ArticlePDF Available

Characterization of Immortalized Dairy Goat Male Germline Stem Cells (mGSCs)

Authors:

Abstract and Figures

Male germline stem cells (mGSCs), in charge for the fertility in male testis, are the only kind of adult stem cells that transmit genetic information to next generation, with promising prospects in germplasm resources preservation and optimization, and production of transgenic animals. Mouse male germline stem cell lines have been established and are valuable for studying the mechanisms of spermatogenesis. However, there is a lack of stable mGSC cell lines in livestock, which restricts the progress of transgenic research and related biotechnology. Here, we firstly established an immortalized dairy goat mGSC cell line to study the biological properties and the signaling pathways associated with mGSCs self-renewal and differentiation. The ectopic factors SV40 large T antigen and Bmi1 genes were transduced into dairy goat mGSCs, and the results showed that the proliferation of these cells that were named mGSCs-I-SB was improved significantly. They maintained the typical characteristics including the expression of mGSC markers, and the potential to differentiate into all three germ layers, sperm-like cells in vitro. Additionally, mGSCs-I-SB survived and differentiated into three germ layer cell types when they were transplanted into chicken embryos. Importantly, the cells also survived in mouse spermatogenesis deficiency model testis which seemed to be the golden standard to examine mGSCs. Conclusively, our results demonstrate that mGSCs-I-SB present the characteristics of mGSCs and may promote the future study on goat mGSCs. J. Cell. Biochem. © 2014 Wiley Periodicals, Inc.
Content may be subject to copyright.
Characterization of Immortalized Dairy Goat male Germline
Stem Cells (mGSCs)
Haijing Zhu, Jing Ma, Rui Du, Liming Zheng, Jiang Wu, Wencong Song, Zhiwei Niu,
Xin He, Enqi Du, Shanting Zhao, and Jinlian Hua*
College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal
Biotechnology of Agriculture Ministry of China, Northwest A&F University, Yangling, Shaanxi 712100, China
ABSTRACT
Male germline stem cells (mGSCs), in charge for the fertility in male testis, are the only kind of adult stem cells that transmit genetic information
to next generation, with promising prospects in germplasm resources preservation and optimization, and production of transgenic animals.
Mouse male germline stem cell lines have been established and are valuable for studying the mechanisms of spermatogenesis. However, there is
a lack of stable mGSC cell lines in livestock, which restricts the progress of transgenic research and related biotechnology. Here, we rstly
established an immortalized dairy goat mGSC cell line to study the biological properties and the signaling pathways associated with mGSCs
self-renewal and differentiation. The ectopic factors SV40 large T antigen and Bmi1 genes were transduced into dairy goat mGSCs, and the
results showed that the proliferation of these cells that were named mGSCs-I-SB was improved signicantly. They maintained the typical
characteristics including the expression of mGSC markers, and the potential to differentiate into all three germ layers, sperm-like cells in vitro.
Additionally, mGSCs-I-SB survived and differentiated into three germ layer cell types when they were transplanted into chicken embryos.
Importantly, the cells also survived in mouse spermatogenesis deciency model testis which seemed to be the golden standard to examine
mGSCs. Conclusively, our results demonstrate that mGSCs-I-SB present the characteristics of mGSCs and may promote the future study on
goat mGSCs. J. Cell. Biochem. 115: 15491560, 2014. ©2014 Wiley Periodicals, Inc.
KEY WORDS: MALE GERMLINE STEM CELLS; IMMORTALIZATION; DAIRY GOAT
Male germline stem cells (mGSCs), also named spermatogo-
nial stem cells (SSCs), localized in the basement of
seminiferous tubules, are able of self-renewal to maintain the
number of mGSCs and differentiation into sperm constitutively
which are mainly in charge of supplying the material basis for male
fertility. Male GSCs have been studied for many years, especially in
mouse and human. Importantly, the immortalized mouse mGSCs
have been obtained and the culture system has been developed
[Feng et al., 2002; Kubota et al., 2004a,b; Hofmann et al., 2005].
Pluripotent stem cells have been obtained from mGSCs in mouse and
human, and are a novel resource for pluripotent cells besides
embryonic stem cells (ESCs) and induced pluripotent stem cells
(iPSCs) [Guan et al., 2006; Conrad et al., 2008]. All these progresses
promoted the study of the mechanisms on self-renewal and
differentiation of mGSCs and their application in agriculture and
life science, such as production of transgenic animals [von
Schonfeldt et al., 2004; Kanatsu-Shinohara et al., 2005, 2006; Li
et al., 2005; Ballow et al., 2006; Naughton et al., 2006; Shinohara
and Kanatasu-Shinohara, 2007; He et al., 2008, 2009; Sikarwar
and Reddy, 2008; Izsvák et al., 2010; Kanatsu-Shinohara and
Shinohara, 2010; Shi et al., 2010; Niu et al., 2011; Zhang et al., 2011;
Suzuki et al., 2012].
As we described previously, dairy goat is important for Chinese
people, and studies on dairy goat mGSCs may improve the
preservation and optimization of the germplasm resources [Zhu
et al., 2013] and provide us a great improvement of quality and
quantity of milk and meat. Up to date, there is a lack of stable culture
system and mGSC cell line in livestock [Luo et al., 2006; Bi et al.,
Grant sponsor: National Natural Science Foundation of China; Grant number: 31272518; Grant sponsor: National
Major Fundamental Research Program of China; Grant number: 2013CB947900; Grant sponsor: Doctoral Fund of
Ministry of Education of China (RFDP); Grant number: 20120204110030; Grant sponsor: Fundamental Research
Funds for the Central Universities; Grant number: QN2011012.
*Correspondence to: Dr Jinlian Hua, College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &
Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China, Northwest A&F University,
Yangling, Shaanxi, China. E-mail: jinlianhua@nwsuaf.edu.cn
Manuscript Received: 19 October 2013; Manuscript Accepted: 27 March 2014
Accepted manuscript online in Wiley Online Library (wileyonlinelibrary.com): 1 April 2014
DOI 10.1002/jcb.24812 ©2014 Wiley Periodicals, Inc. 1549
Journal of Cellular
Biochemistry
ARTICLE
Journal of Cellular Biochemistry 115:1549–1560 (2014)
2007; Aponte et al., 2008; Kaul et al., 2010, 2012; Hua et al., 2011;
Bahadorani et al., 2012; de Barros et al., 2012; Heidari et al., 2012; Li
et al., 2012, 2013; Zhu et al., 2013; McMillan et al., 2013]. The dairy
goat mGSCs cannot survive in vitro under specic culture conditions
for a long period as previously reported by our laboratory [Zhu et al.,
2012]. Thus, it is of great signicance to obtain the immortalized
dairy goat mGSC cell line, which will be critical for the long-term
study of dairy goat mGSCs and other aspects of the cells biology,
including dedifferentiation mGSCs into pluripotent state. Feng et al.
[2002] and Hofmann et al. [2005] have shown that mGSCs could be
immortalized by exogenous factors, such as TERT and Simian virus
40 (SV40) T antigen. These cells shared typical SSC characteristics
including the morphology, expression of mGSCs markers such as
DAZL and GFRa1, the ability of differentiating into haploid sperm
cells and rebuilding seminiferous tubules after transplantation into
mouse spermatogenesis deciency testis.
Immortalization methods in mammals include transduction of
primary cells with vectors carrying viral genes, such as Simian virus 40
(SV40) T antigen, EpsteinBarr virus (EBV), Adenovirus E1A and E1B,
and human Papiloma virus (HPV) E6 and E7. SV40 large T antigen was
regarded as an efcient factor for animal cell immortalization [Lübbe
et al., 1983; Petit et al., 1983; Ozer, 2000], and it has been demonstrated
to be effective for mouse mGSCs immortalization in 2005 [Hofmann
et al., 2005]. Bmi1 is regarded as an marker of mouse mGSCs [Zhang
et al., 2008] and promotes stem cell proliferation [Pietersen et al., 2008].
Being an immortalization factor, Bmi1 has been demonstrated to be
effective in immortalizing somatic cells [Dimri et al., 2002; Saito
et al., 2005]. Conclusively, we believe SV40 large T antigen and Bmi1
are capable of immortalizing dairy goat mGSCs. Here, the lentivirus
particles containing SV40 large T antigen and Bmi1 were transduced
into primary dairy goat mGSCs, and the cells are immortalized and have
been cultured for more than 3 months in vitro. The characteristics of the
immortalized mGSCs were determined through the proliferation test,
marker detection, the differentiation potential assay in vitro and in vivo.
These results suggested that the mGSCs were immortalized by SV40
large T antigen and Bmi1, and these cells will provide unlimited cell
resources for studying male germ cell specication and development.
MATERIALS AND METHODS
ISOLATION AND ENRICHMENT OF MALE GERMLINE STEM CELLS
(mGSCs) FROM GOAT TESTIS
The adult male dairy goats were killed and their testes were collected.
All the procedures were carried on under the supervision of Chinese
Association for Laboratory Animal Science, and approved by
Shaanxi Centre of Stem Cells Engineering & Technology, Northwest
A&F University. Dairy goat mGSCs were isolated and puried as
previously reported [Zhu et al., 2012].
PURIFICATION OF DAIRY GOAT mGSCs
The dairy goat testicular cells were cultured on plates treated with
1mg/ml Fibronectin (Sigma, USA) at 37°C, 5% CO
2
and saturated
humidity for 2 h at the density of 1 10
6
cells/ml [Zhu et al., 2012].
Then the attached cells were dissociated with 0.25% Trypsin
(Invitrogen, USA) for 5 min at 37°C. The cells were collected and
cultured with DMEM/F12 (Invitrogen) supplemented with 10% fetal
bovine serum (FBS, Hyclone, USA), 0.1 mM b-mercaptoethanol
(Sigma) and 2 mM glutamine (Invitrogen).
LENTIVIRUS PREPARATION AND THE IMMORTALIZATION OF DAIRY
GOAT GERMLINE STEM CELLS
Lentivirus production was referred as Anokye-Danso et al. described
[Anokye-Danso et al., 2011]. Briey, HEK293T cells were seeded on
plates 24 h before transfection, then the plasmids containing SV40
large T antigen and Bmi1 along with the plasmids containing pVSVG
and pPAX2 were incubated respectively. The virus-containing
supernatant was collected at 48 h after transfection, ltered to
remove cell debris, and used for transduction. For cell immortaliza-
tion, dairy goat mGSCs were plated at a density of 1 10
5
cells in a
35 mm dish. Twelve hours later, the cells were transduced with virus-
containing supernatant and 10 mg/ml polybrene (Sigma) and
incubated overnight at 37°C and 5% CO
2
. After 24 h, the medium
was discarded and replaced with fresh DMEM/F12 medium
(Invitrogen) supplemented with 10% FBS (Hyclone, USA), 0.1 mM
b-mercaptoethanol (Sigma) and 2 mM glutamine (Invitrogen), then
cultured for more than 1 month until the cells were immortalized.
The immortalized cells were named mGSCs-I-SB.
CELL CYCLE ANALYSIS
For cell cycle analysis, dairy goat mGSCs (5th passage) and
mGSCs-I-SB (10th passage) were cultured for 48 h and then
suspended the cells into single cells and xed them in 70% ice-
cold ethanol for 30 min. After that, cells were incubated with
propidium iodide (PI, Sigma) solution supplemented RNase H
(Beyotime, China) for 20 min. Cell cycle analysis was determined
by ow cytometry as previously reported [He et al., 2008].
POPULATION DOUBLING TIME (PDT) DETERMINATION
The population doubling time (PDT) of dairy goat mGSCs were
estimated according to the protocol described previously [Zhang
et al., 2011]. Briey, cells were serially subcultured, the initial seeding
cell number and the total cell number cultured 48h later were all
counted respectively. PDT was calculated according to the formula,
PDT ¼[log
2
/(logNt0 log N0)] t, where Nt is the number of cells
after t h of culturing, N0 is the number of cells seeded.
5-BROMO-2-DEOXYURIDINE (BrdU) INCORPORATION ASSAY
Dairy goat mGSCs proliferative ability was assessed by BrdU
Incorporation. Dairy goat mGSCs (5th passage) and mGSCs-I-SB
(10th passage) were treated with BrdU (30 mg/ml, Sigma) for 3 h
and then subjected to BrdU immunostaining. Cells were xed with
methanol/acetone (V/V ¼1:1) for 15 min at room temperature and
washed with PBS for three times, incubated with PBS containing
0.1% Triton-100 for 5min. Then the cells were washed three times
with PBS at room temperature. Mouse anti-BrdU (1:100; Santa
Cruz) dissolved in 0.1M PBS (pH 7.4) containing 4% normal goat
serum was added and the cells were incubated overnight at 4°C.
Cells were washed in PBS for three times, and then incubated with
the goat anti-mouse FITC conjugated secondary antibody (1:500,
Chemicon) for 1 h at room temperature. After three washes, cells
were visualized under uorescent microscope and analyzed for
BrdU staining.
1550 CHARACTERIZATION OFIMMORTALIZED DAIRY GOAT mGSCs JOURNAL OFCELLULAR BIOCHEMISTRY
RT-PCR
Total RNA for RT-PCR analysis was extracted from 6th passage dairy
goat mGSCs and 11th passage mGSCs-I-SB cultured under normal
conditions using RNAiso (TaKaRa, Biotech. Co. Ltd.). The cDNA was
synthesized based on 500 mg RNA with a commercially available kit
(TaKaRa, Biotech. Co. Ltd.). The PCR steps included denature at 94°C
for 5 min, followed by repeated cycles 30 s at 95°C, 5558°C for 30 s,
72°C for 3060 s, 35 cycles,and extend at 72°C for 10 min. The primers
were designed basedon the sequences of the open reading frame from
the NCBI GenBank and synthesized by AuGCT Biotechnology
(Beijing). The PCR primers and the length of the amplied products
are shown in Table 1. The PCR products were analyzed by
electrophoresis in 2% agarose (Invitrogen) gel, stained with ethidium
bromide (Invitrogen), and visualized under UV illumination.
IMMUNOFLUORESCENCE STAINING
Dairy goat mGSCs (7th passage) and mGSCs-I-SB (13th passage)
cultured under normal conditions were xed with 4% PFA for 15 min at
room temperature, followed by three washes in cold PBS for 5 min each.
Washed cultures were treated with blocking solution (PBST þ1% BSA)
for a minimum of 30 min and incubated in primary antibodies including
Oct-4 (1:500, Chemicon), Sox2 (1:500, Chemicon), CD49f (1:500,
Chemicon), TERT (1:200, Santa Cruz), Oct4 (1:500, Abcam), Stra8
(1:500, Abcam), Blimp1 (1:200, Santa Cruz), Nanog (1:200, Chemicon).
The appropriate FITC conjugated secondary antibodies were used
according to the manufacturers manual (1:500, Chemicon). The nuclei
of cells were stained by Hoechst 33342. At the same time, the negative
controls were stained with the appropriate uorescent-conjugated
secondary antibodies [Yu et al., 2014].
IN VITRO DIFFERENTIATION OF mGSCs-I-SB
The 15th passage immortalized cells were dissociated into single cell
suspension and resuspended in DMEM (Invitrogen) containing 20%
FBS (Hyclone), 2 mM L-glutamine (Invitrogen), 1% non-essential
amino acids (Millipore), 100 U/ml penicillin and 100 mg/ml
streptomycin at 8001,000 cells/25 ml to form cell clusters. For
further differentiation, the embryoid bodies (EBs) (d3) cultured in
Petri dishes coated with 0.1% gelatin (Sigma) and DMEM medium
containing 20% FBS (Hyclone), 0.1 mM 2-mercaptoethanol (Sigma),
2 mM glutamine (Invitrogen), 1 mM sodium pyruvate (Invitrogen)
and 0.1 mM non-essential amino acids (Invitrogen) for 314 days to
investigate the potentiality of spontaneous differentiation [Zhu
et al., 2012]. And EBs were plated to induce the cells differentiate into
adipocyte, osteoblast and cartilage as previous study [Hua
et al., 2011; Hu et al., 2012; Qiu et al., 2012]. After differentiation,
the cells were determined by histochemistry and immunouores-
cence staining with the antibodies of all three germ layers: NSE
(Ectoderm), Islet1 (Mesoderm and Endoderm), PDX1 (Endoderm)
[Cao et al., 2011; Hua et al., 2011; Qiu et al., 2012].
DETECTION OF THE DIFFERENTIATION POTENTIAL OF
mGSCs-I-SB IN VIVO
The 17th passage of immortalized cells cultured under normal
conditions were transduced with the lentivirus expressing GFP for
12 h, then the cells were dissociated, collected and 0.5 ml working
solution containing the cells with an concentration of 10
7
cells/ml
injected into dorsal neural tube of 2.5 days chicken embryos (E2.5) to
evaluate whether the cells can differentiate into all three germ layers
in vivo. Then the injection hole was sealed with scotch tape and the
egg was returned to incubator until E6 for analysis.
After 3.5 days development, the chicken embryos were xed with
4% PFA for 24 h at 4°C, the oscillation sliced slides were produced
and analyzed through immunouorescence staining. The spinal
cords were coronally section at 80 mm by vibration microtome (VT
1000S, Leica, Germany). The sections were washed three times in
0.1 M PBS for 15 min each and then incubated at 4°C with primary
antibodies for 12 h. All three germ layers markers and the germline
specic markers were detected by immunouorescence staining
assay [Elena de Bellard and Bronner-Fraser, 2005; Boulland
et al., 2010]. Then sections were incubated with secondary antibodies
for 3 h at room temperature and then washed four times for 15 min
each at room temperature. The sections were then counterstained
TABLE 1. The Primers Were Used in Determining the Immortalized Dairy Goat mGSCs
Gene Sense primer Antisense primer Product size (bps) T
m
(°C) Reference sequence
b-Actin GCGGCATCCACGAAACTAC TGATCTCCTTCTGCATCCTGTC 138 58 NM_001101.3
VASA GCTGGCGTAATAGCGAAGAGG GCACAGATGCGTAAGGAGAAAA 107 58 KC189826.1
Gfra1 ATTTTATTACCTGCTGCCA ATTTCAATCATTCCTTCAT 197 51 NM_010279.2
CD117 TCCCAAACCTCAACACCGACAG GTGTAAGTGCCTCCTTCAGTCCC 153 58 NM_013598
CD90 GATCCAGGACTGAGCTCTCGG TCACGGGTCAGACTGAACTCATAC 195 58 NM_006288
CD49f CGAAGCACGAATCCCGAGAC TGCTCTACACGAACAATCGCTTT 235 58 NM_008397
Oct4A GACACCTGGCTTCCGACTTC GCTGAACACCTTCCCAAAGAG 533 59 NM_001265584.1
Bmi1 CCAGAGGGATGGACTGACGA GGGAACTGTGGGTGAGGAGA 147 59 NM_007552.4
Stra8 AAGGACAGCGGGGTTGAC TCGGGTTTTTTTGAGTTGC 170 56 JQ836663.1
PLZF CACCGCAACAGCCAGCACTAT CAGCGTACAGCAGGTCATCCAG 127 58 JX047313.1
NSE AAGGACAAATACGGCAAGG CAGGTCATCACCCACAATC 371 58 JN887466.1
Cyclin D1 TGAACTACCTGGACCGCT CAGGTTCCACTTGAGYTTGT 212 58 NM_053056.2
Sox2 GGCGGCAACCAGAAGAACAG GCATCTTGGGGTTCTCCTGG 109 58 JQ290347.1
Nanog GGAACTGCTGGGGAAAATTA TACAAATCTTCAGGCTGTATGTTG 118 58 FJ970651.1
CDK2 GCCAGGAGTTACTTCTATGC TGGAAGAAAGGGTGAGCC 180 58 NM_001798.3
PCNA AGTGGAGAACTTGGAAATGGAA GAGACAGTGGAGTGGCTTTTGT 167 58 NM_011045.2
exBmi1 TGTTCCCATAGTAACGCCAATA GGCATCAATGAAGTACCCTCC 644 58
SV40 TGACCTCCATAGAAGACACCG CAAATACCTCAGTTGCATCCC 386 58
JOURNAL OFCELLULAR BIOCHEMISTRY CHARACTERIZATION OFIMMORTALIZED DAIRY GOAT mGSCs 1551
with DAPI (1:1,000, Sigma) for 5 min, washed three times for 5 min
each, and nally mounted with a uorescent mounting medium
(Dako, Carpinteria, CA) on glass slides.
TRANSPLANTATION OF THE CELLS INTO RECEIPT
SPERMATOGENESIS DEFICIENCY MOUSE TESTIS
Male GSCs-I-SB (20th passage) were dissociated, and the cells
were collected through centrifuging at 1,500 r/min for 5 min. The
cells were counted and diluted with DMEM/F12 (Invitrogen)
supplemented with 10% FBS, and 10
5
cells/testis were then
transplanted into seminiferous tubules of the eight receipt
spermatogenesis deciency mouse, which were injected with
40 mg/kg weight busulfan (Sigma) and evaluated by previous
study [Choi et al., 2004]. After 2 months, the treated mice were
killed, and the recipients testes were collected. The characteristic
of mGSCs-I-SB was analyzed through supervision for GFP
expression of the seminiferous tubules and parafnslideswhich
were produced from the mouse testis transplanted with the
immortalized cells and untransplanted [Russell et al., 1996]. This
was regarded as the golden standard to determine whether the
putative cells maintain the capabilities of mGSCs which were
established by Brinster and Avarbock [1994] and Brinster and
Zimmermann [1994].
STATISTICAL ANALYSIS
Data are presented as mean SEM. Statistical comparisons were assessed
with analysis of Studentsttest. P<0.05 was considered statistically
difference and P<0.01 was considered signicantly difference.
Fig. 1. Dairy goat mGSCs-I-SB maintain greater proliferation ability compared with mGSCs. A: Growth curve. B: PDT analysis. C: BrdU Incorporation assay. D: Cell cycle analysis
for mGSCs-I-SB and wild mGSCs. E: PCR detection of the exogenous gene integration.
1552 CHARACTERIZATION OFIMMORTALIZED DAIRY GOAT mGSCs JOURNAL OFCELLULAR BIOCHEMISTRY
RESULTS
THE CELLSPROLIFERATION POTENTIAL BEFORE AND AFTER
IMMORTALIZATION
We isolated and enriched dairy goat mGSCs as we described
previously [Zhu et al., 2012]. Then the cells were immortalized by the
lentivirus carrying SV40 large T antigen and Bmi1 genes, and the
mGSCs-I-SB showed an obviously stronger proliferation than wild
mGSCs. Growth curve analysis showed that mGSCs-I-SB had a
higher cell number compared with dairy goat mGSCs (Fig. 1A). PDT
detection showed mGSCs-I-SB hold a lower PDT (28.5 h) to mGSCs
(45 h), which means an improvement of proliferation by immortali-
zation (Fig. 1B). BrdU Incorporation assay showed that over 50% of
mGSCs-I-SB were BrdU positive compared with 20% positive in
Fig. 2. The morphology alteration of mGSCs-I-SB from wild dairy goat mGSCs. The morphology from wild mGSCs (A) (Bar ¼100 mm) to mGSCs-I-SB (B) (Bar ¼50 mm) and
immunouorescence staining for wild mGSCs with antibodies including CD49f, c-Myc, DAZL, Sox2, and Stra8 (C) (Bar ¼200 mm).
JOURNAL OFCELLULAR BIOCHEMISTRY CHARACTERIZATION OFIMMORTALIZED DAIRY GOAT mGSCs 1553
dairy goat mGSCs (Fig. 1C). Also, cell cycle analysis showed that
dairy goat mGSCs to mGSCs-I-SB alteration accompanied an
increase of S phase cell ratio from 37.7% to 55.0% (Fig. 1D).
Moreover, the cells lifespan also showed a great improvement from
less than 10 passages to be easily subcultured up to 30 passages. In
summary, all these results demonstrated that mGSCs-I-SB maintain
a greater potential in proliferation compared to mGSCs.
To evaluate whether the cells were actually immortalized by SV40
large T antigen and Bmi1, we examined the genomic integration of
the exogenous genes. As indicated in Figure 1E, the exogenous
genes-SV40 large T antigen and Bmi1 were positive in mGSCs-I-SB
and negative in mGSCs. This further demonstrated that the
immortalization of dairy goat mGSCs was mediated by SV40 large
T antigen and Bmi1.
THE MARKERS EXPRESSED IN DAIRY GOAT mGSCs-I-SB
The mGSCs-I-SB exhibited spindle-like cell morphology compared to
the epithelioid-like cell morphology before immortalization (Fig. 2A,
B). Whether mGSCs-I-SB expressed the canonical mGSC markers seem
to be important to supply an intuitive evidence for mGSCs. Wedetected
the mGSC markers through RT-PCR and immunouorescencestaining
for the cells before and after immortalization. The immunouores-
cence staining showed dairy goat mGSCs were positive for most of the
male germ cell markers, such as CD49f, DZAL, Stra8, Sox2, andC-Myc
(Fig. 2C) before immortalization, and mGSCs-I-SB were also positive
for CD49f, CD90, Blimp1, C-Myc, Nanog, PLZF, Oct4, Stra8, and TERT
(Fig. 3A). The RT-PCR results showed that the cells were positive for
SSC specic markers such as VASA, GFRa1, CD117, CD90, CD49f,
Stra8, PLZF; they were also positive for pluripotency markers
including Oct4A, Nanog, Sox2; and proliferative markers such as
CDK2, CyclinD1, PCNA, and Bmi1 (Fig. 3B).
THE DIFFERENTIATION POTENTIAL OF mGSCs-I-SB IN VITRO
The cellspluripotency still need to be elucidated although the
associated markers were expressed. Then we examined the
differentiation potential in vitro by inducing the cells differentiation
into osteoblasts, chondrocytes, neural-like cells and adipocytes. The
EBs formed in suspension were collected and cultured under dened
solutions. When the cells were induced for a dened protocol as we
described previously [Hua et al., 2011], the cells displayed positive
for Oil Red O (adipocyte), Alcian blue (chondrocyte), Alizarin Red
(osteoblast) (Fig. 4A), and immunocytochemical staining showed
Fig. 3. Detection of the markers of mGSCs-I-SB. Immunouorescence staining for mGSCs-I-SB with antibodies including CD90, CD49f, Blimp1, c-Myc, Nanog, PLZF, Oct4,
Stra8 and TERT (A) (Bar ¼100 mm). RT-PCR detection for markers: Gfra1, CD117, CD90, CD49f, Vasa, Stra8, Sox2, PLZF, Oct4A, Nanog, CDK2, CyclinD1, and PCNA (B).
1554 CHARACTERIZATION OFIMMORTALIZED DAIRY GOAT mGSCs JOURNAL OFCELLULAR BIOCHEMISTRY
that the cells were differentiated into cells positive for NSE (ectoderm
marker), Islet1 (mesoderm and endoderm marker), PDX1 (endoderm
marker), Scp3 and VASA (germ cell marker) (Fig. 4B). All these
results suggested that mGSCs-I-SB maintain the potential to
differentiate into any of the three germ layers in vitro.
THE DIFFERENTIATION POTENTIAL OF mGSCs-I-SB IN VIVO
To determine the differentiation potential of mGSCs-I-SB in vivo,
the GFP labeled cells were transplanted into chicken embryosdorsal
neural tube under the protocol illustrated in Supplemented Fig. S1.
After 3 days, the GFP positive cells were found in recipients neural
Fig. 4. In vitro differentiation potential determination for mGSCs-I-SB. In vitro, mGSCs-I-SB can form EBs (from left to right, Bar ¼400, 200, 100 mm), then differentiate into
oil red O (Bar ¼100 mm), alcian blue (Bar ¼50 mm) and alizarin red (Bar ¼50 mm) positive cells in appropriate conditions (A). The cells exhibited positive for NSE, Islet1, PDX1,
Scp3, and VASA analyzed by immunouorescence staining (B) (Bar ¼100 mm).
JOURNAL OFCELLULAR BIOCHEMISTRY CHARACTERIZATION OFIMMORTALIZED DAIRY GOAT mGSCs 1555
tube and out everywhere (Fig. 5). The GFP positive cells were also
positive for the markers of all three germ layers including Nestin,
Islet1, and Glut2 (Fig. 5A), and the mGSC markers including CD49f,
GFRa1, and PLZF (Fig. 5B). All these results evidenced that the cells
maintain the differentiation potential to all three germ layers and
male germ cell lineages in vivo.
THE CELLS POTENTIAL TO SURVIVE IN MOUSE IMPAIRED
SEMINIFEROUS TUBULES
The most important characteristic of mGSCs is the potentiality to
reconstitute the impaired seminiferous tubules. The transplantation
assay showed that mGSCs-I-SB maintain the capacity of surviving in
the testis of mouse spermatogenesis deciency models (3 out of 8), and
no tumor structures was observed for at least two months through
supervision of the recipients seminiferous tubules and immunouo-
rescence staining (Fig. 6A,B). All these results indicated that mGSCs-I-
SB maintain the mGSC unique characteristic of surviving in the receipt
mouse spermatogenesis deciency model testis.
THE CELLS CAN BE INDUCED TO DIFFERENTIATE INTO
SPERM-LIKE CELLS
Male GSCs maintain the unique characteristic of differentiating into
sperm cells [Olive and Cuzin, 2005]. To further demonstrate the
immortalized cells maintain the capacity of mGSCs, the cells were
Fig. 5. In vivo differentiation potential of mGSCs-I-SB. When mGSCs-I-SB cells were transplanted and developed in chicken embryo for 3 days, the cells expressed all three germ
layers markers including Nestin, Islet1, Glut2 (A) (Bar ¼400 mm) and CD49f, GFRa1, PLZF (B) (Bar ¼63.5 mm).
1556 CHARACTERIZATION OFIMMORTALIZED DAIRY GOAT mGSCs JOURNAL OFCELLULAR BIOCHEMISTRY
cultured in suspension to form EBs (Fig. 7A). Then the EBs were
induced under 2 10
7
mM RA. After 3 days induction, more round
cells emerged in RA stimulation group than control; and this
tendency seemed to be more obvious when the cells were induced for
7 days. Immunocytochemical staining was carried on to determine
whether the induced cells maintain the characters of male germ cells.
The results showed that the cells were positive for Stra8 (pre-meiosis
marker), Scp3 (meiotic marker) and Acr (post-meiosis marker)
(Fig. 7B). These results showed that mGSCs-I-SB maintain the unique
capacity to differentiate into sperm-like cell (Fig. 7).
DISCUSSION
Previous reports on mGSCs were mainly in the mGSCsestablish-
ment, growth, dedifferentiation, and transplantation [Olive and
Cuzin, 2005]. All these progresses were mainly obtained on mouse
and human. For other species, especially livestock, there is a lack of
stable mGSC line. This greatly restricted the further study on the
utilization of mGSCs in preservation and optimization of germplasm
resources. Thus, the success in establishing immortalized dairy goat
mGSC lines may set a model for other speciesmGSCs.
Up to date, the immortalized germline cells including C-18-4, GC1,
GC2, etc. were established by exogenous SV40T, hTERT, and/or BmI1
[Bellvé et al., 1977]. SV40 large T antigen, a hexamer protein that is a
proto-oncogene derived from the polyoma virus SV40, is capable of
transforming many cell types [Bellvé et al., 1977]. The transforming
activity of T-tag is due in large part to its perturbation of the
retinoblastoma (pRB) and p53 tumor suppressor proteins [Yang
et al., 2007]. SV40 large T antigen has been shown to be the most
simple and reliable agent for thetransformation of many different cell
types in culture. Most importantly, viral genes induceimmortalization
by inactivating the tumor suppressor genes (p53, Rb, and others) that
can cause a replicative senescent state in cells. Recent studies have
also shown that SV40 large T antigen can induce telomerase activity
in the transduced cells. Additionally, T-tag binds to several other
cellular factors, including the transcriptional co-activators p300 and
CBP, which may contribute to its transformation function [Ali and
DeCaprio, 2001]. BMI1 (BMI1 polycomb ring nger oncogene), a
protein which in human is encoded by the BMI1 gene, has been
reported as an oncogene by regulating p16 and p19 that are cell cycle
inhibitor genes[Alkema et al., 1993]. Studies showed thatinhibition of
p16INK4a and introduction of hTERT can immortalize many types of
human cells with little chromosomal instability [Haga et al., 2007].
Fig. 6. Transplantation assay showed mGSCs-I-SB maintain the unique characteristic of mGSCs. After transplantation mGSCs-I-SB into receipt mouse spermatogenesis
deciency testisseminiferous tubules for more than 2 months, the GFP-labeled cells were supervised in the seminiferous tubules (A) (Bar ¼200mm), and the transplanted
mGSCs-I-SB cells existed and also were positive for GFP (B) (Bar¼100 mm).
JOURNAL OFCELLULAR BIOCHEMISTRY CHARACTERIZATION OFIMMORTALIZED DAIRY GOAT mGSCs 1557
Lentiviral vector can be used to integrate both dividing and non-
dividing cells as they can actively pass though nuclei membrane. In
addition, same as retrovirus, lentivirus will integrate into a host cell
genome. Lentivirus is gaining popularity for both in vitro and in vivo
applications of gene transduction. In this study, our results for the
rst time showed that overexpression of ectopic factors-SV40 large
T antigen and Bmi1 using lentivirus system may greatly improve the
proliferation of dairy goat mGSCs analyzed by PDT, BrdU
Incorporation assay and cell cycle. Meanwhile, the immortalized
cells maintain the similar biological characteristics as wild mGSCs
and previous reports on mouse mGSCs [Olive and Cuzin, 2005] when
cultured the cells in similar conditions [Feng et al., 2002]. And, the
cells expressed most of the conserved markers used in clarifying of
mGSCs (Supplemented Figs. S2S4). The immortalized cells also
maintain the capacity to differentiate into all three germ layers as
pluripotent mGSCs did as reported [Olive and Cuzin, 2005].
The golden standard method to examine the mGSCs is to transplant
the cells into the mouse spermatogenesis deciency model testis to
determine whether the cells can reconstitute the seminiferous tubules
which was established by Brinster and Avarbock [1994]. But this
standard is mainly used in mouse and rat. For other species, whether
the cells can survive for a long time is regularly evaluated [Olive and
Cuzin, 2005]. In our study, the immortalized dairy goat mGSCs
xenotransplantated into rodent models seminiferous tubules and
survived in recipients testis for more than 2 months, and this greatly
demonstrate the cells maintain the capabilities of mGSCs.
The chicken embryo is a classical animal model for studying
embryonic and fetal development and for xenotransplantation
experiments to study the behavior of cells in a standardized in vivo
environment [Boulland et al., 2010]. Compared with the mammals,
the main advantages of the chicken embryo are low cost, high
accessibility, ease of surgical manipulation and lack of a fully
developed immune system. Xenotransplantation into chicken
embryos can provide valuable information about cell proliferation,
differentiation and behavior, the responses of cells to signals in
dened embryonic tissue niches, and tumorigenic potential
[Boulland et al., 2010]. Transplanting cells into chicken embryos
can also be a step towards transplantation experiments in other
animal models. Recently the chicken embryo has been used to
evaluate the neurogenic potential of human stem and progenitor
cells following implantation into neural anlage [Boulland
et al., 2010]. In this study, we transplanted the GFP-labeled
mGSCs-I-SB into the developing central nervous system of the
chicken embryo. The results evidenced that mGSCs-I-SB cells can
survive, and meanwhile, and they differentiate into three germ
layers in chicken embryos analyzed by immunouorescence. This
study rst demonstrated chicken embryo was an efcient model to
study the behavior and function of mGSCs.
Male GSCs have the ability to differentiate into spermatogenic cells
through in vivo transplantation or through induction in vitro [Hua
et al., 2011]. The immortalized mGSCs-I-SB were differentiated into
sperm-like cells, and the induced cells were positive for Stra8 (a marker for
pre-meiosis germ cells), Scp3 (a marker for meiosis cells) and Acr (a marker
for post-meiosis cells). In combination with the transplantation assay, the
results support that mGSCs-I-SB maintain the characteristic of mGSCs.
Taken together, our results for the rst time demonstrate that the
immortalized dairy goat mGSCs were obtained through exogenous
expression of SV40 T antigen and Bmi1, and these cells have the
characteristics of mGSCs compared with the mainly existed studies
on mGSCs.
ACKNOWLEDGEMENTS
The authors appreciate Dr. Steven Gao and Dr. Xiang Chen for their
excellent revision and discussion. This work was supported by the
grants from the Program (31272518) of National Natural Science
Foundation of China, National Major Fundamental Research
Program of China (2013CB947900), Doctoral Fund of Ministry of
Fig. 7. The mGSCs-I-SB cells were differentiated into male germ cells in vitro.
A: EBs were formed (from left to right, Bar ¼50, 20 mm), mGSCs-I-SB were
induced to form large, round cells by RA, also, the sperm-like cells were
observed at 7 day induction by RA (D3, Bar ¼100 mm; D7, Bar ¼50 mm).
B: Identication of the mGSCs-I-SB cells induced by RA through
immunouorescence staining. The induced mGSCs-I-SB cells were positive
for STRA8, SCP3 (Bar ¼100 mm) and ACR (Bar ¼200 mm).
1558 CHARACTERIZATION OFIMMORTALIZED DAIRY GOAT mGSCs JOURNAL OFCELLULAR BIOCHEMISTRY
Education of China (RFDP, 20120204110030), the Fundamental
Research Funds for the Central Universities (QN2011012).
REFERENCES
Ali SH, DeCaprio JA. 2001. Cellular transformation by SV40 large T antigen:
Interaction with host proteins. Semin Cancer Biol 11:1523.
Alkema MJ, Wiegant J, Raap AK, Berns A, van Lohuizen M. 1993.
Characterization and chromosomal localization of the human proto-
oncogene BMI-1. Hum Mol Genet 2:15971603.
Anokye-Danso F, Trivedi CM, Juhr D, Gupta M, Cui Z, Tian Y, Zhang Y, Yang
W, Gruber PJ, Epstein JA, Morrisey EE. 2011. Highly efcient miRNA-
mediated reprogramming of mouse and human somatic cells to pluripotency.
Cell Stem Cell 8:376388.
Aponte PM, Soda T, Teerds KJ, Mizrak SC, van de Kant HJ, de Rooij DG. 2008.
Propagation of bovine spermatogonial stem cells in vitro. Reproduction
136:543557.
Bahadorani M, Hosseini SM, Abedi P, Hajian M, Hosseini SE, Vahdati A,
Baharvand H, Nasr-Esfahani MH. 2012. Short-term in-vitro culture of goat
enriched spermatogonial stem cells using different serum concentrations.
J Assist Reprod Genet 29:3946.
Ballow D, Meistrich ML, Matzuk M, Rajkovic A. 2006. Sohlh1 is essential for
spermatogonial differentiation. Dev Biol 294:161167.
Bellvé AR, Cavicchia JC, Millette CF, OBrien DA, Bhatnagar YM, Dym M.
1977. Spermatogenic cells of the prepuberal mouse isolation and
morphological characterization. J Cell Biol 74:6885.
Bi CM, Zhang SQ, Zhang Y, Peng SY, Wang L, An ZX, Qi A, Lv N. 2007.
Immortalization of bovine germ line stem cells by c-myc and hTERT. Anim
Reprod Sci 100:371378.
Boulland JL, Halasi G, Kasumacic N, Glover JC. 2010. Xenotransplantation of
human stem cells into the chicken embryo. J Vis Exp 11(41).pii: 2071. doi:
10.3791/2071.
Brinster RL, Avarbock MR. 1994. Germline transmission of donor haplotype
following spermatogonial transplantation. Proc Natl Acad Sci USA
91:1130311307.
Brinster RL, Zimmermann JW. 1994. Spermatogenesis following male germ-
cell transplantation. Proc Natl Acad Sci USA 91:1129811302.
Cao H, Chu Y, Zhu H, Sun J, Pu Y, Gao Z, Yang C, Peng S, Dou Z, Hua J. 2011.
Characterization of immortalized mesenchymal stem cells derived from
foetal porcine pancreas. Cell Prolif 44:1932.
Choi YJ, Ok DW, Kwon DN, Chung JI, Kim HC, Yeo SM, Kim T, Seo HG, Kim
JH. 2004. Murine male germ cell apoptosis induced by busulfan treatment
correlates with loss of c-kit-expression in a Fas/FasL- and p53-independent
manner. FEBS Lett 575:4151.
Conrad S, Renninger M, Hennenlotter J, Wiesner T, Just L, Bonin M, Aicher W,
Bühring HJ, Mattheus U, Mack A, Wagner HJ, Minger S, Matzkies M, Reppel
M, Hescheler J, Sievert KD, Stenzl A, Skutella T. 2008. Generation of
pluripotent stem cells from adult human testis. Nature 456:344349.
de Barros FR, Worst RA, Saurin GC, Mendes CM, Assumpção ME, Visintin JA.
2012. Alpha-6 integrin expression in bovine spermatogonial cells puried by
discontinuous percoll density gradient. Reprod Domest Anim 47:887890.
Dimri GP, Martinez JL, Jacobs JJ, Keblusek P, Itahana K, Van Lohuizen M,
Campisi J, Wazer DE, Band V. 2002. The Bmi-1 oncogene induces telomerase
activity and immortalizes human mammary epithelial cells. Cancer Res
62:47364745.
Elena de Bellard M, Bronner-Fraser M. 2005. Neural crest migration methods
in the chicken embryo. Methods Mol Biol 294:247267.
Feng LX, Chen Y, Dettin L, Pera RA, Herr JC, Goldberg E, Dym M. 2002.
Generation and in vitro differentiation of a spermatogonial cell line. Science
297:392395.
Guan K, Nayernia K, Maier LS, Wagner S, Dressel R, Lee JH, Nolte J, Wolf F, Li
M, Engel W, Hasenfuss G. 2006. Pluripotency of spermatogonial stem cells
from adult mouse testis. Nature 440:11991203.
Haga K, Ohno S, Yugawa T, Narisawa-Saito M, Fujita M, Sakamoto M,
Galloway DA, Kiyono T. 2007. Efcient immortalization of primary human
cells by p16INK4a-specic short hairpin RNA or Bmi-1, combined with
introduction of hTERT. Cancer Sci 98:147154.
He Z, Jiang J, Kokkinaki M, Golestaneh N, Hofmann MC, Dym M. 2008.
Gdnf upregulates c-Fos transcription via the Ras/Erk1/2 pathway to
promote mouse spermatogonial stem cell proliferation. Stem Cells 26:266
278.
He Z, Jiang J, Kokkinaki M, Dym M. 2009. Nodal signaling via an auto-
crine pathway promotes proliferation of mouse spermatogonial stem/
progenitor cells through Smad2/3 and Oct-4 activation. Stem Cells 27:
25802590.
Heidari B, Rahmati-Ahmadabadi M, Akhondi MM, Zarnani AH, Jeddi-
Tehrani M, Shirazi A, Naderi MM, Behzadi B. 2012. Isolation, identication,
and culture of goat spermatogonial stem cells using c-kit and PGP9. 5
markers. J Assist Reprod Genet 29:10291038.
Hofmann MC, Braydich-Stolle L, Dettin L, Johnson E, Dym M. 2005.
Immortalization of mouse germ line stem cells. Stem Cells 23:200210.
Hu Y, Sun J, Wang J, Wang L, Bai Y, Yu M, Lian Z, Zhang S, Hua J. 2012.
Characterization of female germ-like cells derived from mouse embryonic
stem cells through expression of GFP under the control of Figla promoter.
J Cell Biochem 113:11111121.
Hua J, Zhu H, Pan S, Liu C, Sun J, Ma X, Dong W, Liu W, Li W. 2011.
Pluripotent male germline stem cells from goat fetal testis and their survival
in mouse testis. Cell Reprogram 13:133144.
Izsvák Z, Fröhlich J, Grabundzija I, Shirley JR, Powell HM, Chapman KM,
Ivics Z, Hamra FK. 2010. Generating knockout rats by transposon
mutagenesis in spermatogonial stem cells. Nat Methods 7:443445.
Kanatsu-Shinohara M, Shinohara T. 2010. Germline modication using
mouse spermatogonial stem cells. Methods Enzymol 477:1736.
Kanatsu-Shinohara M, Miki H, Inoue K, Ogonuki N, Toyokuni S, Ogura A,
Shinohara T. 2005. Long-term culture of mouse male germline stem cells
under serum-or feeder-free conditions. Biol Reprod 72:985991.
Kanatsu-Shinohara M, Ikawa M, Takehashi M, Ogonuki N, Miki H, Inoue K,
Kazuki Y, Lee J, Toyokuni S, Oshimura M, Ogura A, Shinohara T. 2006.
Production of knockout mice by random or targeted mutagenesis in
spermatogonial stem cells. Proc Natl Acad Sci USA 103:80188023.
Kaul G, Kaur J, Rafeeqi TA. 2010. Ultrasound guided transplantation of
enriched and cryopreserved spermatogonial cell suspension in goats. Reprod
Domest Anim 45:e249e254.
Kaul G, Kumar S, Kumari S. 2012. Enrichment of CD9þspermatogonial stem
cells from goat (Capra aegagrus hircus) testis using magnetic microbeads.
Stem Cell Disco 2:9299.
Kubota H, Avarbock MR, Brinster RL. 2004. Culture conditions and single
growth factors affect fate determination of mouse spermatogonial stem cells.
Biol Reprod 71:722731.
Kubota H, Avarbock MR, Brinster RL. 2004. Growth factors essential for self-
renewal and expansion of mouse spermatogonial stem cells. Proc Natl Acad
Sci USA 101:1648916494.
Li YF, Guo YL, Li XH, Jin FS, Sun ZY. 2005. Long-term survival of human
spermatogonial stem cells in vitro and its functional identication.
Zhonghua Nan Ke Xue 11:886890,894.
Li S, Shi R, Wang Q, Cai J, Zhang S. 2012. Nanostructure and beta1-integrin
distribution analysis of pigs spermatogonial stem cell by atomic force
microscopy. Gene 495:189193.
Li M, Liu C, Zhu H, Sun J, Yu M, Niu Z, Liu W, Peng S, Hua J. 2013. Expression
pattern of Boule in dairy goat testis and its function in promoting the meiosis
in male germline stem cells (mGSCs). J Cell Biochem 114:294302.
JOURNAL OFCELLULAR BIOCHEMISTRY CHARACTERIZATION OFIMMORTALIZED DAIRY GOAT mGSCs 1559
Luo J, Megee S, Rathi R, Dobrinski I. 2006. Protein gene product 9.5 is a
spermatogonia-specic marker in the pig testis: Application to enrichment
and culture of porcine spermatogonia. Mol Reprod Dev 73:15311540.
Lübbe L, Rudolph M, Scherneck S, Zimmermann W, Geissler E. 1983.
Immortalization of human Lesch-Nyhan-broblasts following infection with
Simian virus. Arch Geschwulstforsch 53:105113.
McMillan M, Andronicos N, Davey R, Stockwell S, Hinch G, Schmoelzl S.
2013. Claudin-8 expression in Sertoli cells and putative spermatogonial stem
cells in the bovine testis. Reprod Fertil Dev, May 15. doi: 10.1071/RD12259.
[Epub ahead of print].
Naughton CK, Jain S, Strickland AM, Gupta A, Milbrandt J. 2006. Glial cell-
line derived neurotrophic factor-mediated RET signaling regulates sper-
matogonial stem cell fate. Biol Reprod 74:314321.
Niu Z, Goodyear SM, Rao S, Wu X, Tobias JW, Avarbock MR, Brinster RL.
2011. MicroRNA-21 regulates the self-renewal of mouse spermatogonial
stem cells. Proc Natl Acad Sci USA 108:1274012745.
Olive V, Cuzin F. 2005. The spermatogonial stem cell: From basic knowledge
to transgenic technology. Int J Biochem Cell Biol 37:246250.
Ozer HL. 2000. SV40-mediated immortalization. Progr Mol Subcell Biol
24:121153.
Petit CA, Gardes M, Feunteun J. 1983. Immortalization of rodent embryo
broblasts by SV40 is maintained by the A gene. Virology 127:7482.
Pietersen AM, Evers B, Prasad AA, Tanger E, Cornelissen-Steijger P, Jonkers
J, van Lohuizen M. 2008. Bmi1 regulates stem cells and proliferation and
differentiation of committed cells in mammary epithelium. Curr Biol
18:10941099.
Qiu P, Bai Y, Liu C, He X, Cao H, Li M, Zhu H, Hua J. 2012. A dose-dependent
function of follicular uid on the proliferation and differentiation of
umbilical cord mesenchymal stem cells (MSCs) of goat. Histochem Cell Biol
138:593603.
Russell LD, Franca LR, Brinster RL. 1996. Ultrastructural observations of
spermatogenesis in mice resulting from transplantation of mouse spermato-
gonia. J Androl 17:603614.
Saito M, Handa K, Kiyono T, Hattori S, Yokoi T, Tsubakimoto T, Harada H,
Noguchi T, Toyoda M, Sato S, Teranaka T. 2005. Immortalization of
cementoblast progenitor cells with Bmi-1 and TERT. J Bone Miner Res 20:
5057.
Shi W, Shi T, Chen Z, Lin J, Jia X, Wang J, Shi H. 2010. Generation of sp3111
transgenic RNAi mice via permanent integration of small hairpin RNAs in
repopulating spermatogonial cells in vivo. Acta Biochim Biophys Sin
42:116221.
Shinohara T, Kanatasu-Shinohara M. 2007. Culture and genetic modication
of mouse male germline stem cells. Tanpakushitsu Kakusan Koso 52-
(16Suppl):20922096.
Sikarwar AP, Reddy KV. 2008. SiRNA-mediated silencing of c-kit in mouse
primary spermatogonial cells induces cell cycle arrest. Oligonucleotides
18:145160.
Suzuki H, Ahn HW, Chu T, Bowden W, Gassei K, Orwig K, Rajkovic A. 2012.
SOHLH1 and SOHLH2 coordinate spermatogonial differentiation. Dev Biol
361:301312.
von Schonfeldt V, Wistuba J, Schlatt S. 2004. Notch-1, c-kit and GFRalpha-1
are developmentally regulated markers for premeiotic germ cells. Cytogenet
Genome Res 105:235239.
Yang G, Rosen DG, Mercado-Uribe I, Colacino JA, Mills GB, Bast RC , Jr.,
Zhou C, Liu J. 2007. Knockdown of p53 combined with expression of the
catalytic subunit of telomerase is sufcient to immortalize primary human
ovarian surface epithelial cells. Carcinogenesis 28:174182.
Yu M, Mu H, Niu Z, Chu Z, Zhu H, Hua J. 2014. MiR-34c enhances mouse
spermatogonial stem cells differentiation by targeting Nanos2. J Cell
Biochem 115:232242.
Zhang S, Li D, Li E, Li L, Wang J, Wang C, Lu J, Zhang X. 2008. Expression
localization of Bmi1 in mice testis. Mol Cell Endocrinol 287:4756.
Zhang S, Sun J, Pan S, Zhu H, Wang L, Hu Y, Wang J, Wang F, Cao H, Yan X,
Hua J. 2011. Retinol (vitamin A) maintains self-renewal of pluripotent male
germline stem cells (mGSCs) from adult mouse testis. J Cell Biochem
112:10091021.
Zhu H, Liu C, Li M, Sun J, Song W, Hua J. 2013. Optimization of the conditions
of isolation and culture of dairy goat male germline stem cells (mGSC). Anim
Reprod Sci 137:4552.
Zhu H, Liu C, Sun J, Li M, Hua J. 2012. Effect of GSK-3 inhibitor on the
proliferation of multipotent male germ line stem cells (mGSCs) derived from
goat testis. Theriogenology 77:19391950.
SUPPORTING INFORMATION
Additional supporting information may be found in the online
version of this article at the publishers web-site.
1560 CHARACTERIZATION OFIMMORTALIZED DAIRY GOAT mGSCs JOURNAL OFCELLULAR BIOCHEMISTRY
... Culture and Preparation of Dairy Goat SSCs. The procedures for isolating and purifying SSCs were in accor-dance with a previous study, and the morphology and function of SSCs we used have been verified [19][20][21]. The procedures for isolating and purifying SSCs are as follows. ...
... The pCDH-Eif2s3y and pCDH lentivirus were collected as described in Materials and Methods. The morphology and function of primary SSCs that we used to verify the direct effects of Eif2s3y have been verified in the past experimental studies [19,21]. The primary cells and pure spermatogonia are shown in Supplemental Figure 1A. ...
Article
Full-text available
The future fertility of males with cancer may be irreversibly compromised by chemotherapy and/or radiotherapy. Spermatogonial stem cell transplantation is believed to be a way to restore fertility in men. However, the survival efficiency of transplanted cells is still low. Eukaryotic translation initiation factor 2 subunit 3 and structural gene Y-linked (Eif2s3y) located on the Y chromosome of male animals is a coding gene of eIF2γ which mainly functions in translation initiation. Recently, the emerging role of Eif2s3y in spermatogenesis has been emphasized in several studies. However, the underlying mechanism is still unclear. In addition, how Eif2s3y functions in large animals remains largely unknown. In this study, we obtained the CDS sequence of the Eif2s3y gene from the testis of dairy goats and found that this gene was highly expressed in the testis and was evolutionarily conserved among different species. Interestingly, overexpression of Eif2s3y promoted the proliferation of spermatogonial stem cells of dairy goats by activating the ERK signaling pathway. In animal experiments, overexpressing Eif2s3y promoted transplanted goat spermatogonial stem cells and produced more colonies after microinjection into the seminiferous tubules of infertile mice. In conclusion, our study highlights an undiscovered role of Eif2s3y in dairy goat reproduction. This finding may provide an important basis for future works regarding male spermatogenic cell restoration and represent a major advance toward surrogate sires becoming a tool for disseminating and regenerating germplasm in all mammals.
Article
Full-text available
Genetic factors play a pivotal role in shaping feed formulation strategies for dairy goats, influencing milk production, composition, and overall productivity through the modulation of genetic parameters and polymorphisms associated with milk protein fractions and yield. Moreover, environmental challenges, including heat stress and feed availability fluctuations, pose significant hurdles in achieving optimal nutrition and performance, necessitating precision livestock farming and the utilization of local by-products to mitigate their impact on herd health and productivity. In addition, nutritional interventions, such as supplement utilization and alternative feed incorporation, offer promising avenues for addressing specific deficiencies and enhancing overall performance, from fatty acid supplementation to herbal feed additives, while balancing rations to meet evolving nutritional needs and ensuring long-term sustainability remains paramount for optimizing herd nutrition, health, and performance in intensive dairy goat production systems. The aim of this study was to explore the multifaceted influences of genetic factors, environmental challenges, and nutritional interventions on feed formulation strategies and overall herd nutrition, health, and performance in intensive dairy goat production systems.
Article
MicroRNAs (miRNAs), as post-transcriptional gene mediators, regulate the biological characteristics of spermatogonial stem cells (SSCs), including proliferation, differentiation and apoptosis. However, the potential roles and mechanisms by which miR-101-5p affected the biological characters of goat SSCs have not been fully elucidated. Herein, we reported that miR-101-5p overexpression decreased cell viability (P < 0.01), arrested cell cycle in the G1 phase (P < 0.05), and aggravated apoptosis of goat SSCs (P < 0.01) compared with negative control (NC), as determined by CCK-8 assay and flow cytometry analysis. Additionally, PCNA protein expression was attenuated by miR-101-5p overexpression (P < 0.05). Notably, the expression of SSCs specific genes Oct4 (P < 0.05), PLZF (P < 0.01) and DAZL (P < 0.01) were decreased in miR-101-5p overexpressed SSCs. Furthermore, the dual luciferase reporter assay showed that, when co-transfected with miR-101-5p mimics, the relative luciferase activity of EZH2 wide-type (WT) was inhibited (P < 0.05) compared with the transfection of EZH2 mutant (MUT). EZH2 expression was negatively correlated with miR-101-5p expression in goat SSCs. Collectively, our data implicates that miR-101-5p overexpression aggravates cell apoptosis, and suppresses cell proliferation of goat SSCs via targeting EZH2, which may impair spermatogenesis.
Article
At present, the knowledge base on characteristics and biology of spermatogonia in livestock is limited in comparison to rodents, yet the importance of studying these cells for comparative species analysis and enhancing reproductive capacity in food animals is high. Previous studies have established that although many core attributes of organ physiology and mechanisms governing essential cellular functions are conserved across eutherians, significant differences exist between mice and higher order mammals. In this chapter, we briefly discuss distinguishing aspects of testicular anatomy and the spermatogenic lineage in livestock and critical considerations for studying spermatogonial stem cell biology in these species.
Article
Full-text available
Spermatogonial stem cells (SSCs) are unique adult stem cells capable of self-renewal and differentiation into sperm. Grouper is a protogynous hermaphroditic fish farmed widely in the tropical and subtropical seas. In this study, we established an SSC line derived from adult testis of orange-spotted grouper, Epinephelus coioides. In the presence of basic fibroblast growth factor (bFGF) and leukemia inhibitory factor (LIF), the cells could be maintained with proliferation and self-renewal over 20 months and 120 passages under in vitro culture conditions. The cells exhibited strong alkaline phosphatase activity and the characteristics of SSCs with the expression of germ cell markers, including Vasa, Dazl, and Plzf, as well as the stem cell markers Nanog, Oct4, and Ssea1. Furthermore, the cultured cells could be induced by 11-ketotestosterone treatment to highly express the meiotic markers Rec8, Sycp3, and Dmc1, and produce some spherical cells, and even sperm-like cells with a tail. The findings of this study suggested that the cultured grouper SSC line would serve as an excellent tool to study the molecular mechanisms behind SSCs self-renewal and differentiation, meiosis during spermatogenesis, and sex reversal in hermaphroditic vertebrates. Moreover, this SSC line has great application value in grouper fish aquaculture, such as germ cell transplantation, genetic manipulation, and disease research.
Article
The busulfan, an alkylating agent, suppresses endogenous spermatogenesis in recipient testes. However, considering a wide variation in the effects of busulfan among animal species, its dosage and route of infusion need optimization to prepare effective and safe recipients. Thus, the current study aimed to create a suitable recipient goat model for germ cell (Gc) transplantation through a single intra-testicular (i.t.) busulfan infusion under ultrasonographic (USG) guidance. As observed through the infusion of trypan blue under USG guidance into mediastinum testis (MT) of pre-pubertal Barbari bucks, 3–5 mL of trypan blue solution could fill almost 80% of seminiferous tubules. Thereafter, in Experiment-1, the effect of different busulfan doses (mg/kg) i.e. 0 [negative control, Group (Gr) 1; 0 mg/kg-MT], 1 (Gr 2; 1 mg/kg-MT), 2 (Gr 3; 2 mg/kg-MT), and 3 (Gr 4; 3 mg/kg-MT) were studied. Further, in Experiment-2, sterilizing effects of busulfan infusion through two different routes [MT or cavum vaginale (CV)] were compared. Following i.t. busulfan treatment, no adverse physiological effects or body weight loss were detected. The histological analyses demonstrate a dose-dependent depletion of Gc with almost complete loss of Gc and spermatogenic activities in Gr 3 and 4, and extensive fibrosis in Gr 4. A considerable suppression of spermatogenesis marked with devoid of endogenous spermatogonial population and absence of significant (P > 0.05) effect on key hematological variables were observed in 2 mg/kg-MT Gr. These findings coupled with the results of significant (P < 0.05) down-regulation of marker genes of undifferentiated spermatogonia (THY-1 and PLZF), Gc pluripotency (UCHL-1, OCT-4, and DDX-4), and adhesion (E-cadherin and β-integrin); up-regulation of apoptotic genes (ID - 4 and BCL-6), and unchanged expression of Sertoli cell marker (vimentin), confirmed the safe and efficient depletion of endogenous Gc in 2 mg/kg-MT Gr. Furthermore, the effect of busulfan infusion on scrotal-testicular biometry, endocrine variables (plasma cortisol and testosterone), and Gc removal was more evident when busulfan was infused into MT than into CV. Overall, the results demonstrated that 2.0 mg/kg is an optimal single dose of busulfan when infused into the MT under USG guidance for the preparation of pre-pubertal recipient bucks. Overall, this study provides a basis to prepare suitable recipients through providing an available niche for efficient Gc transplantation in goats.
Article
Full-text available
RNA N6-methyladenosine (m6A) is essential for many bioprocesses in many species, but its role in goat testis development remains elusive, especially alkB homolog 5 (ALKBH5), one of the m6A demethylases. To this end, nine healthy Haimen goats of different ages were chosen randomly to provide testes. The results showed that the expression level of ALKBH5 was increased significantly (P < 0.05) in the 9-month group compared with the 0-day and 3-month groups, and ALKBH5 was located in goat spermatocytes with the highest expression level compared with Leydig cells and Sertoli cells. Thus, pcDNA3.1-ALKBH5 was constructed to explore the influences of the ALKBH5 increase in goat spermatogonial stem cells (SSC) in vitro. The results showed that the expression level of ALKBH5 in SSC transfected with pcDNA3.1-ALKBH5 (OE_ALKBH5) was significantly increased (P < 0.001) compared with that in SSC transfected with pcDNA3.1-EGFP (EGFP). With ALKBH5 overexpression in SSC, flow cytometry analysis showed that cells at G1 phase were significantly reduced (P < 0.01), while cells at S phase significantly increased (P < 0.01), and cell apoptosis was inhibited. Accordingly, the mRNA degradation of CCND1, CCNE1, and BCL2 was suppressed with ALKBH5 overexpression in SSC after treatment with actinomycin D. Furthermore, the mRNA levels of pluripotency maintenance- and cell differentiation-associated genes were changed between the two groups. Overall, the results indicated the crucial role of ALKBH5 during Haimen goat testis development. The results of this study provide a theoretical basis and technical means for RNA methylation participating in goat testis development.
Article
HT-2 toxin is widely found in moldy crops and is the major metabolite of T-2 toxin, which has been shown to exert various toxic effects in farm animals. However, little is known about the effects of HT-2 toxin on male reproduction, particularly spermatogenesis. This study aims to investigate the toxic effects of HT-2 toxin on goat spermatogonial stem cells (SSCs) and related autophagy-regulated mechanisms. Our results showed that HT-2 toxin exposure resulted in decreased cell viability and proliferation, disrupted SSCs self-renewal, and reduced germ cell-related gene expression. HT-2 toxin exposure also induced oxidative stress and cell apoptosis, as shown by ROS accumulation, increased antioxidant enzyme activity levels, decreased the mitochondrial membrane potential, and increased caspase-9 mRNA and Bcl/bax protein levels. Additionally, HT-2 toxin exposure increased the expression of the autophagy-inducing genes Atg5, Atg7 and Beclin1 and the number of autophagosomes, which indicated that HT-2 toxin induced autophagy in the goat SSCs. Moreover, we also examined a possible mechanism by which HT-2 toxin exposure induced higher expression of AMPK, mTOR and ULK at both the mRNA and protein levels. our results indicated that HT-2 toxin caused apoptosis and autophagy by activating AMPK-mTOR-ULK1 pathway, which further affected SSCs viability.
Article
Orchitis is one of the leading causes of male animal infertility and is associated with inflammatory reactions caused by the bacterium. It has been reported that there is a mutual coupling effect between endoplasmic reticulum stress (ERS) and inflammatory response. Our studies showed that lipopolysaccharide (LPS) could cause testicular damages, apoptosis, ERS, and inflammatory responses in spermatogonial stem cells (SSCs); ERS‐related apoptosis proteins were activated and the expression of ERS genes was significantly upregulated; meanwhile, the expression of Toll‐like receptor 4 and inflammation factors was apparently increased with LPS treatment. Moreover, melatonin (MEL) could rescue testicular damage, and significantly inhibited the expression of ERS‐related apoptosis genes, ERS markers, and inflammatory factors in SSCs and MEL played repairing and anti‐infection roles in LPS‐induced testicular damage. Therefore, MEL may be used as a drug to prevent and control bacterial infections in male reproductive systems. However, the specific molecular mechanism of MEL to resist ERS and inflammatory response remains to be further studied. Melatonin (MEL) could play a significant role in lipopolysaccharide (LPS)‐induced endoplasmic reticulum stress (ERS) and inflammatory responses in spermatogonial stem cells (SSCs). LPS could cause testicular damage and induce apoptosis, ERS, and inflammatory responses in SSCs, while exogenous MEL can inhibit the expression of ERS‐related apoptosis marker genes and inflammatory factors, at the same time, exogenous MEL could also alleviate testicular damage caused by LPS.
Article
Full-text available
The well documented source for adult multipo-tent stem cells is spermatogonial stem cells (SSCs) of mammalian testis. It is foundation of spermatogenesis in the testis throughout adult life by balancing self-renewal and differentiation. SSCs isolation from mammalian testis is difficult because of their scarcity and the lack of well characterized cell surface markers. Thus, the isolation of SSCs is of great interest for explo-ration of spermatogonial physiology and thera-peutic approaches for fertility preservation. CD9 is a surface marker expressed in mouse and rat male germline stem cells. In this study, CD9 positive SSCs were successfully isolated from the goat testis using enzymatic digestion fol-lowed by three step purification: Differential plating, percoll discontinuous density gradient followed by magnetic activated cell sorting (MACS). Percoll discontinuous density gradient showed significant differences in the percentage of CD9 + SSCs across individual fraction. The fraction 36% and 40% gave the highest per-centage of CD9 + SSCs i.e. 82% ± 1.2% and 9.2% ± 1.3% respectively. Magnetic activated cell sort-ing of CD9 + cells in the magnetic fraction of goat testes was in the range of 15% -18% which is upto threefolds. CD9 + SSCs were further recov-ered with appreciable efficiency after immu-nomagnetic isolation by using various bead: cells ratio in which 4:1 ratio gave the highest yield of 69.06 × 10 5 with 18% of CD9 + SSCs. Magnetic activated cell sorting using anti-CD9 antibodies provides an efficient and fast ap-proach as compared to conventional approaches such as differential plating and percoll discon-tinuous density gradient for enrichment strategy for spermatogonial stem cells from goat testes for undertaking research on basic and applied reproductive biology.
Article
Full-text available
Male germline stem cells (mGSC) reside in the basement of seminiferous tubules of the testis and have the capacity of self-renewal and differentiation into sperm throughout the life of animals. Reports on mice and human mGSC have demonstrated that mGSC are an unlimited resource of pluripotent stem cells for sperm production. The conditions of isolation and culture of mouse and human mGSC are well developed; however, the systematic culture conditions of dairy goat mGSC are still deficient although there have been several reports of successful cultures. With the present research, several key elements of isolation and culture of dairy goat mGSC have been determined. Details for the conditions of isolation of dairy testicular spermatogonium cells were optimized, and effects of several extracellular matrix types, ages of dairy goat, and cytokines on enrichment and culture of mGSC were compared. Biological characteristics of the cells were also evaluated by RT-PCR and immunofluorescent staining. The results indicated there is one kind of enzyme cocktail (CTHD (1mg/ml collagenase, 10μg/ml DNase, 1mg/ml hyaluronidase and 1mg/ml trypsin) combined TD (0.25% trypsin and 10mg/ml DNaseI)) that can be used to successfully isolate dairy goat testicular spermatogonium cells efficiently; and fibronectin as well as laminin were efficient extracellular matrix to enrich mGSC among the extracellular matrix types evaluated. Age of dairy goat clearly influenced the cultures of dairy goat mGSC with the efficiency of establishment of an mGSC line being greater if the age of the dairy goat is younger. Some cytokines e.g. BIO (A GSK3 inhibitor, 6-bromoindirubin-3'-oxime) and basic fibroblast growth factor (bFGF) acted positively on the maintenance of proliferation and pluripotency of mGSC. Leukemia inhibitory factor (LIF) might, however, inhibit the proliferation of dairy goat mGSC. These cultured mGSC maintained similar characteristics as mouse and human mGSC. These results provide an efficient system to isolate and culture of dairy goat mGSC.
Article
Full-text available
Introduction Presently the techniques for making transgenic animals are cumbersome, required costly instruments and trained man-power. The ability of spermatogonial stem cells (SSCs) to integrate foreign genes has provided the opportunity for developing alternate methods for generation of transgenic animals. One of the big challenges in this field is development of the methods to identify and purify donor SSCs by antibody mediated cell sorting. Purpose The present study was aimed to identify goat subpopulations of SSCs using polyclonal antibodies against PGP9.5 and c-kit molecular markers as well as the growth characteristics of SSCs during short term culture. Methods One month old goats’ testicular samples were subjected for immunohistochemical and immunocytochemical evaluations. The enzymatically isolated SSCs were cultured in DMEM plus FCS supplemented with (treatment) or without (control) growth factors (GDNF, LIF, FGF, and EGF) for 2 weeks. At the end of culture the morphological characteristics of SSCs colonies and immunocytochemical staining were evaluated. Results The number and size of colonies in treatment groups were significantly (P < 0.01) higher than corresponding values in controls. The presence of PGP 9.5 and c-kit antigens was confirmed in immunocytochemical evaluation. In immunocytochemical evaluation, the proportion of c-kit and PGP9.5 positive cells were significantly (P < 0.001) higher in control and treatment groups, respectively. Conclusions The presence of PGP9.5 and c-kit antigens was confirmed in goat SSCs. Moreover, culture medium supplementation with growth factors could effectively retain the undifferentiation status of SSCs, reflected as a higher population of PGP9.5 positive cells, after short term culture.
Article
Spermatogenesis is the process that involves the division and differentiation of spermatogonial stem cells into spermatozoa. However, the autocrine molecules and signaling pathways controlling their fate remain unknown. This study was designed to identify novel growth factors and signaling pathways that regulate proliferation, differentiation, and survival of spermatogonial stem/progenitor cells. To this end, we have for the first time explored the expression, function, and signaling pathway of Nodal, a member of the transforming growth factor-β superfamily, in mouse spermatogonial stem/progenitor cells. We demonstrate that both Nodal and its receptors are present in these cells and in a spermatogonial stem/progenitor cell line (C18-4 cells), whereas Nodal is undetected in Sertoli cells or differentiated germ cells, as assayed by reverse transcription-polymerase chain reaction, Western blots, and immunocytochemistry. Nodal promotes proliferation of spermatogonial stem/progenitor cells and C18-4 cells, whereas Nodal receptor inhibitor SB431542 blocks their propagation as shown by proliferation and bromodeoxyuridine incorporation assays. Nodal knockdown by RNA interference results in a marked increase of cell apoptosis and a reduction of cell division as indicated by terminal deoxynucleotidyl transferase dUTP nick-end labeling and proliferation assays. Conversely, overexpression of Nodal leads to an increase of cell proliferation. Nodal activates Smad2/3 phosphorylation, Oct-4 transcription, cyclin D1, and cyclin E expression, whereas SB431542 completely abolishes their increase. Together, Nodal was identified as the first autocrine signaling molecule that promotes proliferation of mouse spermatogonial stem/progenitor cells via Smad2/3 and Oct-4 activation. This study thus provides novel and important insights into molecular mechanisms regulating proliferation and survival of spermatogonial stem/progenitor cells. STEM CELLS 2009;27:2580–2590
Article
MiRNAs are expressed in many mammalian cells, acting specific roles in regulating gene expression or mediating special mRNAs cleavage by targeting their 3'UTR. Some miRNAs are essential and important for animal development. However, it is still unclear what the relationship is between miR-34c and mammalian spermatogonial stem cells (SSCs). We found that a conserved microRNA-34c through its target-Nanos2, regulating SSCs' differentiation in mouse. Immunohistochemistry analysis of Nanos2 and miR-34c FISH results revealed the opposite expression trends between them. Seven bioinformatics websites and programs predicted that miR-34c has interaction sites in Nanos2's 3' untranslated region (3'UTR). Dual-luciferase reporter vector and mutated dual-luciferase reporter vector analysis validated that they are interacted. After transfection miR-34c mimics into mouse SSCs, or miR-34c lentiviral vector in vitro co-cultivation with seminiferous tubules, and western blot analysis demonstrated that miR-34c over-expression could suppress Nanos2 expression in post-transcription level. Our experiments identified that miR-34c may promote meiosis process by interacting with Nanos2 leading up-regulation of Stra8 in mouse spermatogonial stem cells. J. Cell. Biochem. © 2013 Wiley Periodicals, Inc.
Article
The proto-oncogene bml -1 is frequently activated by Moloney murine leukemia proviral insertions in E μ-mcy transgenic mice1,2 Using a mouse bmi -1 cDNA probe a transcript of 3.3 kb was detected on Northern blots of human Burkitts lymphoma cell lines. We have Isolated and sequenced cDNA clones from a human erythroleukemia cell line (K562) derived cDNA library, using different mouse bml -l cDNA fragments as a probe. Analysis of genomic BMI-1 sequences reveals a gene structure which is very similar to that of the mouse, consisting of at least 10 exons. The human cDNA is 3203 bp in length and shows 86&percnt; identify to the mouse nucleotide sequence. The open reading frame encodes a protein of 326 amino acids which shares 98&percnt; Identity to the amino acid sequence of mouse bmi-1 protein. In vitro translation experiments show that human cDNA derived RNA translates into a protein with a mobility of 44–46 kD on SDS polyacrylamide gels. Fluorescence In situ hybridization (FISH) on metaphase chromosome spreads located the human BMI-1 gene to the short arm of chromosome 10 (10p13), a region known to be involved in translocations in various leukemias.
Article
Adhesion molecules are expressed by both adult and embryonic stem cells, with different classes of adhesion molecules involved in cell-membrane and intercellular contacts. In this study the expression of the adhesion molecule claudin-8 (CLDN8), a tight-junction protein, was investigated as a potential marker for undifferentiated spermatogonia in the bovine testis. We found that CLDN8 was expressed by both spermatogonia and a subset of Sertoli cells in the bovine testis. We also showed co-expression of GFRα1 in testis cells with CLDN8 and with Dolichos biflorus agglutinin- fluorescein isothiocyanate (DBA-FITC) staining. We observed co-enrichment of spermatogonia and CLDN8-expressing Sertoli cells in DBA-FITC-assisted magnetic-activated cell sorting (MACS), an observation supported by results from fluorescence-activated cell sorting analysis, which showed CLDN8-expressing cells were over-represented in the MACSpositive cell fraction, leading to the hypothesis that CLDN8 may play a role in the spermatogonial stem-cell niche.
Article
Boule is a conserved gene in meiosis, which encodes RNA binding protein required for spermatocyte meiosis. Deletion of Boule was found to block meiosis in spermatogenesis which contribute to infertility. Up to date, the expression and function of Boule in the goat testis are not known. The objectives of this study were to investigate the expression pattern of Boule in dairy goat testis and their function in male germline stem cells (mGSCs). The results first revealed that the expression level of Boule in adult testes was significantly higher than younger and immature goats, and azoospermia and male intersex testis. Over-expression of Boule promoted the expression of meiosis-related genes in dairy goat mGSCs. The expression of Stra8 was up-regulated by over-expression of Boule analysed by western blotting and Luciferase reporter assay. While, Cdc25a, the downstream regulator of Boule, was found not to affect the expression of Stra8, and our data illustrated that Cdc25a did not regulate meiosis via Stra8. The expression of Stra8 and Boule was up-regulated by RA induction. Taken together, results suggest the Boule plays an important role in dairy goat spermatogenesis and that over-expression of Boule may promote spermatogenesis and meiosis in dairy goat. J. Cell. Biochem. © 2012 Wiley Periodicals, Inc.