• No results found

Cleavage pattern and survivin expression in porcine embryos by somatic cell nuclear transfer

N/A
N/A
Protected

Academic year: 2021

Share "Cleavage pattern and survivin expression in porcine embryos by somatic cell nuclear transfer"

Copied!
10
0
0

Loading.... (view fulltext now)

Full text

(1)

Cleavage pattern and survivin expression in porcine embryos by

somatic cell nuclear transfer

Yubyeol Jeon

a

, Se Heon Jeong

a

, Dibyendu Biswas

a

, Eui Man Jung

b

, Eui Bae Jeung

b

,

Eun Song Lee

c

, Sang-Hwan Hyun

a,

*

aLaboratory of Veterinary Embryology and Biotechnology, College of Veterinary Medicine, Chungbuk National University, Cheongju-361-763, Chungbuk, South Korea

bLaboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju-361-763, Chungbuk, South Korea

cSchool of Veterinary Medicine Kangwon National University Chunchon 200-701, Kangwon, South Korea Received 20 November 2010; received in revised form 28 March 2011; accepted 3 April 2011

Abstract

Mammalian embryos produced in vitro show a high rate of early developmental failure. Numerous somatic cell nuclear transfer (SCNT) embryos undergo arrest and show abnormal gene expression in the early developmental stages. The purpose of this study was to analyze porcine SCNT embryo development and investigate the cause of porcine SCNT embryo arrest. The temporal cleavage pattern of porcine SCNT embryos was analyzed first, and the blastocyst origin at early developmental stage was identified. To investigate markers of arrest in the cleavage patterns of preimplantation SCNT embryos, the expression of survivin—the smallest member of the inhibitor of apoptosis (IAP) gene family, which suppresses apoptosis and regulates cell division—was compared between embryos showing normal cleavage and arrested embryos.

A total of 511 SCNT embryos were used for cleavage pattern analysis. Twenty-four hours post activation (hpa), embryos were classified into five groups based on the cleavage stage as follows; 1-cell, 2-cell, 4-cell, 8-cell and fragmentation (frag). In addition, 48 hpa embryos were more strictly classified into 15 groups based on the cleavage stage of 24 hpa; 1–1 cell (24 hpa– 48 hpa), 1–2 cell, 1– 4 cell, 1– 8 cell, 1 cell-frag, 2–2 cell, 2– 4 cell, 2– 8 cell, 2 cell-frag, 4 – 4 cell, 4 – 8 cell, 4 cell-frag, 8 – 8 cell, 8 cell-frag, and frag-frag. These groups were cultured until 7 d post activation, and were evaluated for blastocyst formation. At 24 hpa, the proportion of 2-cell stage was significantly higher (44.5%) than those in the other cleavage stages (1-cell: 13.4%; 4-cell: 17.9%; 8-cell: 10.3%; and frag: 13.9%). At 48 hpa, the proportion of embryos in the 2– 4 cell stage was significantly higher (32.4%) than those in the other cleavage stages (2– 8 cell: 8.2%; 4 – 8 cell: 12.1%; and frag-frag: 13.9%). Some embryos arrested at 48 hpa (1–1 cell: 5.8%; 2–2 cell: 2.8%; 4 – 4 cell: 3.8%; 8 – 8 cell: 6.5%; and total arrested embryos: 18.9%). Blastocyst formation rates were higher in 2-4 cell cleavage group (20.2%) than in other groups. SCNT embryos in 2– 4 cell stage showed stable developmental competence. In addition, we investigated survivin expression in porcine SCNT embryos during the early developmental stages. The levels of survivin mRNA in 2-cell, 4-cell stage SCNT embryos were significantly higher than those of arrested embryos. Survivin protein expression showed a similar pattern to that of survivin mRNA. Normally cleaving embryos showed higher survivin protein expression levels than arrested embryos. These observations suggested that 2– 4 cell cleaving embryos at 48 hpa have high developmental competence, and that embryonic arrest, which may be influenced by survivin expression in porcine SCNT embryos.

© 2011 Elsevier Inc. All rights reserved.

Keywords: SCNT; Cleavage pattern; Embryo arrest; Survivin

* Corresponding author. Tel.:⫹82-43-261-3393; fax: ⫹82-43-267-3150.

E-mail address:shhyun@cbu.ac.kr(S.H. Hyun).

Theriogenology 76 (2011) 1187–1196

www.theriojournal.com

(2)

1. Introduction

The somatic cell nuclear transfer (SCNT) technique is a useful tool for biomedical studies. Specifically, pig cloning by SCNT can be used for xenotransplantaion and the development of animal models for human dis-eases [1,2]. Indeed, pigs share many characteristics with humans, such as anatomy, physiology and body size [3]. However, unsolved problems exist with the current cloning technology. For example, animal clon-ing has low efficiency and cloned animal can display abnormal development [4 –7]. One critical factor of cloned embryo developmental failures is aberrant epi-genetic reprogramming [8]. For a donor nucleus to support development into a clone, the nucleus must be reprogrammed into a state compatible with normal em-bryonic development[9]. However, aberrant epigenetic reprogramming often occurs due to abnormal gene ex-pression or silencing. Numerous aberrations have been observed, ranging from suppression of gene expression to de novo overexpression or more frequently to sig-nificant up- or down-regulation of a specific gene[10 – 15]. Therefore, cloning efficiency after embryo transfer (ET) is still very low.

The ability to reliably and noninvasively predict which early embryos have the ability to develop to full-term would constitute a major breakthrough in em-bryo-based biotechnologies[16]. This could lead to the successful production of cloned animals by ET embryo selection. Embryos that exhibit continuing develop-mental competence could be selected by cleavage pat-tern analysis. In vitro-produced mammalian embryos have a high frequency of early developmental failure. Approximately 10 –15% of IVF embryos permanently arrest in mitosis at the 2- to 4-cell cleavage stage[17]. SCNT embryos are similar. The reasons for this em-bryo demise remain unclear. Arrested emem-bryos derived from IVF do not display apoptotic characteristics[18 – 23], while SCNT arrested embryos do so,[24], proba-bly because nuclear reprogramming of the differenti-ated somatic cell genome occurs gradually over two or three cell cycles or aberrant nuclear reprogramming

[25]. Aberrant nuclear reprogramming causes abnormal gene expression, including anti-apoptic and cell prolif-eration genes.

Survivin, an apoptotic protein inhibitor containing a single baculovirus, inhibits apoptotic protein repeat do-mains[26]. It is a member of the chromosomal passen-ger complex (CPC), which is implicated in kineto-chore attachment, bipolar spindle formation, and cytokinesis [27]. Survivin is a bifunctional protein that suppresses apoptosis and regulates cell division

[28]. It is not expressed in most normal adult tissues, but is highly expressed in solid and hematological malignancies, where it has been linked to increased angiogenesis and tumorigenesis[29,30]. During human, bovine, and murine embryonic development, survivin is ubiquitously expressed [31]. Survivin dsRNA-targeted embryos have decreased developmental rates as blasto-cysts and an increased in the apoptosis index [28]. This study demonstrated that survivin is critical for normal embryonic development.

The aim of this study was to analyze the temporal developmental pattern of porcine SCNT embryos and to investigate survivin expression-related embryonic arrest in porcine SCNT embryos. First, the temporal SCNT embryo developmental pattern was analyzed and the origin of successfully developed blastocysts was identified. Then, to determine whether survivin expres-sion was related to embryonic arrest, survivin mRNA and protein expression was compared between normal embryos and arrested embryos.

2. Materials and methods

2.1. Chemicals

All chemicals were purchased from Sigma-Aldrich Chemical Company (St. Louis, MO, USA) unless stated otherwise.

2.2. Ovary collection, recovery, and in vitro oocyte maturation

(3)

mm follicles of prepubertal gilt ovaries. After centrifugation at 1600⫻ g for 30 min, the supernatants were collected and filtered sequentially through 1.2 and 0.45␮M syringe filters (Gelman Sciences, Ann Arbor, MI, USA). The prepared pFF was then stored at –20 °C until use. For maturation, the selected COCs were washed three times in oocyte maturation medium containing hormone supplements, and approxi-mately 50–60 oocytes were transferred into each well of a 4-well Nunc dish (Nunc, Roskilde, Denmark) containing 500 ␮l of culture medium and equilibrated at least 2 h with 5% CO2at 39 °C in a humidified atmosphere. After 20–22 h of

maturation with hormones, the oocytes were washed two times in a maturation medium without hormone supplements and then cultured for 20–22 h without hormone supplements at 39 °C under 5% CO2in air.

2.3. Donor cell preparation

Fibroblasts were isolated from adult pig’s ear. The hair and other soft tissues were removed using iris scissors and watchmaker’s forceps and discarded. After washing twice with dPBS (Invitrogen, Carlsbad, CA, USA), the tissue was minced with a surgical blade on a 100 mm culture dish. The minced ear tissues were dissociated in DMEM (Invitrogen) supplemented with 0.1% (w/v) trypsin and 1 mM EDTA (Invitrogen) for 1–2 h. Trypsinzed cells were washed once by centrifugation at 300 ⫻ g for 10 min and subsequently seeded into 100 mm plastic culture dishes. Seeded cells were then cultured for 6–8 d in DMEM supplemented with 10% (v/v) fetal bovine serum (FBS; Invitrogen), 1 mM sodium pyruvate, 1% (v/v) non-essential amino acids (Invitrogen), and 10 mg/ml penicillin-streptomycin solution at 39 °C in a humidified atmosphere of 5% CO2and 95% air. After

re-moving the unattached clumps of cells or explants, the at-tached cells were further cultured until confluent. Subcultur-ing was done at intervals of 5–7 d by trypsinization for 2 min using 0.1% trypsin and 0.02% EDTA. The cells were then stored in freezing medium in liquid nitrogen after two pas-sages. The freezing medium consisted of 70% (v/v) DMEM, 10% (v/v) DMSO, and 20% (v/v) FBS. Prior to SCNT, the cells were thawed and subsequently cultured in 10% FBS with DMEM for 3–4 d until 80% confluence. The individual cells were retrieved from the monolayer by trypsinization for ⬃1 min and subsequently used for SCNT.

2.4. Micromanipulation for somatic cell nuclear transfer, fusion, and activation

The After 40 h of IVM, a denuded oocytes were incubated for 5 min in a manipulation medium (calci-um-free TLH-BSA) containing 5 mg/mL Hoechst 33342 and then washed twice with fresh manipulation medium, The washed oocytes were transferred into a

drop of manipulation medium containing 5 mg/mL cytochalasin B and were overlaid with mineral oil. Oocytes were enucleated by aspirating the PB and MII chromosomes using a 16␮m glass pipette (Humagen, Charlottesville, VA). After enucleation, using a fine injecting pipette, a 12–15 ␮m trypsinized fetal fibro-blast with a smooth cell surface was transferred into the perivitelline space of an enucleated oocyte. The cou-plets were equilibrated with 0.28 M mannitol solution containing 0.5 mM HEPES, 0.1 mM CaCl2, and MgSO4 for 2–3 min and transferred to a fusion cham-ber containing two electrodes overlaid with mannitol solution. Membrane fusion was induced by applying an alternating current (AC) field of 2V cycling at 1 MHz for 2 sec, followed by two pulses of 160V/mm direct current (DC) for 60␮sec using a cell fusion generator (LF101; NepaGene, Chiba, Japan). Activated oocytes were washed 3– 4 times with TLH-BSA. After 1 h, the fusion was checked, and fused, properly shaped oocytes were washed 3– 4 times and further cultured with PZM3 media covered with prewarmed mineral oil and then incubated under 5% O2, 5% CO2, and 90% N2 at 39 °C. Cleavage and blastocyst formation were evalu-ated under a stereomicroscope at 24, 48 and 168 h post activation (hpa), respectively.

2.5. Classification of cleavage pattern

The embryos were assessed for cleavage on day 1 (24 hpa), day 2 (48 hpa) and for blastocyst development on day 7. The day of SCNT was day 0. At 24 hpa, embryos were classified into five groups based on the cleavage state as follows; 1 cell, 2 cell, 4 cell, 8 cell and fragmentation (frag). At 48 hpa, embryos were more strictly classified into fifteen groups based on the cleav-age state of 24 hpa; 1–1 cell (24hpa– 48hpa), 1–2 cell, 1– 4 cell, 1– 8 cell, 1 cell-frag, 2–2 cell, 2– 4 cell, 2– 8 cell, 2 cell-frag, 4 – 4 cell, 4 – 8 cell, 4 cell-frag, 8 – 8 cell, 8 cell-frag, and frag-frag (seeFig. 1). These groups were cultured until 7 d post activation, and were eval-uated for blastocyst formation.

2.6. Semiquantitative PCR

(4)

measuring the absorbance at 260 nm. First strand com-plementary DNA (cDNA) was prepared by subjecting total RNA (1␮g) to reverse transcription using mMLV reverse transcriptase (Invitrogen Co.) and random primers (9-mers; TaKaRa Bio Inc., Otsu, Shiga, Japan). To determine the conditions for logarithmic phase PCR amplification for target mRNA, aliquots (1 ␮g) were amplified using different numbers of cycles. The 1A

gene was PCR-amplified to rule out the possibility of RNA degradation and was used to control for variation in mRNA concentrations in the RT reaction. A linear relationship between PCR product band visibility and number of amplification cycles was observed for target mRNAs. The 1A and survivin genes were quantified using 28 and 30 cycles, respectively. The cDNA was amplified in a 20 ␮l PCR reaction containing 1 U Taq polymerase (iNtRON Bio Inc.), 2 mM dNTP, and 0.1␮M specific primers. PCR reactions were denatured at 95 °C for 30 sec, annealed at 58 °C for 30 sec, and extended at 72 °C for 30 sec. PCR products (8␮l) were fractionated on a 2.3% agarose gel, stained with ethidium bromide, and photographed under UV illumination. The photograph was scanned and analyzed using a Gel Doc EQ system (Bio-rad Laboratories, Inc.). The oligo primers for all PCR reactions are indicated inTable 1.

2.7. Immunofluorescence

Oocytes and embryos at different stages [metaphase II (MII), normal cleaved embryo (1-cell, 2-cell, 4-cell and 8-cell), abnormal developmental embryo (arrested embryo and fragmented embryo) and blastocyst] (see

Fig. 2) were fixed with 4% formaldehyde for 20 min and permeabilized with 0.2% Triton X-100 for 10 min. Embryos were then incubated first with porcine sur-vivin antibodies (1ug/ml) (Abcam, Cambridge, UK) for 1 h and then with FITC-labeled secondary antibodies (diluted 1:1000) (Abcam, Cambridge, UK). Hoechst Fig 1. Cleavage pattern of porcine SCNT embryos classified by time

flow.

(5)

33342 (0.5 mg/L) (Sigma) was used to stain nuclei. Stained oocytes and embryo were mounted under a cover slip. Slides were examined using fluorescence microscope.

2.8. Statistical analysis

The statistical analysis was conducted using SPSS Inc. software (PASW Statistics 17). A one-way analysis of variance with Duncan multiple-range test was used to assess day1 and day2 embryo cleavage patterns, and developmental rate of blastocysts. All data are pre-sented as mean⫾ SEM. Differences at P ⬍ 0.05 were considered significant.

3. Results

A total of 617 oocytes were used and SCNT was 6 times replicated for the analysis of embryo cleavage pat-tern. From among used oocytes, 511 (82.4%) oocytes were fused. 376 of 511 fused embryos developed to cleav-age stcleav-age (72.3%). And 62 of 511 fused embryos devel-oped to blastocyst stage (12.1%) (Table 2).

3.1. Cleavage assessment at 24 hour post activation and blastocyst development at day 7

At 24 hpa, the 2-cell stage proportion was signifi-cantly higher (44.5%) than other cleavage stages

(1-cell: 13.4%, 4-(1-cell: 17.9%, 8-(1-cell: 10.3% and frag: 13.9%) (Table 3). Each blastomere of the 2-cell stage embryo showed most clear. At day 7, the blastocyst formation rate from the 2-cell stage (18.0%) was sig-nificantly higher than other groups (P⬍ 0.05). 3.2. Cleavage assessment at 48 hour post activation and blastocyst development at day 7

At 48 hpa, 2– 4 cell stage proportion was signifi-cantly higher (32.4%) than other cleavage stages (2– 8 cell: 8.2%, 4 – 8 cell: 12.1% and frag-frag: 13.9%).(Table 4) Some embryos arrested at 48 hpa (1–1 cell: 5.8%, 2–2 cell: 2.8%, 4 – 4 cell: 3.8%, 8 – 8 cell: 6.5% and total arrested embryos: 18.9%). Blas-tocyst formation rates were higher in the 2– 4 cell stage group (20.2%) than in other groups, but not significantly.

3.3. Expression of survivin mRNA in different porcine SCNT embryo developmental stage

Survivin mRNA was detected in all stage of em-bryos. Expression of survivin mRNA in normal cleaved (2, 4-cell) stage of SCNT embryos were significantly higher than arrested embryos (Fig. 3). Survivin mRNA in arrested embryos expressed not significantly differ-ent with that of fragmdiffer-ented embryos.

Table 1

Sequences of the oligonucleotide primers and probe used in RT-PCR.

Primer for Sequence Accession

No. Survivin

Forward 5=-GAC GAC GAC CCC ATA GAA GA-3=

AF195781.1 Reverse 3=-TTT GAC GTT TCT TTC

AGG CG-5= 1-A

Forward 5=-CAC CGT AGG AGG TCT AAC G-3=

AP003428.1 Reverse 3=-GTA TCG TCG AGG TAT

TCC G-5=

Table 2

In vitro development of reconstructed oocytes followed by SCNT in pigs.

Rep. No. of examined oocytes No. (mean%⫾ SEM) fused oocytes No. (mean%⫾ SEM) of embryo developed to

ⱖ 2-cell Blastocyst

6 617 511 (82.4⫾ 3.0) 376 (72.3⫾ 4.9) 62 (12.1⫾ 0.4)

Table 3

Cleavage pattern and blastocyst formation of at 24 hour post activation (hpa). Classified group at 24 hpa Rate of cleavage assessment at 24 hpa (Mean⫾ SEM) Rate of BL from classified group at 24 hpa (Mean⫾ SEM) 1-cell 13.4⫾ 1.5a 7.4⫾ 1.8a,b 2-cell 44.5⫾ 5.0b 18.0⫾ 2.9c 4-cell 17.9⫾ 1.6a 12.6⫾ 4.4b,c 8-cell 10.3⫾ 1.8a 13.3⫾ 2.4b,c Frag. 13.9⫾ 3.0a 2.2⫾ 2.2a Six replicates.

(6)

3.4. Expression of survivin protein in different porcine SCNT embryo developmental stage

The observed immunoreactivity of oocytes and embryos in all stages was predominantly localized to the cytoplasm. At the 2- to 8-cell stage, embryos were more stained than at other stages (Fig. 4). Survivin protein expression patterns were similar to mRNA expression patterns. Arrested embryos had lower survivin protein expression than 2- and 4-cell stage embryos (Fig. 4).

4. Discussion

Animal cloning by nuclear transfer is very ineffi-cient[32]. Particularly in pigs, a large number of SCNT embryos arrest or fragment during the preimplantation stage. However, the causes of this phenomenon are unclear. This study was performed to identify the ori-gins of successfully developed blastocysts by analyzing SCNT embryo cleavage patterns and investigating the cause of abnormal development.

At 24 hpa, about 73% of SCNT embryos were cleaved (2-cell: 44.5%, 4-cell: 17.9%, 8-cell: 10.3%). Among these cleaved embryos, 4-cell and 8-cell stage embryos had a cleavage velocity that was too fast. According to Booth’s report after 1 day of culture, the 2-cell stage is the optimal cleavage stage [16]. Our study also showed a similar cleavage pattern.

Blasto-cysts derived from 2-cell stage embryos were signifi-cantly higher than those from other cleavage stages. At 48 hpa, the 2– 4 cell stage proportion was significantly higher than other cleavage stages. Blastocyst formation rates were higher in the 2– 4-cell stage group (20.2%) than other groups. These data suggest that the 2– 4-cell stage group embryos at 48 hpa had high developmental competence. Porcine IVF embryos, however, are a little different. In a previous report[33], at 48 h post-insem-ination, 4-cell and 8-cell embryos had high develop-mental competence. IVF embryo developdevelop-mental veloc-ity is faster than in SCNT embryos. Indeed, SCNT embryos had more exposure to external stress and spent more time in the nuclear remodeling stage as compare to IVF embryos. Some SCNT embryos had the same cleavage pattern at 24 hpa and 48 hpa. These embryo’s cleavage pattern was the same at 72 hpa. Their devel-opment was arrested. At 48 hpa, 18.9% of embryos were arrested. This percentage in SCNT arrested em-bryos was similar to that of IVF arrested emem-bryos. This is not a desirable phenomenon; the reason for this embryo demise is unclear, but abnormal nuclear repro-gramming and reactive oxygen species (ROS) are likely involved in embryo arrest [25,34]. Aberrant nuclear reprogramming causes abnormal gene expression in-cluding anti-apoptic and cell proliferation genes. Apop-tosis, a mechanism to rid the early embryo of unwanted or damaged cells, if extensive, can also lead to embryo arrest and developmental failure[18,21]. Thus, proper anti-apototic gene expression must be successful during development. Cell proliferation is also an important factor in embryo development. High cell number pre-implatation embryos have a high developmental com-petence. Furthermore, if proliferative ability is lost, the embryo will arrest. ROS are implicated in the induction of apoptosis[35]and cellular senescence[36]. In vitro culture conditions and exogenous factors contribute to elevated ROS production in embryos[37], which leads to increased oxidative stress [37]. Intracellular ROS levels are significantly more abundant in 2- to 4-cell arrested embryos[17].

This study was aimed to determine whether abnor-mal nuclear reprogramming could induce embryo ar-rest. Abnormally expressed anti-apoptic or cell prolif-eration gene and were chosen that could be induced by aberrant nuclear reprogramming. One candidate gene was survivin. Survivin has two functions: apoptosis inhibition and cellular division regulation [38]. In a recent study using a murine model, survivin was re-ported to be an essential anti-apoptic gene and ex-pressed in preimplantation mouse embryos at all stages

Table 4

Cleavage pattern and blastocyst formation of at 48 hour post activation (hpa). Classified Group at 48 hpa (Day 2) [day1–day2] Rate of cleavage assessment at 48 hpa (Mean⫾ SEM ) Rate of BL from classified group at 48 hpa (mean⫾ SEM) 1–1 5.8⫾ 1.4a,b,c 0a 1–2 2.4⫾ 0.6a,b 12.5⫾ 8.5a,b,c 1–4 2.4⫾ 0.7a,b 13.9⫾ 6.3a,b,c 1–8 0.6⫾ 0.4a 0a 1–f 2.3⫾ 0.9a,b 0a 2–2 2.8⫾ 1.1a,b,c 0a 2–4 32.4⫾ 5.0f 20.2⫾ 1.8c 2–8 8.2⫾ 0.9c,d 11.7⫾ 5.1a,b,c 2–f 1.2⫾ 0.6a,b 0a 4–4 3.8⫾ 0.6a,b,c 5.6⫾ 5.6a,b,c 4–8 12.1⫾ 1.4d,e 14.7⫾ 6.0a,b,c 4–f 2.0⫾ 1.0a,b 0a 8–8 6.5⫾ 1.5b,c 17.4⫾ 7.6b,c 8–f 3.8⫾ 0.6a,b,c 12.5⫾ 8.5a,b,c f–f 13.9⫾ 3.0e 2.2⫾ 2.2a,b Six replicates.

(7)

of development [39]. In this study, survivin mRNA expression was detected at all embryonic developmen-tal stages. Survivin mRNA was expressed in mature oocytes, and gradually increased after activation. Ex-pression of survivin mRNA in SCNT embryo 2-cell stage at 24 hpa and 4-cell stage at 48 hpa was signifi-cantly higher than that of arrested oocytes. Further-more, survivin mRNA expression in arrested and frag-mented embryos was similar, suggesting that arrested embryos had abnormal survivin expression similar to abnormally fragmented embryos. When survivin mRNA expression levels were compared in normally cleaved embryos and abnormally developed embryos (i.e., fragmented and arrested), significant differences existed between them. Survivin protein expression pat-terns were similar. Arrested embryos had weak survivin protein expression. In other words, abnormally devel-oped embryos had a low survivin expression level. These results demonstrate that analysis of survivin ex-pression level may assist in determining embryo qual-ity. Similarly, several studies have been reported in other species. In bovines, survivin dsRNA-targeted em-bryos had decreased developmental rates as blastocysts and an increase in the apoptosis index [28]. In mice, over 80% of embryos injected with a survivin siRNA exhibited abnormalities in development and cell divi-sion [40]. Knockout of mouse survivin results in

em-bryonic lethality and survivin null embryos exhibit gross preimplantation stage abnormalities, such as giant nuclei, micronuclei formation, and irregular nuclear morphology [41]. Finally, development of mouse embryos treated with antisense survivin oli-gonucleotides was arrested at the morula or blasto-cyst stage with disruption of tubulin formation and abnormal nuclei [39].

(8)

reprogramming caused abnormal survivin expression and abnormal survivin expression caused embryo arrest were suspected.

In this study, 511 SCNT embryos were analyzed to trace normal SCNT development. Results suggest that the origins of successful development occur at an early developmental stage. The origin at 24 hpa and 48 hpa each are 2-cell, 4-cell stage. After 48 hpa, cleavage pattern assessment was difficult because embryos were compacted and could not divide any further. Survivin expression was significantly different between nor-mally cleaved embryos and arrested embryos. These results indicate that survivin expression is important to porcine SCNT embryo development. Thus, survivin may be a good candidate marker for embryo quality. Survivin is one factor involved in embryo development. However, survivin function and mechanism during em-bryo development remains unclear. In further studies, survivin function must be determined and more inves-tigation into other developmental factors is necessary.

Acknowledgments

This work was supported by a grant (#20070301034040) from the BioGreen 21 Program, Rural Development Admin-istration, Republic of Korea.

References

[1] Dai Y, Vaught TD, Boone J, Chen SH, Phelps CJ, Ball S, Monahan JA, Jobst PM, McCreath KJ, Lamborn AE, Cowell-Lucero JL, Wells KD, Colman A, Polejaeva IA, Ayares DL. Targeted disruption of the alpha1,3-galactosyltransferase gene in cloned pigs. Nat Biotechnol 2002;20:251–5.

[2] Lai L, Kolber-Simonds D, Park KW, Cheong HT, Greenstein JL, Im GS, Samuel M, Bonk A, Rieke A, Day BN, Murphy CN, Carter DB, Hawley RJ, Prather RS. Production of alpha-1,3-galactosyltransferase knockout pigs by nuclear transfer cloning. Science 2002;295:1089 –92.

[3] Tucker A, Belcher C, Moloo B, Bell J, Mazzulli T, Humar A, Hughes A, McArdle P, Talbot A. The production of transgenic pigs for potential use in clinical xenotransplantation: baseline clinical pathology and organ size studies. Xenotransplantation 2002;9:203– 8.

(9)

[4] Estrada JL, Collins B, York A, Bischoff S, Sommer J, Tsai S, Petters RM, Piedrahita JA. Successful cloning of the Yucatan minipig using commercial/occidental breeds as oocyte donors and embryo recipients. Cloning Stem Cells 2008;10:287–96. [5] Koo OJ, Park HJ, Kwon DK, Kang JT, Jang G, Lee BC. Effect

of recipient breed on delivery rate of cloned miniature pig. Zygote 2009;17:203–7.

[6] Wakai T, Sugimura S, Yamanaka K, Kawahara M, Sasada H, Tanaka H, Ando A, Kobayashi E, Sato E. Production of viable cloned miniature pig embryos using oocytes derived from do-mestic pig ovaries. Cloning Stem Cells 2008;10:249 – 62. [7] Zhao J, Ross JW, Hao Y, Spate LD, Walters EM, Samuel MS,

Rieke A, Murphy CN, Prather RS. Significant improvement in cloning efficiency of an inbred miniature pig by histone deacetylase inhibitor treatment after somatic cell nuclear trans-fer. Biol Reprod 2009;81:525–30.

[8] Lee E, Lee SH, Kim S, Jeong YW, Kim JH, Koo OJ, Park SM, Hashem MA, Hossein MS, Son HY, Lee CK, Hwang WS, Kang SK, Lee BC. Analysis of nuclear reprogramming in cloned miniature pig embryos by expression of Oct-4 and Oct-4 related genes. Biochem Biophys Res Commun 2006;348:1419 –28. [9] Rideout WM, 3rd, Eggan K, Jaenisch R. Nuclear cloning and

epigenetic reprogramming of the genome. Science 2001;293: 1093– 8.

[10] Daniels R, Hall V, Trounson AO. Analysis of gene transcription in bovine nuclear transfer embryos reconstructed with granulosa cell nuclei. Biol Reprod 2000;63:1034 – 40.

[11] Humpherys D, Eggan K, Akutsu H, Friedman A, Hochedlinger K, Yanagimachi R, Lander ES, Golub TR, Jaenisch R. Abnor-mal gene expression in cloned mice derived from embryonic stem cell and cumulus cell nuclei. Proc Natl Acad Sci U S A 2002;99:12889 –94.

[12] Rizos D, Lonergan P, Boland MP, Arroyo-Garcia R, Pintado B, de la Fuente J, Gutierrez-Adan A. Analysis of differential mes-senger RNA expression between bovine blastocysts produced in different culture systems: implications for blastocyst quality. Biol Reprod 2002;66:589 –95.

[13] Wrenzycki C, Herrmann D, Keskintepe L, Martins A, Jr., Siri-sathien S, Brackett B, Niemann H. Effects of culture system and protein supplementation on mRNA expression in pre-implanta-tion bovine embryos. Hum Reprod 2001;16:893–901. [14] Wrenzycki C, Herrmann D, Lucas-Hahn A, Lemme E, Korsawe

K, Niemann H. Gene expression patterns in in vitro-produced and somatic nuclear transfer-derived preimplantation bovine embryos: relationship to the large offspring syndrome? Anim Reprod Sci 2004;82:593– 603.

[15] Wrenzycki C, Wells D, Herrmann D, Miller A, Oliver J, Tervit R, Niemann H. Nuclear transfer protocol affects messenger RNA expression patterns in cloned bovine blastocysts. Biol Reprod 2001;65:309 –17.

[16] Booth PJ, Watson TJ, Leese HJ. Prediction of porcine blas-tocyst formation using morphological, kinetic, and amino acid depletion and appearance criteria determined during the early cleavage of in vitro-produced embryos. Biol Reprod 2007;77:765–79.

[17] Betts DH, Madan P. Permanent embryo arrest: molecular and cellular concepts. Mol Hum Reprod 2008;14:445–53. [18] Betts DH, King WA. Genetic regulation of embryo death and

senescence. Theriogenology 2001;55:171–91.

[19] Byrne AT, Southgate J, Brison DR, Leese HJ. Analysis of apoptosis in the preimplantation bovine embryo using TUNEL. J Reprod Fertil 1999;117:97–105.

[20] Gjorret JO, Knijn HM, Dieleman SJ, Avery B, Larsson LI, Maddox-Hyttel P. Chronology of apoptosis in bovine embryos produced in vivo and in vitro. Biol Reprod 2003;69:1193–200. [21] Hardy K, Spanos S, Becker D, Iannelli P, Winston RM, Stark J. From cell death to embryo arrest: mathematical models of human preimplantation embryo development. Proc Natl Acad Sci U S A 2001;98:1655– 60.

[22] Matwee C, Betts DH, King WA. Apoptosis in the early bovine embryo. Zygote 2000;8:57– 68.

[23] Matwee C, Kamaruddin M, Betts DH, Basrur PK, King WA. The effects of antibodies to heat shock protein 70 in fertilization and embryo development. Mol Hum Reprod 2001;7:829 –37. [24] Gjorret JO, Wengle J, Maddox-Hyttel P, King WA.

Chronolog-ical appearance of apoptosis in bovine embryos reconstructed by somatic cell nuclear transfer from quiescent granulosa cells. Reprod Domest Anim 2005;40:210 – 6.

[25] King WA, Shepherd DL, Plante L, Lavoir MC, Looney CR, Barnes FL. Nucleolar and mitochondrial morphology in bovine embryos reconstructed by nuclear transfer. Mol Reprod Dev 1996;44:499 –506.

[26] Altieri DC. Molecular circuits of apoptosis regulation and cell division control: the survivin paradigm. J Cell Biochem 2004; 92:656 – 63.

[27] Ruchaud S, Carmena M, Earnshaw WC. Chromosomal passengers: conducting cell division. Nat Rev Mol Cell Biol 2007;8:798–812. [28] Park SY, Kim EY, Jeon K, Cui XS, Lee WD, Kim NH, Park SP,

Lim JH. Survivin acts as anti-apoptotic factor during the devel-opment of bovine pre-implantation embryos. Mol Reprod Dev 2007;74:582–90.

[29] Altieri DC. Validating survivin as a cancer therapeutic target. Nat Rev Cancer 2003;3:46 –54.

[30] de Graaf AO, de Witte T, Jansen JH. Inhibitor of apoptosis proteins: new therapeutic targets in hematological cancer? Leu-kemia 2004;18:1751–9.

[31] Adida C, Crotty PL, McGrath J, Berrebi D, Diebold J, Altieri DC. Developmentally regulated expression of the novel cancer anti-apoptosis gene survivin in human and mouse differentia-tion. Am J Pathol 1998;152:43–9.

[32] Zhao LW, Yang XY, Guan PF, Fu J, Li H, Zhou YY, Huang SZ, Zeng YT, Zeng FY. Improved efficiency of bovine somatic cell nuclear transfer by optimizing operational procedures. J Reprod Dev 2009;55:542– 6.

[33] Jeon Y, Hong SM, Hyun SH. Prediction of Developmental Ability of In Vitro Fertilized Porcine Embryos by Analysis of Early Cleavage Pattern. J Embryo Trans 2009;24:65–70. [34] Favetta LA, St John EJ, King WA, Betts DH. High levels of

p66shc and intracellular ROS in permanently arrested early embryos. Free Radic Biol Med 2007;42:1201–10.

[35] Stone JR, Yang S. Hydrogen peroxide: a signaling messenger. Antioxid Redox Signal 2006;8:243–70.

[36] Passos JF, Saretzki G, Ahmed S, Nelson G, Richter T, Peters H, Wappler I, Birket MJ, Harold G, Schaeuble K, Birch-Machin MA, Kirkwood TB, von Zglinicki T. Mitochondrial dysfunction accounts for the stochastic heterogeneity in telomere-dependent senescence. PLoS Biol 2007;5:e110.

[37] Guerin P, El Mouatassim S, Menezo Y. Oxidative stress and protec-tion against reactive oxygen species in the pre-implantaprotec-tion embryo and its surroundings. Hum Reprod Update 2001;7:175–89. [38] Jeon K, Kim EY, Tae JC, Lee CH, Lee KS, Kim YO, Jeong DK, Cho

(10)

[39] Kawamura K, Sato N, Fukuda J, Kodama H, Kumagai J, Tan-ikawa H, Nakamura A, Honda Y, Sato T, Tanaka T. Ghrelin inhibits the development of mouse preimplantation embryos in vitro. Endocrinology 2003;144:2623–33.

[40] Malter H SN, Cohen J. Interference with survivin gene function in mouse embryos: Towards a moleclular model for human embryo fragmentation. Fertil Steril 2003;80:79.

[41] Uren AG, Wong L, Pakusch M, Fowler KJ, Burrows FJ, Vaux DL, Choo KH. Survivin and the inner centromere protein INCENP show similar cell-cycle localization and gene knock-out phenotype. Curr Biol 2000;10:1319 –28.

[42] Skoufias DA, Mollinari C, Lacroix FB, Margolis RL. Human survivin is a kinetochore-associated passenger protein. J Cell Biol 2000;151:1575– 82.

[43] Wheatley SP, Carvalho A, Vagnarelli P, Earnshaw WC. INCENP is required for proper targeting of Survivin to the centromeres and the anaphase spindle during mitosis. Curr Biol 2001;11:886 –90.

[44] Kelly AE, Sampath SC, Maniar TA, Woo EM, Chait BT, Funabiki H. Chromosomal enrichment and activation of the aurora B pathway are coupled to spatially regulate spindle assembly. Dev Cell 2007;12:31– 43.

[45] Sampath SC, Ohi R, Leismann O, Salic A, Pozniakovski A, Funabiki H. The chromosomal passenger complex is required for chromatin-induced microtubule stabilization and spindle as-sembly. Cell 2004;118:187–202.

[46] Kelly AE, Ghenoiu C, Xue JZ, Zierhut C, Kimura H, Funabiki H. Survivin reads phosphorylated histone H3 threonine 3 to activate the mitotic kinase Aurora B. Science 2010;330:235–9. [47] Xing X, Magnani L, Lee K, Wang C, Cabot RA, Machaty Z. Gene expression and development of early pig embryos pro-duced by serial nuclear transfer. Mol Reprod Dev 2009;76: 555– 63.

References

Related documents

CAUSES AND REPORTED CASES OF ABSOLUTE MACROCEPHALY WITH NORMAL INTELLIGENCE AND.

Our results allow users to construct reusable application processing elements specified in DMIL [7] (language for data mining and integration, which is being designed within

DCs have been shown to be required for de novo priming of naive donor-reactive T cells through acquisition of graft DC–derived exosomes expressing graft class I and class II

phosphoramidate oligonucleotides on K562 cells ELISA- based MTT assay demonstrating number of the cell in 8 µM concentration. Optical density decreased significantly in both

The model of external friction using artificial neural networks ensures a higher accuracy in predicting the friction force value than the theoretical-experimental model.. In order

Rasis are Rasis other than Gemini, Virgo, Libra, Aquarius and the first half of Sagittarius) be Signs ascending with Mercury and Saturn be in Angles, or in aspect to the Ascendant,

To gain insight into the factors that affect the emergence of entrepreneurship and into the economic outcomes of various types of entrepreneurship, the significance of