ArticlePDF Available

Binding and Inactivation of the Germ Cell-Specific Protein Phosphatase PP1 2 by sds22 During Epididymal Sperm Maturation

Authors:

Abstract and Figures

Testis- and sperm-specific protein phosphatase, PP1gamma2, is a key enzyme regulating sperm function. Its activity decreases during sperm maturation in the epididymis. Inhibition of PP1gamma2 leads to motility initiation and stimulation. Our laboratory is focused on identifying mechanisms responsible for the decline in PP1gamma2 activity during sperm motility initiation in the epididymis. Previously, using immuno-affinity chromatography, we showed that a mammalian homologue of yeast sds22 is bound to PP1gamma2 in motile caudal spermatozoa (Huang Z, et al. Biol Reprod 2002; 67:1936-1942). The objectives of this study were to determine: 1) stoichiometry of PP1gamma2-sds22 binding and 2) whether PP1gamma2 in immotile caput epididymal spermatozoa is bound to sds22. The enzyme from caudal and caput sperm extracts was purified by column chromatography. Immunoreactive PP1gamma2 and sds22 from both caudal and caput spermatozoa were found in the flow-through fraction of a DEAE-cellulose column. However, PP1gamma2 from caudal spermatozoa was inactive, whereas in caput spermatozoa it was active. The DEAE-cellulose flow-through fractions were next passed through a SP-sepharose column. Caudal sperm sds22 and PP1gamma2 coeluted in the gradient fraction. In contrast, caput sperm sds22 and PP1gamma2 were separated in the flow-through and gradient fractions, respectively. Further purification through a Superose 6 column showed that PP1gamma2-sds22 complex from caudal sperm was 88 kDa in size. Caput sperm sds22 and PP1gamma2 eluted at 60 kDa and 39 kDa, respectively. SDS-PAGE of these purified fractions revealed that in caudal sperm, the 88-kDa species is composed of sds22 (43 kDa) and PP1gamma2 (39 kDa), suggesting a 1:1 complex between these two proteins. PP1gamma2 bound to sds22 in this complex was inactive. Caput sperm sds22 eluting as a 60-kDa species was found to be associated with a 17-kDa protein (p17). This suggests that dissociation of sds22 from p17 or some other posttranslational modification of sds22 is required for its binding and inactivation of PP1gamma2. Studies are currently underway to determine the mechanisms responsible for development of sds22 binding to PP1gamma2 during epididymal sperm maturation.
Content may be subject to copyright.
1572
BIOLOGY OF REPRODUCTION 69, 1572–1579 (2003)
Published online before print 25 June 2003.
DOI 10.1095/biolreprod.103.018739
Binding and Inactivation of the Germ Cell-Specific Protein Phosphatase PP1g2
by sds22 During Epididymal Sperm Maturation
1
Sanjay Mishra, Payaningal R. Somanath, Zaohua Huang, and Srinivasan Vijayaraghavan
2
Biological Sciences Department, Kent State University, Kent, Ohio 44242
ABSTRACT
Testis- and sperm-specific protein phosphatase, PP1g2, is a
key enzyme regulating sperm function. Its activity decreases
during sperm maturation in the epididymis. Inhibition of PP1g2
leads to motility initiation and stimulation. Our laboratory is
focused on identifying mechanisms responsible for the decline
in PP1g2 activity during sperm motility initiation in the epidid-
ymis. Previously, using immuno-affinity chromatography, we
showed that a mammalian homologue of yeast sds22 is bound
to PP1g2 in motile caudal spermatozoa (Huang Z, et al. Biol
Reprod 2002; 67:1936–1942). The objectives of this study were
to determine: 1) stoichiometry of PP1g2-sds22 binding and 2)
whether PP1g2 in immotile caput epididymal spermatozoa is
bound to sds22. The enzyme from caudal and caput sperm ex-
tracts was purified by column chromatography. Immunoreactive
PP1g2 and sds22 from both caudal and caput spermatozoa were
found in the flow-through fraction of a DEAE-cellulose column.
However, PP1g2 from caudal spermatozoa was inactive, where-
as in caput spermatozoa it was active. The DEAE-cellulose flow-
through fractions were next passed through a SP-sepharose col-
umn. Caudal sperm sds22 and PP1g2 coeluted in the gradient
fraction. In contrast, caput sperm sds22 and PP1g2 were sepa-
rated in the flow-through and gradient fractions, respectively.
Further purification through a Superose 6 column showed that
PP1g2-sds22 complex from caudal sperm was 88 kDa in size.
Caput sperm sds22 and PP1g2 eluted at 60 kDa and 39 kDa,
respectively. SDS-PAGE of these purified fractions revealed that
in caudal sperm, the 88-kDa species is composed of sds22 (43
kDa) and PP1g2 (39 kDa), suggesting a 1:1 complex between
these two proteins. PP1g2 bound to sds22 in this complex was
inactive. Caput sperm sds22 eluting as a 60-kDa species was
found to be associated with a 17-kDa protein (p17). This sug-
gests that dissociation of sds22 from p17 or some other post-
translational modification of sds22 is required for its binding
and inactivation of PP1g2. Studies are currently underway to
determine the mechanisms responsible for development of
sds22 binding to PP1g2 during epididymal sperm maturation.
gamete biology, phosphatases, signal transduction, sperm matu-
ration, testis
INTRODUCTION
We discovered that testis-specific protein phosphatase,
PP1
g
2, is the predominant protein phosphatase in sperma-
tozoa [1, 2]. High PP1
g
2 activity results in low motility,
1
Supported by NIH grant RO1 HD38520.
2
Correspondence: Srinivasan Vijayaraghavan, Biological Sciences Depart-
ment, Kent State University, Kent, Ohio 44242-0001. FAX: 330 672 3713;
e-mail: svijayar@kent.edu
Received: 29 April 2003.
First decision: 21 May 2003.
Accepted: 17 June 2003.
Q 2003 by the Society for the Study of Reproduction, Inc.
ISSN: 0006-3363. http://www.biolreprod.org
and low PP1
g
2 activity is associated with vigorous motility
[2]. This decline in PP1
g
2 activity during sperm maturation
is not due to a decline in the amount of the enzyme but to
a decrease in its catalytic activity. We found that the protein
phosphatase inhibitors, okadaic acid and calyculin A, ini-
tiate and stimulate motility of epididymal spermatozoa. The
enzyme PP1
g
2 is involved in the regulation of spermatozoa
from all mammalian species studied so far, including hu-
man [1–3].
At least four different somatic cell serine/threonine phos-
phatase types, PP1, PP2A, PP2B, and PP2C, are known [4
6]. Four catalytic subunit isoforms of PP1, encoded by three
genes, are PP1
a
, PP1
g
1, PP1
g
2, and PP1
d
[6–8]. PP1
g
1
and PP1
g
2 are alternatively spliced isoforms generated
from a single gene [6–8]. Although PP1
g
1 is ubiquitous,
PP1
g
2 is present only in germ cells and spermatozoa [8,
9]. These two PP1 variants are identical in all respects ex-
cept that PP1
g
2 has a unique 21 amino acid carboxy ter-
minus extension. It is noteworthy that the carboxy terminus
extension of PP1
g
2, which is lacking in somatic cell PP1
isoforms, is conserved in all mammalian spermatozoa we
have studied so far, suggesting sperm-specific roles for this
amino acid sequence domain. Disruption of the PP1
g
gene
in mice causes sterility in males because of arrest of sper-
matogenesis at the spermatid stage [9]. This observation
shows that other isoforms of PP1 can compensate for the
lack of PP1
g
1 in somatic cells and for the lack of PP1
g
2
in germ cells until the final stages of spermatogenesis. Tes-
tis-specific PP1
g
2 is indispensable in the final stages of
spermatogenesis.
Our studies are focused on understanding mechanisms
regulating PP1
g
2 and determination of the basis for the
decline in PP1
g
2 activity during epididymal sperm matu-
ration. Protein phosphatases, in general, are regulated by
their binding and targeting proteins [10, 11]. Following
identification of PP1
g
2, we speculated that one or more
somatic cell protein regulators of PP1 might be present in
spermatozoa. Our initial efforts focused on proteins I1 and
I2, which are ubiquitous PP1 inhibitors found in all somatic
cells [11]. Surprisingly, sperm do not contain any detectable
I1 or I2 suggesting that mechanisms regulating PP1
g
2
could be unique (unpublished data, [2]).
We first used immunoaffinity chromatography to isolate
PP1
g
2 and its associated proteins [12]. Antibodies to PP1
g
2
immunoprecipitate sds22, a mammalian homologue of the
yeast PP1-binding protein [12]. Sds22 is a prototypic mem-
ber of a family of proteins containing leucine-rich repeats
[13]. Human sds22 contains 11 repeats of a leucine-rich 22
amino acid segment. The leucine-rich repeat is a structural
motif used in such diverse molecular recognition processes
as signal transduction, cell adhesion, cell development and
RNA processing [13]. In caudal epididymal spermatozoa,
sds22-bound PP1
g
2 is catalytically inactive [12].
In the immunoprecipitation study, PP1
g
2 antibodies
1573
REGULATION OF PP1
g
2 BY sds22
were found to bind to only a portion of the total PP1
g
2in
sperm extracts [12]. Coimmunoprecipitation suggested, but
did not prove, that sds22 was bound to PP1
g
2. Stoichi-
ometry of PP1
g
2 binding to sds22 was not known. It was
also necessary to determine why PP1
g
2 activity is lower in
caudal, compared with caput spermatozoa. Therefore, the
present study was undertaken to purify PP1
g
2 and sds22,
by column chromatography, from both caput and caudal
spermatozoa and to understand how sds22 may regulate
PP1
g
2 activity during sperm maturation and motility initi-
ation. These studies show that in caudal spermatozoa, a
proportion of PP1
g
2 present as a heterodimer with sds22,
is inactive. In caput epididymal spermatozoa, sds22 is not
bound to PP1
g
2. Caput sperm sds22 may be bound to a
17-kDa protein, p17. A significant proportion of caput
sperm PP1
g
2 is therefore in its catalytically active state.
These studies suggest that dissociation of sds22 from p17
or some other modification of sds22 is followed by its bind-
ing and inactivation of PP1
g
2. Because inhibition of PP1
g
2
activity induces motility, binding and inactivation of PP1
g
2
by sds22 during sperm maturation in the epididymis is pre-
sumably one of the biochemical events responsible for mo-
tility initiation.
MATERIALS AND METHODS
Preparation of Sperm Extracts
Testes with intact tunica from mature bulls were obtained from a local
slaughterhouse. Spermatozoa were isolated from caudal and caput epidid-
ymis and washed as previously described [3] in buffer A (10 mM Tris-
HCl [pH 7.2] containing 120 mM NaCl, 10 mM KCl, 5 mM MgSO
4
).
Sperm pellets were suspended in a homogenization buffer (buffer B) (10
mM Tris [pH 7.2] containing 1 mM EDTA, 1 mM EGTA, 10 mM ben-
zamidine-HCl, 1 mM PMSF, 0.01 mM N-p-tosyl-
L
-phenylalanine chloro-
methyl ketone [TPCK], and 5 mM
b
-mercaptoethanol). The sperm sus-
pension was sonicated with three 5-sec bursts of a Biosonic II sonicator
(Bronwell Scientific, Rochester, NY) at maximum setting. The sperm son-
icate was then centrifuged at 16000
3
g for 10 min. The 16 000
3
g
supernatants were supplemented with 10% glycerol and stored at
2
20
8
C
until further use for column procedures.
Column Chromatography
All column procedures were conducted at 4
8
C. Total protein in caudal
and caput sperm extracts and fractions obtained from column chromatog-
raphy was measured with Coomassie brilliant blue dye reagent (Bio-Rad
laboratories, Hercules, CA) as described previously [14]. Caudal sperm
extract (50 ml prepared from 5
3
10
10
spermatozoa in buffer B) was
passed through a DEAE-cellulose (0.5cm
3
13 cm) column pre-equili-
brated with buffer C (buffer B with 0.05 M KCl and additional protease
inhibitors-pepstatin A [1
m
g/ml], aprotinin [2
m
g/m], and leupeptin [0.5
m
g/ml]). The column was washed with 20 ml of buffer C followed by
elution with a linear gradient of 0.05–0.65 M KCl in buffer C. Flow-
through and gradient fractions (0.185–0.35 M KCl) containing PP1
g
2 ac-
tivity and PP1
g
2 and/or sds22 immunoreactivity were pooled separately.
The pooled fractions were concentrated to 12.5 ml using Centricon-10
filter (Millipore Corp., Bedford, MA). A SP-sepharose column (5 ml pre-
packed, Pharmacia, Piscataway, NJ) was pre-equilibrated with buffer C.
The DEAE-cellulose flow-through fraction was applied to this SP-sephar-
ose column. The column was then washed with 10 ml buffer C followed
by elution with a linear gradient of 0.05–0.65 M KCl in buffer C. Fractions
containing sds22 and PP1
g
2 were pooled, concentrated to 1 ml, and ap-
plied in five batches to Superose 6 (24 ml, prepacked high-resolution
FPLC, Pharmacia, Piscataway, NJ) column pre-equilibrated with buffer C.
The elution was performed with buffer C. Immunoreactive fractions of all
five batches were pooled, concentrated to 1 ml, and applied to Mono S (1
ml, prepacked high-resolution FPLC, Pharmacia, Piscataway, NJ) column.
The column was washed with 5 ml buffer C followed by elution with a
linear gradient of 0.05–0.65 M KCl in buffer C. Immunoreactive fractions
(0.140–0.215 M KCl) containing sds22-PP1
g
2 complex were pooled, and
concentrated to 0.5 ml. For purification of sds22 and PP1
g
2 from caput
epididymal spermatozoa, we followed the same protocol as described
above for caudal sperm, except that 50 ml of extract derived from 1
3
10
10
spermatozoa was used. The final step for purifying caput sds22-p17
complex through a heparin sepharose (1 ml, prepacked Pharmacia, Pis-
cataway, NJ) column was similar to the protocol used for Mono S column
chromatography. Purified and concentrated caudal sds22-PP1
g
2 complex
and caput sds22 and free PP1
g
2 fractions were stored at
2
20
8
C until
further use.
Protein Phosphatase Assay
Preparation of radiolabeled phosphorylase a, and its use as a substrate
for measurement of PP1 activity is based on a protocol described previ-
ously [3]. The substrate, aliquots from sperm extracts, and fractions ob-
tained after column chromatography were incubated (in a total volume of
40
m
l) at 30
8
Cwith1mMMn
2
1
and with or without inhibitors for 10
min. At the end of this incubation, the reaction was terminated with 180
m
l of 20% trichloro acetic acid, after which the tubes were centrifuged for
5 min at 12000
3
g at 4
8
C. The supernatants were analyzed for
32
PO
4
released from phosphorylase a. One unit of enzyme activity was defined
as the amount of enzyme that catalyzed the release of 1 nmol of
32
PO
4
/
min under conditions of the enzyme assay. This assay is specific for the
protein phosphatases PP1 and PP2A [3].
SDS-PAGE and Western Blot Analysis
Sperm extracts (30–50
m
g) and aliquots from fractions obtained from
column chromatography (2–10
m
g) were separated by SDS-PAGE through
12% acrylamide slab gels based on the protocol of Laemmli [15]. One
milliliter of soluble extracts (prepared from 10
9
cells) contain approxi-
mately 1 mg/ml and 1.9 mg/ml protein for caudal and caput sperm, re-
spectively. After electrophoresis, proteins were electrophoretically trans-
ferred to Immobilon-P, PVDF membrane (Millipore Corp.). Nonspecific
protein binding sites on the membrane were blocked with 5% nonfat dry
milk in Tris buffered saline (TBS; 25 mM Tris-HCl, pH 7.4, 150 mM
NaCl). The blots were washed twice for 15 min each with TBS containing
0.1% Tween 20 (TTBS) and then incubated with anti-PP1
g
2 (1:5000) or
anti-sds22 (1:2000). Both antibodies were commercially prepared (Zymed
Laboratories, San Francisco, CA) by using a synthetic carboxyl terminus
extension of PP1
g
2 (22 amino acids of the carboxyl terminus) and amino
acid residues 329–342 of sds22 as antigens. Antibodies were affinity pu-
rified with the synthetic peptides conjugated to a sulfo-link column (Pierce,
Rockford, IL). After washing, the blots were incubated with the appropri-
ate secondary antibody conjugated to horseradish peroxidase at 1:2000
dilution for 1 h at room temperature. Blots were washed with TTBS twice
15 min each and four times 5 min each. Blots were then developed with
an enhanced chemiluminescence kit (Amersham, Piscataway, NJ). Protein
composition of purified samples was also assessed by Coomassie blue or
silver staining following SDS-PAGE. Coomassie staining was done with
0.1% Coomassie brilliant blue R250 in 40% methanol and 10% acetic
acid. Silver staining was performed using a commercial kit (Bio-Rad Lab-
oratories) as per instructions provided by the manufacturer.
RESULTS
Purification of PP1
g
2 and sds22 From Caudal
Epididymal Spermatozoa
We used DEAE-cellulose, SP-sepharose, high-resolution
Superose 6, and Mono S columns for purification of PP1
g
2
and sds22 from caudal spermatozoa. Column fractions were
analyzed for PP1
g
2 and sds22 immunoreactivity and for
PP1
g
2 catalytic activity. Following at least three pilot pu-
rification runs, 50 ml of sperm extracts prepared from 5
3
10
10
bovine caudal epididymal spermatozoa were used to
isolate enough PP1
g
2 and sds22 for biochemical studies.
Specific activity of PP1
g
2 in the pooled extracts was 1.71
U/mg protein. A summary of the purification steps is shown
in Table 1.
The sperm extract was first passed through a DEAE-
cellulose column. A portion of the enzyme was found in
the flow-through fraction, and the rest was absorbed in the
column. In addition to PP1
g
2, the DEAE-cellulose flow-
through fraction also contained immunoreactive sds22. The
enzyme in the DEAE-cellulose flow-through fraction was
inactive whereas that released from the column by a salt
1574
MISHRA ET AL.
TABLE 1. Summary of sds22-PP1
g
2 purification from caudal epididymal sperm extracts.*
Purification step Protein (mg) Total activity (U)
Specific activity
(U/mg protein)
Caudal sperm extract 152 260 1.71
DEAE-cellulose flow-through 65 Inactive
DEAE-cellulose gradient (0.185–0.350 M KC1)
(PP1
g
2 without sds22) 71 155 2.18
SP sepharose gradient (0.125–0.275 M KC1)
of DEAE-cellulose flow-through 21 Inactive
Superose 6 of SP-sepharose gradient 1.05 Inactive
Mono S gradient
(0.140–0.215 M KC1) of SP-sepharose gradient 0.525 Inactive
* Table summarizes purification of three batches of 50 ml each of caudal sperm extract as outlined under
Materials
and Methods
. The total number of cells corresponding to 153 mg protein is 1.5 3 10
11
. Activity of PP1
g
2 in caudal
sperm extracts and fractions obtained after column procedures was measured in the presence of 1 mM Mn
2
1
.
FIG. 1. Western blot analysis showing caudal sperm PP1g2-sds22 com-
plex during different stages of purification through column chromatogra-
phy. The same blot was probed with PP1g2 and sds22 antibodies. Lane
1 is caudal extract (30 mg); lane 2 is DEAE-cellulose flow-through fraction;
lane 3 is SP-sepharose flow-through of DEAE-cellulose flow-through frac-
tion; and lane 4 is SP-sepharose gradient of DEAE-cellulose flow-through
fraction. In lanes 2–4, 25 ml each of the concentrated column fractions
as described in
Materials and Methods
were loaded.
FIG. 2. Molecular weight determination of the caudal sperm PP1g2-
sds22 complex. A) Western blot of the fractions (25 ml each) showing
PP1g2-sds22 complex eluted in the fractions 22–29 in Superose 6. B)
Elution profile of PP1g2-sds22 complex from caudal sperm extracts in
Superose 6. PP1g2-sds22 complex eluted in Superose 6 at 88 kDa. Cal-
ibration of the column for its void volume was performed by running blue
dextran. Standardization of the column was done by resolving a mixture
of aldolase (5 mg/ml), albumin (5 mg/ml), ovalbumin (10 mg/ml), and
chymotrypsinogen (10 mg/ml).
gradient (0.185–0.35 M KCl) had a specific activity of 2.18
U/mg protein (Table 1). Details of purification of PP1
g
2
released from the DEAE-cellulose column are not outlined
in this report. The focus in this report is on PP1
g
2 asso-
ciated with sds22. The DEAE-cellulose flow-through frac-
tion containing PP1
g
2 and sds22 was concentrated and
passed through a SP-sepharose column. Here PP1
g
2 and
sds22 were bound in the column. All the bound PP1
g
2 and
sds22 were released by a salt gradient between 0.125–0.275
M KCl. The presence of sds22 and PP1
g
2 in the DEAE-
cellulose and SP-sepharose column fractions is shown in
Figure 1 (lanes 2 and 4).
The SP-sepharose salt gradient fractions containing both
PP1
g
2 and sds22 were concentrated and passed through a
Superose 6 column. In the Superose 6 column, sds22 and
PP1
g
2 coeluted as a 88-kDa species in fractions 26, 27,
and 28 (Fig. 2). Coelution of sds22 and PP1
g
2 through
these three columns, DEAE-cellulose, SP-sepharose, and
Superose 6, suggested that they could be complexed. To
confirm their association, we further purified sds22 and
PP1
g
2 containing fractions through a Mono S column.
Sds22 and PP1
g
2 were absorbed on the Mono S column
and were coeluted in a 0.140–0.215 M KCl gradient. These
fractions containing PP1
g
2 and sds22 were pooled and con-
centrated and analyzed by SDS-PAGE and Western blot.
Coomassie blue and silver staining following SDS-PAGE
(Fig. 3A) showed that the complex eluting from Mono S
column contains two major protein bands at 43 kDa and
39 kDa. These two protein bands were confirmed to be
1575
REGULATION OF PP1
g
2 BY sds22
FIG. 3. Caudal sperm PP1g2 is bound to protein sds22. A) Coomassie
blue (lane 1) and silver stained gels (lane 2) showing PP1g2 and sds22
after SDS-PAGE of purified PP1g2-sds22 complex obtained after Mono S
column chromatography (2.5 mg). Silver staining was performed on the
same gel following Coomassie blue. Lane 3 shows protein molecular
weight markers. B) Western blots showing immunoreactive PP1g2 (left
panel) and sds22 (right panel) in caudal extracts (lane 1: 30 mg protein)
and purified PP1g2-sds22 complex (lane 2: 2.5 mg protein).
FIG. 4. Western blot analysis showing caput sperm PP1g2 and sds22
during different stages of purification through column chromatography.
Blot was probed with both PP1g2 and sds22 antibodies. Lane 1 is caput
extract (50 mg); lane 2 is DEAE-cellulose flow-through fraction; lane 3 is
SP-sepharose flow-through of DEAE-cellulose flow-through fraction; and
lane 4 is SP-sepharose gradient of DEAE-cellulose flow-through fraction.
In lanes 2–4, 25 ml each of the concentrated column fractions as de-
scribed in
Materials and Methods
were loaded.
sds22 and PP1
g
2, respectively, by Western blot analysis
(Fig. 3B, lane 2 in the left and right panels). All column
fractions containing sds22 and PP1
g
2 had virtually no pro-
tein phosphatase activity (Table 1).
Purification of PP1
g
2 and sds22 From Caput
Epididymal Spermatozoa
Next, we subjected caput sperm extracts to essentially
the same purification scheme used for caudal sperm ex-
tracts. Caput sperm extracts have higher PP1
g
2-specific ac-
tivity: 7.26 U/mg protein, compared with 1.71 U/mg protein
in caudal sperm. A summary of the purification of sds22
and PP1
g
2 from caput sperm extracts is shown in Table 2.
Similar to caudal sperm extracts, PP1
g
2 in caput sperm
extracts also separated into flow-through and gradient frac-
tions on DEAE-cellulose. In addition to PP1
g
2, the DEAE
-cellulose flow-through fraction also contained immunore-
active sds22. However, unlike caudal sperm (Table 1),
PP1
g
2 in the DEAE-cellulose flow-through fraction of ca-
put sperm extracts was catalytically active: 14.7U/mg pro-
tein (Table 2). The enzyme released by a salt gradient
(0.17–0.305 M KCl) from the DEAE-cellulose column had
a specific activity of 9.6U/mg protein. Further purification
of DEAE-bound pool of PP1
g
2 is not outlined here. The
DEAE-cellulose flow-through fraction, containing sds22
and PP1
g
2, was next passed through a SP-sepharose col-
umn. In this column, sds22 was found in the flow through
fraction, but PP1
g
2 was absorbed by the matrix and was
eluted by a salt gradient (0.41–0.5 M KCl). Specific activity
of enzyme eluting from SP-sepharose was 19.8 U/mg pro-
tein (Table 2). Western blot (Fig. 4) shows the fractionation
of sds22 and PP1
g
2 through DEAE-cellulose and SP-se-
pharose columns. It can be seen that sds22 and PP1
g
2 pres-
ent together in the DEAE-cellulose flow-through fraction
(Fig. 4, lane 2) are separated in the SP-sepharose column
(Fig. 4, lanes 3 and 4).
The SP-sepharose column fractions containing sds22 and
PP1
g
2 were concentrated and passed through a Superose 6
column. Caput sperm sds22 eluted as a 60-kDa protein,
fractions 31, 32, and 33 (Fig. 5A). This 60-kDa protein was
next passed through a heparin sepharose column, in which
immunoreactive sds22 was found in the flow-through frac-
tion. SDS-PAGE followed by Coomassie blue and silver
staining shows that heparin Sepharose-purified 60-kDa spe-
cies is composed of two major protein bands at 43 kDa and
1576
MISHRA ET AL.
TABLE 2. Summary of sds22 and PP1
g
2 purification from caput epididymal sperm extracts.*
Purification step Protein (mg) Total activity (U)
Specific activity
(U/mg protein) Recovery (%)
Purification
fold
Caput sperm extract 54.6 396 7.26 100 1
DEAE-cellulose flow-through
(sds221PP1
g
2) 21.5 31.5 14.65 72 2
DEAE-cellulose gradient
(0.17–0.305 M KC1) (PP1
g
2
without sds22) 26.6 255 9.6 64 1.3
SP sepharose flow-through of
DEAE cellulose flow-through
(free sds22) 11.8 Inactive
Superose 6 (sds22) 1.1 Inactive
Heparin sepharose flow-through
(sd22) 0.49 Inactive
SP sepharose gradient (0.410–
0.5 M KC1) of DEAE cellu-
lose flow-through
(free PP1
g
2) 5.2 103 19.81 26 2.7
Superose 6 (free PP1
g
2) 0.15 85 567 22 78
* Table summarizes purification of three batches of 50 ml each of caput sperm extract as outlined under
Materials and Methods
. The total number of
cells corresponding to 54.6 mg protein is 3 3 10
10
. Activity of PP1
g
2 in caput sperm extracts and fractions obtained after column procedures was
measured in the presence of 1 mM Mn
2
1
. Experimental details are given in
Materials and Methods
.
17 kDa (p17) (Fig. 5, B, C, and D). The SP-sepharose frac-
tions containing PP1
g
2 eluted through the Superose 6 col-
umn at about 39 kDa: a molecular size in close correspon-
dence to the calculated size based on its amino acid se-
quence (39 kDa) (Fig. 5A). Identities of purified PP1
g
2 and
sds22 were also confirmed by Western blot analysis (Fig.
5D).
Caput Sperm sds22-p17 Complex Does Not Bind
to PP1
g
2
Caudal but not caput sperm sds22 is bound to PP1
g
2. A
logical question is whether the lack of sds22-PP1
g
2 binding
is due to some modification in PP1
g
2 in caput spermatozoa.
If this were the case, purified caput sperm sds22 should
bind to recombinant PP1
g
2 expressed in bacteria. We in-
vestigated whether the purified caput sperm sds22-p17
complex can inhibit recombinant PP1
g
2 in vitro. Varying
concentrations of heparin sepharose-purified sds22-p17
complex (0 to 1
m
g) were preincubated with PP1
g
2 (Table
2 and Fig. 5D) followed by measurement of protein phos-
phatase activity. Catalytic activity of PP1
g
2 activity was
not significantly inhibited at any concentration of sds22
(Fig. 6). Both calyculin A (5 nM) and protein I2 (10 ng),
known inhibitors of PP1, do inhibit catalytic activity of the
recombinant enzyme (Fig. 6), showing that the recombinant
enzyme is capable of binding to these inhibitors. Identical
results to those shown in Figure 6 were also obtained when
recombinant PP1
a
or PP1
g
2 purified from caput sperm ex-
tracts (Superose 6 eluate; Fig. 5A) instead of recombinant
PP1
g
2 was used (data not shown).
DISCUSSION
In somatic cells, PP1 isoforms are regulated by several
ubiquitous and cell-specific regulatory and targeting pro-
teins [11]. Germ cell-specific protein phosphatase PP1
g
2is
the predominant serine/threonine phosphatase in sperma-
tozoa [1–3]. The activity of this enzyme is inversely cor-
related with motility: low activity in vigorously motile cau-
dal spermatozoa and high activity in immotile caput sper-
matozoa [2]. Inhibition of the enzyme with okadaic acid
and calyculin A causes motility initiation and stimulation
[1–3]. Our focus was to understand how PP1
g
2 is regulated
and how the activity of the enzyme declines during epidid-
ymal sperm maturation.
Antibodies to PP1
g
2 coprecipitate sds22 from sperm
extracts suggesting that sds22 is bound to PP1
g
2 [12]. One
of the objectives of this study was to confirm this obser-
vation by column purification techniques and determine
stoichiometry of the PP1
g
2-sds22 complex. Caudal sperm
sds22 copurifies with PP1
g
2 through four different col-
umns: DEAE-cellulose, SP-sepharose, Superose 6, and
Mono S. The fact that sds22 and PP1
g
2 copurify in col-
umn chromatography and immunoprecipitation [12]
strongly supports the conclusion that sds22 is complexed
to PP1
g
2. The apparent molecular weight of the sds22 and
PP1
g
2 complex, determined by Superose 6 column chro-
matography, is 88 kDa. This is in close correspondence
with a molecular weight of 82 kDa, for a 1:1 PP1
g
2-sds22
complex, calculated on the basis of their amino acid se-
quences.
Our data do not rule out the possibility that some of
caudal sperm sds22 may exist as a homodimer (86 kDa).
It is, however, unlikely that this sds22 will coelute with the
sds22-PP1
g
2 complex through the four different column
matrices. Furthermore, free sds22 (i.e., sds22 not bound to
PP1
g
2) exists as a 60-kDa species in caput spermatozoa
(Fig. 5). It is noteworthy that immunoreactive sds22 is not
detected in any other column fraction except those also con-
taining PP1
g
2 (Table 1 and Fig. 1), suggesting that all of
the sds22 present in caudal sperm extracts is bound to
PP1
g
2 (i.e., sds22 in sperm extracts is a PP1
g
2 regulatory
protein).
In accordance with our earlier observations made with
crude caudal sperm extracts [12], we also determined that
column-purified sds22-PP1
g
2 complex does not also bind
to microcystin (data not shown). Microcystin can bind to
PP1 and PP2A catalytic subunits alone or when the cata-
lytic subunits are associated with a variety of their regula-
tory proteins [16]. In particular, the sds22 complex with the
somatic cell PP1 isoform, PP1
a
, can bind to microcystin
[17]. The inability of the sperm sds22-PP1
g
2 complex to
bind to microcystin suggests that sds22 masks the micro-
cystin-binding site on PP1
g
2. Purified PP1
g
2-sds22 com-
plex and the complex present in the different column frac-
tions are virtually devoid of catalytic activity (Table 1). The
1577
REGULATION OF PP1
g
2 BY sds22
FIG. 5. Molecular weight determination of the caput sperm PP1g2 and
sds22. A) Elution profile of sds22-p17 complex and PP1g2 from caput
sperm extracts in Superose 6. PP1g2 and sds22-p17 complex eluted in
Superose 6 at 39 kDa and 60 kDa, respectively. Calibration of the column
for its void volume was performed by running blue dextran. Standardi-
zation of the column was performed as described in Figure 2. B and C)
Coomassie blue and silver stained gels showing sds22 and p17 after SDS-
PAGE of purified sds22-p17 complex (5 mg) obtained after heparin se-
pharose column chromatography. C) Western blots showing immunore-
active PP1g2 and sds22 in caput extract (50 mg protein), purified PP1g2
fraction and sds22-p17 complex (5 mg).
FIG. 6. Effect of caput sperm sds22-p17 complex on catalytic activity of
recombinant PP1g2. The indicated amounts of sds22-p17 complex puri-
fied from caput sperm extracts, 5 nM calyculin A or 10 ng of I2 were
added to recombinant PP1g2 (150 mU) and incubated at 308C for 10
minutes. After this incubation, the phosphatase assay was initiated with
phosphorylase
a
as described in
Materials and Methods.
Results are ex-
pressed as mean 6 SEM (n 5 3).
sds22-PP1
g
2 complex isolated by immunoaffinity chro-
matography from crude sperm extracts was also inactive
[12]. Thus sds22 is a PP1
g
2 inhibitor in spermatozoa. It is
noteworthy that spermatozoa do not contain the ubiquitous
somatic cell PP1 inhibitors I1 [2] and I2 (unpublished data).
Data presented in this report provide strong evidence that
in motile caudal spermatozoa PP1
g
2, present as a 1:1 com-
plex with sds22, is catalytically inactive.
The next objective of this study was to examine wheth-
er sds22 in caput epididymal spermatozoa is also bound
to PP1
g
2. Data in Table 2 and Figure 4 clearly show that
caput sperm sds22 is not bound to PP1
g
2. Sds22 in caput
sperm extracts is present in the flow-through fraction of
SP-sepharose column, which does not contain any detect-
able PP1
g
2 (Fig. 4). The enzyme PP1
g
2, free of any de-
tectable sds22, is released by a salt gradient from SP-se-
pharose (Fig. 4). Unlike caudal sperm extracts, PP1
g
2
from caput spermatozoa in the DEAE cellulose flow-
though fraction and SP-sepharose gradient fractions is cat-
alytically active. The specific activity of caput sperm
PP1
g
2, not bound to sds22, following Superose 6 column
purification, is 567 U/mg protein. The enzyme at this stage
of purification (78-fold purification, compared with crude
extracts, Table 2) is not expected to be homogenous. Our
goal here was not purification of the enzyme to homoge-
neity but to assess whether the enzyme was bound to
sds22 or some other regulatory protein. The apparent mo-
lecular weight of the active subunit of the enzyme eluting
through the Superose 6 column is 39 kDa in correspon-
dence with the calculated molecular weight (39 kDa)
based on its amino acid sequence. In caput sperm, active
PP1
g
2 is not bound to sds22 or any other regulatory pro-
tein. One of the reasons for the high protein phosphatase
activity in caput extracts is due to this 39-kDa free cata-
lytic subunit form of PP1
g
2.
Our studies also show that a portion of PP1
g
2 in both
caput and caudal sperm extracts is bound to DEAE-cellu-
lose. Specific activities of PP1
g
2 released from DEAE-cel-
lulose are 9.6 and 2.18 U/mg protein in caput and caudal
sperm, respectively (Tables 1 and 2). We have not described
further purification of this DEAE-cellulose bound form of
sperm PP1
g
2 in this study. These results have been pre-
sented elsewhere [18, 19].
One of the important questions emerging from this study
is why sds22 from caput spermatozoa is not bound to
PP1
g
2. Caput sperm sds22 elutes as a 60-kDa species
through the Superose 6 column (Fig. 5A). This 60-kDa spe-
cies consists of sds22 (43 kDa) and another 17-kDa protein
(p17) (Fig. 5, B and C). This suggests that sds22 may exist
as a complex with p17 in caput spermatozoa. It is therefore
possible that p17, bound to sds22, prevents sds22 from
binding to PP1
g
2. Caput sperm sds22 complexed to p17 is
also unable to inhibit recombinant PP1
g
2 in vitro (Fig. 6),
1578
MISHRA ET AL.
presumably because it cannot bind to it. Partially purified
caput sperm PP1
g
2 behaves like recombinant PP1
g
2 with
respect to inhibition by calyculin A and inhibitor I2 and
binding to microcystin. Thus, the reason for inability of
sds22 to bind to PP1
g
2 probably is not due to a modifi-
cation of the PP1
g
2 subunit but most likely is due to the
fact p17 bound to sds22 prevents formation of the sds22-
PP1
g
2 complex. Dissociation of the sds22-p17 complex
and sds22-PP1
g
2 complex formation may be caused by
phosphorylation or some other chemical modification of
sds22 or p17. Alternatively p17 may be proteolyzed during
epididymal sperm maturation (i.e., p17 may be absent in
caudal spermatozoa). Molecular characterization of p17,
development of antibodies against p17, and determination
of the status of p17 in caudal spermatozoa will help us to
explore mechanisms underlying sds22 dissociation from
p17 followed by binding to PP1
g
2 during epididymal
sperm development.
Our data conclusively demonstrate that purified caput
sds22 is free of PP1
g
2. However, coelution of p17 and
sds22 does not necessarily imply that they are complexed
to each other (Fig. 5). It could be argued that p17 may not
be bound to sds22 but is a protein contaminant coeluting
with sds22 in the Superose 6 column. Other mechanisms
may be responsible for the development of sds22 binding
to PP1
g
2. It may be possible that sds22 phosphorylation
may be necessary for its binding to PP1
g
2 (i.e., sds22 may
be phosphorylated in caudal but not caput spermatozoa).
Insights into these questions might have been provided if
we were able to immunoprecipitate sds22 from sperm ex-
tracts. However, antibody preparations against two regions
in the carboxy terminus region of sds22 were unsuitable for
immunoprecipitation [12].
Whatever the reason for the lack of binding of sds22 to
PP1
g
2 may be, our data clearly show that all of sds22 in
caudal sperm extracts is bound to PP1
g
2, whereas in caput
sperm extracts, sds22 is free of any detectable PP1
g
2. A
consequence of this difference is that a proportion of caput
sperm PP1
g
2 is in its free (39 kDa) catalytically active
form, with a specific activity of 567 U/mg protein after
partial purification through Superose 6 column (Table 2).
Thus, one of the reasons for the higher PP1
g
2 activity in
caput than in caudal spermatozoa is that PP1
g
2isnot
bound to sds22.
Although sds22 is present in yeast and is ubiquitous in
animal cells [20], several features of sperm sds22 are
unique. In somatic cells, sds22 is localized to the nucleus
and only a small proportion of cellular PP1 is bound to
sds22 [17]. In spermatozoa, sds22 and sds22-bound PP1
g
2
are cytoplasmic [12]. Unlike complexes of sds22 with so-
matic cell isoforms of PP1 [16], sperm PP1
g
2-sds22 com-
plex does not bind to microcystin [12]. This suggests that
PP1
g
2 binding to sds22 may involve a unique amino acid
domain in PP1
g
2. Finally, in yeast, PP1 binding to sds22
activates the enzyme [20], whereas in spermatozoa, sds22-
bound PP1
g
2 is inactive. These differences suggest that
unique isoform- or sperm-specific mechanisms may operate
with respect to sds22 regulation of PP1
g
2. Furthermore, the
significance of sds22-bound PP1 in somatic cells is unclear
because it is not known whether free sds22 is found in
somatic cells. It is therefore also unclear whether sds22
binding to PP1 is regulated. Our studies are the first to show
that a significant change in the composition of sds22 occurs
during sperm development.
Studies described here clearly show that a portion of
PP1
g
2 in motile caudal epididymal spermatozoa is inac-
tive and present as a heterodimer with sds22. In immotile
caput spermatozoa, sds22 is not bound to PP1
g
2 but is
either free or bound to a 17-kDa protein. Therefore, a por-
tion of caput sperm PP1
g
2 is in its catalytically active and
free form. It is therefore very likely that the reason for the
higher PP1
g
2 activity in caput, compared with caudal
spermatozoa [1–3] is due to the inability of sds22 to bind
and inactivate PP1
g
2. That is, the ability of sds22 to bind
to PP1
g
2 develops during sperm maturation. We hypoth-
esize that the change in binding partners of sds22 from
p17 to PP1
g
2 and the development of the binding capacity
of sds22 for PP1
g
2 are key biochemical events responsible
for the decline in protein phosphatase activity during ep-
ididymal sperm maturation and motility initiation. Studies
are underway to determine molecular mechanisms and in-
tracellular signals regulating sds22 binding to PP1
g
2 dur-
ing sperm development.
ACKNOWLEDGEMENTS
We thank Dr. Balwant Khatra, University of California Long Beach,
for his generous supply of phosphorylase b and phosphorylase kinase. We
also thank Shannan Jack, Brian Sapola, John Ferrara, and Kimberly Mey-
ers for their assistance in the laboratory and useful discussions.
REFERENCES
1. Smith GD, Wolf DP, Trautman KC, da Cruz e Silva EF, Greengard P,
Vijayaraghavan S. Primate sperm contain protein phosphatase1, a bio-
chemical mediator of motility. Biol Reprod 1996; 54:719–727.
2. Smith GD, Wolf DP, Trautman KC, Vijayaraghavan S. Motility po-
tential of macaque epididymal sperm: the role of protein phosphatase
and glycogen synthase kinase-3 activities. J Androl 1996; 20:47–53.
3. Vijayaraghavan S, Stephens DT, Trautman K, Smith GD, Khatra B,
da Cruz e Silva EF, Greengard P. Sperm motility development in the
epididymis is associated with decreased glycogen synthase kinase-3
and protein phosphatase 1 activity. Biol Reprod 1996; 54:709–718.
4. Honkanen RE, Golden T. Regulators of serine/threonine protein phos-
phatases at the dawn of a clinical era? Curr Med Chem 2002; 9:2055–
2075.
5. Mumby MC, Walter G. Protein serine/threonine phosphatases: struc-
ture, regulation, and functions in cell growth. Physiol Rev 1993; 73:
673–99.
6. Wera S, Hemmings BA. Serine/threonine protein phosphatases. Bio-
chem J 1995; 311:17–29.
7. Sasaki K, Shima H, Kitagawa Y, Irino S, Sugimura T, Nagao M.
Identification of members of the protein phosphatase 1 gene family in
the rat and enhanced expression of protein phosphatase 1
a
gene in rat
hepatocellular carcinomas. Jpn J Cancer Res 1990; 81:1272–1280.
8. Kitagawa Y, Sasaki K, Shima H, Shibuya M, Sugimura T, Nagao M.
Protein phosphatases possibly involved in rat spermatogenesis. Bio-
chem Biophys Res Commun 1990; 31:230–235.
9. Varmuza S, Jurisicova A, Okano K, Hudson J, Boekelheide K, Shipp
EB. Spermiogenesis is impaired in mice bearing a targeted mutation
in the protein phosphatase 1c gamma gene. Dev Biol 1999; 205:98–
110.
10. Cohen PT. Protein phosphatase 1- targeted in many directions. J Cell
Sci 2002; 115:241–256.
11. Oliver CJ, Shenolikar S. Physiologic importance of protein phospha-
tase inhibitors. Front Biosci 1998; 3:D961–D972.
12. Huang Z, Khatra B, Bollen M, Carr DW, Vijayaraghavan S. Sperm
PP1
g
2 is regulated by a homologue of the yeast protein phosphatase
binding protein sds22. Biol Reprod 2002; 67:1936–1942.
13. Kobe B, Deisenhofer J. A structural basis of the interactions between
leucine-rich repeats and protein ligands. Nature 1995; 374:183–186.
14. Bradford MM. A rapid and sensitive method for the quantitation of
microgram quantities of protein utilizing the principle of protein-dye
binding. Anal Biochem 1976; 72:248–254.
15. Laemmli UK. Cleavage of structural proteins during the assembly of
the head of bacteriophage T4. Nature 1970; 227:680685.
16. Meek S, Morrice N, MacKintosh C. Microcystin affinity purification
of plant protein phosphatases: PP1C, PP5 and a regulatory A-subunit
of PP2A. FEBS Lett 1999; 457:494498.
17. Tran HT, Bridges D, Ulke A, Moorhead GB. Detection of multiple
1579
REGULATION OF PP1
g
2 BY sds22
splice variants of the nuclear protein phosphatase 1 regulator sds22 in
rat liver nuclei. Biochem Cell Biol 2002; 80:811–815.
18. Somanath PR, Mishra S, Huang Z, Vijayaraghavan S. Protein phos-
phatase PP1
g
2 localized in the sperm head is phosphorylated and
bound to protein 14-3-3. Biol Reprod 2003; 68(Suppl 1):18 (abstract).
19. Mishra S, Somanath PR, Vijayaraghavan S. Distinct pools of PP1
g
2
are regulated by sds22, protein 14-3-3 and hsp90 within spermatozoa.
In: Late Breaking Abstracts, Annual Meeting of the American Society
of Biochemistry and Molecular Biology; 2003; San Diego, CA. Ab-
stract LB270.
20. MacKelvie SH, Andrews PD, Stark MJ. The Saccharomyces cerevi-
siae gene sds22 encodes a potential regulator of the mitotic function
of yeast type 1 protein phosphatase. Mol Cell Biol 1995; 15:3777–
3785.
... In sperm, PP1γ2 activity is mainly regulated by three specific inhibitors, protein phosphatase inhibitor 2 (PPP1R2, also known as I2), protein phosphatase 1 regulatory subunit 7 (PPP1R7, also known as SDS22) and protein phosphatase 1 regulatory subunit 11 (PPP1R11, also known as I3), whose association with PP1γ2 varies during epididymal sperm maturation [7,50,51]. In brief, PP1γ2 is solely bound to I3 in immotile spermatozoa of the caput epididymis, while in caudal spermatozoa it is bound to all three inhibitors. ...
... The PK that phosphorylates PP2A is still unclear. Active GSK3 phosphorylates the PP1γ2 inhibitor I2 at Thr73, which results in active PP1γ2, in a complex with only I3, since SDS22 is bound to p17 [51,57]. PP1γ2 not only contributes to GSK3 dephosphorylation, but also to key Ser and Thr residues dephosphorylation [4,57]. ...
... Highly phosphorylated GSK3 at Ser residues is inactive and incapable of phosphorylating I2, which can inhibit PP1γ2 [57]. Simultaneously, SDS22 is free from its interaction with p17, being in a complex with PP1γ2 as well [51]. The decrease in the activity of the Ser/Thr PPs, which causes a notable decrease in dephosphorylation, and an increase in the number of phosphorylated residues due to PKA activity, was observed in caudal spermatozoa [1,19,67]. ...
Article
Full-text available
Male fertility relies on the ability of spermatozoa to fertilize the egg in the female reproductive tract (FRT). Spermatozoa acquire activated motility during epididymal maturation; however, to be capable of fertilization, they must achieve hyperactivated motility in the FRT. Extensive research found that three protein phosphatases (PPs) are crucial to sperm motility regulation, the sperm-specific protein phosphatase type 1 (PP1) isoform gamma 2 (PP1γ2), protein phosphatase type 2A (PP2A) and protein phosphatase type 2B (PP2B). Studies have reported that PP activity decreases during epididymal maturation, whereas protein kinase activity increases, which appears to be a requirement for motility acquisition. An interplay between these PPs has been extensively investigated; however, many specific interactions and some inconsistencies remain to be elucidated. The study of PPs significantly advanced following the identification of naturally occurring toxins, including calyculin A, okadaic acid, cyclosporin, endothall and deltamethrin, which are powerful and specific PP inhibitors. This review aims to overview the protein phosphorylation-dependent biochemical pathways underlying sperm motility acquisition and hyperactivation, followed by a discussion of the PP inhibitors that allowed advances in the current knowledge of these pathways. Since male infertility cases still attain alarming numbers, additional research on the topic is required, particularly using other PP inhibitors.
... This maturation involves the migration of the cytoplasmic droplet, located on the mid-piece to travel from the proximal position to the distal position. Also while moving through the epididymal duct; rat sperm acquire the ability of increased progressive motility [3,4]. Maturation of sperm predominantly takes place while sperm move from caput to the corpus sections of the epididymis. ...
... The ultimate goal in reproduction is to produce pregnancies and the method that will produce the best results is always going to be natural mating or at least the use of ejaculated semen. However, in many cases natural mating is not an option and ejaculated semen is unavailable, due to difficulty of handling the animal, death prior to collection or obstructive azoospermia preventing ejaculation [3]. Whether or not seminal plasma is beneficial to sperm, it does apparently serve a physiological purpose. ...
Article
Fertilization, an important phase of reproduction, is physiologically the union of the male and female gamete. This succession involves the fusion of an oocyte with a sperm, leading to generation of a single diploid cell, the zygote, from which a new individual organism develops. The elucidation of the physiological, biochemical and molecular mechanisms of fertilization has enthralled researchers of relevance for several years. This editorial embodies this fascinating succession at physiological, biochemical and molecular level.
... PP1γ2 is testis-/spermatozoa-specific, and is known to be essential for spermatogenesis, sperm motility, and male fertility (Kitagawa et al. 1990;Shima et al. 1993a, b;da Cruz e Silva et al. 1995;Smith et al. 1996Smith et al. , 1999Vijayaraghavan et al. 1996;Fardilha et al. 2011). PP1γ2 interacts with SDS22 (also known as protein phosphatase 1, regulatory inhibitor subunit 7, PPP1R7), leading to a decline of phosphatase activity of PP1γ2 and, subsequently, increased sperm mobility (Huang et al. 2002;Mishra et al. 2003). Biochemical and bioinformatics analyses revealed that the SDS22-PP1γ2 interaction occurs through the sixth leucine rich-repeat (LRR) of SDS22 (Choy et al. 2019). ...
Article
Full-text available
Preferentially expressed antigen in melanoma (PRAME) is a cancer/testis antigen (CTA) that is predominantly expressed in normal male gonad tissues and a variety of tumors. PRAME proteins are present in the acrosome and sperm tail, but their role in sperm function is unknown. The objective of this study was to examine the function of the bovine Y-linked PRAME (PRAMEY) during spermatozoal capacitation, the acrosome reaction (AR), and fertilization. Freshly ejaculated spermatozoa were induced to capacitate and undergo AR in vitro. Western blotting results revealed a decrease in the PRAMEY protein in capacitated spermatozoa, and the release of the PRAMEY protein from the acrosome during the AR, suggesting its involvement in sperm capacitation and AR. IVF was performed using in vitro matured bovine oocytes and cauda epididymal spermatozoa either treated with PRAMEY antibody, rabbit IgG, or DPBS. Sperm-egg binding and early embryos were examined at 6 and 45 h post IVF, respectively. The number of spermatozoa that bound per oocyte was nearly two-fold greater in the PRAMEY antibody treatment group (34.4) when compared to both the rabbit IgG (17.6) and DPBS (18.1) controls (P < 0.01). Polyspermy rate in the antibody-treated group (18.9%) was three-fold greater than the rabbit IgG control (6.0%) (P < 0.01). The results indicate that PRAMEY may play a role in anti-polyspermy defense. This study thus provides the initial evidence for the involvement of the PRAME protein family in sperm function and fertilization.
... They speculated that SDS22 could act as the third interactor of multiple PP1 complexes, thereby exhibiting diverse functions [47]. In addition to cell division, SDS22 has been implicated in multiple processes such as cell shape and polarity [48], epithelium integrity [49], sperm motility [50,51] and plant immunity [52][53][54][55], and is now receiving increasing interest as a therapeutic target in cancer studies [56,57]. ...
Article
Full-text available
Protein phosphatase 1 (PP1) is a key enzyme for Plasmodium development. However, the detailed mechanisms underlying its regulation remain to be deciphered. Here, we report the functional characterization of the Plasmodium berghei leucine-rich repeat protein 1 (PbLRR1), an orthologue of SDS22, one of the most ancient and conserved PP1 interactors. Our study shows that PbLRR1 is expressed during intra-erythrocytic development of the parasite, and up to the zygote stage in mosquitoes. PbLRR1 can be found in complex with PbPP1 in both asexual and sexual stages and inhibits its phosphatase activity. Genetic analysis demonstrates that PbLRR1 depletion adversely affects the development of oocysts. PbLRR1 interactome analysis associated with phospho-proteomics studies identifies several novel putative PbLRR1/PbPP1 partners. Some of these partners have previously been characterized as essential for the parasite sexual development. Interestingly, and for the first time, Inhibitor 3 (I3), a well-known and direct interactant of Plasmodium PP1, was found to be drastically hypophosphorylated in PbLRR1-depleted parasites. These data, along with the detection of I3 with PP1 in the LRR1 interactome, strongly suggest that the phosphorylation status of PbI3 is under the control of the PP1–LRR1 complex and could contribute (in)directly to oocyst development. This study provides new insights into previously unrecognized PbPP1 fine regulation of Plasmodium oocyst development through its interaction with PbLRR1.
... Members of the PRAME gene family encode leucine-rich repeat (LRR) proteins that fold into a horseshoe shape, which provides a versatile structural framework for the formation of protein-protein interactions (Kobe and Kajava, 2001;Epping et al., 2005;Wadelin et al., 2010). Protein phosphatase 1, regulatory (inhibitor) subunit 7 (PPP1R7), also known as SDS22, is another LRR protein and interacts with protein phosphatase 1 catalytic subunit gamma isozyme (PPP1CC), also known as PP1γ2, in bovine cauda epididymal spermatozoa (Huang et al., 2002;Mishra et al., 2003). As a testis/ spermatozoa-specific phosphatase, PP1γ2 acts as an important regulator of sperm motility, and male fertility (Smith et al., 1996;Fardilha et al., 2011). ...
Article
Full-text available
The preferentially expressed antigen in melanoma, Y-linked (PRAMEY) is a cancer/testis antigen expressed predominantly in bovine spermatogenic cells, playing an important role in germ cell formation. To better understand PRAMEY’s function during spermatogenesis, we studied the dynamics of PRAMEY isoforms by Western blotting (WB) with PRAMEY-specific antibodies. The PRAMEY protein was assessed in the bovine testicular and epididymal spermatozoa, fluid and tissues, and as well as in ejaculated semen. The protein was further examined, at a subcellular level in sperm head and tail, as well as in the subcellular components, including the cytosol, nucleus, membrane, and mitochondria. RNA expression of PRAMEY was also evaluated in testis and epididymal tissues. Our WB results confirmed the previously reported four isoforms of PRAMEY (58, 30, 26, and 13 kDa) in the bovine testis and spermatozoa. We found that testicular spermatozoa expressed the 58 and 30 kDa isoforms. As spermatozoa migrated to the epididymis, they expressed two additional isoforms, 26 and 13 kDa. Similarly, the 58 and 30 kDa isoforms were detected only in the testis fluid, while all four isoforms were detected in fluid from the cauda epididymis. Tissue evaluation indicated a significantly higher expression of the 58 and 13 kDa isoforms in the cauda tissue when compared to both the testis and caput tissue (p < 0.05). These results indicated that testis samples (spermatozoa, fluid, and tissue) expressed predominantly the 58 and 30 kDa PRAMEY isoforms, suggesting their involvement in spermatogenesis. In contrast, the 26 kDa isoform was specific to epididymal sperm and the 13 kDa isoform was marked in samples derived from the cauda epididymis, suggesting their involvement in sperm maturation. Results from the sperm head and tail experiments indicated that the 13 kDa isoform increased 4-fold in sperm tails from caput to cauda, suggesting this isoform may have a significant role in tail function. Additionally, the 13 kDa isoform increased significantly (p < 0.05) in the cytosol during epididymal passage and tended to increase in other subcellular components. The expression of PRAMEY in the sperm subcellular components during epididymal maturation suggests the involvement of PRAMEY, especially the 13 kDa isoform, in sperm motility.
... The sources of images are from [42,44,89,90]. mobility of caudal sperm [99,100]. Protein sequence and structure analyses reveal that the sixth LRR of SDS22 is highly conserved among bovine, mouse, and human orthologs. ...
Article
The precise mechanisms of the reproductive physiological processes, such as labor initiation, are poorly understood. Oxytocin (OT) is one of the well-known uterotonics and is clinically adopted as a medication to facilitate childbirth. Vasopressin (VP), a posterior pituitary hormone similar to OT, has also been proposed to be involved in the reproductive physiology. In this study, we found that a total deficiency of V1a receptor subtype (V1aR) in mice resulted in a reduced number of pups, delayed labor initiation, and increased post-delivery hemorrhage compared with those in wild-type mice. Among the VP receptor subtypes, only V1aR was found to be expressed in the murine uterus and its distribution pattern was different from that of the oxytocin receptor (OTR); V1aR expression was mainly distributed in the circular myometrium, whereas OTR was strongly expressed in both the circular and longitudinal myometrium. The maximum contractile force of the circular myometrium, induced by VP or OT, was attenuated in the pregnant uterus of Avpr1a-deficient mice. Contrarily, while OT expression was decreased in the Avpr1a-deficient uterus, OTR expression was significantly increased. These results suggest that V1aR deficiency not only reduces the uterine contractile force but also perturbs the expression of genes responsible for the reproductive physiology. Therefore, V1aR is necessary to exert the maximum contraction of the circular myometrium to deliver pups. This study revealed an important role of V1aR in physiological contraction and term parturition in mice.
... In caput spermatozoa, bovine sperm PPP1R7 and PPP1CC2 do not directly interact. Instead, PPP1R7 is associated with a 17 kDa protein (p17) [65], resulting in free and active PPP1. Consequently, PPP1 dephosphorylates key proteins for sperm motility, resulting in immotile spermatozoa. ...
Chapter
The sperm cell is unique in its function. It is the only human cell that must leave the body where it is produced and fulfills its goal in a different organism being, thus, a highly specialized cell. Sperm cells are produced in the testis, acquire motility during the epididymis journey and fertilize the oocyte in the female reproductive system. Moreover, since these cells are virtually transcriptionally silent, they rely exclusively on protein-protein interactions and post translational modifications to control signaling pathways. These sperm cell unique features are reflected in sperm-specific signaling. Several sperm-specific/enriched proteins are responsible for controlling sperm functions such as sperm motility and acrosome reaction. In this chapter, we describe the signaling events that characterize sperm motility and acrosome reaction as well as the unique proteins that control such events.
... Conversely, the removal of phosphate groups by phosphatases has also been implicated in epididymal sperm maturation (Huang and Vijayaraghavan, 2004;Vijayaraghavan et al., 2007;Silva et al., 2014;Goswami et al., 2019). On the other hand, some studies have shown that sperm phosphoprotein phosphatase 1 catalytic subunit gamma 2 (PPP1CC2, also named PP1γ2) activity declines during epididymal maturation and is important in regulating sperm motility (Vijayaraghavan et al., 1996;Mishra et al., 2003;Goswami et al., 2019). ...
Article
Full-text available
After leaving the testis, mammalian sperm undergoes a sequential maturation process in the epididymis followed by capacitation during its movement through the female reproductive tract. These phenotypic changes are associated with modification of protein phosphorylation and membrane remodeling, which is requisite for sperm to acquire forward motility and induce fertilization. However, the molecular mechanisms underlying sperm maturation and capacitation are still not fully understood. Herein, we show that PPP3R2, a testis-specific regulatory subunit of protein phosphatase 3 (an isoform of calcineurin in testis), is essential for sperm maturation and capacitation. Knockout of Ppp3r2 in mice leads to male sterility due to sperm motility impairment and morphological defects. One very noteworthy change includes increases in sperm membrane stiffness. Moreover, PPP3R2 regulates sperm maturation and capacitation via: (1) modulation of membrane diffusion barrier function at the annulus; (2) facilitation of cholesterol efflux during sperm capacitation. Taken together, PPP3R2 plays a critical role in modulating cholesterol efflux and mediating dynamic control of membrane remodeling during sperm maturation and capacitation.
... Further analysis indicated that PRAMEY interacts with the protein phosphatase 1 gamma isoform 2 (PP1γ2) ( Zhao et al. 2013). The latter is a key component for regulation of spermatozoa motility and male fertility (Mishra et al. 2003, Fardilha et al. 2011). ...
Article
Full-text available
In higher eukaryotes, the activity and specificity of the type 1 protein serine-threonine phosphatase (PP1) catalytic subunit is thought to be controlled by its association with a number of regulatory or targeting subunits. Here we describe the characterization of a gene encoding one such potential polypeptide in the yeast Saccharomyces cerevisiae. The gene which we have isolated (termed SDS22) encodes a product with a high degree of sequence identity to the fission yeast sds22 protein, a known regulator of the mitotic function of PP1 in Schizosaccharomyces pombe. Using two different criteria, we have demonstrated that Sds22p and the catalytic subunit of PP1 (Glc7p) interact in yeast cells. We have also generated a temperature-sensitive allele of GLC7 (glc7-12) which causes a block to the completion of mitosis at the restrictive temperature. Additional copies of SDS22 lead to allele-specific suppression of the glc7-12 mutant, strongly suggesting that the interaction between the two proteins is of functional significance. Sds22p is therefore likely to be the second example of a PP1 regulatory subunit identified in S. cerevisiae.
Article
Full-text available
We isolated four kings of cDNA clones of isotypes of catalytic subunits of protein phosphatase 1 (PP-1) from rat liver and testis cDNA libraries. For the cloning, cDNA fragments of dis2ml and dis2m2, which encode mouse PP-1 catalytic subunit, were used as probes. Two of the four isotypes were thought to be derived from the same gene and produced by alternative splicing. Based on the comparative study of their nucleotide and deduced amino acid sequences with those reported, these cDNA clones were named rat PP-1 alpha, PP-1 gamma 1, PP-1 gamma 2 and PP-1 delta. The deduced amino acid sequences of these four cDNA clones showed about 90% identity. Their amino-terminal regions were highly conserved, and their differences were mainly in the carboxy-terminal regions. Furthermore, several amino acids located in the middle regions of the peptides were conserved in all the isotypes of the catalytic subunits of PP-1, PP-2A, PP-2B and PP-2C. These conserved regions are suggested to be the functional domains of the catalytic subunits of protein phosphatases. Rat hepatocellular carcinomas induced by a food mutagen, 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline showed increased expression of PP-1 alpha, but no increased expression of PP-1 gamma 1, PP-1 gamma 2 or PP-1 delta. Involvement of PP-1 alpha in hepatocarcinogenesis or in hepatic cell proliferation was suspected.
Article
A protein determination method which involves the binding of Coomassie Brilliant Blue G-250 to protein is described. The binding of the dye to protein causes a shift in the absorption maximum of the dye from 465 to 595 nm, and it is the increase in absorption at 595 nm which is monitored. This assay is very reproducible and rapid with the dye binding process virtually complete in approximately 2 min with good color stability for 1 hr. There is little or no interference from cations such as sodium or potassium nor from carbohydrates such as sucrose. A small amount of color is developed in the presence of strongly alkaline buffering agents, but the assay may be run accurately by the use of proper buffer controls. The only components found to give excessive interfering color in the assay are relatively large amounts of detergents such as sodium dodecyl sulfate, Triton X-100, and commercial glassware detergents. Interference by small amounts of detergent may be eliminated by the use of proper controls.
Article
Sperm motility initiation, capacitation, and hyperactivation are modulated by an interplay of intracellular Ca2+, cAMP, and pH. Mechanisms by which these mediators alter sperm function have not been elucidated but may involve reversible alterations in regulatory protein phosphorylation. Studies were designed 1) to investigate the influence of the protein phosphatase (PP) inhibitor calyculin A (CA) on human sperm motility and 2) to characterize the CA-sensitive PP and its endogenous regulators in human and rhesus monkey sperm. CA (50 nM) treatment of human sperm resulted in an increase in percentage motility and an acceleration in mean path velocity. Inhibition of either protein phosphatase-1 (PP1) or protein phosphatase-2A (PP2A) could be responsible for this motility stimulation, since both of these phosphatases are sensitive to nanomolar quantities of CA. PP activity in human (n = 4) and rhesus monkey (n = 4) sperm sonicates was measured using [32P]-phosphorylase-a, the preferred substrate for PP1 and PP2A, in the absence of divalent cations. Human (6.2 +/- 4.5 x 10(-2) mU/10(6) sperm) and monkey (4.3 +/- 0.8 x 10(-2) mU/10(6) sperm) sonicates contained activity tentatively identified as PP1 on the basis of inhibition profiles in okadaic acid (OA) and CA. Western blot analysis with antibodies against various isoforms of PP1 subsequently documented the presence of PP1 gamma 2 in human and monkey sperm. PP1 activity in most tissues is regulated by the heat-stable inhibitors I1 or I2. Sperm sonicates contained inhibitor activity similar to I2 as well as glycogen synthase kinase-3 (GSK-3) activity, which is involved in the activation of the PP1-I2 complex. These results indicate, for the first time, that human and rhesus monkey sperm contain PP1 and regulators of PP1 and that inhibition of PP1 activity by CA can enhance motility.
Article
Serine/threonine phosphatase PP1γ2 is a testis-specific protein phosphatase isoform in spermatozoa. This enzyme appears to play a key role in motility initiation and stimulation. Catalytic activity of PP1γ2 is higher in immotile compared with motile spermatozoa. Inhibition of PP1γ2 activity causes both motility initiation and motility stimulation. Protein phosphatases, in general, are regulated by their binding proteins. The objective of this article is to understand the mechanisms by which PP1γ2 is regulated, first by identifying its regulatory proteins. We had previously shown that a portion of bovine sperm PP1γ2 is present in the cytosolic fraction of sperm sonicates. We purified PP1γ2 from soluble bovine sperm extracts by immunoaffinity chromatography. Gel electrophoresis of the purified enzyme showed that it was complexed to a protein 43 Mr × 10⁻³ in size. Microsequencing revealed that this protein is a mammalian homologue of sds22, which is a yeast PP1 binding protein. Phosphatase activity measurements showed that PP1γ2 complexed to sds22 is catalytically inactive. The complex cannot be activated by limited proteolysis. The complex is unable to bind to microcystin sepharose. This suggests that sds22 may block the microcystin binding site in PP1γ2. A proportion of PP1γ2 in sperm extracts, which is presumably not complexed to sds22, is catalytically active. Fluorescence immunocytochemistry was used to determine the intrasperm localization of PP1γ2 and sds22. Both proteins are present in the tail. They are also present in distinct locations in the head. Our data suggest that PP1γ2 binding to sds22 inhibits its catalytic activity. Mechanisms regulating sds22 binding to PP1γ2 are likely to be important in understanding the biochemical basis underlying development and regulation of sperm function.
Article
We isolated four kinds of cDNA clones of isotypes of catalytic subunits of protein phosphatase 1 (PP-1) from rat liver and testis cDNA libraries. For the cloning, cDNA fragments of dis2ml and dis2m2, which encode mouse PP-1 catalytic subunits, were used as probes. Two of the four isotypes were thought to be derived from the same gene and produced by alternative splicing. Based on the comparative study of their nucleotide and deduced amino acid sequences with those reported, these cDNA clones were named rat PP-1α, PP-1γ1, PP-1γ2 and PP-δ. The deduced amino acid sequences of these four cDNA clones showed about 90% identity. Their amino-terminal regions were highly conserved, and their differences were mainly in the carboxy-terminal regions. Furthermore, several amino acids located in the middle regions of the peptides were conserved in all the isotypes of the catalytic subunits of PP-1, PP-2A, PP-2B and PP-2C. These conserved regions are suggested to be the functional domains of the catalytic subunits of protein phosphatases. Rat hepatocellular carcinomas induced by a food mutagen, 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline showed increased expression of PP-1α, but no increased expression of PP-1γ1, PP-1γ2 or PP-1δ. Involvement of PP-1α in hepatocarcinogenesis or in hepatic cell proliferation was suspected.
Article
The expression of mRNAs for catalytic subunits of serine/threonine protein phosphatases 1 (PP-1) and 2A (PP-2A) in various rat tissues were examined. Four kinds of probes were used to detect mRNAs for two isotypes of PP-1 (dis2m1 and dis2m2), and two isotypes of PP-2A (PP-2Aα and PP-2Aβ). mRNAs for all of these four catalytic subunits were expressed in almost all tissues at substantial levels. They were expressed in two different sizes in most tissues. Remarkable evidence is that the smaller sized mRNAs of dis2m1 and PP-2Aβ, 1.8 kb and 1.4 kb in length, respectively, were specifically highly expressed in testis. Both these smaller sized mRNAs began to be expressed at the age when meiosis started and were detected in testicular germ cells at the pachytene stage of meiotic prophase. Protein phosphatases which have peptides encoded by dis2m1 and PP-2Aβ as catalytic subunits may play important roles in spermatogenesis.
Article
A protein determination method which involves the binding of Coomassie Brilliant Blue G-250 to protein is described. The binding of the dye to protein causes a shift in the absorption maximum of the dye from 465 to 595 nm, and it is the increase in absorption at 595 nm which is monitored. This assay is very reproducible and rapid with the dye binding process virtually complete in approximately 2 min with good color stability for 1 hr. There is little or no interference from cations such as sodium or potassium nor from carbohydrates such as sucrose. A small amount of color is developed in the presence of strongly alkaline buffering agents, but the assay may be run accurately by the use of proper buffer controls. The only components found to give excessive interfering color in the assay are relatively large amounts of detergents such as sodium dodecyl sulfate, Triton X-100, and commercial glassware detergents. Interference by small amounts of detergent may be eliminated by the use of proper controls.
Article
Using an improved method of gel electrophoresis, many hitherto unknown proteins have been found in bacteriophage T4 and some of these have been identified with specific gene products. Four major components of the head are cleaved during the process of assembly, apparently after the precursor proteins have assembled into some large intermediate structure.