken through a dehydration series ending in xylene
and dried. An laser capture microdissection microscope and
laser set up (Advanced Optical Microscopy Facility, University
Health Network) was used to microdissect and capture medial
and adventitial layer regions of carotids (or bladder smooth
muscle layer) from the stained and fixed sections. RNA was
extracted with the PicoPure kit, and qRT-PCR was carried out
as described above.
Tritiated Thymidine Uptake Assays—Primary VSMC were
seeded into 4 replicate wells of 6-well plates, allowed to grow
overnight, and then serum-starved (0.1% serum; DMEM) for
24 h. Serum-synchronized cells were then stimulated with 10%
serum and 50 ng/ml PDGF-BB for 24 h. Cells were given triti-
ated thymidine for the last 1 h before harvest. Cells were lysed
and processed for liquid scintillation counting.
BrdU-PI Flow Cytometry—Primary VSMC were seeded into
T-75 flasks, allowed to grow overnight, and then serum-starved
(0.1% serum; DMEM) for 24 h. Synchronized cells were then
stimulated with 10% serum and 50 ng/ml PDGF-BB for 24 h.
Cells were labeled with BrdU for the last 1 h before harvest.
Cells were harvested, fixed in 70% ethanol, and processed for
anti-BrdU FITC-conjugated antibody staining as well as pro-
pidium iodide staining and used for flow cytometry.
Microarray Analysis—Primary VSMC from the carotid
arteries of P4KO and P4WT mice were grown in quadruplicate
T-75 flasks, serum-starved for 24 h, serum- and PDGF-BB-
stimulated for 24 h, Hoechst-stained for the last 1 h before
harvest, and then flow-sorted to obtain the G0/G1 subpopula-
tion from all 8 samples (n ϭ 4 for each genotype). Total RNA
was extracted from the 8 samples and used for microarray anal-
ysis. In a second microarray, cells were starved for 24 h and
serum- and PDGF-BB-stimulated for 24 h (G1/S stage), and
RNA was extracted from P4WT cells, P4KO cells, PMCA4b-
rescued P4KO cells, and vector-rescued P4KO cells. In a third
experiment, RNA was extracted from P4KO (n ϭ 4), and
P4KOϩPMCA4a-rescued (n ϭ 4) cells was used for microarray
analysis. RNA was submitted to The Center for Applied
Genomics (SickKids Hospital, Toronto, ON) for microarray
analyses. Table 1 and supplemental Tables S2–S8 show genes
increased or decreased in expression or which are known to be
implicated in G1 phase arrest or are part of the NFAT pathway
and were seen to be strongly modulated in P4KO cells as com-
pared with P4WT cells. The preprocessing includes three steps:
background correction (performed in GenomeStudio soft-
ware), quantile normalization (16), and log2 transformation of
normalized data. The last two steps were performed in lumi R
package (17). The Center for Applied Genomics used LIMMA
(linear models for microarray data) (18) to identify differentially
expressed gene signatures under different conditions. Briefly
speaking, it starts by fitting a linear model for each gene in the
data, then an empirical Bayes method is used to moderate the
standard errors for estimating the moderated t-statistics for
each gene, which shrinks the standard errors toward a common
value. This test is similar to an analysis of variance method for
each gene except that the residual standard deviations are mod-
erated across genes to ensure more stable inference for each
gene. The moderated standard deviations are a compromise
TABLE 1
Summary of microarray results
Cdkn2b, cyclin-dependent kinase inhibitor 2b.
Comparison
Gene modulated
(-fold change; p value)
Early G1 P4KO vs. P4WT Dcn (ϩ69.1; p ϭ 8.95E-09)
Rgs16 (ϩ39.4; p ϭ 6.38E-09)
E2f2 (Ϫ3.6; p ϭ 4.02E-05)
Late G1/S P4KO vs. P4WT Dcn (ϩ9.3; p ϭ 3.24E-20)
Rgs16 (ϩ15.1; p ϭ 2.00E-18)
PMCA4a-rescued vs. P4KO Tfap2b (Ϫ44.6; p ϭ 2.5E-10)
Dcn (Ϫ1.27; p ϭ NS)
Rgs16 (ϩ1.1; p ϭ NS)
PMCA4b-rescued vs. P4KO Cdkn2b (p15) (-50.4; p ϭ 8.51E-24)
Dcn (Ϫ2.4; p ϭ 1.59E-13)
Rgs16 (Ϫ18.5; p ϭ 3.07E-19)
PMCA4 Efflux Activity Regulates Cell Cycle in VSMC
MARCH 7, 2014•VOLUME 289•NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 7223
atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
between the individual gene-wise standard deviations and an
overall pooled standard deviation. We evaluated the false dis-
covery rate using the Benjamini multiple testing procedure
(19). Our PMCA4 microarray data have been placed in the
PubMed microarray database GEO under accession number
GSE38320.
P4KO Electroporated with PMCA4 cDNA—Electroporation
was carried out with the 4D-Nucleofector (Lonza; Mississauga,
ON), and transfection efficiencies of ϳ70% were regularly
achieved (supplemental Figs. S4 and S5). Transfected cells were
selected with the appropriate antibiotic (puromycin or G418),
and the success of the drug selection was validated by RT-PCR
(supplemental Fig. S6). P4KO cells were electroporated with
either wild-type mouse PMCA4a, wild-type mouse PMCA4b,
human PMCA4b mutant lacking a carboxyl-terminal PDZ
binding domain (PMCA4b-⌬PDZ), or human PMCA4b-Asp
mutant (catalytic site point mutant with 10% efflux activity
compared with wild-type) or with vector where all constructs
carried an antibiotic resistance cassette. Electroporated cells
were allowed to recover for 24 h and then were subjected to 14
days of antibiotic selection before being used for BrdU-PI flow
cytometry, microarray, or Western blots.
Statistics—qRT-PCR data, flow cytometry data, and IHC
staining quantification are expressed as the mean Ϯ S.E. and
represent results from at least two separate experiments (each
having n Ն 3 samples per group). Statistical analyses were per-
formed using Student’s t test. Statistical significance was
defined as p Յ 0.05.
RESULTS
Cloning Mouse PMCA4 Splice Variants—cDNA constructs
encoding PMCA4a, PMCA4b, and the novel splice variant
termed PMCA4e were cloned from total RNA extracted from
mouse bladder, the murine VSMC cell line MOVAS, and
mouse brain, respectively. Sequence analysis showed that the
PMCA4e cDNA clone contains a novel exon-20 sequence that
is 10 nucleotide longer than the evolutionarily conserved 181-
nucleotide sequence of other mammalian PMCA4a variants.
These additional 10 nucleotide result in a premature stop
codon being created in the mature, spliced PMCA4e mRNA.
The predicted amino acid sequences at the C terminus of the
three cloned splice variants are shown in supplemental Fig.
S1A. The sequence of the extra 10 nucleotides included in the
mouse PMCA4e splice variant novel exon-20 is conserved in
the genomes of man, chimpanzee, monkey, dog, cat, rabbit,
mouse, and rat (supplemental Fig. S1C), which suggests a
unique functional role for PMCA4e. PHI-BLAST analysis
showed that other members of the PMCA family (viz. PMCA1
and PMCA3) also express splice variants similar to the
PMCA4e variant we have cloned and share the GSE peptide
motif at their carboxyl tails (supplemental Fig. S1C).
PMCA4 Splice Variant-specific Antibodies—The PDZ bind-
ing domain lies within the last seven amino acids of the carboxyl
terminus of mouse PMCA4b (viz. PSLETPV), which is absent
from the mouse PMCA4a tail. Two peptides were designed
(Aves Labs); one coding for the PMCA4b carboxyl tail and the
other coding for an exon-20-specific epitope expected in both
PMCA4a and PMCA4e variants (supplemental Fig. S1B), which
corresponds to the carboxyl end of the calmodulin binding
motif.
Hearts from homozygous Neyses laboratory-generated
PMCA4 knock-out adult mice (PMCA4Ϫ/Ϫ
) and littermate
wild-type controls (PMCA4ϩ/ϩ
) as well as C57/BL6/J mice
were used in genomic PCR (14). This confirmed the genetic
deletion of PMCA4 (exons 2–3) in knock-out hearts, with the
816-bp PMCA4 (exon3–4)-specific band identified only in
hearts from PMCA4ϩ/ϩ
and C57BL6/J mice (supplemental Fig.
S2A). Next, Western blots revealed that the PMCA4b antibody
recognized a ϳ130-kDa protein from brain, stomach, and heart
of C57BL6/J mice but did not bind to protein extracts made
from PMCA4Ϫ/Ϫ
heart (supplemental Fig. S2B). Similarly, the
PMCA4a/e antibody detected a ϳ130-kDa protein in extracts
from C57BL6/J brain and stomach but not heart from C57BL6/J
mice, presumably because of very low or absent PMCA4a/e
expression in the normal mouse heart.
PMCA4 Splice Variants Show Differential mRNA and Protein
Expression Levels in Specific Mouse Organs—Having previously
demonstrated a functional consequence of PMCA1a and
PMCA4b overexpression (10, 11), we were interested in explor-
ing the regulation of alternative splicing at splice site C in the
PMCA4 pre-mRNA that generates PMCA4a, -4b, and -4e
splice variants. To separate the consequences of transcriptional
regulation (via the endogenous PMCA4 promoter) from those
of alternative splicing modulation, the relative proportions of
each PMCA4 splice variant were calculated from absolute copy
number data obtained by quantitative RT-PCR. We reasoned
that promoter up- or down-regulation would increase the abso-
lute copy numbers of all alternatively spliced transcripts (so
that analysis using absolute copy numbers would tend to reflect
the effects of promoter regulation), whereas changes in the rel-
ative proportions of each PMCA4 splice variant would accu-
rately reflect regulation of alternative splicing. Others engaged
in studies of alternative splicing have also used this strategy
(20).
In this manner we determined the relative proportions of
PMCA4 splice variants in various mouse organs (Fig. 1A). We
found that mouse organs could be grouped into three catego-
ries: organs predominantly expressing PMCA4e (brain), organs
predominantly expressing PMCA4a (bladder), and organs pre-
dominantly expressing PMCA4b (all other organs tested),
excluding liver, which showed negligible PMCA4 expression.
We then used our PMCA4b and PMCA4a/e antibodies to
probe PMCA4 splice variant proteins (Fig. 1B) and found that
the organs surveyed also displayed distinct PMCA4a/e and
PMCA4b expression profiles.
Regulation of PMCA4 Alternative Splicing in Proliferating
VSMC in Vivo—We next searched for a disease model in which
PMCA4 alternative splicing may be modulated. When we
examined mouse carotid arteries at day 9 post wire denudation
injury, we found that the roughly equal proportion of PMC4a
and PMCA4b splice variants observed in uninjured controls
was dramatically altered, with preferential expression of
PMCA4b at the expense of PMCA4a splice variant in injured
arteries (Fig. 2A). Western blot analysis showed that this shift in
alternative splicing was also manifest at the protein level (Fig.
2B). Laser capture microdissection coupled with qRT-PCR
PMCA4 Efflux Activity Regulates Cell Cycle in VSMC
7224 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289•NUMBER 10•MARCH 7, 2014
atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
showed that injured media and remodeled adventitia have a
higher proportion of the PMCA4b transcript as compared with
uninjured media, whereas a sample from bladder smooth mus-
cle expressed a higher proportion of PMCA4a variant (supple-
mental Fig. S3).
IHC confocal imaging showed the expression of a smooth
muscle marker (sm22␣) in uninjured and injured carotid artery
cross-sections, whereas trichrome staining showed increased
collagen deposition in remodeled adventitia (Fig. 3A). IHC
staining with a proliferation marker (proliferating cell nuclear
antigen) was only visible in injured arteries. Consistent with
qRT-PCR and Western blot, IHC showed a decrease in
PMCA4a/e and an increase in PMCA4b staining in injured
carotids as compared with uninjured arteries that was further
corroborated by quantifying immunofluorescent intensities
with PMCA4 splice variant-specific antibodies in injured and
uninjured medial layers (Fig. 3B).
PMCA4 functional knock-out mice kindly provided by Gary
Shull (Cincinnati, OH) lack exon-11, which encodes the central
catalytic loop (81 amino acids; residues 440–520; see “Materials
and Methods”) (12), resulting in a protein product devoid of any
Ca2ϩ
efflux activity (12). In the current manuscript, these mice
are referred to as P4KO mice, with their wild-type littermates
termed P4WT. Carotid artery cross-sections from P4WT and
P4KO mice at day 9 post-injury were used for morphometry
(Fig. 3, C and D). There was a 2-fold decrease in the adventitia:
media ratio of injured P4KO arteries compared with P4WT
arteries. This decreased remodeling in P4KO mice suggested
two interrelated possibilities: (i) absence of functional PMCA4
impairs VSMC proliferation in vivo, and (ii) increased propor-
tion of PMCA4b observed in injured arteries may play a physi-
ologically relevant role in VSMC proliferation.
PMCA4 Knock-out Causes G1 Phase Arrest, Which Can Only
Be Rescued by PMCA4 with Intact Ca2ϩ
Efflux Activity—To
probe the correlation between relative proportions of PMCA4
splice variants and VSMC proliferation, we isolated primary
carotid VSMC from P4WT and P4KO mice. [3
H]Thymidine
uptake assays using cell cycle-synchronized VSMC cultures
showed an ϳ70% reduction in G1 to S phase cell cycle progres-
sion of P4KO VSMC compared with P4WT cells (Fig. 4A). Sim-
ilarly, BrdU-PI flow cytometry showed an ϳ30% increase in
G1-stage P4KO cells as compared with P4WT (Fig. 4B), sug-
gesting a G1 stage arrest.
Importantly, electroporation of either mouse PMCA4a or
PMCA4b expression constructs rescued the cell cycle pheno-
type of P4KO cells (Fig. 4B), i.e. PMCA4a- or PMCA4b-rescued
P4KO VSMC manifest a 40–50% increase in S phase cells and
an ϳ50% reduction in G1 phase cells. Western blots confirmed
(supplemental Fig. S7) that as a result of these electroporations,
PMCA4a was overexpressed 1.5-fold in P4KOϩ4a cells,
whereas PMCA4b was overexpressed 1.8-fold in P4KOϩ4b
FIGURE 1. Distinct PMCA4 splice variant expression levels in mouse
organs. A, qRT-PCR-determined relative proportions of PMCA4 splice vari-
ants in various organs of C57BL6/J mice. Bars represent mean relative propor-
tionforeachsplicevariantmRNAextractedfromtheorgansofthreeC57BL6/J
mice, normalized for cDNA conversion efficiency (EGFP) and loading (␤2-mi-
croglobulin).LivergavenoPMCA4splicevariantcopiesuponamplification.B,
immunoblots of microsomal protein extracts from various organs of
C57BL6/J mice. Microsomal extracts were made from the organs of three
C57BL6/J mice and resulted in nine separate immunoblots (three each for
PMCA4-a/e, PMCA4b, and GAPDH); representative blots are shown. Br, brain;
U,uterus;bl,bladder;M,skeletalmuscle;K,kidney;S,stomach;H,heart;L,liver.
FIGURE 2. PMCA4 splice variant mRNA and protein expression levels are
regulated after arterial injury. A, qRT-PCR-determined relative proportions
of PMCA4 spice variants in uninjured and injured carotid arteries from male
C57BL6/J mice (n ϭ 4). p values for uninjured versus injured, PMCA4a ϭ 0.001
and PMCA4b ϭ 0.01, by Students’ t test. B, immunoblots of protein extracts
made from uninjured and injured mouse carotid arteries. Representative
blots from two independent experiments are shown.
PMCA4 Efflux Activity Regulates Cell Cycle in VSMC
MARCH 7, 2014•VOLUME 289•NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 7225
atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
cells as compared with P4KO cells. Although expression of a
PMCA4b mutant lacking the PDZ binding domain was still able
to rescue the G1 arrest of P4KO cells, expression of a PMCA4b-
Asp mutant (with only 10% of the Ca2ϩ
efflux activity of wild-
type PMCA4) failed to rescue the G1 phase arrest of P4KO cells
(Fig. 4B).
In a separate series of experiments assessing cell cycle pro-
gression at multiple time points after serum stimulation of
G0-synchronized P4WT, P4KO, and PMCA4a- or PMCA4b-
rescued cell populations, we confirmed that P4KO cells suffer
from a G1 phase arrest that is completely alleviated by either
PMCA4a or PMCA4b cDNA expression (Fig. 5). Together
these data show that whereas specific PDZ binding domain-
mediated PMCA4b/nNOS (21) or PMCA4b/CASK interac-
tions (22) are not necessary for PMCA4b to regulate cell cycle,
the presence of Ca2ϩ
efflux activity is required for PMCA4 to
mediate G1-to-S phase progression in VSMC.
Microarray Analysis and Validation of Identified Targets—
Early G1 stage P4KO and P4WT cells were collected via flow
sorting (by gating for G0/G1 phase Hoechst-stained cells) and
used for microarray analysis. This unbiased exploration
revealed that Decorin (ϩ69.1-fold; p ϭ 8.9E-09) and Rgs16
(ϩ39.4-fold; p ϭ 6.3E-09; Fig. 6A and supplemental Tables 2
and 3) were markedly increased in P4KO cells as compared with
P4WT cells. Decorin is a known inhibitor of the VSMC cell
cycle and implicated in G1 phase arrest (for review, see Ref. 23).
Decorin is known to reduce cyclin D1 expression via the epi-
dermal growth factor receptor (EGFR) or NFAT pathways (24–
26), with NFAT activation also known to inhibit Rgs16 expres-
sion (27) (see the proposed model, supplemental Fig. S8). This
FIGURE 3. Immunofluorescence-based quantification revealed regulated PMCA4 splicing in VSMC of injured carotid arteries. A, confocal images of
uninjured and injured carotid artery cross-sections from male C57BL6/J mice stained for PMCA4a/e, PMCA4b, smooth muscle marker (sm22␣), proliferation
marker (proliferating cell nuclear antigen), and collagen deposition (trichrome). Scale bars in panel A trichrome images ϭ 500 ␮m, in confocal images ϭ 20 ␮m,
and in panel D confocal images ϭ 50 ␮m. B, confocal microscopy Z-stack quantification of PMCA4a/e staining. Each bar represents a mean of 3 mouse carotid
artery cross-sections in which ϳ40–70 cells or cell clusters were used to quantify fluorescent intensity through 20–40 focal planes of 5-␮m-thick sections. C,
morphometric analysis of injured carotid arteries in PMCA4 wild-type (P4WT) and PMCA4 knock-out (P4KO) mice (n ϭ 4). Carotid artery cross-sections were
immunostained for PMCA4a/e with media and adventitia captured as regions of interest for area measurements on FluoView microscope (40ϫ objective) and
software (Olympus, Mountain View, CA). The ratio of adventitia:media areas are decreased in injured carotid arteries of P4KO mice (P4WT ϭ 3.1 Ϯ 0.5 versus
PMKO ϭ 1.4 Ϯ 0.1; p ϭ 0.01; n ϭ 4). D, representative immunofluorescent micrographs of P4WT and P4KO injured carotid arteries immunostained for
PMCA4a/e show reduced medial thickening in P4KO mice.
PMCA4 Efflux Activity Regulates Cell Cycle in VSMC
7226 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289•NUMBER 10•MARCH 7, 2014
atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
suggested that the high Rgs16 expression observed in P4KO
cells (as compared with P4WT cells) may be due to reduced
NFAT expression or activation in P4KO cells. Consistent with
this, immunoblot analysis showed that Decorin and Rgs16 lev-
els were increased, and Cyclin D1 levels were decreased in
P4KO cells as compared with P4WT cells (Fig. 7).
Similar results were observed in vivo. First, Decorin-staining
intensity trended 18% lower in medial layers of injured as com-
pared with uninjured carotid arteries of P4WT mice (0.82 Ϯ
0.05 versus 1.00 Ϯ 0.06; n ϭ 3, p ϭ 0.1), suggesting injury-
induced decreases in Decorin expression accompanied the
reduced PMCA4a:PMCA4b expression ratio. Second, this pat-
tern was lost in P4KO mice, in which carotid artery denudation
injury failed to reduce Decorin expression (1.10 Ϯ 0.03 versus
1.00 Ϯ 0.04; n ϭ 3, p ϭ NS), supporting a role for PMCA4 in
regulating Decorin expression in vivo. Indeed, similar to the
microarray-documented difference in Decorin expression
observed in primary VSMC cultures from P4KO versus P4WT
mice, levels of Decorin expression were significantly higher in
injured carotid arteries of P4KO animals as compared with
P4WT (1.10 Ϯ 0.03 versus 0.82 Ϯ 0.05; n ϭ 3, p ϭ 0.01) (data not
shown).
Because Decorin and Rgs16 have both been implicated in the
NFAT pathway, we next examined mRNA levels of NFAT facil-
itator proteins Rcan1, Rcan2, and the NFAT target E2F2 in our
microarray results (supplemental Table S4). Rcan2 was reduced
Ϫ6.6-fold (p ϭ 3.8E-08) and E2F2 was decreased Ϫ3.6-fold (p ϭ
4.0E-05) in P4KO as compared with P4WT cells, suggesting
diminished NFAT action. To examine this more directly, cyto-
plasmic protein extracts were probed for NFATc3 levels and
found to be significantly decreased in P4KO cells as compared
with P4WT cells (Fig. 7).
PMCA4a rescue of P4KO cells followed by late G1/S microar-
ray analysis (Fig. 6, B and C) suggested some reversal of the
expression levels of Decorin (PMCA4a rescue versus P4KO ϭ
Ϫ1.27-fold; p ϭ NS) and Rgs16 (PMCA4a rescue versus
P4KO ϭ ϩ1.1; p ϭ NS); however, these changes did not reach
statistical significance (supplemental Table S5). Rather,
PMCA4a-rescued P4KO cells showed a sharp decline in the
expression of a known anti-proliferative transcription factor
viz. AP-2␤ (PMCA4a rescue versus P4KO ϭ 44.6; p ϭ 2.5E-10)
(supplemental Table S5). AP-2␤ inhibits the activity of AP-2␣
(28) and also represses c-Myc transactivation (29). AP-2␣ is
also known to bind to the ErbB2 promoter (EGFR family mem-
ber) and promote proliferation via the EGFR pathway (30) acti-
vated by vessel injury in vivo (31). AP-2␤ is also implicated in
VSMC proliferation in vivo by its role in the closure of the
ductus arteriosus (32).
Additional microarray analysis using RNA harvested from
late G1/S-stage P4KO, P4WT, and PMCA4b-rescued- versus
vector-transfected-P4KO cells showed that Decorin was down-
regulated Ϫ2.4-fold (p ϭ 1.59E-13) in PMCA4b-rescued versus
FIGURE4.Ca2؉
effluxactivityofPMCA4isrequiredtorescuetheG1 arrest
of functional PMCA4 ablation. A, [3
H]thymidine uptake at the late G1/S-
stage was assayed in cell cycle-synchronized PMCA4 wild-type (P4WT) and
PMCA4knock-out(P4KO)primaryVSMC(nϭ3experiments).P4KOcellsshow
a 64% reduction in thymidine uptake as compared with P4WT cells (*, p ϭ
2.8E-08, Student’s t test). B, flow cytometric determination of cell cycle distri-
butions of P4WT and P4KO primary VSMC. P4KO cells were electroporated
with expression constructs encoding mouse PMCA4a, mouse PMCA4b,
human PMCA4b-Asp mutant (10% Ca2ϩ
efflux activity compared with wild-
type), or human PMCA4b-⌬PDZ mutant (lacking PDZ-binding domain) and
G0-synchronized by serum starvation. After synchronized serum stimulation
for 22 h, the cell cycle distribution of each population was determined by
BrdU-PI flow cytometry (n ϭ 3 independent experiments with multiple repli-
cates for all samples). *p ϭ 0.004 for P4WT versus P4KO; §, p ϭ 1.85E-6 for
PMCA4a-rescued P4KO versus P4KO; ϱ, p ϭ 0.001 for PMCA4b-rescued P4KO
versus P4KO; †, p ϭ 2.9E-16 for huP4b-Asp-rescued P4KO versus PMCA4b-
rescued P4KO; ‡, p ϭ 0.0001 for huP4b-⌬PDZ-rescued P4KO versus P4KO.
Student’s t test was used for all comparisons.
FIGURE 5. Correction of the impaired cell cycle progression of PMCA4
knock-out VSMC. Cell cycle distributions of P4WT, P4KO, or P4KO VSMC elec-
troporated with mouse PMCA4a or PMCA4b cDNA were determined by
BrdU-PI flow cytometry (n ϭ 2 independent experiments with multiple repli-
cates for all samples) at various time points in the cell cycle using serum-
synchronized populations.
PMCA4 Efflux Activity Regulates Cell Cycle in VSMC
MARCH 7, 2014•VOLUME 289•NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 7227
atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
P4KO cells (down Ϫ15.3-fold in PMCA4b-rescued versus con-
trol P4KO cells; p ϭ 5.5E-22), whereas it remained up-regulated
ϩ9.3-fold (p ϭ 3.24E-20) in P4KO versus P4WT G1/S stage
cells (Fig. 6D; and supplemental Tables S6–S8). Rgs16 was also
decreased Ϫ18.5-fold (p ϭ 3.07E-19) in PMCA4b-rescued ver-
sus P4KO cells (Ϫ3.9-fold in PMCA4b-rescued versus vector-
control P4KO cells; p ϭ 9.2E-14), whereas it continued to be
increased ϩ15.1-fold (p ϭ 2.0E-18) in P4KO versus P4WT at
the G1/S stage (Fig. 6D; and supplemental Tables S6–S8). Inter-
estingly, Cdkn2b (cyclin-dependent kinase inhibitor 2b; also
known as p15 or Ink4b) was markedly decreased in PMCA4b-
rescued cells as compared with P4KO cells (PMCA4b versus
P4KO ϭ Ϫ50.4; p ϭ 8.51E-24). p15 is a cyclin-dependent kinase
inhibitor that forms a complex with CDK4 and prevents activa-
tion of Cyclin D1 (33).
Immunoblot analysis using late G1/S-stage cytoplasmic pro-
tein extracts showed that Rgs16 was increased in P4KO (and
control vector-rescued P4KO) cells versus P4WT cells but
decreased in PMCA4b-rescued P4KO cells (Fig. 7). Western
blots also showed that the decrease in NFATc3 expression
observed in P4KO cells was reversed by rescue with PMCA4b
(Fig. 7C). Together, these data clearly demonstrate that
PMCA4 splice variants rescue common Decorin, Rgs16, and
NFATc3 alterations observed in VSMC lacking any PMCA4
expression (P4KO) and also that expression of PMCA4a and
PMCA4b result in the regulation of unique downstream
mediators.
DISCUSSION
In the current study we present evidence that PMCA4 gene
deletion impairs cell cycle progression and causes a G1 phase
arrest in VSMC. Furthermore, quiescent VSMC in vivo have
roughly equal proportions of PMCA4a and PMCA4b splice
variants, whereas proliferating VSMC in vivo appear to have
relatively higher levels of PMCA4b than PMCA4a expression.
This suggested differential roles for these two PMCA4 splice
variants in the VSMC cell cycle. However, when P4KO VSMC
were electroporated with either PMCA4a or PMCA4b cDNA,
cells are rescued from the G1 phase arrest, and altered expres-
sion levels of Decorin and Rgs16 are corrected. Also, our data
show that whereas the PDZ binding domain unique to
PMCA4b (i.e. absent in PMCA4a) is not needed for this effect,
Ca2ϩ
efflux activity is required to rescue the G1 phase arrest of
P4KO cells. This observation suggest that the Ca2ϩ
efflux activ-
ity of PMCA4 is critical to G1-to-S-phase progression in VSMC
but leaves open the possibility that other domains within the
PMCA4 protein may also regulate the VSMC cell cycle.
FIGURE6.PutativedownstreamcellcyclemediatorsofPMCA4splicevari-
ants identified by microarray. RNA isolated from cell cycle-synchronized,
Hoechst-stained, and flow-sorted G0/G1 (A) and late G1/S-synchronized (B)
populations of P4WT and P4KO VSMC was used for microarray analysis.
PMCA4a (C)- and PMCA4b (D)-rescued P4KO VSMC at late G1/S were also
subjected to microarray analysis versus P4KO VSMC at the same cell cycle
stage. Bar graphs show -fold change in expression in pair-wise comparisons
between various cell cycle stage and genotype samples for Decorin, Rgs16,
AP2-␤, and p15 mRNA levels as determined by microarray analysis.
FIGURE 7. Validation of putative downstream cell cycle mediators of
PMCA4 by immunoblot. A, protein extracts were made from late G1/S-syn-
chronized P4WT, P4KO, and P4KO VSMC electroporated with mouse PMCA4a,
mouse PMCA4b, human PMCA4b-Asp mutant (10% of normal efflux activity),
or human PMCA4b-⌬PDZ mutant (lacking the PDZ binding domain) and
probed for Decorin, Rgs16, Cyclin D1, and GAPDH (n ϭ 2 separate experi-
ments). B, P4WT and P4KO primary cells were serum synchronized for 48 h
and harvested (G0) or stimulated with serum ϩ PDGF-BB for 24 h (G1/S) for
Western blots (n ϭ 2) for Decorin, Cyclin D1, and GAPDH. C, protein extracts
from late G1/S-synchronized VSMC (P4WT, P4KO, P4KOϩVector, and
P4KOϩmouse PMCA4b) were immunoblotted (n ϭ 2) for NFATc3, Rgs16 and
GAPDH. Representative immunoblots are shown.
PMCA4 Efflux Activity Regulates Cell Cycle in VSMC
7228 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289•NUMBER 10•MARCH 7, 2014
atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
Prior to out current results, a possible mechanism through
which PMCA4 alternatively spliced domains could regulate cell
cycle included PMCA4b binding to the tumor suppresser
RASSF1 (34), which mediates G1 cell cycle arrest via p21
expression (35) or via p53 and p16 accumulation (36). Instead of
directly testing the involvement of such a candidate, we used an
unbiased, open-ended microarray strategy to discover putative
PMCA4 targets. This analysis strongly implicated two proteins
(Decorin, Rgs16) previously not known as PMCA4-regulated
targets capable of modulating cell cycle. Both of these proteins
are markedly increased upon PMCA4 gene deletion, which is
reversed by expression of PMCA4a or PMCA4b (but not vector
DNA) in P4KO cells. Interestingly, both these PMCA4 target
proteins participate in the NFAT pathway. Decorin is an estab-
lished natural onco-suppressive factor and a novel ligand for
EGFR (37, 38), which is increased in VSMC in vivo after vascu-
lar injury (31) and triggers a signaling cascade involving Akt and
terminating in cyclin D1 induction (39). Decorin can inhibit
EGFR, has been implicated in G1 phase arrest (by inducing p21
expression), and is deregulated in many types of human cancers
(23, 24, 40). Decorin is also known to inhibit the VSMC cell
cycle by triggering Myc degradation (41) and can also inhibit
FosL1 (42). Under normal conditions FosL1 and Jun dimerize
to form AP-1, which can induce the NFAT family of transcrip-
tion factors (43), leading to cyclin D1 expression (26). Impor-
tant confirmation of our in vitro microarray and Western blot
results regarding Decorin included the novel finding that the
decrease in Decorin expression observed in injured carotid
arteries of P4WT mice is lost in P4KO mice, which manifest
significantly higher levels of Decorin expression after injury.
Rgs16, the other putative PMCA4 target identified here, is
known to block cell cycle progression by inhibiting the MAPK
and Akt pathways and is also modulated by the NFAT pathway.
Rgs16 overexpression in MCF7 breast cancer cells inhibits
EGFR-mediated proliferation and Akt phosphorylation,
whereas shRNA-mediated extinction of Rgs16 augments cell
growth (44). Transient as well as stable Rgs16 expression in
CHO cells can attenuate IP3 production and p38 MAPK acti-
vation by platelet-activating factor receptor (45). In T cells,
PKC activation induces TNF␣, which increases Rgs16 expres-
sion. Importantly, the calcineurin/NFAT pathway can repress
this effect on Rgs16 (27).
Our results suggest that PMCA4a (supplemental Fig. S8) as
well as PMCA4b (supplemental Fig. S9) function to limit
Decorin and Rgs16 expression (as well as increasing NFATc3
and cyclin D1 expression) and that PMCA4a may additionally
function via inhibiting AP-2␤ expression (which de-represses
c-Myc and induces EGFR expression), whereas PMCA4b may
work via decreasing the CDK4 inhibitor p15, allowing cyclin D1
to activate CDK4.
It is indeed surprising that although PMCA4a and PMCA4b
proteins show very different expression patterns in quiescent
and injured carotid arteries in vivo, both these splice variants
rescue the G1 arrest prevalent in proliferating primary VSMC
derived from the carotid arteries of P4KO mice. Potential
explanations include the following. (i) Our rescue of P4KO cells
was with the PMCA4x/a splice variant, whereas the PMCA4a
splice variant actually expressed in quiescent carotid artery
media may be PMCA4z/a. RNA-Seq of injured and uninjured
carotid arteries would be required to explore this possibility as
would the cloning and rescue by a PMCA4z/a (and PMCA4z/b)
splice variant in vitro. (ii) The P4KO mice we have employed
express “non-functional” PMCA4a and PMCA4b. As such,
endogenous non-functional PMCA4b proteins may form mul-
timeric complexes with exogenous PMCA4a enabling the non-
functional PMCA4b molecules to regain normal Ca2ϩ
efflux
activity within complexes functionally defined by the protein-
protein interactions of PMCA4b. In this scenario, although
P4KO cells were electroporated with PMCA4a, the cells were
able to regenerate “functional” PMCA4b pumps associated
with a normal Ca2ϩ
efflux activity. (iii) Electroporation of
PMCA4a into de-differentiated, proliferating P4KO VSMC in
culture may not mimic differentiated, quiescent VSMC in vivo,
where a balance between PMCA4a and PMCA4b expression
levels might be the critical factor driving the differentiated,
non-proliferative state. In this explanation, expressing
PMCA4a alone in de-differentiated, proliferating VSMC may
not be enough to regain the quiescent, non-proliferative state.
To achieve the latter, a fine balance in the expression levels and
possibly differential subcellular localization of the two splice
variants of PMCA4a and PMCA4b might be what is required.
The eukaryotic cell cycle is known to be regulated by Ca2ϩ
signals at various check points within the cycle (for review, see
Ref. 1). Both voltage- and ligand-gated Ca2ϩ
transporters are
known to control cell cycle checkpoints by modulating intra-
cellular Ca2ϩ
signals or by hitherto unknown mechanisms
involving macromolecular complexes with growth factor
receptors, cell adhesion molecules, or cytoplasmic regulatory
proteins (1). The latter may or may not depend on the Ca2ϩ
transport function of these transporters but do involve the par-
ticipation of their non-catalytic domains in signaling cascades
that modulate cell cycle checkpoints. For example, the voltage-
gated potassium channel subunit hERG1 (Kcnh2 or Kv11.1)
regulates proliferation by a mechanism involving its recruit-
ment into multiprotein membrane complexes including integ-
rins and growth factor receptors (46). Other examples where
Ca2ϩ
channel domains not involved in the Ca2ϩ
transport func-
tion regulate transcription via a protein signaling cascade
include Cav1.2 (Cacna1c), the L-type Ca2ϩ
channel 250-kDa
␣1C subunit that forms the Ca2ϩ
transporting transmembrane
pore. An endogenously cleaved 15-kDa carboxyl-terminal frag-
ment of Cav1.2 (calcium channel-associated transcription reg-
ulator (or CCAT)) translocates to the nucleus, binds to a
nuclear protein, associates with endogenous promoters, and
regulates the expression of a wide variety of endogenous genes
including the bone morphogenetic protein inhibitor follistatin-
like-1 (47, 48) protein and thus de-represses stem cell quies-
cence (45, 46). In a separate study involving the point mutation
of the Cav1.2 calmodulin binding domain, it was shown that
Ca2ϩ
/calmodulin binding to Cav1.2 enables specific activation
of the Ras/MAPK pathway, CREB activation, and CREB-driven
transcription (47). Importantly, it was discovered that the abil-
ity of Cav1.2 to generate a sustained [Ca2ϩ
]i rise was not needed
for it to activate the Ras/MAPK/CREB pathway leading to gene
transcription (47).
PMCA4 Efflux Activity Regulates Cell Cycle in VSMC
MARCH 7, 2014•VOLUME 289•NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 7229
atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
Further studies focused on elucidating the molecular mech-
anisms that allow PMCA4 to regulate the VSMC cell cycle are
needed. The current study supplies the impetus for researchers
examining PMCAs in other tissue systems to explore the roles
of PMCA4a and PMCA4b in other cellular processes. The only
other known example of physiological consequences of a
PMCA4a/PMCA4b switch in vivo is the observed increase in
relative PMCA4a splice variant expression (and decreased rel-
ative PMCA4b levels) during functional sperm maturation (49).
In that model of differentiation, there was a steady shift from
predominantly PMCA4b expression in the testis, caput, and
corpus epididymis to predominantly PMCA4a expression in
the cauda epididymis. It is interesting to note that differentia-
tion (associated with lack of cell proliferation) appears to
require an increase in the relative level of PMCA4a expression
in this model of sperm maturation. This correlates with the
observed decrease in relative PMCA4a expression in prolifer-
ating VSMC in vivo reported here and is reminiscent of the
2.5-fold reduction in rate of cell proliferation resulting from
transient overexpression of PMCA1a splice variant in mouse
VSMC (10) as well as the high relative proportion of PMCA4a
variant in bladder smooth muscle cells (SMCs) (quiescent
SMCs) and the roughly equal relative proportions of PMCA4a
and PMCA4b in uninjured carotid artery SMCs (quiescent
VSMC).
REFERENCES
1. Becchetti, A. (2011) Ion channels and transporters in cancer. 1. Ion chan-
nels and cell proliferation in cancer. Am. J. Physiol. Cell Physiol. 301,
C255–C265
2. Lipskaia, L., and Lompré, A. M. (2004) Alteration in temporal kinetics of
Ca2ϩ
signaling and control of growth and proliferation. Biol. Cell 96,
55–68
3. Carafoli, E., and Brini, M. (2000) Calcium pumps. Structural basis for and
mechanism of calcium transmembrane transport. Curr. Opin. Chem. Biol.
4, 152–161
4. Strehler, E. E., and Zacharias, D. A. (2001) Role of alternative splicing in
generating isoform diversity among plasma membrane calcium pumps.
Physiol. Rev. 81, 21–50
5. Keeton, T. P., Burk, S. E., and Shull, G. E. (1993) Alternative splicing of
exons encoding the calmodulin-binding domains and C termini of plasma
membrane Ca2ϩ
-ATPase isoforms 1, 2, 3, and 4. J. Biol. Chem. 268,
2740–2748
6. Santiago-García, J., Mas-Oliva, J., Saavedra, D., and Zarain-Herzberg, A.
(1996) Analysis of mRNA expression and cloning of a novel plasma mem-
brane Ca2ϩ
-ATPase splice variant in human heart. Mol. Cell. Biochem.
155, 173–182
7. Afroze, T., and Husain, M. (2000) c-Myb-binding sites mediate G1/S-
associated repression of the plasma membrane Ca2ϩ
-ATPase-1 promoter.
J. Biol. Chem. 275, 9062–9069
8. Afroze, T., Yang, L. L., Wang, C., Gros, R., Kalair, W., Hoque, A. N.,
Mungrue, I. N., Zhu, Z., and Husain, M. (2003) Calcineurin-independent
regulation of plasma membrane Ca2ϩ
ATPase-4 in the vascular smooth
muscle cell cycle. Am. J. Physiol. Cell Physiol. 285, C88–C95
9. Husain, M., Bein, K., Jiang, L., Alper, S. L., Simons, M., and Rosenberg,
R. D. (1997) c-Myb-dependent cell cycle progression and Ca2ϩ
storage in
cultured vascular smooth muscle cells. Circ. Res. 80, 617–626
10. Husain, M., Jiang, L., See, V., Bein, K., Simons, M., Alper, S. L., and Rosen-
berg, R. D. (1997) Regulation of vascular smooth muscle cell proliferation
by plasma membrane Ca2ϩ
-ATPase. Am. J. Physiol. 272, C1947–C1959
11. Gros, R., Afroze, T., You, X. M., Kabir, G., Van Wert, R., Kalair, W., Hoque,
A. E., Mungrue, I. N., and Husain, M. (2003) Plasma membrane calcium
ATPase overexpression in arterial smooth muscle increases vasomotor
responsiveness and blood pressure. Circ. Res. 93, 614–621
12. Okunade, G. W., Miller, M. L., Pyne, G. J., Sutliff, R. L., O’Connor, K. T.,
Neumann, J. C., Andringa, A., Miller, D. A., Prasad, V., Doetschman, T.,
Paul, R. J., and Shull, G. E. (2004) Targeted ablation of plasma membrane
Ca2ϩ
-ATPase (PMCA) 1 and 4 indicates a major housekeeping function
for PMCA1 and a critical role in hyperactivated sperm motility and male
fertility for PMCA4. J. Biol. Chem. 279, 33742–33750
13. Hummler, E., Cole, T. J., Blendy, J. A., Ganss, R., Aguzzi, A., Schmid, W.,
Beermann, F., and Schütz, G. (1994) Targeted mutation of the CREB gene.
Compensation within the CREB/ATF family of transcription factors. Proc.
Natl. Acad. Sci. U.S.A. 91, 5647–5651
14. Schuh, K., Cartwright, E. J., Jankevics, E., Bundschu, K., Liebermann, J.,
Williams, J. C., Armesilla, A. L., Emerson, M., Oceandy, D., Knobeloch,
K. P., and Neyses, L. (2004) Plasma membrane Ca2ϩ
ATPase 4 is required
for sperm motility and male fertility. J. Biol. Chem. 279, 28220–28226
15. You, X. M., Mungrue, I. N., Kalair, W., Afroze, T., Ravi, B., Sadi, A. M.,
Gros, R., and Husain, M. (2003) Conditional expression of a dominant-
negative c-Myb in vascular smooth muscle cells inhibits arterial remodel-
ing after injury. Circ. Res. 92, 314–321
16. Bolstad, B. M., Irizarry, R. A., Astrand, M., and Speed, T. P. (2003) A
comparison of normalization methods for high density oligonucleotide
array data based on variance and bias. Bioinformatics 19, 185–193
17. Du, P., Kibbe, W. A., and Lin, S. M. (2008) lumi. A pipeline for processing
Illumina microarray. Bioinformatics 24, 1547–1548
18. Smyth, G. K. (2004) Linear models and empirical bayes methods for as-
sessing differential expression in microarray experiments. Stat. Appl.
Genet Mol. Biol. 3, Article3
19. Reiner, A., Yekutieli, D., and Benjamini, Y. (2003) Identifying differentially
expressed genes using false discovery rate controlling procedures. Bioin-
formatics 19, 368–375
20. Xie, J., Jan, C., Stoilov, P., Park, J., and Black, D. L. (2005) A consensus
CaMK IV-responsive RNA sequence mediates regulation of alternative
exons in neurons. RNA 11, 1825–1834
21. Oceandy, D., Stanley, P. J., Cartwright, E. J., and Neyses, L. (2007) The
regulatory function of plasma-membrane Ca2ϩ
-ATPase (PMCA) in the
heart. Biochem. Soc. Trans. 35, 927–930
22. Schuh, K., Uldrijan, S., Gambaryan, S., Roethlein, N., and Neyses, L. (2003)
Interaction of the plasma membrane Ca2ϩ
pump 4b/CI with the Ca2ϩ
/
calmodulin-dependent membrane-associated kinase CASK. J. Biol. Chem.
278, 9778–9783
23. Ständer, M., Naumann, U., Wick, W., and Weller, M. (1999) Transform-
ing growth factor-␤ and p-21. Multiple molecular targets of decorin-me-
diated suppression of neoplastic growth. Cell Tissue Res. 296, 221–227
24. Hu, Y., Sun, H., Owens, R. T., Wu, J., Chen, Y. Q., Berquin, I. M., Perry, D.,
O’Flaherty, J. T., and Edwards, I. J. (2009) Decorin suppresses prostate
tumor growth through inhibition of epidermal growth factor and andro-
gen receptor pathways. Neoplasia 11, 1042–1053
25. Iozzo, R. V., Moscatello, D. K., McQuillan, D. J., and Eichstetter, I. (1999)
Decorin is a biological ligand for the epidermal growth factor receptor.
J. Biol. Chem. 274, 4489–4492
26. Karpurapu, M., Wang, D., Van Quyen, D., Kim, T. K., Kundumani-Srid-
haran, V., Pulusani, S., and Rao, G. N. (2010) Cyclin D1 is a bona fide target
gene of NFATc1 and is sufficient in the mediation of injury-induced vas-
cular wall remodeling. J. Biol. Chem. 285, 3510–3523
27. Fong, C. W., Zhang, Y., Neo, S. Y., and Lin, S. C. (2000) Specific induction
of RGS16 (regulator of G-protein signalling 16) mRNA by protein kinase C
in CEM leukaemia cells is mediated via tumour necrosis factor ␣ in a
calcium-sensitive manner. Biochem. J. 352, 747–753
28. Buettner, R., Kannan, P., Imhof, A., Bauer, R., Yim, S. O., Glockshuber, R.,
Van Dyke, M. W., and Tainsky, M. A. (1993) An alternatively spliced
mRNA from the AP-2 gene encodes a negative regulator of transcriptional
activation by AP-2. Mol. Cell. Biol. 13, 4174–4185
29. Gaubatz, S., Imhof, A., Dosch, R., Werner, O., Mitchell, P., Buettner, R.,
and Eilers, M. (1995) Transcriptional activation by Myc is under negative
control by the transcription factor AP-2. EMBO J. 14, 1508–1519
30. Han, C. Y., Lim, S. C., Choi, H. S., and Kang, K. W. (2008) Induction of
ErbB2 by ultraviolet A irradiation. Potential role in malignant transforma-
tion of keratinocytes. Cancer Sci. 99, 502–509
PMCA4 Efflux Activity Regulates Cell Cycle in VSMC
7230 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289•NUMBER 10•MARCH 7, 2014
atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
31. Singh, N. K., Wang, D., Kundumani-Sridharan, V., Van Quyen, D., Niu, J.,
and Rao, G. N. (2011) 15-Lipoxygenase-1-enhanced Src-Janus kinase
2-signal transducer and activator of transcription 3 stimulation and
monocyte chemoattractant protein-1 expression require redox-sensitive
activation of epidermal growth factor receptor in vascular wall remodel-
ing. J. Biol. Chem. 286, 22478–22488
32. Zhao, F., Bosserhoff, A. K., Buettner, R., and Moser, M. (2011) A heart-
hand syndrome gene: Tfap2b plays a critical role in the development and
remodeling of mouse ductus arteriosus and limb patterning. PLoS ONE 6,
e22908
33. Frost, S. J., Simpson, D. J., and Farrell, W. E. (2001) Decreased proliferation
and cell cycle arrest in neoplastic rat pituitary cells is associated with
transforming growth factor-␤1-induced expression of p15/INK4B. Mol.
Cell. Endocrinol. 176, 29–37
34. Armesilla, A. L., Williams, J. C., Buch, M. H., Pickard, A., Emerson, M.,
Cartwright, E. J., Oceandy, D., Vos, M. D., Gillies, S., Clark, G. J., and
Neyses, L. (2004) Novel functional interaction between the plasma mem-
brane Ca2ϩ
pump 4b and the proapoptotic tumor suppressor Ras-associ-
ated factor 1 (RASSF1). J. Biol. Chem. 279, 31318–31328
35. Kim, D. I., Lee, S. J., Lee, S. B., Park, K., Kim, W. J., and Moon, S. K. (2008)
Requirement for Ras/Raf/ERK pathway in naringin-induced G1 cell-cycle
arrest via p21WAF1 expression. Carcinogenesis 29, 1701–1709
36. Serrano, M., Lin, A. W., McCurrach, M. E., Beach, D., and Lowe, S. W.
(1997) Oncogenic ras provokes premature cell senescence associated with
accumulation of p53 and p16INK4a. Cell 88, 593–602
37. De Luca, A., Santra, M., Baldi, A., Giordano, A., and Iozzo, R. V. (1996)
Decorin-induced growth suppression is associated with up-regulation of
p21, an inhibitor of cyclin-dependent kinases. J. Biol. Chem. 271,
18961–18965
38. Moscatello, D. K., Santra, M., Mann, D. M., McQuillan, D. J., Wong, A. J.,
and Iozzo, R. V. (1998) Decorin suppresses tumor cell growth by activating
the epidermal growth factor receptor. J. Clin. Invest. 101, 406–412
39. Liu, G., Hitomi, H., Hosomi, N., Shibayama, Y., Nakano, D., Kiyomoto, H.,
Ma, H., Yamaji, Y., Kohno, M., Ichihara, A., Itoh, H., and Nishiyama, A.
(2011) Prorenin induces vascular smooth muscle cell proliferation and
hypertrophy via epidermal growth factor receptor-mediated extracellular
signal-regulated kinase and Akt activation pathway. J. Hypertens. 29,
696–705
40. Schönherr, E., Levkau, B., Schaefer, L., Kresse, H., and Walsh, K. (2001)
Decorin-mediated signal transduction in endothelial cells. Involvement of
Akt/protein kinase B in up-regulation of p21(WAF1/CIP1) but not
p27(KIP1). J. Biol. Chem. 276, 40687–40692
41. Buraschi, S., Pal, N., Tyler-Rubinstein, N., Owens, R. T., Neill, T., and
Iozzo, R. V. (2010) Decorin antagonizes Met receptor activity and down-
regulates ␤-catenin and Myc levels. J. Biol. Chem. 285, 42075–42085
42. Tharaux, P. L., Chatziantoniou, C., Fakhouri, F., and Dussaule, J. C. (2000)
Angiotensin II activates collagen I gene through a mechanism involving
the MAP/ER kinase pathway. Hypertension 36, 330–336
43. Huang, H., Chang, E. J., Ryu, J., Lee, Z. H., Lee, Y., and Kim, H. H. (2006)
Induction of c-Fos and NFATc1 during RANKL-stimulated osteoclast
differentiation is mediated by the p38 signaling pathway. Biochem. Bio-
phys. Res. Commun. 351, 99–105
44. Liang, G., Bansal, G., Xie, Z., and Druey, K. M. (2009) RGS16 inhibits
breast cancer cell growth by mitigating phosphatidylinositol 3-kinase sig-
naling. J. Biol. Chem. 284, 21719–21727
45. Zhang, Y., Neo, S. Y., Han, J., Yaw, L. P., and Lin, S. C. (1999) RGS16
attenuates G␣q-dependent p38 mitogen-activated protein kinase activa-
tion by platelet-activating factor. J. Biol. Chem. 274, 2851–2857
46. Pillozzi, S., and Arcangeli, A. (2010) Physical and functional interaction
between integrins and hERG1 channels in cancer cells. Adv. Exp. Med.
Biol. 674, 55–67
47. Dolmetsch, R. E., Pajvani, U., Fife, K., Spotts, J. M., and Greenberg, M. E.
(2001) Signaling to the nucleus by an L-type calcium channel-calmodulin
complex through the MAP kinase pathway. Science 294, 333–339
48. Yucel, G., Altindag, B., Gomez-Ospina, N., Rana, A., Panagiotakos, G.,
Lara, M. F., Dolmetsch, R., and Oro, A. E. (2013) State-dependent signal-
ing by Cav1.2 regulates hair follicle stem cell function. Genes Dev. 27,
1217–1222
49. Brandenburger, T., Strehler, E. E., Filoteo, A. G., Caride, A. J., Aumüller,
G., Post, H., Schwarz, A., and Wilhelm, B. (2011) Switch of PMCA4 splice
variants in bovine epididymis results in altered isoform expression during
functional sperm maturation. J. Biol. Chem. 286, 7938–7946
PMCA4 Efflux Activity Regulates Cell Cycle in VSMC
MARCH 7, 2014•VOLUME 289•NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 7231
atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
1
Online Supplementary Information
TABLE S-I: PRIMERS FOR CLONING, GENOMIC PCR & QUANTITATIVE RT-PCR
Name Sequence Comments
EGFP-forward ACACCCTGGTGAACCGCAT 50 nM; Ta=60ºC
EGFP-reverse CCATGATATAGACGTTGTGGCTGTT 50 nM; Ta=60ºC
Microglobulin-
forward
GCTATCCAGAAAACCCCTCAA 50 nM; Ta=60ºC
Microglobulin-
reverse
CATGTCTCGATCCCAGTAGACGGT 50 nM; Ta=60ºC
PMCA4a forward TCAACACCTTGATGTAAAACTGGTTCCTAGC 300 nM;
Ta=69ºC
PMCA4a reverse GACGACTCTGATTGCCCACAGGAG 300 nM;
Ta=69ºC
PMCA4b reverse TATGGAACAGTTTGACGACTCTGATCTGAG 300 nM;
Ta=69ºC
PMCA4b forward AGTGACAAAGAGGACATCAGCAGGGATACTGAG 300 nM;
Ta=69ºC
PMCA4e forward ACACCTTGATGTAAAACTGGTTCCTAGCTC 300 nM;
Ta=69ºC
PMCA4e reverse AACAGTTTGACGACTCTGATTCTCACTCAC 300 nM;
Ta=69ºC
5’UTR forward GCTACTCTGAAACTTATTGCACGAG Ta=60ºC
Mid-cDNA reverse CTTCACTGGCCATGGGCTCAATG Ta=60ºC
Mid-cDNA forward TCATTGAGCCCATGGCCAGTGAAG Ta=60ºC
3’UTR reverse CCACCGTCACATACAGATGG Ta=60 ºC
PMCA4-exon-3-
forward
(Genomic PCR)
AACGTGATACCTCCAAAAAGGCCCAAG Ta=65ºC
Amplicon = 816
bp
PMCA4-exon-4-
reverse
(Genomic PCR)
AATTTTTGCTCCAGTTCGATGCGACTC Ta=65ºC
Amplicon = 816
bp
Table-S-I Legend: Note: f, forward; r, reverse; Ta, annealing temperature; EGFP, enhanced green fluorescent
protein; all qRT-PCR primer pairs generated single amplicon peaks.
2
TABLE S-II: Genes Modulated in P4KO vs. P4WT cells (Early G1 Stage)
Fold Change p-value Symbol Definition
47.5 2.39E-09 Casp1 Caspase 1 (ICE); down regulates c-myc; inflammatory.
39.4 6.38E-09 Rgs16 Regulator of G-protein signaling 16 (Rgs16); inhibits
p38MAPK; NFAT suppresses Rgs16
21.2 6.38E-09 Gpnmb Glycoprotein (transmembrane) nmb (Gpnmb)
69.1 8.95E-09 Dcn Decorin; tumor suppressor; reduces tumor
metastases
14.2 1.24E-08 Adm Adrenomedullin (Adm)
30.6 1.24E-08 Pdpn Podoplanin; transmembrane glycoprotein
20.9 1.49E-08 Lce1f Late cornified envelope 1F
40.2 1.54E-08 LOC100048554 PREDICTED: similar to monocyte chemoattractant
protein-2 (MCP-2).
23.5 1.54E-08 Serpinb2 Serine (or cysteine) peptidase inhibitor, clade B, member
2 (Serpinb2)
-6.6 3.81E-08 Rcan2 Regulator of Calcineurin 2 mRNA
-3.6 4.02E-05 E2f2 E2F transcription factor 2 mRNA
3
TABLE S-III: Genes Known to be Implicated in G1 Phase Arrest
(P4KO vs. P4WT cells in early G1 stage)
Fold Change Symbol Definition
69.1 Dcn decorin (Dcn); tumor suppressor; reduces tumor metastases
5.2 Igf1 insulin-like growth factor 1; regulates MAPK/Akt pathway
5 Itga5 integrin alpha 5 (fibronectin receptor alpha)
4.2 Egfr epidermal growth factor receptor; tumor metastases
3.9 Fgf2 fibroblast growth factor 2
3.4 Ngf nerve growth factor
3.3 Dusp1 dual specificity phosphatase 1; ER stress induced apoptosis
2.8 Csf1 colony stimulating factor 1; induces NFATc1 degradation
2.1 Akap12 A kinase (PRKA) anchor protein 12; cell growth-related protein
2.1 Cdkn2A cyclin-dependent kinase inhibitor 2A; p16(INK4a); tumor
suppressor
-3.6 E2f2 E2F transcription factor 2; tumor suppressor
-3.5 Mcm7 minichromosome maintenance deficient 7; chromosomal DNA
replication factor
-3.2 Mybl2 Myeloblastosis oncogene-like 2 (Mybl2); B-Myb; S and G2/M
phase regulation
-3 Notch1 Notch gene homolog 1; tumor suppressor effect in myeloid
leukemia
-2.8 Rbl1 retinoblastoma-like 1 (p107); cell cycle regulation
-2.5 Pim1 proviral integration site 1; regulates S phase entry
-2.2 Mtbp Mdm2, transformed 3T3 cell double minute p53 binding protein;
Myc regulator
-2.2 Cdkn1B cyclin-dependent kinase inhibitor 1B; p27 or Kip1; regulates cell
cycle exit
-2.1 Cdkn1C cyclin-dependent kinase inhibitor 1C (p57 or Kip2); regulator of
cell cycle exit
TABLE S-IV: NFAT Facilitator Proteins, NFAT pathway modulators and NFAT Targets
(P4KO vs. P4WT in early G1 stage)
Fold Change p-value Gene Name Definition
69.1 8.95E-09 Dcn Decorin (Dcn), mRNA.
39.4 6.38E-09 Rgs16
Regulator of G-protein signaling 16 (Rgs16),
mRNA.
-6.6 3.81E-08 Rcan2
Regulator of calcineurin 2 (Rcan2), transcript variant
1, mRNA.
-1.8 0.002574978 Rcan1
Regulator of calcineurin 1 (Rcan1), transcript variant
2, mRNA.
-3.6 4.02E-05 E2f2 E2F transcription factor 2 (E2f2), mRNA.
4
TABLE S-V: Genes Modulated in PMCA4a-Rescued P4KO at Late G1/S Stage
(PMCA4a-rescued P4KO vs. P4KO)
FoldChange p-value Symbol Definition
-44.6 2.5E-10 Tfap2b
AP-2β, Activating enhancer-binding protein 2
beta, transcript variant 2 (inhibits Myc
transactivation, inhibits cell cycle)
-15.5 4.7E-09 Angpt2 Angiopoietin 2 (disrupts vascular remodeling)
-24.7 8.02E-09 Pcdh17 Protocadherin (cell-cell connections)
-8.2 4.09E-08 Tfap2a
AP-2α, Activating enhancer-binding protein 2
alpha
(transcription activator/repressor; mutations cause
branchio-oculo facial syndrome)
-1.27 NS Dcn Decorin
+1.1 NS Rgs16 Rgs16
+4.9 1.68E-07 Tnxb
Tenascin XB (matrix maturation during wound
healing)
+7.8 2.15E-07 Tnfrsf11b
Tumor necrosis factor receptor superfamily,
member 11b (vascular calcification)
+4.8 2.74E-07 Slitrk5
SLIT and NTRK-like family, member 5
(axonogenesis)
+9.1 5.56E-07 Igf2bp3
Insulin-like growth factor 2 mRNA binding
protein 3 (represses translation of insulin-like
growth factor-2)
+4.9 7.26E-07 Rragb
Ras-related GTP binding B (Ras-homologous
GTPases; signal transducers)
5
TABLE S-VI: Genes Modulated in PMCA4b-Rescued P4KO at Late G1/S Stage
(P4b-rescued P4KO vs. P4KO)
Fold Change p-value GeneName Definition
24.4 1.85E-25 Ptprd
Protein tyrosine phosphatase, receptor type, D
(Ptprd), transcript variant a, mRNA.
14.9 1.97E-21 Gjb3 Gap Junction protein beta 3 mRNA
32.0 2.03E-21 Anxa8 Annexin A8 mRNA
-136.8 6.80E-24 Dkk3
Dickkopf homolog 3 (Xenopus laevis) (Dkk3),
mRNA.
-50.4 8.51E-24 Cdkn2b
Cyclin-dependent kinase inhibitor 2B (p15;
inhibits CDK4) (Cdkn2b), mRNA.
-13.4 8.72E-24 Loxl1 Lysyl oxidase-like 1 (Loxl1), mRNA.
-23.9 8.72E-24 Fgf10 Fibroblast growth factor 10 (Fgf10), mRNA.
-22.3 1.37E-22 Pdgfra
Platelet derived growth factor receptor, alpha
polypeptide (Pdgfra), transcript variant 1, mRNA.
-9.2 4.87E-22 Sema3f
Sema domain, immunoglobulin domain (Ig), short
basic domain, secreted, (semaphorin) 3 F
(Sema3f), mRNA.
-2.4 1.59E-13 Dcn Decorin (Dcn), mRNA.
-18.5 3.07E-19 Rgs16
Regulator of G-protein signaling 16 (Rgs16),
mRNA.
TABLE S-VII: Genes Modulated in P4KO at Late G1/S Stage
(P4KO vs. P4WT)
Fold Change p-value GeneName Definition
39.4 1.20E-24 Thy1 Thymus cell antigen 1, theta (Thy1), mRNA.
33.0 7.76E-22 Ly6a
Lymphocyte antigen 6 complex, locus A (Ly6a),
mRNA.
64.7 1.12E-20 Serpinf1
Serine (or cysteine) peptidase inhibitor, clade F,
member 1 (Serpinf1), mRNA.
5.2 2.00E-20 Igfbp4 Insulin-like growth factor binding protein 4
7.4 2.82E-20 Pcdh21 Protocadherin 21 (Pcdh21), mRNA.
9.3 3.24E-20 Dcn Decorin (Dcn), mRNA.
4.6 5.75E-20 Nnmt
Nicotinamide N-methyltransferase (Nnmt),
mRNA.
7.6 6.68E-20 Spon2
Spondin 2, extracellular matrix protein (Spon2),
mRNA.
15.1 2.00E-18 Rgs16
Mus musculus regulator of G-protein signaling
16 (Rgs16), mRNA.
-7.2 6.68E-20 Dlx1 Distal-less homeobox 1 mRNA
-5.8 3.12E-17 Tuft1 Tuftelin 1 mRNA
6
TABLE S-VIII: Genes Modulated in PMCA4b-Rescued P4KO at Late G1/S Stage
(PMCA4b-rescued P4KO vs. Vector-control P4KO)
FoldChange p-value Symbol Definition
22.5 1.48E-24 Pacrg Park2 co-regulated mRNA
9.0 5.59E-21 Gpc1 Glypican 1 mRNA
-28.0 1.29E-24 Ogn Osteoglycin (Ogn), mRNA.
-15.3 1.29E-24 Ppap2b
Phosphatidic acid phosphatase type 2B (Ppap2b),
mRNA.
-17.4 3.56E-24 Atp1b1
ATPase, Na+/K+ transporting, beta 1 polypeptide
(Atp1b1), mRNA.
-7.3 4.31E-22 Dusp1 Dual specificity phosphatase 1 (Dusp1), mRNA.
-15.3 5.55E-22 Dcn Decorin (Dcn), mRNA.
-22.5 5.69E-22 Cdkn2b
Cyclin-dependent kinase inhibitor 2B (p15,
inhibits CDK4) (Cdkn2b), mRNA.
-7.7 8.59E-22 Loxl1 Lysyl oxidase-like 1 (Loxl1), mRNA.
-7.9 9.58E-22 Ier3 Immediate early response 3 (Ier3), mRNA.
-3.9 9.25E-14 Rgs16
Regulator of G-protein signaling 16 (Rgs16),
mRNA.
A
PMCA4-b (alternatively spliced variant) = VASHSDSPLPSLETPV
PMCA4-a (classical exon-20 of 181 nt) = AVTSPPVGNQSRQTVP
PMCA4-e (novel exon-20 of 191 nt) = AVTSPPVGSE
B
Peptide used for PMCA4b Antibody = SHSDSPLPSLETPV
Peptide used for PMCA4a/e Antibody = QTEAPLKRVRENMTQHLD
Figure S-I. (A) Predicted amino acid sequences at the C-terminus of the cloned splice
variants. Bold letters denote exon-20 or exon-21 encoded residues. (B) Sequences of
peptides used to generate chicken polyclonal Abs against PMCA4b and PMCA4a/e splice
variants. Bold letters denote residues conserved between mouse and rat PMCA4. (C)
Schematic representation and relevant sequences of alternatively spliced PMCA genes.
Lines show introns, rectangles are exons, and filled triangles are in-frame stop codons.
Sequences at the carboxy tails of resulting splice variants are displayed. Bold letters show
residues expressed as a result of exon inclusion or exclusion. NB: alternative splicing at site
C of PMCA1 and PMCA3 is more complex than shown. The schematic emphasizes instead
the similarities between ‘e’ variants of PMCA1, PMCA3 and PMCA4.
AAA
PMCA3e
PMCA3a
PMCA3b
PMCA1b
PMCA1a
PMCA1 pre-mRNA
poly A site
Stop codon
PMCA3 pre-mRNA
VVFSSSTASTTVGSEW (mouse)
VVFSSSTASTPVGSEW (rat)
VVFSSSTASTTVGFEW (human)
STASTTVGYPSGECIS (mouse)
TSSSPGSPLHSLETSL (mouse)
VTLSAAKPPSAAGSES (mouse)
VTLSAAKPTSAAGSES (rat)
AILSAANPTSAAGSES (human)
AILSAANPTSAAGSES (dog)
LSTATANTSPAPGSEST (tetraodon)
AKPPSAAGNPGGESIP (mouse)
FSSRPGSPLHSMETSL (mouse)
SSAISSAVTSPPVGSE (mouse)
SPSTSSAVTPPPVGSE (rat)
PTTSVPAVSSPPMGSE (human)
PTTSVPAVSSPPMGSE (chimp)
TTSVPAAVSSPTLGSE (dog)
AVTSPPVGNQSRQTVP (mouse)
VASHSDSPLPSLETPV (mouse)
PMCA4a
PMCA4b
PMCA4 pre-mRNA
PMCA4e
PMCA1e
C
M.W.ladder
PMCA4+/+
C57/Bl6
PMCA4-/-
816 bp band
specific for
PMCA4+/+
A
130 kDa
Anti-PMCA4a/e Anti-PMCA4b
a b c d a b c dB
Figure S-II. Validation of PMCA4a/e and PMCA4b specific antibodies (A) Genomic PCR
using exon-3 and exon-4 specific primers confirmed that the Neyses lab-generated PMCA4-
/- mouse hearts (in which exons-2 and -3 have been deleted) lack these PMCA4 exons from
their genome. (B) Immunoblots of microsomal protein extracts made from (a) C57bl6 heart,
(b) PMCA4-/- mouse heart, (c) C57bl6 stomach and (d) C57bl6 brain were probed with
affinity purified chicken polyclonal antibodies raised against peptides specific for PMCA4b
and PMCA4a/e. Note that the PMCA4a/e antibody did not give a band for normal mouse
heart (lane a) presumably because of negligible PMCA4a expression in the heart.
Relative Levels of PMCA4 splice variant (%) in LCM samples
Bladder SMC
Injured
Medial SMC
Remodeled
Adventitial SMC
0 20 40 60 80 100 120
PMCA4b
PMCA4a
Uninjured
Medial SMC
Figure S-III. Laser capture microdissection (LCM) was used to obtain uninjured
media (n=1), injured media (n=3) and remodeled adventitia regions (n=2) from mouse
carotid artery cross-sections (8-10 wk old male; day-9 post injury). Bladder smooth
muscle region (n=1) was used to show that the LCM protocol could demonstrate high
relative PMCA4a abundance observed in Fig1A. These microdissected regions were
used to isolate RNA, make cDNA and carry out qRT-PCR for microglobulin
(housekeeping gene) and PMCA4a and PMCA4b to calculate microglobulin-normalized
relative proportions of PMCA4a:4b splice variants in the various arterial regions.
Figure S-IV. Optimization of Electroporation Parameters. maxGFP plasmid, mouse
primary carotid artery smooth muscle cells, Lonza P1 Nucleofection Mix and Lonza 4D
Nucleofector were used.
CA-137 electroporation parameters: 67% efficiency
Mean of 4 fields of view; > 100 cells counted; not much cell death
CM-137 electroporation parameters: 100% efficiency
Mean of 3 fields of view ~50 cells counted; significant cell death
Negative Control: No electroporation parameters used; cells
incubated with maxGFP plasmid in cuvette
Figure S-V. Validation of electroporation of PMCA4 cDNA into VSMCs. EGFP-tagged
human PMCA4b-∆-PDZ cDNA construct was electroporated into P4KO primary carotid artery
smooth muscle cells using the Lonza 4D Nucleofector. Electroporated cells were seeded on
cover slips, allowed to recover for 24 h, fixed with paraformaldehyde, washed with PBS,
mounted on slides and used for confocal imaging.
EGFP-huPMCA4b-∆-PDZ construct electroporated into P4KO cells
Unelectroporated P4KO cells
DNAladder
PMCA4a
electroporated
Vector
electroporated
P4KOcells
NoTemplate
DNAladder
PMCA4a specific
amplicon band:
RT-PCR primers
designed to
detect PMCA4
exon9-exon10
junction which is
absent from
P4KO cells
Figure S-VI. RT-PCR validation of electroporation of PMCA4 cDNA into VSMCs. P4KO
cells carry a genetic deletion whereby exon-9 has been removed and exon-8 splices to exon-
10 in mature PMCA4 mRNA. Mouse PMCA4a cDNA construct encodes exon-9 joined to
exon-10. RT-PCR primers designed to detect this exon-9-exon-10 junction were used to
screen cDNA made from P4KO cells, P4KO cells rescued with vector and P4KO cells
rescued with mouse PMCA4a cDNA (cells were subjected to puromycin selection for 14 days
after electroporation). Agarose gel electrophoresis showed exon-9-exon-10 junction only in
those P4KO cells which received PMCA4a cDNA.
Figure S-VII. PMCA4a/e, PMCA4b and cleaved Caspase-3 Western blots. Whole cell
protein extracts made from P4WT, P4KO, P4KO+4a and P4KO+4b rescued primary carotid
VSMCs were used for immunoblots probed with anti-PMCA4a/e, anti-PMCA4b or anti-
cleaved Caspase-3 (apoptosis marker) antibodies. LiCor’s IR680 fluorescent dye conjugated
secondary antibodies (anti-chicken for PMCA4a/e and PMCA4b and anti-rabbit for cleaved
Caspase-3) were used and blots were imaged with the Odyssey fluorescent scanner (LiCor,
NE). PMCA4a was overexpressed 1.5-fold in P4KO+4a cells while PMCA4b was
overexpressed 1.8-fold in P4KO+4b cells as compared to P4KO cells. There was no cleaved
Caspase-3 band visible which signifies absence of any apoptosis in these cells.
PMCA4a/e
PMCA4b
P4WT P4KO P4KO+4a P4KO+4b
Cleaved Caspase-3
17 kDa
(expected band)
P4WT
P4KO
P4KO+4a
P4KO+4b
Protein
Ladder
Protein
Ladder
Figure S-VIII. Schematic showing role of proteins modulated by PMCA4a
overexpression in PMCA4 knockout cells. AP-2β represses Myc transactivation
and also inhibits AP-2 α from binding to the EGFR promoter and enhancing
proliferation. PMCA4a overexpression in PMCA4 knockout VSMC de-represses Myc
activity and allows AP-2 α to induce EGFR expression which leads to enhanced
proliferation. PMCA4a effects on Decorin and Rgs16 omitted for simplicity.
plasma membrane
AP-2 α
?
?
AP-2 β
PMCA4a
PI3K
p85
PI3K
Akt Signaling
Cell Cycle
GRB2
SOS
Ras
Cyclin D1
GSK3
FoxO
β -Catenin
Retinoblastoma protein
E2F2 repressor complexes
MAPK
Signaling
EGFR
EGFR is upregulated in
VSMC in vivo following
vascular injury
ErbB2 (EGFR)
promoter
c-Myc
EGFR
AP-2 α
plasma membrane
PI3K
p85
PI3K
Akt Signaling
Cell Cycle
GRB2
SOS
Ras
MAPK
Signaling
p15
(inhibits
Cyclin D1/Cdk4
and
Cyclin D1/Cdk6
complexes)
FosL1
AP-1
(FosL1 + Jun)
NFAT
Rgs16
Cyclin D1
GSK3
FoxO
β -Catenin
Retinoblastoma protein
E2F2 repressor complexes
Figure S-IX. Schematic showing role of proteins modulated by PMCA4b
overexpression in PMCA4 knockout cells. PMCA4b overexpression in
PMCA4 knockout VSMC represses Rgs16 and Decorin, which in turn regulates
NFAT and Cyclin D1 expression in the early G1 phase of the cell cycle.
PMCA4b
EGFR is
upregulated
in VSMC
in vivo
following
vascular
injury
EGFR
DecorinPMCA4b
?
?
??
?
?
Cyclin D1 promoter
NFAT

More Related Content

What's hot

Characterization of the human HCN1 channel and its inhibition by capsazepine
Characterization of the human HCN1 channel and its inhibition by capsazepineCharacterization of the human HCN1 channel and its inhibition by capsazepine
Characterization of the human HCN1 channel and its inhibition by capsazepineShahnaz Yusaf
 
Poster Filippa Pettersson ICDT 2010
Poster Filippa Pettersson ICDT 2010Poster Filippa Pettersson ICDT 2010
Poster Filippa Pettersson ICDT 2010FilippaP
 
In vitro transcription and transfection of HCV genomic replicon
In vitro transcription and transfection of HCV genomic repliconIn vitro transcription and transfection of HCV genomic replicon
In vitro transcription and transfection of HCV genomic repliconBinodGupta27
 
Семинар ДНК 16/05/2014 Сибэнзим
Семинар ДНК 16/05/2014 СибэнзимСеминар ДНК 16/05/2014 Сибэнзим
Семинар ДНК 16/05/2014 СибэнзимRuslan Titov
 
Irvin PLoS Genet 2014
Irvin PLoS Genet 2014Irvin PLoS Genet 2014
Irvin PLoS Genet 2014Jordan Irvin
 
Generation of MRP2 Efflux Transporter Knock-Out in HepaRG Cell Line
Generation of MRP2 Efflux Transporter Knock-Out in HepaRG Cell LineGeneration of MRP2 Efflux Transporter Knock-Out in HepaRG Cell Line
Generation of MRP2 Efflux Transporter Knock-Out in HepaRG Cell Linemdmitc
 
Culture of Renal Proximal Tubule Epithelial Cell Line SA7K Using Extracellula...
Culture of Renal Proximal Tubule Epithelial Cell Line SA7K Using Extracellula...Culture of Renal Proximal Tubule Epithelial Cell Line SA7K Using Extracellula...
Culture of Renal Proximal Tubule Epithelial Cell Line SA7K Using Extracellula...mdmitc
 
Multiplex energy metabolism
Multiplex energy metabolismMultiplex energy metabolism
Multiplex energy metabolismMatt Sanderson
 
NAGLU_IGF2 in vitro studies
NAGLU_IGF2 in vitro studiesNAGLU_IGF2 in vitro studies
NAGLU_IGF2 in vitro studiesShih-hsin Kan
 
Dissertation final complete1
Dissertation final complete1Dissertation final complete1
Dissertation final complete1Patrick Newton
 
Multi-domain Challenges of Fc Fusion Proteins and Bispecific Antibodies
Multi-domain Challenges of Fc Fusion Proteins and Bispecific AntibodiesMulti-domain Challenges of Fc Fusion Proteins and Bispecific Antibodies
Multi-domain Challenges of Fc Fusion Proteins and Bispecific AntibodiesJin Di, Ph.D.
 
Fluorescence activated cell sorted assay for Gaucher's disease
Fluorescence activated cell sorted assay for Gaucher's diseaseFluorescence activated cell sorted assay for Gaucher's disease
Fluorescence activated cell sorted assay for Gaucher's diseaseMayank Sagar
 
Glucocorticoids modulate micro rna expression and processing during lymphocyt...
Glucocorticoids modulate micro rna expression and processing during lymphocyt...Glucocorticoids modulate micro rna expression and processing during lymphocyt...
Glucocorticoids modulate micro rna expression and processing during lymphocyt...wangdong2336
 
Antibody-Producing Cell Lines Development
Antibody-Producing Cell Lines DevelopmentAntibody-Producing Cell Lines Development
Antibody-Producing Cell Lines Developmentcreativebiogene1
 
Regulation of atp7 a gene expression by the grx1 as an inducer in menkes d...
Regulation of atp7 a gene expression by the    grx1 as an inducer in menkes d...Regulation of atp7 a gene expression by the    grx1 as an inducer in menkes d...
Regulation of atp7 a gene expression by the grx1 as an inducer in menkes d...Pranamee Sarma
 
FINAL poster ORD
FINAL poster ORDFINAL poster ORD
FINAL poster ORDJoe Cameron
 

What's hot (19)

Characterization of the human HCN1 channel and its inhibition by capsazepine
Characterization of the human HCN1 channel and its inhibition by capsazepineCharacterization of the human HCN1 channel and its inhibition by capsazepine
Characterization of the human HCN1 channel and its inhibition by capsazepine
 
Poster Filippa Pettersson ICDT 2010
Poster Filippa Pettersson ICDT 2010Poster Filippa Pettersson ICDT 2010
Poster Filippa Pettersson ICDT 2010
 
In vitro transcription and transfection of HCV genomic replicon
In vitro transcription and transfection of HCV genomic repliconIn vitro transcription and transfection of HCV genomic replicon
In vitro transcription and transfection of HCV genomic replicon
 
Семинар ДНК 16/05/2014 Сибэнзим
Семинар ДНК 16/05/2014 СибэнзимСеминар ДНК 16/05/2014 Сибэнзим
Семинар ДНК 16/05/2014 Сибэнзим
 
Irvin PLoS Genet 2014
Irvin PLoS Genet 2014Irvin PLoS Genet 2014
Irvin PLoS Genet 2014
 
Generation of MRP2 Efflux Transporter Knock-Out in HepaRG Cell Line
Generation of MRP2 Efflux Transporter Knock-Out in HepaRG Cell LineGeneration of MRP2 Efflux Transporter Knock-Out in HepaRG Cell Line
Generation of MRP2 Efflux Transporter Knock-Out in HepaRG Cell Line
 
Culture of Renal Proximal Tubule Epithelial Cell Line SA7K Using Extracellula...
Culture of Renal Proximal Tubule Epithelial Cell Line SA7K Using Extracellula...Culture of Renal Proximal Tubule Epithelial Cell Line SA7K Using Extracellula...
Culture of Renal Proximal Tubule Epithelial Cell Line SA7K Using Extracellula...
 
Cristina Clement L08 presentation YI APS 2015 Orlando Florida
Cristina Clement L08 presentation YI APS 2015 Orlando FloridaCristina Clement L08 presentation YI APS 2015 Orlando Florida
Cristina Clement L08 presentation YI APS 2015 Orlando Florida
 
Multiplex energy metabolism
Multiplex energy metabolismMultiplex energy metabolism
Multiplex energy metabolism
 
NAGLU_IGF2 in vitro studies
NAGLU_IGF2 in vitro studiesNAGLU_IGF2 in vitro studies
NAGLU_IGF2 in vitro studies
 
12263-184470-1-PB
12263-184470-1-PB12263-184470-1-PB
12263-184470-1-PB
 
Dissertation final complete1
Dissertation final complete1Dissertation final complete1
Dissertation final complete1
 
Multi-domain Challenges of Fc Fusion Proteins and Bispecific Antibodies
Multi-domain Challenges of Fc Fusion Proteins and Bispecific AntibodiesMulti-domain Challenges of Fc Fusion Proteins and Bispecific Antibodies
Multi-domain Challenges of Fc Fusion Proteins and Bispecific Antibodies
 
Fluorescence activated cell sorted assay for Gaucher's disease
Fluorescence activated cell sorted assay for Gaucher's diseaseFluorescence activated cell sorted assay for Gaucher's disease
Fluorescence activated cell sorted assay for Gaucher's disease
 
Glucocorticoids modulate micro rna expression and processing during lymphocyt...
Glucocorticoids modulate micro rna expression and processing during lymphocyt...Glucocorticoids modulate micro rna expression and processing during lymphocyt...
Glucocorticoids modulate micro rna expression and processing during lymphocyt...
 
Antibody-Producing Cell Lines Development
Antibody-Producing Cell Lines DevelopmentAntibody-Producing Cell Lines Development
Antibody-Producing Cell Lines Development
 
BBRC ndrg2
BBRC ndrg2BBRC ndrg2
BBRC ndrg2
 
Regulation of atp7 a gene expression by the grx1 as an inducer in menkes d...
Regulation of atp7 a gene expression by the    grx1 as an inducer in menkes d...Regulation of atp7 a gene expression by the    grx1 as an inducer in menkes d...
Regulation of atp7 a gene expression by the grx1 as an inducer in menkes d...
 
FINAL poster ORD
FINAL poster ORDFINAL poster ORD
FINAL poster ORD
 

Similar to PMCA4 Regulates VSMC Cell Cycle Progression

Regulation Of Atp7 A Gene Expression By The Grx1 As An Inducer In Menkes D...
Regulation Of Atp7 A Gene Expression By The    Grx1 As An Inducer In Menkes D...Regulation Of Atp7 A Gene Expression By The    Grx1 As An Inducer In Menkes D...
Regulation Of Atp7 A Gene Expression By The Grx1 As An Inducer In Menkes D...pranamees
 
Nucl. Acids Res.-2014-Di Lorenzo-8297-309
Nucl. Acids Res.-2014-Di Lorenzo-8297-309Nucl. Acids Res.-2014-Di Lorenzo-8297-309
Nucl. Acids Res.-2014-Di Lorenzo-8297-309Alessandra Di Lorenzo
 
20081216 05袁國芳 紅麴菌基因體計畫及基因研究
20081216 05袁國芳 紅麴菌基因體計畫及基因研究20081216 05袁國芳 紅麴菌基因體計畫及基因研究
20081216 05袁國芳 紅麴菌基因體計畫及基因研究Monascus2008
 
P68 RNA helicase unwinds the human let-7 microRNA precursor duplex and is req...
P68 RNA helicase unwinds the human let-7 microRNA precursor duplex and is req...P68 RNA helicase unwinds the human let-7 microRNA precursor duplex and is req...
P68 RNA helicase unwinds the human let-7 microRNA precursor duplex and is req...David W. Salzman
 
Aditya - Cell Cycle Article Presentation.pptx
Aditya - Cell Cycle Article Presentation.pptxAditya - Cell Cycle Article Presentation.pptx
Aditya - Cell Cycle Article Presentation.pptxArPratiwiHasanuddin1
 
Modulation of MMP and ADAM gene expression in human chondrocytes by IL-1 and OSM
Modulation of MMP and ADAM gene expression in human chondrocytes by IL-1 and OSMModulation of MMP and ADAM gene expression in human chondrocytes by IL-1 and OSM
Modulation of MMP and ADAM gene expression in human chondrocytes by IL-1 and OSMpjtkoshy
 
Na f activates map ks and induces apoptosis in odontoblast-like
Na f activates map ks and induces apoptosis in odontoblast-likeNa f activates map ks and induces apoptosis in odontoblast-like
Na f activates map ks and induces apoptosis in odontoblast-likeGanesh Murthi
 
Discovery and Mechanistic Study of Mycobacterium tuberculosis PafA Inhibitors...
Discovery and Mechanistic Study of Mycobacterium tuberculosis PafA Inhibitors...Discovery and Mechanistic Study of Mycobacterium tuberculosis PafA Inhibitors...
Discovery and Mechanistic Study of Mycobacterium tuberculosis PafA Inhibitors...Dr.Shuaib Ahmad
 
TRISTETRAPROLIN - its role in inflammation and cancer
TRISTETRAPROLIN - its role in inflammation and cancerTRISTETRAPROLIN - its role in inflammation and cancer
TRISTETRAPROLIN - its role in inflammation and cancerHimadri Nath
 
Medicilon KRAS-targeted Drugs R&D Service.pdf
Medicilon KRAS-targeted Drugs R&D Service.pdfMedicilon KRAS-targeted Drugs R&D Service.pdf
Medicilon KRAS-targeted Drugs R&D Service.pdfmedicilonz
 

Similar to PMCA4 Regulates VSMC Cell Cycle Progression (20)

Regulation Of Atp7 A Gene Expression By The Grx1 As An Inducer In Menkes D...
Regulation Of Atp7 A Gene Expression By The    Grx1 As An Inducer In Menkes D...Regulation Of Atp7 A Gene Expression By The    Grx1 As An Inducer In Menkes D...
Regulation Of Atp7 A Gene Expression By The Grx1 As An Inducer In Menkes D...
 
pap paper pdf
pap paper pdfpap paper pdf
pap paper pdf
 
CcP2APX_Biochem_2008
CcP2APX_Biochem_2008CcP2APX_Biochem_2008
CcP2APX_Biochem_2008
 
Nucl. Acids Res.-2014-Di Lorenzo-8297-309
Nucl. Acids Res.-2014-Di Lorenzo-8297-309Nucl. Acids Res.-2014-Di Lorenzo-8297-309
Nucl. Acids Res.-2014-Di Lorenzo-8297-309
 
Vaillencourt
VaillencourtVaillencourt
Vaillencourt
 
20081216 05袁國芳 紅麴菌基因體計畫及基因研究
20081216 05袁國芳 紅麴菌基因體計畫及基因研究20081216 05袁國芳 紅麴菌基因體計畫及基因研究
20081216 05袁國芳 紅麴菌基因體計畫及基因研究
 
P68 RNA helicase unwinds the human let-7 microRNA precursor duplex and is req...
P68 RNA helicase unwinds the human let-7 microRNA precursor duplex and is req...P68 RNA helicase unwinds the human let-7 microRNA precursor duplex and is req...
P68 RNA helicase unwinds the human let-7 microRNA precursor duplex and is req...
 
Momen A et al 2014 J Receptors Signal Transduction
Momen A et al 2014  J Receptors Signal TransductionMomen A et al 2014  J Receptors Signal Transduction
Momen A et al 2014 J Receptors Signal Transduction
 
Aditya - Cell Cycle Article Presentation.pptx
Aditya - Cell Cycle Article Presentation.pptxAditya - Cell Cycle Article Presentation.pptx
Aditya - Cell Cycle Article Presentation.pptx
 
Modulation of MMP and ADAM gene expression in human chondrocytes by IL-1 and OSM
Modulation of MMP and ADAM gene expression in human chondrocytes by IL-1 and OSMModulation of MMP and ADAM gene expression in human chondrocytes by IL-1 and OSM
Modulation of MMP and ADAM gene expression in human chondrocytes by IL-1 and OSM
 
Na f activates map ks and induces apoptosis in odontoblast-like
Na f activates map ks and induces apoptosis in odontoblast-likeNa f activates map ks and induces apoptosis in odontoblast-like
Na f activates map ks and induces apoptosis in odontoblast-like
 
Polymophism in the promoter
Polymophism in the promoterPolymophism in the promoter
Polymophism in the promoter
 
Discovery and Mechanistic Study of Mycobacterium tuberculosis PafA Inhibitors...
Discovery and Mechanistic Study of Mycobacterium tuberculosis PafA Inhibitors...Discovery and Mechanistic Study of Mycobacterium tuberculosis PafA Inhibitors...
Discovery and Mechanistic Study of Mycobacterium tuberculosis PafA Inhibitors...
 
TRISTETRAPROLIN - its role in inflammation and cancer
TRISTETRAPROLIN - its role in inflammation and cancerTRISTETRAPROLIN - its role in inflammation and cancer
TRISTETRAPROLIN - its role in inflammation and cancer
 
Thesis
ThesisThesis
Thesis
 
KL PhD Presentation
KL PhD PresentationKL PhD Presentation
KL PhD Presentation
 
article
articlearticle
article
 
00b49517a6e686b6be000000
00b49517a6e686b6be00000000b49517a6e686b6be000000
00b49517a6e686b6be000000
 
FXR PCSK9
FXR PCSK9FXR PCSK9
FXR PCSK9
 
Medicilon KRAS-targeted Drugs R&D Service.pdf
Medicilon KRAS-targeted Drugs R&D Service.pdfMedicilon KRAS-targeted Drugs R&D Service.pdf
Medicilon KRAS-targeted Drugs R&D Service.pdf
 

PMCA4 Regulates VSMC Cell Cycle Progression

  • 1. and Mansoor Husain MomenMansoor Tanwir, Taha Tabish, Abdul Talat Afroze, Ge Yang, Amir Khoshbin, Vascular Smooth Muscle Cells Mediates Cell Cycle Progression in ATPase-4 (PMCA4)2+Membrane Ca Calcium Efflux Activity of Plasma Gene Regulation: doi: 10.1074/jbc.M113.533638 originally published online January 21, 2014 2014, 289:7221-7231.J. Biol. Chem. 10.1074/jbc.M113.533638Access the most updated version of this article at doi: .JBC Affinity SitesFind articles, minireviews, Reflections and Classics on similar topics on the Alerts: When a correction for this article is posted• When this article is cited• to choose from all of JBC's e-mail alertsClick here Supplemental material: http://www.jbc.org/content/suppl/2014/01/21/M113.533638.DC1.html http://www.jbc.org/content/289/10/7221.full.html#ref-list-1 This article cites 48 references, 30 of which can be accessed free at atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/DownloadedfromatTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
  • 2. Calcium Efflux Activity of Plasma Membrane Ca2؉ ATPase-4 (PMCA4) Mediates Cell Cycle Progression in Vascular Smooth Muscle Cells*□S Received for publication,November 11, 2013, and in revised form, January 10, 2014 Published, JBC Papers in Press,January 21, 2014, DOI 10.1074/jbc.M113.533638 Talat Afroze‡ , Ge Yang§ , Amir Khoshbin‡ , Mansoor Tanwir‡ , Taha Tabish‡ , Abdul Momen‡ , and Mansoor Husain‡§¶ʈ1 From the ‡ Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada, § Institute of Medical Sciences, University of Toronto, Toronto, Ontario M5S 1A8, Canada, and ¶ Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario M5G 1L7, and ʈ Department of Medicine, University of Toronto, Toronto, Ontario M5G 2C4, Canada Background: PMCA4 actions in vascular smooth muscle are not understood. Results: Alternative splicing of PMCA4 changes during vessel injury. Cell cycle arrest and downstream effectors of PMCA4 deletion are rescued by PMCA4a, PMCA4b, and PMCA4b unable to bind PDZ- proteins but not by inactive PMCA4. Conclusion: Ca2ϩ efflux activity of PMCA4 regulates G1 progression. Significance: PMCA4 Ca2ϩ efflux regulates cell cycle. We explored the role played by plasma membrane calcium ATPase-4 (PMCA4) and its alternative splice variants in the cell cycle of vascular smooth muscle cells (VSMC). A novel variant (PMCA4e) was discovered. Quantitative real-time-PCR-quanti- fied PMCA4 splice variant proportions differed in specific organs. The PMCA4a:4b ratio in uninjured carotid arteries (ϳ1:1) was significantly reduced by wire denudation injury (to ϳ1:3) by modulation of alternative splicing, as confirmed by novel antibodies against PMCA4a/e and PMCA4b. Laser cap- ture microdissection localized this shift to the media and adven- titia. Primary carotid VSMC from PMCA4 knock-out (P4KO) mice showed impaired [3 H]thymidine incorporation and G1 phase arrest as compared with wild type (P4WT). Electropora- tion of expression constructs encoding PMCA4a, PMCA4b, and a PMCA4b mutant lacking PDZ binding rescued this phenotype of P4KO cells, whereas a mutant with only 10% of normal Ca2؉ efflux activity could not. Microarray of early G1-synchronized VSMC showed 39-fold higher Rgs16 (NFAT (nuclear factor of activated T-cells) target; MAPK inhibitor) and 69-fold higher Decorin (G1 arrest marker) expression in P4KO versus P4WT. Validation by Western blot also revealed decreased levels of Cyclin D1 and NFATc3 in P4KO. Microarrays of P4KO VSMC rescued by PMCA4a or PMCA4b expression showed reversal of perturbed Rgs16, Decorin, and NFATc3 expression levels. How- ever, PMCA4a rescue caused a 44-fold reduction in AP-2␤, a known anti-proliferative transcription factor, whereas PMCA4b rescue resulted in a 50-fold reduction in p15 (Cyclin D1/Cdk4 inhibitor). We conclude that Ca2؉ efflux activity of PMCA4 underlies G1 progression in VSMC and that PMCA4a and PMCA4b differentially regulate specific downstream mediators. Progression through the cell cycle involves Ca2ϩ transport- ers including Ca2ϩ channels and Ca2ϩ efflux pumps (for review, see Ref. 1), and sarco-endoplasmic reticulum Ca2ϩ ATPase-2 (SERCA2) splice variants are differentially expressed at various stages of the cell cycle (for review, see Ref. 2). However, whether and how plasma membrane Ca2ϩ ATPases (PMCAs),2 a family of low capacity, high affinity plasma membrane-associated Ca2ϩ efflux pumps encoded by four genes (PMCA1–4) (for review, see Refs. 3 and 4) also participated in this process was not known. Alternative splicing at the amino (N) and carboxyl (C) termi- nal sites of PMCA4 pre-mRNA can produce x or z splice vari- ants at site N and a, b, or d splice variants at site C, each leading to the expression of a different protein (4–6). PMCA4 is expressed later in development than PMCA1 but is expressed more or less ubiquitously in adult mammalian organs (4). PMCA gene transcription is controlled by c-Myb, leading to changes in the free intracellular Ca2ϩ concentration ([Ca2ϩ ]i) during the cell cycle progression of vascular smooth muscle cells (VSMC) (7–9). Our initial observations in VSMC showed that inhibiting c-Myb expression or function prevented cell cycle progression and lowered [Ca2ϩ ]i (10). We found these changes to be medi- ated by increased PMCA1 and PMCA4 expression. Indeed, * This work was supported by operating grants (to M. H.) from the Canadian Institutes of Health Research (MOP-64352) and the Heart and Stroke Foun- dation of Ontario (T-5254 and NA6713). □S This article contains supplemental Tables S1–S8 and Figs. S1–S9. The nucle- otide sequence(s) reported in this paper has been submitted to the Gen- BankTM/EBI Data Bank with accession number(s) JF683384, EU493095, and EU493094 PMCA4 microarray data have been placed in the PubMed microarray database GEO under accession number GSE38320. 1 A Clinician-Scientist of the Canadian Institutes of Health Research (CL- 42617), currently a Career Investigator of the Heart and Stroke Foundation of Ontario (CI-5503). To whom correspondence should be addressed: TMDT-3-909, 200 Elizabeth St., Toronto, ON, Canada, M5G-2C4. Tel.: 416- 581-7489; E-mail: mansoor.husain@uhn.ca. 2 The abbreviations used are: PMCA, plasma membrane Ca2ϩ ATPases; qRT, quantitative real-time; VSMC, vascular smooth muscle cells; EGFP, enhanced GFP; IHC, immunohistochemistry; NFAT, nuclear factor of acti- vated T-cells; CREB, cAMP-response element-binding protein; PI, pro- pidium iodide; NS, not significant; Rgs16, regulator of G-protein signaling- 16; Dcn, Decorin. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 289, NO. 10, pp. 7221–7231, March 7, 2014 © 2014 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A. MARCH 7, 2014•VOLUME 289•NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 7221 atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
  • 3. transient overexpression of PMCA1a in VSMC elevated the Ca2ϩ efflux rate, decreased the [Ca2ϩ ]i, and reduced the rate of cell proliferation by over 2.5-fold (10). Whereas PMCA4b over- expression in mice increased blood pressure and myogenic tone (11), mice with functional knock-out of PMCA4 showed impaired phasic contractions and apoptosis in portal vein smooth muscle in vitro provided the mice were also heterozy- gous for the PMCA1 gene deletion (12). Together, these find- ings suggested that PMCA4 plays a role in VSMC biology. We now present evidence that the relative proportions of PMCA4a and PMCA4b splice variants are regulated after wire denudation injury of carotid arteries in vivo. Quiescent VSMC in vivo have roughly equal proportions of PMCA4a and PMCA4b splice variants until arterial injury leads to a signifi- cant decrease in the ratio of PMCA4a to PMCA4b. Mice lacking functional PMCA4 (P4KO) show reduced remodeling after injury in vivo, although primary VSMC isolated from such mice manifest reduced cell proliferation and a G1 phase arrest that can be rescued by either the PMCA4a or PMCA4b splice vari- ant alone. A PMCA4b mutant lacking the carboxyl-terminal PDZ binding domain also rescued the G1 phase arrest of P4KO VSMC, whereas a PMCA4b mutant with only 10% of the Ca2ϩ efflux activity of the wild-type pump could not. Microarray data show that functional PMCA4 ablation caused strong increases in Decorin (a G1 arrest marker) and regulator of G-protein sig- naling-16 (Rgs16; mitogen-activated protein kinase (MAPK) inhibitor, nuclear factor of activated T-cells (NFAT) target) expression, although rescue with either PMCA4a or PMCA4b restored normal Decorin and Rgs16 expression levels. Having said this, PMCA4a and PMCA4b rescues did differ in their global outcomes, with PMCA4a causing decreased expression of the transcription factor AP-2␤ (an inhibitor of c-Myc, AP-2␣, and cell cycle), whereas PMCA4b caused decreased expression of p15 (Cdkn2b (cyclin-dependent kinase inhibitor 2b)), an inhibitor of Cyclin D1/Cdk4. Together, these findings lead us to conclude that the Ca2ϩ efflux activity of PMCA4 mediates G1 phase cell cycle progression in VSMC and that PMCA4a and PMCA4b appear to work through different effec- tor proteins to do so. MATERIALS AND METHODS Mice and Cell Culture—C57BL6/J mice were purchased (Charles River Laboratories, Wilmington, MA). Frozen PMCA4 knock-out hearts (lacking exons 2 and 3) were a gift from Ludwig Neyses (Manchester, UK) (13, 14). Because dele- tion of exons 2–3 disrupts the reading frame of the resulting PMCA4 locus, these mice do not produce any PMCA4 protein (supplemental Fig. S2, panel B). PMCA4 functional knock-out mice distinct from the Neyses laboratory mice and lacking exon-11 (encoding codons 440–520, including aspartate 466, which is the catalytic phosphorylation site; GenBankTM acces- sion number NM_001167949.2) were a kind gift from Gary Shull (12). These exon-11-deficient mice express a mutant PMCA4 mRNA as a consequence of exon-10 splicing to exon-12 (12), which preserves the reading frame and enables these mice to produce functionless mutant PMCA4a and PMCA4b proteins (devoid of a catalytic site). These mice are referred to as P4KO mice, with their wild-type littermates denoted as P4WT. Protocols for mouse tissue harvest and carotid artery wire denudation injury have been described (15) and were approved by the Animal Care Committee of Univer- sity Health Network. Primary VSMC cultures were maintained in Dulbecco’s modified Eagle’s medium (DMEM) containing 10% fetal calf serum (Wisent; Montreal, Quebec, Canada), 50 ng/ml rat recombinant PDGF-BB (Sigma), and 1% penicillin- streptomycin in humidified air containing 5% CO2 at 37 °C. Cloning of PMCA4 cDNA Variants—PMCA4a, PMCA4b, and PMCA4e cDNA were cloned from total RNA extracted from mouse bladder, a mouse VSMC cell line (MOVAS; ATCC number CRL-2797), and mouse brain of C57 BL6/J mice, respectively (NucleoSpin RNA II kit; Takara-Clontech, Moun- tain View, CA) and PCR-cloned using mouse PMCA4-specific primers (see supplement Table S1) targeting either the 5Ј or 3Ј half of PMCA4 mRNA into pGemT-Easy vector (Promega). Clones containing the 5Ј and 3Ј halves of PMCA4 were digested with BamHI, gel-eluted, and ligated to generate full-length cDNA clones for each splice variant and sequenced. These cDNA sequences have been deposited in GenBankTM under accession numbers JF683384 (PMCA4a), EU493095 (PMCA4b), and EU493094 (PMCA4e). Antibody Generation and Validation—Peptides (supple- mental Fig. IB) specific for PMCA4b and PMCA4a/e carboxyl termini were designed (Aves Labs; Tigard, OR) and used to inject chickens. Polyclonal chicken antibodies with expected specificity for PMCA4 splice variants were collected and affin- ity-purified on columns containing the immobilized peptide antigen. Western Blotting—Protein extracts made from primary VSMC and various mouse organs including uninjured and injured carotid arteries were resolved on mini-PAGE gels (Novex; Invitrogen), blotted, and hybridized to various anti- bodies and visualized either by HRP-conjugated secondary antibodies (ECL kit; PerkinElmer Life Sciences) or by near infrared dye conjugated secondary antibodies using the Odys- sey laser scanner (LiCor, Lincoln, NE). qRT-PCR—Total RNA was extracted from various organs of C57BL6/J mice with the PicoPure RNA extraction kit (Invitro- gen). After DNase treatment and the addition of 1.0 pg of EGFP RNA to each sample (reverse transcription efficiency control), cDNA was synthesized, and qRT-PCR was carried out with primers specific for EGFP (cDNA synthesis efficiency), ␤2-mi- croglobulin (housekeeping gene), PMCA4a, PMCA4b, and PMCA4e splice variants (see supplemental Table S1) using SYBR GREEN master mix in an ABI HT-7900 (Invitrogen) thermal cycler. qRT-PCR primers for PMCA4 splice variants were first used to gel elute amplified products from each primer pair, clone them into pGemT-Easy vector, and sequence them to confirm the identity of the amplicon for each splice variant. Next, the PMCA4a, PMCA4b, and PMCA4e primer pairs were tested under various amplification conditions to measure amplification efficiency. Amplification conditions were chosen where the individual Ct values for each splice variant differed by Ͻ5% from the mean Ct value for all three variants. This ensured that amplification efficiency would not vary significantly among the PCR reactions for the three PMCA4 splice variants. The amplification conditions employed consisted of a hot start PMCA4 Efflux Activity Regulates Cell Cycle in VSMC 7222 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289•NUMBER 10•MARCH 7, 2014 atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
  • 4. of 94 °C for 10 min followed by 50 cycles of 94 °C for 15 s (dena- turation), 69 °C for 30 s (annealing), and 72 °C for 30 s (exten- sion). Gel eluted amplicons for EGFP, microglobulin, and the three PMCA4 splice variants were diluted to generate copy number standards containing 1 ϫ 106 , 1 ϫ 105 , 1 ϫ 104 , 1 ϫ 103 , 100, and 10 copies/␮l. The absolute copy number for each par- ticular mRNA was calculated via the absolute quantification method (Protocol 4304965; Applied Biosystems) that employs Ct values of copy number standards to calculate absolute copy numbers of each template in each sample. The absolute copy numbers of PMCA4 splice variants were then normalized to the copies of EGFP (cDNA synthesis efficiency control) and micro- globulin (housekeeping gene control) cDNA in each sample. Finally, normalized copy numbers of each PMCA4 splice vari- ant in each sample were summed, and copy numbers of each splice variant were divided by this sum to obtain the relative proportion of each splice variant in each sample. Immunohistochemistry (IHC), Immunofluorescent Staining, and Morphometry—Mouse carotid artery cross-sections (para- formaldehyde-fixed, paraffin-embedded) were used for immu- nostaining with various antibodies and fluorophore-conju- gated secondary antibodies (Jackson ImmunoResearch and Invitrogen) and visualized by confocal imaging on the Olympus Fluoview microscope using Fluoview software (Advanced Opti- cal Microscopy Facility, University Health Network). Regions of interest were drawn on the images, and fluorescence inten- sity was quantified in the regions of interest using Fluoview software. Quantification was done on at least two different regions of interests of both uninjured and injured carotid artery sections embedded in one paraffin section (three uninjured and three injured arteries per section), immunostained together on one slide, and imaged via z-stacks using the same laser intensity for all sections. For carotid artery morphometry, sections from P4KO and P4WT mice (day 9 post injury) were immunostained with anti-PMCA4a/e antibody, and the area of the entire media and entire remodeled adventitia was measured at 40ϫ resolu- tion on the Olympus Fluoview microscope using Fluoview software. Laser Capture Microdissection—Mouse carotid arteries (uninjured and injured) or mouse bladder fresh-frozen in OCT (Optical Cutting Temperature Compound, Sakura Finetek, Torrance, CA) were sectioned (10 ␮m thick) and mounted on laser capture microdissection microscope slides. Sections were lightly stained with Histogene (Invitrogen), fixed with 70% eth- anol, and taken through a dehydration series ending in xylene and dried. An laser capture microdissection microscope and laser set up (Advanced Optical Microscopy Facility, University Health Network) was used to microdissect and capture medial and adventitial layer regions of carotids (or bladder smooth muscle layer) from the stained and fixed sections. RNA was extracted with the PicoPure kit, and qRT-PCR was carried out as described above. Tritiated Thymidine Uptake Assays—Primary VSMC were seeded into 4 replicate wells of 6-well plates, allowed to grow overnight, and then serum-starved (0.1% serum; DMEM) for 24 h. Serum-synchronized cells were then stimulated with 10% serum and 50 ng/ml PDGF-BB for 24 h. Cells were given triti- ated thymidine for the last 1 h before harvest. Cells were lysed and processed for liquid scintillation counting. BrdU-PI Flow Cytometry—Primary VSMC were seeded into T-75 flasks, allowed to grow overnight, and then serum-starved (0.1% serum; DMEM) for 24 h. Synchronized cells were then stimulated with 10% serum and 50 ng/ml PDGF-BB for 24 h. Cells were labeled with BrdU for the last 1 h before harvest. Cells were harvested, fixed in 70% ethanol, and processed for anti-BrdU FITC-conjugated antibody staining as well as pro- pidium iodide staining and used for flow cytometry. Microarray Analysis—Primary VSMC from the carotid arteries of P4KO and P4WT mice were grown in quadruplicate T-75 flasks, serum-starved for 24 h, serum- and PDGF-BB- stimulated for 24 h, Hoechst-stained for the last 1 h before harvest, and then flow-sorted to obtain the G0/G1 subpopula- tion from all 8 samples (n ϭ 4 for each genotype). Total RNA was extracted from the 8 samples and used for microarray anal- ysis. In a second microarray, cells were starved for 24 h and serum- and PDGF-BB-stimulated for 24 h (G1/S stage), and RNA was extracted from P4WT cells, P4KO cells, PMCA4b- rescued P4KO cells, and vector-rescued P4KO cells. In a third experiment, RNA was extracted from P4KO (n ϭ 4), and P4KOϩPMCA4a-rescued (n ϭ 4) cells was used for microarray analysis. RNA was submitted to The Center for Applied Genomics (SickKids Hospital, Toronto, ON) for microarray analyses. Table 1 and supplemental Tables S2–S8 show genes increased or decreased in expression or which are known to be implicated in G1 phase arrest or are part of the NFAT pathway and were seen to be strongly modulated in P4KO cells as com- pared with P4WT cells. The preprocessing includes three steps: background correction (performed in GenomeStudio soft- ware), quantile normalization (16), and log2 transformation of normalized data. The last two steps were performed in lumi R package (17). The Center for Applied Genomics used LIMMA (linear models for microarray data) (18) to identify differentially expressed gene signatures under different conditions. Briefly speaking, it starts by fitting a linear model for each gene in the data, then an empirical Bayes method is used to moderate the standard errors for estimating the moderated t-statistics for each gene, which shrinks the standard errors toward a common value. This test is similar to an analysis of variance method for each gene except that the residual standard deviations are mod- erated across genes to ensure more stable inference for each gene. The moderated standard deviations are a compromise TABLE 1 Summary of microarray results Cdkn2b, cyclin-dependent kinase inhibitor 2b. Comparison Gene modulated (-fold change; p value) Early G1 P4KO vs. P4WT Dcn (ϩ69.1; p ϭ 8.95E-09) Rgs16 (ϩ39.4; p ϭ 6.38E-09) E2f2 (Ϫ3.6; p ϭ 4.02E-05) Late G1/S P4KO vs. P4WT Dcn (ϩ9.3; p ϭ 3.24E-20) Rgs16 (ϩ15.1; p ϭ 2.00E-18) PMCA4a-rescued vs. P4KO Tfap2b (Ϫ44.6; p ϭ 2.5E-10) Dcn (Ϫ1.27; p ϭ NS) Rgs16 (ϩ1.1; p ϭ NS) PMCA4b-rescued vs. P4KO Cdkn2b (p15) (-50.4; p ϭ 8.51E-24) Dcn (Ϫ2.4; p ϭ 1.59E-13) Rgs16 (Ϫ18.5; p ϭ 3.07E-19) PMCA4 Efflux Activity Regulates Cell Cycle in VSMC MARCH 7, 2014•VOLUME 289•NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 7223 atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
  • 5. between the individual gene-wise standard deviations and an overall pooled standard deviation. We evaluated the false dis- covery rate using the Benjamini multiple testing procedure (19). Our PMCA4 microarray data have been placed in the PubMed microarray database GEO under accession number GSE38320. P4KO Electroporated with PMCA4 cDNA—Electroporation was carried out with the 4D-Nucleofector (Lonza; Mississauga, ON), and transfection efficiencies of ϳ70% were regularly achieved (supplemental Figs. S4 and S5). Transfected cells were selected with the appropriate antibiotic (puromycin or G418), and the success of the drug selection was validated by RT-PCR (supplemental Fig. S6). P4KO cells were electroporated with either wild-type mouse PMCA4a, wild-type mouse PMCA4b, human PMCA4b mutant lacking a carboxyl-terminal PDZ binding domain (PMCA4b-⌬PDZ), or human PMCA4b-Asp mutant (catalytic site point mutant with 10% efflux activity compared with wild-type) or with vector where all constructs carried an antibiotic resistance cassette. Electroporated cells were allowed to recover for 24 h and then were subjected to 14 days of antibiotic selection before being used for BrdU-PI flow cytometry, microarray, or Western blots. Statistics—qRT-PCR data, flow cytometry data, and IHC staining quantification are expressed as the mean Ϯ S.E. and represent results from at least two separate experiments (each having n Ն 3 samples per group). Statistical analyses were per- formed using Student’s t test. Statistical significance was defined as p Յ 0.05. RESULTS Cloning Mouse PMCA4 Splice Variants—cDNA constructs encoding PMCA4a, PMCA4b, and the novel splice variant termed PMCA4e were cloned from total RNA extracted from mouse bladder, the murine VSMC cell line MOVAS, and mouse brain, respectively. Sequence analysis showed that the PMCA4e cDNA clone contains a novel exon-20 sequence that is 10 nucleotide longer than the evolutionarily conserved 181- nucleotide sequence of other mammalian PMCA4a variants. These additional 10 nucleotide result in a premature stop codon being created in the mature, spliced PMCA4e mRNA. The predicted amino acid sequences at the C terminus of the three cloned splice variants are shown in supplemental Fig. S1A. The sequence of the extra 10 nucleotides included in the mouse PMCA4e splice variant novel exon-20 is conserved in the genomes of man, chimpanzee, monkey, dog, cat, rabbit, mouse, and rat (supplemental Fig. S1C), which suggests a unique functional role for PMCA4e. PHI-BLAST analysis showed that other members of the PMCA family (viz. PMCA1 and PMCA3) also express splice variants similar to the PMCA4e variant we have cloned and share the GSE peptide motif at their carboxyl tails (supplemental Fig. S1C). PMCA4 Splice Variant-specific Antibodies—The PDZ bind- ing domain lies within the last seven amino acids of the carboxyl terminus of mouse PMCA4b (viz. PSLETPV), which is absent from the mouse PMCA4a tail. Two peptides were designed (Aves Labs); one coding for the PMCA4b carboxyl tail and the other coding for an exon-20-specific epitope expected in both PMCA4a and PMCA4e variants (supplemental Fig. S1B), which corresponds to the carboxyl end of the calmodulin binding motif. Hearts from homozygous Neyses laboratory-generated PMCA4 knock-out adult mice (PMCA4Ϫ/Ϫ ) and littermate wild-type controls (PMCA4ϩ/ϩ ) as well as C57/BL6/J mice were used in genomic PCR (14). This confirmed the genetic deletion of PMCA4 (exons 2–3) in knock-out hearts, with the 816-bp PMCA4 (exon3–4)-specific band identified only in hearts from PMCA4ϩ/ϩ and C57BL6/J mice (supplemental Fig. S2A). Next, Western blots revealed that the PMCA4b antibody recognized a ϳ130-kDa protein from brain, stomach, and heart of C57BL6/J mice but did not bind to protein extracts made from PMCA4Ϫ/Ϫ heart (supplemental Fig. S2B). Similarly, the PMCA4a/e antibody detected a ϳ130-kDa protein in extracts from C57BL6/J brain and stomach but not heart from C57BL6/J mice, presumably because of very low or absent PMCA4a/e expression in the normal mouse heart. PMCA4 Splice Variants Show Differential mRNA and Protein Expression Levels in Specific Mouse Organs—Having previously demonstrated a functional consequence of PMCA1a and PMCA4b overexpression (10, 11), we were interested in explor- ing the regulation of alternative splicing at splice site C in the PMCA4 pre-mRNA that generates PMCA4a, -4b, and -4e splice variants. To separate the consequences of transcriptional regulation (via the endogenous PMCA4 promoter) from those of alternative splicing modulation, the relative proportions of each PMCA4 splice variant were calculated from absolute copy number data obtained by quantitative RT-PCR. We reasoned that promoter up- or down-regulation would increase the abso- lute copy numbers of all alternatively spliced transcripts (so that analysis using absolute copy numbers would tend to reflect the effects of promoter regulation), whereas changes in the rel- ative proportions of each PMCA4 splice variant would accu- rately reflect regulation of alternative splicing. Others engaged in studies of alternative splicing have also used this strategy (20). In this manner we determined the relative proportions of PMCA4 splice variants in various mouse organs (Fig. 1A). We found that mouse organs could be grouped into three catego- ries: organs predominantly expressing PMCA4e (brain), organs predominantly expressing PMCA4a (bladder), and organs pre- dominantly expressing PMCA4b (all other organs tested), excluding liver, which showed negligible PMCA4 expression. We then used our PMCA4b and PMCA4a/e antibodies to probe PMCA4 splice variant proteins (Fig. 1B) and found that the organs surveyed also displayed distinct PMCA4a/e and PMCA4b expression profiles. Regulation of PMCA4 Alternative Splicing in Proliferating VSMC in Vivo—We next searched for a disease model in which PMCA4 alternative splicing may be modulated. When we examined mouse carotid arteries at day 9 post wire denudation injury, we found that the roughly equal proportion of PMC4a and PMCA4b splice variants observed in uninjured controls was dramatically altered, with preferential expression of PMCA4b at the expense of PMCA4a splice variant in injured arteries (Fig. 2A). Western blot analysis showed that this shift in alternative splicing was also manifest at the protein level (Fig. 2B). Laser capture microdissection coupled with qRT-PCR PMCA4 Efflux Activity Regulates Cell Cycle in VSMC 7224 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289•NUMBER 10•MARCH 7, 2014 atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
  • 6. showed that injured media and remodeled adventitia have a higher proportion of the PMCA4b transcript as compared with uninjured media, whereas a sample from bladder smooth mus- cle expressed a higher proportion of PMCA4a variant (supple- mental Fig. S3). IHC confocal imaging showed the expression of a smooth muscle marker (sm22␣) in uninjured and injured carotid artery cross-sections, whereas trichrome staining showed increased collagen deposition in remodeled adventitia (Fig. 3A). IHC staining with a proliferation marker (proliferating cell nuclear antigen) was only visible in injured arteries. Consistent with qRT-PCR and Western blot, IHC showed a decrease in PMCA4a/e and an increase in PMCA4b staining in injured carotids as compared with uninjured arteries that was further corroborated by quantifying immunofluorescent intensities with PMCA4 splice variant-specific antibodies in injured and uninjured medial layers (Fig. 3B). PMCA4 functional knock-out mice kindly provided by Gary Shull (Cincinnati, OH) lack exon-11, which encodes the central catalytic loop (81 amino acids; residues 440–520; see “Materials and Methods”) (12), resulting in a protein product devoid of any Ca2ϩ efflux activity (12). In the current manuscript, these mice are referred to as P4KO mice, with their wild-type littermates termed P4WT. Carotid artery cross-sections from P4WT and P4KO mice at day 9 post-injury were used for morphometry (Fig. 3, C and D). There was a 2-fold decrease in the adventitia: media ratio of injured P4KO arteries compared with P4WT arteries. This decreased remodeling in P4KO mice suggested two interrelated possibilities: (i) absence of functional PMCA4 impairs VSMC proliferation in vivo, and (ii) increased propor- tion of PMCA4b observed in injured arteries may play a physi- ologically relevant role in VSMC proliferation. PMCA4 Knock-out Causes G1 Phase Arrest, Which Can Only Be Rescued by PMCA4 with Intact Ca2ϩ Efflux Activity—To probe the correlation between relative proportions of PMCA4 splice variants and VSMC proliferation, we isolated primary carotid VSMC from P4WT and P4KO mice. [3 H]Thymidine uptake assays using cell cycle-synchronized VSMC cultures showed an ϳ70% reduction in G1 to S phase cell cycle progres- sion of P4KO VSMC compared with P4WT cells (Fig. 4A). Sim- ilarly, BrdU-PI flow cytometry showed an ϳ30% increase in G1-stage P4KO cells as compared with P4WT (Fig. 4B), sug- gesting a G1 stage arrest. Importantly, electroporation of either mouse PMCA4a or PMCA4b expression constructs rescued the cell cycle pheno- type of P4KO cells (Fig. 4B), i.e. PMCA4a- or PMCA4b-rescued P4KO VSMC manifest a 40–50% increase in S phase cells and an ϳ50% reduction in G1 phase cells. Western blots confirmed (supplemental Fig. S7) that as a result of these electroporations, PMCA4a was overexpressed 1.5-fold in P4KOϩ4a cells, whereas PMCA4b was overexpressed 1.8-fold in P4KOϩ4b FIGURE 1. Distinct PMCA4 splice variant expression levels in mouse organs. A, qRT-PCR-determined relative proportions of PMCA4 splice vari- ants in various organs of C57BL6/J mice. Bars represent mean relative propor- tionforeachsplicevariantmRNAextractedfromtheorgansofthreeC57BL6/J mice, normalized for cDNA conversion efficiency (EGFP) and loading (␤2-mi- croglobulin).LivergavenoPMCA4splicevariantcopiesuponamplification.B, immunoblots of microsomal protein extracts from various organs of C57BL6/J mice. Microsomal extracts were made from the organs of three C57BL6/J mice and resulted in nine separate immunoblots (three each for PMCA4-a/e, PMCA4b, and GAPDH); representative blots are shown. Br, brain; U,uterus;bl,bladder;M,skeletalmuscle;K,kidney;S,stomach;H,heart;L,liver. FIGURE 2. PMCA4 splice variant mRNA and protein expression levels are regulated after arterial injury. A, qRT-PCR-determined relative proportions of PMCA4 spice variants in uninjured and injured carotid arteries from male C57BL6/J mice (n ϭ 4). p values for uninjured versus injured, PMCA4a ϭ 0.001 and PMCA4b ϭ 0.01, by Students’ t test. B, immunoblots of protein extracts made from uninjured and injured mouse carotid arteries. Representative blots from two independent experiments are shown. PMCA4 Efflux Activity Regulates Cell Cycle in VSMC MARCH 7, 2014•VOLUME 289•NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 7225 atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
  • 7. cells as compared with P4KO cells. Although expression of a PMCA4b mutant lacking the PDZ binding domain was still able to rescue the G1 arrest of P4KO cells, expression of a PMCA4b- Asp mutant (with only 10% of the Ca2ϩ efflux activity of wild- type PMCA4) failed to rescue the G1 phase arrest of P4KO cells (Fig. 4B). In a separate series of experiments assessing cell cycle pro- gression at multiple time points after serum stimulation of G0-synchronized P4WT, P4KO, and PMCA4a- or PMCA4b- rescued cell populations, we confirmed that P4KO cells suffer from a G1 phase arrest that is completely alleviated by either PMCA4a or PMCA4b cDNA expression (Fig. 5). Together these data show that whereas specific PDZ binding domain- mediated PMCA4b/nNOS (21) or PMCA4b/CASK interac- tions (22) are not necessary for PMCA4b to regulate cell cycle, the presence of Ca2ϩ efflux activity is required for PMCA4 to mediate G1-to-S phase progression in VSMC. Microarray Analysis and Validation of Identified Targets— Early G1 stage P4KO and P4WT cells were collected via flow sorting (by gating for G0/G1 phase Hoechst-stained cells) and used for microarray analysis. This unbiased exploration revealed that Decorin (ϩ69.1-fold; p ϭ 8.9E-09) and Rgs16 (ϩ39.4-fold; p ϭ 6.3E-09; Fig. 6A and supplemental Tables 2 and 3) were markedly increased in P4KO cells as compared with P4WT cells. Decorin is a known inhibitor of the VSMC cell cycle and implicated in G1 phase arrest (for review, see Ref. 23). Decorin is known to reduce cyclin D1 expression via the epi- dermal growth factor receptor (EGFR) or NFAT pathways (24– 26), with NFAT activation also known to inhibit Rgs16 expres- sion (27) (see the proposed model, supplemental Fig. S8). This FIGURE 3. Immunofluorescence-based quantification revealed regulated PMCA4 splicing in VSMC of injured carotid arteries. A, confocal images of uninjured and injured carotid artery cross-sections from male C57BL6/J mice stained for PMCA4a/e, PMCA4b, smooth muscle marker (sm22␣), proliferation marker (proliferating cell nuclear antigen), and collagen deposition (trichrome). Scale bars in panel A trichrome images ϭ 500 ␮m, in confocal images ϭ 20 ␮m, and in panel D confocal images ϭ 50 ␮m. B, confocal microscopy Z-stack quantification of PMCA4a/e staining. Each bar represents a mean of 3 mouse carotid artery cross-sections in which ϳ40–70 cells or cell clusters were used to quantify fluorescent intensity through 20–40 focal planes of 5-␮m-thick sections. C, morphometric analysis of injured carotid arteries in PMCA4 wild-type (P4WT) and PMCA4 knock-out (P4KO) mice (n ϭ 4). Carotid artery cross-sections were immunostained for PMCA4a/e with media and adventitia captured as regions of interest for area measurements on FluoView microscope (40ϫ objective) and software (Olympus, Mountain View, CA). The ratio of adventitia:media areas are decreased in injured carotid arteries of P4KO mice (P4WT ϭ 3.1 Ϯ 0.5 versus PMKO ϭ 1.4 Ϯ 0.1; p ϭ 0.01; n ϭ 4). D, representative immunofluorescent micrographs of P4WT and P4KO injured carotid arteries immunostained for PMCA4a/e show reduced medial thickening in P4KO mice. PMCA4 Efflux Activity Regulates Cell Cycle in VSMC 7226 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289•NUMBER 10•MARCH 7, 2014 atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
  • 8. suggested that the high Rgs16 expression observed in P4KO cells (as compared with P4WT cells) may be due to reduced NFAT expression or activation in P4KO cells. Consistent with this, immunoblot analysis showed that Decorin and Rgs16 lev- els were increased, and Cyclin D1 levels were decreased in P4KO cells as compared with P4WT cells (Fig. 7). Similar results were observed in vivo. First, Decorin-staining intensity trended 18% lower in medial layers of injured as com- pared with uninjured carotid arteries of P4WT mice (0.82 Ϯ 0.05 versus 1.00 Ϯ 0.06; n ϭ 3, p ϭ 0.1), suggesting injury- induced decreases in Decorin expression accompanied the reduced PMCA4a:PMCA4b expression ratio. Second, this pat- tern was lost in P4KO mice, in which carotid artery denudation injury failed to reduce Decorin expression (1.10 Ϯ 0.03 versus 1.00 Ϯ 0.04; n ϭ 3, p ϭ NS), supporting a role for PMCA4 in regulating Decorin expression in vivo. Indeed, similar to the microarray-documented difference in Decorin expression observed in primary VSMC cultures from P4KO versus P4WT mice, levels of Decorin expression were significantly higher in injured carotid arteries of P4KO animals as compared with P4WT (1.10 Ϯ 0.03 versus 0.82 Ϯ 0.05; n ϭ 3, p ϭ 0.01) (data not shown). Because Decorin and Rgs16 have both been implicated in the NFAT pathway, we next examined mRNA levels of NFAT facil- itator proteins Rcan1, Rcan2, and the NFAT target E2F2 in our microarray results (supplemental Table S4). Rcan2 was reduced Ϫ6.6-fold (p ϭ 3.8E-08) and E2F2 was decreased Ϫ3.6-fold (p ϭ 4.0E-05) in P4KO as compared with P4WT cells, suggesting diminished NFAT action. To examine this more directly, cyto- plasmic protein extracts were probed for NFATc3 levels and found to be significantly decreased in P4KO cells as compared with P4WT cells (Fig. 7). PMCA4a rescue of P4KO cells followed by late G1/S microar- ray analysis (Fig. 6, B and C) suggested some reversal of the expression levels of Decorin (PMCA4a rescue versus P4KO ϭ Ϫ1.27-fold; p ϭ NS) and Rgs16 (PMCA4a rescue versus P4KO ϭ ϩ1.1; p ϭ NS); however, these changes did not reach statistical significance (supplemental Table S5). Rather, PMCA4a-rescued P4KO cells showed a sharp decline in the expression of a known anti-proliferative transcription factor viz. AP-2␤ (PMCA4a rescue versus P4KO ϭ 44.6; p ϭ 2.5E-10) (supplemental Table S5). AP-2␤ inhibits the activity of AP-2␣ (28) and also represses c-Myc transactivation (29). AP-2␣ is also known to bind to the ErbB2 promoter (EGFR family mem- ber) and promote proliferation via the EGFR pathway (30) acti- vated by vessel injury in vivo (31). AP-2␤ is also implicated in VSMC proliferation in vivo by its role in the closure of the ductus arteriosus (32). Additional microarray analysis using RNA harvested from late G1/S-stage P4KO, P4WT, and PMCA4b-rescued- versus vector-transfected-P4KO cells showed that Decorin was down- regulated Ϫ2.4-fold (p ϭ 1.59E-13) in PMCA4b-rescued versus FIGURE4.Ca2؉ effluxactivityofPMCA4isrequiredtorescuetheG1 arrest of functional PMCA4 ablation. A, [3 H]thymidine uptake at the late G1/S- stage was assayed in cell cycle-synchronized PMCA4 wild-type (P4WT) and PMCA4knock-out(P4KO)primaryVSMC(nϭ3experiments).P4KOcellsshow a 64% reduction in thymidine uptake as compared with P4WT cells (*, p ϭ 2.8E-08, Student’s t test). B, flow cytometric determination of cell cycle distri- butions of P4WT and P4KO primary VSMC. P4KO cells were electroporated with expression constructs encoding mouse PMCA4a, mouse PMCA4b, human PMCA4b-Asp mutant (10% Ca2ϩ efflux activity compared with wild- type), or human PMCA4b-⌬PDZ mutant (lacking PDZ-binding domain) and G0-synchronized by serum starvation. After synchronized serum stimulation for 22 h, the cell cycle distribution of each population was determined by BrdU-PI flow cytometry (n ϭ 3 independent experiments with multiple repli- cates for all samples). *p ϭ 0.004 for P4WT versus P4KO; §, p ϭ 1.85E-6 for PMCA4a-rescued P4KO versus P4KO; ϱ, p ϭ 0.001 for PMCA4b-rescued P4KO versus P4KO; †, p ϭ 2.9E-16 for huP4b-Asp-rescued P4KO versus PMCA4b- rescued P4KO; ‡, p ϭ 0.0001 for huP4b-⌬PDZ-rescued P4KO versus P4KO. Student’s t test was used for all comparisons. FIGURE 5. Correction of the impaired cell cycle progression of PMCA4 knock-out VSMC. Cell cycle distributions of P4WT, P4KO, or P4KO VSMC elec- troporated with mouse PMCA4a or PMCA4b cDNA were determined by BrdU-PI flow cytometry (n ϭ 2 independent experiments with multiple repli- cates for all samples) at various time points in the cell cycle using serum- synchronized populations. PMCA4 Efflux Activity Regulates Cell Cycle in VSMC MARCH 7, 2014•VOLUME 289•NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 7227 atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
  • 9. P4KO cells (down Ϫ15.3-fold in PMCA4b-rescued versus con- trol P4KO cells; p ϭ 5.5E-22), whereas it remained up-regulated ϩ9.3-fold (p ϭ 3.24E-20) in P4KO versus P4WT G1/S stage cells (Fig. 6D; and supplemental Tables S6–S8). Rgs16 was also decreased Ϫ18.5-fold (p ϭ 3.07E-19) in PMCA4b-rescued ver- sus P4KO cells (Ϫ3.9-fold in PMCA4b-rescued versus vector- control P4KO cells; p ϭ 9.2E-14), whereas it continued to be increased ϩ15.1-fold (p ϭ 2.0E-18) in P4KO versus P4WT at the G1/S stage (Fig. 6D; and supplemental Tables S6–S8). Inter- estingly, Cdkn2b (cyclin-dependent kinase inhibitor 2b; also known as p15 or Ink4b) was markedly decreased in PMCA4b- rescued cells as compared with P4KO cells (PMCA4b versus P4KO ϭ Ϫ50.4; p ϭ 8.51E-24). p15 is a cyclin-dependent kinase inhibitor that forms a complex with CDK4 and prevents activa- tion of Cyclin D1 (33). Immunoblot analysis using late G1/S-stage cytoplasmic pro- tein extracts showed that Rgs16 was increased in P4KO (and control vector-rescued P4KO) cells versus P4WT cells but decreased in PMCA4b-rescued P4KO cells (Fig. 7). Western blots also showed that the decrease in NFATc3 expression observed in P4KO cells was reversed by rescue with PMCA4b (Fig. 7C). Together, these data clearly demonstrate that PMCA4 splice variants rescue common Decorin, Rgs16, and NFATc3 alterations observed in VSMC lacking any PMCA4 expression (P4KO) and also that expression of PMCA4a and PMCA4b result in the regulation of unique downstream mediators. DISCUSSION In the current study we present evidence that PMCA4 gene deletion impairs cell cycle progression and causes a G1 phase arrest in VSMC. Furthermore, quiescent VSMC in vivo have roughly equal proportions of PMCA4a and PMCA4b splice variants, whereas proliferating VSMC in vivo appear to have relatively higher levels of PMCA4b than PMCA4a expression. This suggested differential roles for these two PMCA4 splice variants in the VSMC cell cycle. However, when P4KO VSMC were electroporated with either PMCA4a or PMCA4b cDNA, cells are rescued from the G1 phase arrest, and altered expres- sion levels of Decorin and Rgs16 are corrected. Also, our data show that whereas the PDZ binding domain unique to PMCA4b (i.e. absent in PMCA4a) is not needed for this effect, Ca2ϩ efflux activity is required to rescue the G1 phase arrest of P4KO cells. This observation suggest that the Ca2ϩ efflux activ- ity of PMCA4 is critical to G1-to-S-phase progression in VSMC but leaves open the possibility that other domains within the PMCA4 protein may also regulate the VSMC cell cycle. FIGURE6.PutativedownstreamcellcyclemediatorsofPMCA4splicevari- ants identified by microarray. RNA isolated from cell cycle-synchronized, Hoechst-stained, and flow-sorted G0/G1 (A) and late G1/S-synchronized (B) populations of P4WT and P4KO VSMC was used for microarray analysis. PMCA4a (C)- and PMCA4b (D)-rescued P4KO VSMC at late G1/S were also subjected to microarray analysis versus P4KO VSMC at the same cell cycle stage. Bar graphs show -fold change in expression in pair-wise comparisons between various cell cycle stage and genotype samples for Decorin, Rgs16, AP2-␤, and p15 mRNA levels as determined by microarray analysis. FIGURE 7. Validation of putative downstream cell cycle mediators of PMCA4 by immunoblot. A, protein extracts were made from late G1/S-syn- chronized P4WT, P4KO, and P4KO VSMC electroporated with mouse PMCA4a, mouse PMCA4b, human PMCA4b-Asp mutant (10% of normal efflux activity), or human PMCA4b-⌬PDZ mutant (lacking the PDZ binding domain) and probed for Decorin, Rgs16, Cyclin D1, and GAPDH (n ϭ 2 separate experi- ments). B, P4WT and P4KO primary cells were serum synchronized for 48 h and harvested (G0) or stimulated with serum ϩ PDGF-BB for 24 h (G1/S) for Western blots (n ϭ 2) for Decorin, Cyclin D1, and GAPDH. C, protein extracts from late G1/S-synchronized VSMC (P4WT, P4KO, P4KOϩVector, and P4KOϩmouse PMCA4b) were immunoblotted (n ϭ 2) for NFATc3, Rgs16 and GAPDH. Representative immunoblots are shown. PMCA4 Efflux Activity Regulates Cell Cycle in VSMC 7228 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289•NUMBER 10•MARCH 7, 2014 atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
  • 10. Prior to out current results, a possible mechanism through which PMCA4 alternatively spliced domains could regulate cell cycle included PMCA4b binding to the tumor suppresser RASSF1 (34), which mediates G1 cell cycle arrest via p21 expression (35) or via p53 and p16 accumulation (36). Instead of directly testing the involvement of such a candidate, we used an unbiased, open-ended microarray strategy to discover putative PMCA4 targets. This analysis strongly implicated two proteins (Decorin, Rgs16) previously not known as PMCA4-regulated targets capable of modulating cell cycle. Both of these proteins are markedly increased upon PMCA4 gene deletion, which is reversed by expression of PMCA4a or PMCA4b (but not vector DNA) in P4KO cells. Interestingly, both these PMCA4 target proteins participate in the NFAT pathway. Decorin is an estab- lished natural onco-suppressive factor and a novel ligand for EGFR (37, 38), which is increased in VSMC in vivo after vascu- lar injury (31) and triggers a signaling cascade involving Akt and terminating in cyclin D1 induction (39). Decorin can inhibit EGFR, has been implicated in G1 phase arrest (by inducing p21 expression), and is deregulated in many types of human cancers (23, 24, 40). Decorin is also known to inhibit the VSMC cell cycle by triggering Myc degradation (41) and can also inhibit FosL1 (42). Under normal conditions FosL1 and Jun dimerize to form AP-1, which can induce the NFAT family of transcrip- tion factors (43), leading to cyclin D1 expression (26). Impor- tant confirmation of our in vitro microarray and Western blot results regarding Decorin included the novel finding that the decrease in Decorin expression observed in injured carotid arteries of P4WT mice is lost in P4KO mice, which manifest significantly higher levels of Decorin expression after injury. Rgs16, the other putative PMCA4 target identified here, is known to block cell cycle progression by inhibiting the MAPK and Akt pathways and is also modulated by the NFAT pathway. Rgs16 overexpression in MCF7 breast cancer cells inhibits EGFR-mediated proliferation and Akt phosphorylation, whereas shRNA-mediated extinction of Rgs16 augments cell growth (44). Transient as well as stable Rgs16 expression in CHO cells can attenuate IP3 production and p38 MAPK acti- vation by platelet-activating factor receptor (45). In T cells, PKC activation induces TNF␣, which increases Rgs16 expres- sion. Importantly, the calcineurin/NFAT pathway can repress this effect on Rgs16 (27). Our results suggest that PMCA4a (supplemental Fig. S8) as well as PMCA4b (supplemental Fig. S9) function to limit Decorin and Rgs16 expression (as well as increasing NFATc3 and cyclin D1 expression) and that PMCA4a may additionally function via inhibiting AP-2␤ expression (which de-represses c-Myc and induces EGFR expression), whereas PMCA4b may work via decreasing the CDK4 inhibitor p15, allowing cyclin D1 to activate CDK4. It is indeed surprising that although PMCA4a and PMCA4b proteins show very different expression patterns in quiescent and injured carotid arteries in vivo, both these splice variants rescue the G1 arrest prevalent in proliferating primary VSMC derived from the carotid arteries of P4KO mice. Potential explanations include the following. (i) Our rescue of P4KO cells was with the PMCA4x/a splice variant, whereas the PMCA4a splice variant actually expressed in quiescent carotid artery media may be PMCA4z/a. RNA-Seq of injured and uninjured carotid arteries would be required to explore this possibility as would the cloning and rescue by a PMCA4z/a (and PMCA4z/b) splice variant in vitro. (ii) The P4KO mice we have employed express “non-functional” PMCA4a and PMCA4b. As such, endogenous non-functional PMCA4b proteins may form mul- timeric complexes with exogenous PMCA4a enabling the non- functional PMCA4b molecules to regain normal Ca2ϩ efflux activity within complexes functionally defined by the protein- protein interactions of PMCA4b. In this scenario, although P4KO cells were electroporated with PMCA4a, the cells were able to regenerate “functional” PMCA4b pumps associated with a normal Ca2ϩ efflux activity. (iii) Electroporation of PMCA4a into de-differentiated, proliferating P4KO VSMC in culture may not mimic differentiated, quiescent VSMC in vivo, where a balance between PMCA4a and PMCA4b expression levels might be the critical factor driving the differentiated, non-proliferative state. In this explanation, expressing PMCA4a alone in de-differentiated, proliferating VSMC may not be enough to regain the quiescent, non-proliferative state. To achieve the latter, a fine balance in the expression levels and possibly differential subcellular localization of the two splice variants of PMCA4a and PMCA4b might be what is required. The eukaryotic cell cycle is known to be regulated by Ca2ϩ signals at various check points within the cycle (for review, see Ref. 1). Both voltage- and ligand-gated Ca2ϩ transporters are known to control cell cycle checkpoints by modulating intra- cellular Ca2ϩ signals or by hitherto unknown mechanisms involving macromolecular complexes with growth factor receptors, cell adhesion molecules, or cytoplasmic regulatory proteins (1). The latter may or may not depend on the Ca2ϩ transport function of these transporters but do involve the par- ticipation of their non-catalytic domains in signaling cascades that modulate cell cycle checkpoints. For example, the voltage- gated potassium channel subunit hERG1 (Kcnh2 or Kv11.1) regulates proliferation by a mechanism involving its recruit- ment into multiprotein membrane complexes including integ- rins and growth factor receptors (46). Other examples where Ca2ϩ channel domains not involved in the Ca2ϩ transport func- tion regulate transcription via a protein signaling cascade include Cav1.2 (Cacna1c), the L-type Ca2ϩ channel 250-kDa ␣1C subunit that forms the Ca2ϩ transporting transmembrane pore. An endogenously cleaved 15-kDa carboxyl-terminal frag- ment of Cav1.2 (calcium channel-associated transcription reg- ulator (or CCAT)) translocates to the nucleus, binds to a nuclear protein, associates with endogenous promoters, and regulates the expression of a wide variety of endogenous genes including the bone morphogenetic protein inhibitor follistatin- like-1 (47, 48) protein and thus de-represses stem cell quies- cence (45, 46). In a separate study involving the point mutation of the Cav1.2 calmodulin binding domain, it was shown that Ca2ϩ /calmodulin binding to Cav1.2 enables specific activation of the Ras/MAPK pathway, CREB activation, and CREB-driven transcription (47). Importantly, it was discovered that the abil- ity of Cav1.2 to generate a sustained [Ca2ϩ ]i rise was not needed for it to activate the Ras/MAPK/CREB pathway leading to gene transcription (47). PMCA4 Efflux Activity Regulates Cell Cycle in VSMC MARCH 7, 2014•VOLUME 289•NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 7229 atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
  • 11. Further studies focused on elucidating the molecular mech- anisms that allow PMCA4 to regulate the VSMC cell cycle are needed. The current study supplies the impetus for researchers examining PMCAs in other tissue systems to explore the roles of PMCA4a and PMCA4b in other cellular processes. The only other known example of physiological consequences of a PMCA4a/PMCA4b switch in vivo is the observed increase in relative PMCA4a splice variant expression (and decreased rel- ative PMCA4b levels) during functional sperm maturation (49). In that model of differentiation, there was a steady shift from predominantly PMCA4b expression in the testis, caput, and corpus epididymis to predominantly PMCA4a expression in the cauda epididymis. It is interesting to note that differentia- tion (associated with lack of cell proliferation) appears to require an increase in the relative level of PMCA4a expression in this model of sperm maturation. This correlates with the observed decrease in relative PMCA4a expression in prolifer- ating VSMC in vivo reported here and is reminiscent of the 2.5-fold reduction in rate of cell proliferation resulting from transient overexpression of PMCA1a splice variant in mouse VSMC (10) as well as the high relative proportion of PMCA4a variant in bladder smooth muscle cells (SMCs) (quiescent SMCs) and the roughly equal relative proportions of PMCA4a and PMCA4b in uninjured carotid artery SMCs (quiescent VSMC). REFERENCES 1. Becchetti, A. (2011) Ion channels and transporters in cancer. 1. Ion chan- nels and cell proliferation in cancer. Am. J. Physiol. Cell Physiol. 301, C255–C265 2. Lipskaia, L., and Lompré, A. M. (2004) Alteration in temporal kinetics of Ca2ϩ signaling and control of growth and proliferation. Biol. Cell 96, 55–68 3. Carafoli, E., and Brini, M. (2000) Calcium pumps. Structural basis for and mechanism of calcium transmembrane transport. Curr. Opin. Chem. Biol. 4, 152–161 4. Strehler, E. E., and Zacharias, D. A. (2001) Role of alternative splicing in generating isoform diversity among plasma membrane calcium pumps. Physiol. Rev. 81, 21–50 5. Keeton, T. P., Burk, S. E., and Shull, G. E. (1993) Alternative splicing of exons encoding the calmodulin-binding domains and C termini of plasma membrane Ca2ϩ -ATPase isoforms 1, 2, 3, and 4. J. Biol. Chem. 268, 2740–2748 6. Santiago-García, J., Mas-Oliva, J., Saavedra, D., and Zarain-Herzberg, A. (1996) Analysis of mRNA expression and cloning of a novel plasma mem- brane Ca2ϩ -ATPase splice variant in human heart. Mol. Cell. Biochem. 155, 173–182 7. Afroze, T., and Husain, M. (2000) c-Myb-binding sites mediate G1/S- associated repression of the plasma membrane Ca2ϩ -ATPase-1 promoter. J. Biol. Chem. 275, 9062–9069 8. Afroze, T., Yang, L. L., Wang, C., Gros, R., Kalair, W., Hoque, A. N., Mungrue, I. N., Zhu, Z., and Husain, M. (2003) Calcineurin-independent regulation of plasma membrane Ca2ϩ ATPase-4 in the vascular smooth muscle cell cycle. Am. J. Physiol. Cell Physiol. 285, C88–C95 9. Husain, M., Bein, K., Jiang, L., Alper, S. L., Simons, M., and Rosenberg, R. D. (1997) c-Myb-dependent cell cycle progression and Ca2ϩ storage in cultured vascular smooth muscle cells. Circ. Res. 80, 617–626 10. Husain, M., Jiang, L., See, V., Bein, K., Simons, M., Alper, S. L., and Rosen- berg, R. D. (1997) Regulation of vascular smooth muscle cell proliferation by plasma membrane Ca2ϩ -ATPase. Am. J. Physiol. 272, C1947–C1959 11. Gros, R., Afroze, T., You, X. M., Kabir, G., Van Wert, R., Kalair, W., Hoque, A. E., Mungrue, I. N., and Husain, M. (2003) Plasma membrane calcium ATPase overexpression in arterial smooth muscle increases vasomotor responsiveness and blood pressure. Circ. Res. 93, 614–621 12. Okunade, G. W., Miller, M. L., Pyne, G. J., Sutliff, R. L., O’Connor, K. T., Neumann, J. C., Andringa, A., Miller, D. A., Prasad, V., Doetschman, T., Paul, R. J., and Shull, G. E. (2004) Targeted ablation of plasma membrane Ca2ϩ -ATPase (PMCA) 1 and 4 indicates a major housekeeping function for PMCA1 and a critical role in hyperactivated sperm motility and male fertility for PMCA4. J. Biol. Chem. 279, 33742–33750 13. Hummler, E., Cole, T. J., Blendy, J. A., Ganss, R., Aguzzi, A., Schmid, W., Beermann, F., and Schütz, G. (1994) Targeted mutation of the CREB gene. Compensation within the CREB/ATF family of transcription factors. Proc. Natl. Acad. Sci. U.S.A. 91, 5647–5651 14. Schuh, K., Cartwright, E. J., Jankevics, E., Bundschu, K., Liebermann, J., Williams, J. C., Armesilla, A. L., Emerson, M., Oceandy, D., Knobeloch, K. P., and Neyses, L. (2004) Plasma membrane Ca2ϩ ATPase 4 is required for sperm motility and male fertility. J. Biol. Chem. 279, 28220–28226 15. You, X. M., Mungrue, I. N., Kalair, W., Afroze, T., Ravi, B., Sadi, A. M., Gros, R., and Husain, M. (2003) Conditional expression of a dominant- negative c-Myb in vascular smooth muscle cells inhibits arterial remodel- ing after injury. Circ. Res. 92, 314–321 16. Bolstad, B. M., Irizarry, R. A., Astrand, M., and Speed, T. P. (2003) A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics 19, 185–193 17. Du, P., Kibbe, W. A., and Lin, S. M. (2008) lumi. A pipeline for processing Illumina microarray. Bioinformatics 24, 1547–1548 18. Smyth, G. K. (2004) Linear models and empirical bayes methods for as- sessing differential expression in microarray experiments. Stat. Appl. Genet Mol. Biol. 3, Article3 19. Reiner, A., Yekutieli, D., and Benjamini, Y. (2003) Identifying differentially expressed genes using false discovery rate controlling procedures. Bioin- formatics 19, 368–375 20. Xie, J., Jan, C., Stoilov, P., Park, J., and Black, D. L. (2005) A consensus CaMK IV-responsive RNA sequence mediates regulation of alternative exons in neurons. RNA 11, 1825–1834 21. Oceandy, D., Stanley, P. J., Cartwright, E. J., and Neyses, L. (2007) The regulatory function of plasma-membrane Ca2ϩ -ATPase (PMCA) in the heart. Biochem. Soc. Trans. 35, 927–930 22. Schuh, K., Uldrijan, S., Gambaryan, S., Roethlein, N., and Neyses, L. (2003) Interaction of the plasma membrane Ca2ϩ pump 4b/CI with the Ca2ϩ / calmodulin-dependent membrane-associated kinase CASK. J. Biol. Chem. 278, 9778–9783 23. Ständer, M., Naumann, U., Wick, W., and Weller, M. (1999) Transform- ing growth factor-␤ and p-21. Multiple molecular targets of decorin-me- diated suppression of neoplastic growth. Cell Tissue Res. 296, 221–227 24. Hu, Y., Sun, H., Owens, R. T., Wu, J., Chen, Y. Q., Berquin, I. M., Perry, D., O’Flaherty, J. T., and Edwards, I. J. (2009) Decorin suppresses prostate tumor growth through inhibition of epidermal growth factor and andro- gen receptor pathways. Neoplasia 11, 1042–1053 25. Iozzo, R. V., Moscatello, D. K., McQuillan, D. J., and Eichstetter, I. (1999) Decorin is a biological ligand for the epidermal growth factor receptor. J. Biol. Chem. 274, 4489–4492 26. Karpurapu, M., Wang, D., Van Quyen, D., Kim, T. K., Kundumani-Srid- haran, V., Pulusani, S., and Rao, G. N. (2010) Cyclin D1 is a bona fide target gene of NFATc1 and is sufficient in the mediation of injury-induced vas- cular wall remodeling. J. Biol. Chem. 285, 3510–3523 27. Fong, C. W., Zhang, Y., Neo, S. Y., and Lin, S. C. (2000) Specific induction of RGS16 (regulator of G-protein signalling 16) mRNA by protein kinase C in CEM leukaemia cells is mediated via tumour necrosis factor ␣ in a calcium-sensitive manner. Biochem. J. 352, 747–753 28. Buettner, R., Kannan, P., Imhof, A., Bauer, R., Yim, S. O., Glockshuber, R., Van Dyke, M. W., and Tainsky, M. A. (1993) An alternatively spliced mRNA from the AP-2 gene encodes a negative regulator of transcriptional activation by AP-2. Mol. Cell. Biol. 13, 4174–4185 29. Gaubatz, S., Imhof, A., Dosch, R., Werner, O., Mitchell, P., Buettner, R., and Eilers, M. (1995) Transcriptional activation by Myc is under negative control by the transcription factor AP-2. EMBO J. 14, 1508–1519 30. Han, C. Y., Lim, S. C., Choi, H. S., and Kang, K. W. (2008) Induction of ErbB2 by ultraviolet A irradiation. Potential role in malignant transforma- tion of keratinocytes. Cancer Sci. 99, 502–509 PMCA4 Efflux Activity Regulates Cell Cycle in VSMC 7230 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289•NUMBER 10•MARCH 7, 2014 atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
  • 12. 31. Singh, N. K., Wang, D., Kundumani-Sridharan, V., Van Quyen, D., Niu, J., and Rao, G. N. (2011) 15-Lipoxygenase-1-enhanced Src-Janus kinase 2-signal transducer and activator of transcription 3 stimulation and monocyte chemoattractant protein-1 expression require redox-sensitive activation of epidermal growth factor receptor in vascular wall remodel- ing. J. Biol. Chem. 286, 22478–22488 32. Zhao, F., Bosserhoff, A. K., Buettner, R., and Moser, M. (2011) A heart- hand syndrome gene: Tfap2b plays a critical role in the development and remodeling of mouse ductus arteriosus and limb patterning. PLoS ONE 6, e22908 33. Frost, S. J., Simpson, D. J., and Farrell, W. E. (2001) Decreased proliferation and cell cycle arrest in neoplastic rat pituitary cells is associated with transforming growth factor-␤1-induced expression of p15/INK4B. Mol. Cell. Endocrinol. 176, 29–37 34. Armesilla, A. L., Williams, J. C., Buch, M. H., Pickard, A., Emerson, M., Cartwright, E. J., Oceandy, D., Vos, M. D., Gillies, S., Clark, G. J., and Neyses, L. (2004) Novel functional interaction between the plasma mem- brane Ca2ϩ pump 4b and the proapoptotic tumor suppressor Ras-associ- ated factor 1 (RASSF1). J. Biol. Chem. 279, 31318–31328 35. Kim, D. I., Lee, S. J., Lee, S. B., Park, K., Kim, W. J., and Moon, S. K. (2008) Requirement for Ras/Raf/ERK pathway in naringin-induced G1 cell-cycle arrest via p21WAF1 expression. Carcinogenesis 29, 1701–1709 36. Serrano, M., Lin, A. W., McCurrach, M. E., Beach, D., and Lowe, S. W. (1997) Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593–602 37. De Luca, A., Santra, M., Baldi, A., Giordano, A., and Iozzo, R. V. (1996) Decorin-induced growth suppression is associated with up-regulation of p21, an inhibitor of cyclin-dependent kinases. J. Biol. Chem. 271, 18961–18965 38. Moscatello, D. K., Santra, M., Mann, D. M., McQuillan, D. J., Wong, A. J., and Iozzo, R. V. (1998) Decorin suppresses tumor cell growth by activating the epidermal growth factor receptor. J. Clin. Invest. 101, 406–412 39. Liu, G., Hitomi, H., Hosomi, N., Shibayama, Y., Nakano, D., Kiyomoto, H., Ma, H., Yamaji, Y., Kohno, M., Ichihara, A., Itoh, H., and Nishiyama, A. (2011) Prorenin induces vascular smooth muscle cell proliferation and hypertrophy via epidermal growth factor receptor-mediated extracellular signal-regulated kinase and Akt activation pathway. J. Hypertens. 29, 696–705 40. Schönherr, E., Levkau, B., Schaefer, L., Kresse, H., and Walsh, K. (2001) Decorin-mediated signal transduction in endothelial cells. Involvement of Akt/protein kinase B in up-regulation of p21(WAF1/CIP1) but not p27(KIP1). J. Biol. Chem. 276, 40687–40692 41. Buraschi, S., Pal, N., Tyler-Rubinstein, N., Owens, R. T., Neill, T., and Iozzo, R. V. (2010) Decorin antagonizes Met receptor activity and down- regulates ␤-catenin and Myc levels. J. Biol. Chem. 285, 42075–42085 42. Tharaux, P. L., Chatziantoniou, C., Fakhouri, F., and Dussaule, J. C. (2000) Angiotensin II activates collagen I gene through a mechanism involving the MAP/ER kinase pathway. Hypertension 36, 330–336 43. Huang, H., Chang, E. J., Ryu, J., Lee, Z. H., Lee, Y., and Kim, H. H. (2006) Induction of c-Fos and NFATc1 during RANKL-stimulated osteoclast differentiation is mediated by the p38 signaling pathway. Biochem. Bio- phys. Res. Commun. 351, 99–105 44. Liang, G., Bansal, G., Xie, Z., and Druey, K. M. (2009) RGS16 inhibits breast cancer cell growth by mitigating phosphatidylinositol 3-kinase sig- naling. J. Biol. Chem. 284, 21719–21727 45. Zhang, Y., Neo, S. Y., Han, J., Yaw, L. P., and Lin, S. C. (1999) RGS16 attenuates G␣q-dependent p38 mitogen-activated protein kinase activa- tion by platelet-activating factor. J. Biol. Chem. 274, 2851–2857 46. Pillozzi, S., and Arcangeli, A. (2010) Physical and functional interaction between integrins and hERG1 channels in cancer cells. Adv. Exp. Med. Biol. 674, 55–67 47. Dolmetsch, R. E., Pajvani, U., Fife, K., Spotts, J. M., and Greenberg, M. E. (2001) Signaling to the nucleus by an L-type calcium channel-calmodulin complex through the MAP kinase pathway. Science 294, 333–339 48. Yucel, G., Altindag, B., Gomez-Ospina, N., Rana, A., Panagiotakos, G., Lara, M. F., Dolmetsch, R., and Oro, A. E. (2013) State-dependent signal- ing by Cav1.2 regulates hair follicle stem cell function. Genes Dev. 27, 1217–1222 49. Brandenburger, T., Strehler, E. E., Filoteo, A. G., Caride, A. J., Aumüller, G., Post, H., Schwarz, A., and Wilhelm, B. (2011) Switch of PMCA4 splice variants in bovine epididymis results in altered isoform expression during functional sperm maturation. J. Biol. Chem. 286, 7938–7946 PMCA4 Efflux Activity Regulates Cell Cycle in VSMC MARCH 7, 2014•VOLUME 289•NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 7231 atTORONTOWESTERNHOSPITALonJune2,2014http://www.jbc.org/Downloadedfrom
  • 13. 1 Online Supplementary Information TABLE S-I: PRIMERS FOR CLONING, GENOMIC PCR & QUANTITATIVE RT-PCR Name Sequence Comments EGFP-forward ACACCCTGGTGAACCGCAT 50 nM; Ta=60ºC EGFP-reverse CCATGATATAGACGTTGTGGCTGTT 50 nM; Ta=60ºC Microglobulin- forward GCTATCCAGAAAACCCCTCAA 50 nM; Ta=60ºC Microglobulin- reverse CATGTCTCGATCCCAGTAGACGGT 50 nM; Ta=60ºC PMCA4a forward TCAACACCTTGATGTAAAACTGGTTCCTAGC 300 nM; Ta=69ºC PMCA4a reverse GACGACTCTGATTGCCCACAGGAG 300 nM; Ta=69ºC PMCA4b reverse TATGGAACAGTTTGACGACTCTGATCTGAG 300 nM; Ta=69ºC PMCA4b forward AGTGACAAAGAGGACATCAGCAGGGATACTGAG 300 nM; Ta=69ºC PMCA4e forward ACACCTTGATGTAAAACTGGTTCCTAGCTC 300 nM; Ta=69ºC PMCA4e reverse AACAGTTTGACGACTCTGATTCTCACTCAC 300 nM; Ta=69ºC 5’UTR forward GCTACTCTGAAACTTATTGCACGAG Ta=60ºC Mid-cDNA reverse CTTCACTGGCCATGGGCTCAATG Ta=60ºC Mid-cDNA forward TCATTGAGCCCATGGCCAGTGAAG Ta=60ºC 3’UTR reverse CCACCGTCACATACAGATGG Ta=60 ºC PMCA4-exon-3- forward (Genomic PCR) AACGTGATACCTCCAAAAAGGCCCAAG Ta=65ºC Amplicon = 816 bp PMCA4-exon-4- reverse (Genomic PCR) AATTTTTGCTCCAGTTCGATGCGACTC Ta=65ºC Amplicon = 816 bp Table-S-I Legend: Note: f, forward; r, reverse; Ta, annealing temperature; EGFP, enhanced green fluorescent protein; all qRT-PCR primer pairs generated single amplicon peaks.
  • 14. 2 TABLE S-II: Genes Modulated in P4KO vs. P4WT cells (Early G1 Stage) Fold Change p-value Symbol Definition 47.5 2.39E-09 Casp1 Caspase 1 (ICE); down regulates c-myc; inflammatory. 39.4 6.38E-09 Rgs16 Regulator of G-protein signaling 16 (Rgs16); inhibits p38MAPK; NFAT suppresses Rgs16 21.2 6.38E-09 Gpnmb Glycoprotein (transmembrane) nmb (Gpnmb) 69.1 8.95E-09 Dcn Decorin; tumor suppressor; reduces tumor metastases 14.2 1.24E-08 Adm Adrenomedullin (Adm) 30.6 1.24E-08 Pdpn Podoplanin; transmembrane glycoprotein 20.9 1.49E-08 Lce1f Late cornified envelope 1F 40.2 1.54E-08 LOC100048554 PREDICTED: similar to monocyte chemoattractant protein-2 (MCP-2). 23.5 1.54E-08 Serpinb2 Serine (or cysteine) peptidase inhibitor, clade B, member 2 (Serpinb2) -6.6 3.81E-08 Rcan2 Regulator of Calcineurin 2 mRNA -3.6 4.02E-05 E2f2 E2F transcription factor 2 mRNA
  • 15. 3 TABLE S-III: Genes Known to be Implicated in G1 Phase Arrest (P4KO vs. P4WT cells in early G1 stage) Fold Change Symbol Definition 69.1 Dcn decorin (Dcn); tumor suppressor; reduces tumor metastases 5.2 Igf1 insulin-like growth factor 1; regulates MAPK/Akt pathway 5 Itga5 integrin alpha 5 (fibronectin receptor alpha) 4.2 Egfr epidermal growth factor receptor; tumor metastases 3.9 Fgf2 fibroblast growth factor 2 3.4 Ngf nerve growth factor 3.3 Dusp1 dual specificity phosphatase 1; ER stress induced apoptosis 2.8 Csf1 colony stimulating factor 1; induces NFATc1 degradation 2.1 Akap12 A kinase (PRKA) anchor protein 12; cell growth-related protein 2.1 Cdkn2A cyclin-dependent kinase inhibitor 2A; p16(INK4a); tumor suppressor -3.6 E2f2 E2F transcription factor 2; tumor suppressor -3.5 Mcm7 minichromosome maintenance deficient 7; chromosomal DNA replication factor -3.2 Mybl2 Myeloblastosis oncogene-like 2 (Mybl2); B-Myb; S and G2/M phase regulation -3 Notch1 Notch gene homolog 1; tumor suppressor effect in myeloid leukemia -2.8 Rbl1 retinoblastoma-like 1 (p107); cell cycle regulation -2.5 Pim1 proviral integration site 1; regulates S phase entry -2.2 Mtbp Mdm2, transformed 3T3 cell double minute p53 binding protein; Myc regulator -2.2 Cdkn1B cyclin-dependent kinase inhibitor 1B; p27 or Kip1; regulates cell cycle exit -2.1 Cdkn1C cyclin-dependent kinase inhibitor 1C (p57 or Kip2); regulator of cell cycle exit TABLE S-IV: NFAT Facilitator Proteins, NFAT pathway modulators and NFAT Targets (P4KO vs. P4WT in early G1 stage) Fold Change p-value Gene Name Definition 69.1 8.95E-09 Dcn Decorin (Dcn), mRNA. 39.4 6.38E-09 Rgs16 Regulator of G-protein signaling 16 (Rgs16), mRNA. -6.6 3.81E-08 Rcan2 Regulator of calcineurin 2 (Rcan2), transcript variant 1, mRNA. -1.8 0.002574978 Rcan1 Regulator of calcineurin 1 (Rcan1), transcript variant 2, mRNA. -3.6 4.02E-05 E2f2 E2F transcription factor 2 (E2f2), mRNA.
  • 16. 4 TABLE S-V: Genes Modulated in PMCA4a-Rescued P4KO at Late G1/S Stage (PMCA4a-rescued P4KO vs. P4KO) FoldChange p-value Symbol Definition -44.6 2.5E-10 Tfap2b AP-2β, Activating enhancer-binding protein 2 beta, transcript variant 2 (inhibits Myc transactivation, inhibits cell cycle) -15.5 4.7E-09 Angpt2 Angiopoietin 2 (disrupts vascular remodeling) -24.7 8.02E-09 Pcdh17 Protocadherin (cell-cell connections) -8.2 4.09E-08 Tfap2a AP-2α, Activating enhancer-binding protein 2 alpha (transcription activator/repressor; mutations cause branchio-oculo facial syndrome) -1.27 NS Dcn Decorin +1.1 NS Rgs16 Rgs16 +4.9 1.68E-07 Tnxb Tenascin XB (matrix maturation during wound healing) +7.8 2.15E-07 Tnfrsf11b Tumor necrosis factor receptor superfamily, member 11b (vascular calcification) +4.8 2.74E-07 Slitrk5 SLIT and NTRK-like family, member 5 (axonogenesis) +9.1 5.56E-07 Igf2bp3 Insulin-like growth factor 2 mRNA binding protein 3 (represses translation of insulin-like growth factor-2) +4.9 7.26E-07 Rragb Ras-related GTP binding B (Ras-homologous GTPases; signal transducers)
  • 17. 5 TABLE S-VI: Genes Modulated in PMCA4b-Rescued P4KO at Late G1/S Stage (P4b-rescued P4KO vs. P4KO) Fold Change p-value GeneName Definition 24.4 1.85E-25 Ptprd Protein tyrosine phosphatase, receptor type, D (Ptprd), transcript variant a, mRNA. 14.9 1.97E-21 Gjb3 Gap Junction protein beta 3 mRNA 32.0 2.03E-21 Anxa8 Annexin A8 mRNA -136.8 6.80E-24 Dkk3 Dickkopf homolog 3 (Xenopus laevis) (Dkk3), mRNA. -50.4 8.51E-24 Cdkn2b Cyclin-dependent kinase inhibitor 2B (p15; inhibits CDK4) (Cdkn2b), mRNA. -13.4 8.72E-24 Loxl1 Lysyl oxidase-like 1 (Loxl1), mRNA. -23.9 8.72E-24 Fgf10 Fibroblast growth factor 10 (Fgf10), mRNA. -22.3 1.37E-22 Pdgfra Platelet derived growth factor receptor, alpha polypeptide (Pdgfra), transcript variant 1, mRNA. -9.2 4.87E-22 Sema3f Sema domain, immunoglobulin domain (Ig), short basic domain, secreted, (semaphorin) 3 F (Sema3f), mRNA. -2.4 1.59E-13 Dcn Decorin (Dcn), mRNA. -18.5 3.07E-19 Rgs16 Regulator of G-protein signaling 16 (Rgs16), mRNA. TABLE S-VII: Genes Modulated in P4KO at Late G1/S Stage (P4KO vs. P4WT) Fold Change p-value GeneName Definition 39.4 1.20E-24 Thy1 Thymus cell antigen 1, theta (Thy1), mRNA. 33.0 7.76E-22 Ly6a Lymphocyte antigen 6 complex, locus A (Ly6a), mRNA. 64.7 1.12E-20 Serpinf1 Serine (or cysteine) peptidase inhibitor, clade F, member 1 (Serpinf1), mRNA. 5.2 2.00E-20 Igfbp4 Insulin-like growth factor binding protein 4 7.4 2.82E-20 Pcdh21 Protocadherin 21 (Pcdh21), mRNA. 9.3 3.24E-20 Dcn Decorin (Dcn), mRNA. 4.6 5.75E-20 Nnmt Nicotinamide N-methyltransferase (Nnmt), mRNA. 7.6 6.68E-20 Spon2 Spondin 2, extracellular matrix protein (Spon2), mRNA. 15.1 2.00E-18 Rgs16 Mus musculus regulator of G-protein signaling 16 (Rgs16), mRNA. -7.2 6.68E-20 Dlx1 Distal-less homeobox 1 mRNA -5.8 3.12E-17 Tuft1 Tuftelin 1 mRNA
  • 18. 6 TABLE S-VIII: Genes Modulated in PMCA4b-Rescued P4KO at Late G1/S Stage (PMCA4b-rescued P4KO vs. Vector-control P4KO) FoldChange p-value Symbol Definition 22.5 1.48E-24 Pacrg Park2 co-regulated mRNA 9.0 5.59E-21 Gpc1 Glypican 1 mRNA -28.0 1.29E-24 Ogn Osteoglycin (Ogn), mRNA. -15.3 1.29E-24 Ppap2b Phosphatidic acid phosphatase type 2B (Ppap2b), mRNA. -17.4 3.56E-24 Atp1b1 ATPase, Na+/K+ transporting, beta 1 polypeptide (Atp1b1), mRNA. -7.3 4.31E-22 Dusp1 Dual specificity phosphatase 1 (Dusp1), mRNA. -15.3 5.55E-22 Dcn Decorin (Dcn), mRNA. -22.5 5.69E-22 Cdkn2b Cyclin-dependent kinase inhibitor 2B (p15, inhibits CDK4) (Cdkn2b), mRNA. -7.7 8.59E-22 Loxl1 Lysyl oxidase-like 1 (Loxl1), mRNA. -7.9 9.58E-22 Ier3 Immediate early response 3 (Ier3), mRNA. -3.9 9.25E-14 Rgs16 Regulator of G-protein signaling 16 (Rgs16), mRNA.
  • 19. A PMCA4-b (alternatively spliced variant) = VASHSDSPLPSLETPV PMCA4-a (classical exon-20 of 181 nt) = AVTSPPVGNQSRQTVP PMCA4-e (novel exon-20 of 191 nt) = AVTSPPVGSE B Peptide used for PMCA4b Antibody = SHSDSPLPSLETPV Peptide used for PMCA4a/e Antibody = QTEAPLKRVRENMTQHLD
  • 20. Figure S-I. (A) Predicted amino acid sequences at the C-terminus of the cloned splice variants. Bold letters denote exon-20 or exon-21 encoded residues. (B) Sequences of peptides used to generate chicken polyclonal Abs against PMCA4b and PMCA4a/e splice variants. Bold letters denote residues conserved between mouse and rat PMCA4. (C) Schematic representation and relevant sequences of alternatively spliced PMCA genes. Lines show introns, rectangles are exons, and filled triangles are in-frame stop codons. Sequences at the carboxy tails of resulting splice variants are displayed. Bold letters show residues expressed as a result of exon inclusion or exclusion. NB: alternative splicing at site C of PMCA1 and PMCA3 is more complex than shown. The schematic emphasizes instead the similarities between ‘e’ variants of PMCA1, PMCA3 and PMCA4. AAA PMCA3e PMCA3a PMCA3b PMCA1b PMCA1a PMCA1 pre-mRNA poly A site Stop codon PMCA3 pre-mRNA VVFSSSTASTTVGSEW (mouse) VVFSSSTASTPVGSEW (rat) VVFSSSTASTTVGFEW (human) STASTTVGYPSGECIS (mouse) TSSSPGSPLHSLETSL (mouse) VTLSAAKPPSAAGSES (mouse) VTLSAAKPTSAAGSES (rat) AILSAANPTSAAGSES (human) AILSAANPTSAAGSES (dog) LSTATANTSPAPGSEST (tetraodon) AKPPSAAGNPGGESIP (mouse) FSSRPGSPLHSMETSL (mouse) SSAISSAVTSPPVGSE (mouse) SPSTSSAVTPPPVGSE (rat) PTTSVPAVSSPPMGSE (human) PTTSVPAVSSPPMGSE (chimp) TTSVPAAVSSPTLGSE (dog) AVTSPPVGNQSRQTVP (mouse) VASHSDSPLPSLETPV (mouse) PMCA4a PMCA4b PMCA4 pre-mRNA PMCA4e PMCA1e C
  • 21. M.W.ladder PMCA4+/+ C57/Bl6 PMCA4-/- 816 bp band specific for PMCA4+/+ A 130 kDa Anti-PMCA4a/e Anti-PMCA4b a b c d a b c dB Figure S-II. Validation of PMCA4a/e and PMCA4b specific antibodies (A) Genomic PCR using exon-3 and exon-4 specific primers confirmed that the Neyses lab-generated PMCA4- /- mouse hearts (in which exons-2 and -3 have been deleted) lack these PMCA4 exons from their genome. (B) Immunoblots of microsomal protein extracts made from (a) C57bl6 heart, (b) PMCA4-/- mouse heart, (c) C57bl6 stomach and (d) C57bl6 brain were probed with affinity purified chicken polyclonal antibodies raised against peptides specific for PMCA4b and PMCA4a/e. Note that the PMCA4a/e antibody did not give a band for normal mouse heart (lane a) presumably because of negligible PMCA4a expression in the heart.
  • 22. Relative Levels of PMCA4 splice variant (%) in LCM samples Bladder SMC Injured Medial SMC Remodeled Adventitial SMC 0 20 40 60 80 100 120 PMCA4b PMCA4a Uninjured Medial SMC Figure S-III. Laser capture microdissection (LCM) was used to obtain uninjured media (n=1), injured media (n=3) and remodeled adventitia regions (n=2) from mouse carotid artery cross-sections (8-10 wk old male; day-9 post injury). Bladder smooth muscle region (n=1) was used to show that the LCM protocol could demonstrate high relative PMCA4a abundance observed in Fig1A. These microdissected regions were used to isolate RNA, make cDNA and carry out qRT-PCR for microglobulin (housekeeping gene) and PMCA4a and PMCA4b to calculate microglobulin-normalized relative proportions of PMCA4a:4b splice variants in the various arterial regions.
  • 23. Figure S-IV. Optimization of Electroporation Parameters. maxGFP plasmid, mouse primary carotid artery smooth muscle cells, Lonza P1 Nucleofection Mix and Lonza 4D Nucleofector were used. CA-137 electroporation parameters: 67% efficiency Mean of 4 fields of view; > 100 cells counted; not much cell death CM-137 electroporation parameters: 100% efficiency Mean of 3 fields of view ~50 cells counted; significant cell death Negative Control: No electroporation parameters used; cells incubated with maxGFP plasmid in cuvette
  • 24. Figure S-V. Validation of electroporation of PMCA4 cDNA into VSMCs. EGFP-tagged human PMCA4b-∆-PDZ cDNA construct was electroporated into P4KO primary carotid artery smooth muscle cells using the Lonza 4D Nucleofector. Electroporated cells were seeded on cover slips, allowed to recover for 24 h, fixed with paraformaldehyde, washed with PBS, mounted on slides and used for confocal imaging. EGFP-huPMCA4b-∆-PDZ construct electroporated into P4KO cells Unelectroporated P4KO cells
  • 25. DNAladder PMCA4a electroporated Vector electroporated P4KOcells NoTemplate DNAladder PMCA4a specific amplicon band: RT-PCR primers designed to detect PMCA4 exon9-exon10 junction which is absent from P4KO cells Figure S-VI. RT-PCR validation of electroporation of PMCA4 cDNA into VSMCs. P4KO cells carry a genetic deletion whereby exon-9 has been removed and exon-8 splices to exon- 10 in mature PMCA4 mRNA. Mouse PMCA4a cDNA construct encodes exon-9 joined to exon-10. RT-PCR primers designed to detect this exon-9-exon-10 junction were used to screen cDNA made from P4KO cells, P4KO cells rescued with vector and P4KO cells rescued with mouse PMCA4a cDNA (cells were subjected to puromycin selection for 14 days after electroporation). Agarose gel electrophoresis showed exon-9-exon-10 junction only in those P4KO cells which received PMCA4a cDNA.
  • 26. Figure S-VII. PMCA4a/e, PMCA4b and cleaved Caspase-3 Western blots. Whole cell protein extracts made from P4WT, P4KO, P4KO+4a and P4KO+4b rescued primary carotid VSMCs were used for immunoblots probed with anti-PMCA4a/e, anti-PMCA4b or anti- cleaved Caspase-3 (apoptosis marker) antibodies. LiCor’s IR680 fluorescent dye conjugated secondary antibodies (anti-chicken for PMCA4a/e and PMCA4b and anti-rabbit for cleaved Caspase-3) were used and blots were imaged with the Odyssey fluorescent scanner (LiCor, NE). PMCA4a was overexpressed 1.5-fold in P4KO+4a cells while PMCA4b was overexpressed 1.8-fold in P4KO+4b cells as compared to P4KO cells. There was no cleaved Caspase-3 band visible which signifies absence of any apoptosis in these cells. PMCA4a/e PMCA4b P4WT P4KO P4KO+4a P4KO+4b Cleaved Caspase-3 17 kDa (expected band) P4WT P4KO P4KO+4a P4KO+4b Protein Ladder Protein Ladder
  • 27. Figure S-VIII. Schematic showing role of proteins modulated by PMCA4a overexpression in PMCA4 knockout cells. AP-2β represses Myc transactivation and also inhibits AP-2 α from binding to the EGFR promoter and enhancing proliferation. PMCA4a overexpression in PMCA4 knockout VSMC de-represses Myc activity and allows AP-2 α to induce EGFR expression which leads to enhanced proliferation. PMCA4a effects on Decorin and Rgs16 omitted for simplicity. plasma membrane AP-2 α ? ? AP-2 β PMCA4a PI3K p85 PI3K Akt Signaling Cell Cycle GRB2 SOS Ras Cyclin D1 GSK3 FoxO β -Catenin Retinoblastoma protein E2F2 repressor complexes MAPK Signaling EGFR EGFR is upregulated in VSMC in vivo following vascular injury ErbB2 (EGFR) promoter c-Myc EGFR AP-2 α
  • 28. plasma membrane PI3K p85 PI3K Akt Signaling Cell Cycle GRB2 SOS Ras MAPK Signaling p15 (inhibits Cyclin D1/Cdk4 and Cyclin D1/Cdk6 complexes) FosL1 AP-1 (FosL1 + Jun) NFAT Rgs16 Cyclin D1 GSK3 FoxO β -Catenin Retinoblastoma protein E2F2 repressor complexes Figure S-IX. Schematic showing role of proteins modulated by PMCA4b overexpression in PMCA4 knockout cells. PMCA4b overexpression in PMCA4 knockout VSMC represses Rgs16 and Decorin, which in turn regulates NFAT and Cyclin D1 expression in the early G1 phase of the cell cycle. PMCA4b EGFR is upregulated in VSMC in vivo following vascular injury EGFR DecorinPMCA4b ? ? ?? ? ? Cyclin D1 promoter NFAT