Distributions of Mitochondria and the Cytoskeleton in Hamster Embryos Developed In Vivo and In Vitro

Similar documents
Effect of Temperature Decline on the Cytoskeletal Organization of the Porcine Oocyte

Supplemental Information. Fluorescence-based visualization of autophagic activity predicts mouse embryo

Cytoskeletal Organization of Porcine Oocytes Aged and Activated Electrically or by Sperm

Characterization of Anti-Hamster ZP-0 Monoclonal Antibody

Effects of Centrifugation and Lipid Removal on the Cryopreservation of in Vitro Produced Bovine Embryos at the Eight-Cell Stage

T H E J O U R N A L O F C E L L B I O L O G Y

Interval between PMSG Priming and hcg Injection in Superovulation of the Mongolian Gerbil

Effects of Glutamine, Glycine and Taurine on the Development of In Vitro Fertilized Bovine Zygotes in a Chemically Defined Medium

Preimplantation genetic diagnosis: polar body and embryo biopsy

A Precise Bicoid Gradient is Nonessential During Cycles for Precise Patterning in the Drosophila Blastoderm

The subcortical maternal complex controls symmetric division of mouse zygotes by

Supplementary material Legends to Supplementary Figures Figure S1. Figure S2. Figure S3.

(FITC) or rhodamine blue isothiocyanate (RBITC) for use in mixed egg-transfer experiments. Both FITC and RBITC bind to the zona pellucida

Effect of Bovine Follicular Fluid Added to the Maturation Medium on Sperm Penetration in Pig Oocytes Matured In Vitro

Karen L.P. McNally, Amy S. Fabritius, Marina L. Ellefson, Jonathan R. Flynn, Jennifer A. Milan, and Francis J. McNally

16 Effect of cell surface N-linked oligosaccharide chains on the compaction of preimplantation mouse embryos

A comparison of the effects of estrus cow. nuclear maturation of bovine oocytes

Instructions for Use. APO-AB Annexin V-Biotin Apoptosis Detection Kit 100 tests

Keywords: ATP, blastocysts, fertilization, high-polarized mitochondria, human, implantation

Effect of Warming on the Survivability and Fertilizability of Vitrified Matured Bovine Oocytes

Summary. Mouse eggs were fertilized in vitro, in the presence and

Instructions for use

Mouse sperm extraction:

Inhibitory Effect of Iodoacetate on Developmental Competence of Porcine Early Stage Embryos In Vitro

T H E J O U R N A L O F C E L L B I O L O G Y

Alterations of the cytoskeleton and polyploidy induced by cryopreservation of metaphase II mouse oocytes

Effect of the Well of the Well (WOW) System on In Vitro Culture for Porcine Embryos after Intracytoplasmic Sperm Injection

Fluorescence Expression by Bovine Embryos after Pronuclear Microinjection with the EGFP Gene

Supplemental Materials Molecular Biology of the Cell

Effect of Leukemia Inhibiton Factor (LIF) on in vitro maturation and fertilization of matured cattle oocytes

Serafino et al. Thymosin α1 activates complement receptor-mediated phagocytosis in human monocyte-derived macrophages. SUPPLEMENTARY FIGURES

Adenosine Triphosphate Content in Human Unfertilized Oocytes, Undivided Zygotes and Embryos Unsuitable for Transfer or Cryopreservation

Fluorescence Microscopy

Calcium and ph Sensitivity of Myofibrils Isolated From Red and White Porcine Muscles

Ultrarapid freezing of early cleavage stage human embryos and eight-cell mouse embryos*

Imaging of Chromosomes at Nanometer-Scale Resolution, Using Soft X-Ray Microscope

MII. Supplement Figure 1. CapZ β2. Merge. 250ng. 500ng DIC. Merge. Journal of Cell Science Supplementary Material. GFP-CapZ β2 DNA

The incidence of cytoplasmic fragmentation in mouse embryos in vitro is not affected by inhibition of caspase activity

Proteomic profiling of small-molecule inhibitors reveals dispensability of MTH1 for cancer cell survival

Non-invasive methods of embryo selection

Title. CitationJapanese Journal of Veterinary Research, 62(3): 135- Issue Date DOI. Doc URL. Type. File Information. surrounding oocytes

Microtubule and microfilament organization in maturing human oocytes

For the rapid, sensitive and accurate measurement of apoptosis in various samples.

Cells & Cell Transport. Cells

Tanimoto et al., http :// /cgi /content /full /jcb /DC1

SUPPLEMENTARY MATERIAL. Sample preparation for light microscopy

Evaluating the Quality of Human Embryos with a Measurement of Oxygen Consumption by Scanning Electrochemical Microscopy

Degree of Cortical Granule Exocytosis in in vitro- matured Porcine Oocytes Induced by. Different Artificial Stimulators

Limited recovery of meiotic spindles in living human oocytes after cooling rewarming observed using polarized light microscopy

Supplemental Information. Autophagy in Oncogenic K-Ras. Promotes Basal Extrusion. of Epithelial Cells by Degrading S1P. Current Biology, Volume 24

Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran. Received 23 January 2013; revised 10 April 2013; accepted 13 April 2013

Annexin V-PE Apoptosis Detection Kit

Title. Author(s)TEKELI, Tevfik; KWEON, Oh Kyeong; KANAGAWA, Hiroshi. CitationJapanese Journal of Veterinary Research, 35(4): 283-


General Biology. Overview: The Key Roles of Cell Division The continuity of life is based upon the reproduction of cells, or cell division

Influence of maturation culture period on the development of canine oocytes after in vitro maturation and fertilization

Foundational questions Oocyte-derived functional mediators of early embryonic development (EST and candidate gene) JY-1 Nobox Importin 8 Oocyte and cu

klp-18 (RNAi) Control. supplementary information. starting strain: AV335 [emb-27(g48); GFP::histone; GFP::tubulin] bleach

Classification of Morphological Changes Based on the Number of Cleavage Divisions in Bovine Embryos

Downregulation of angiotensin type 1 receptor and nuclear factor-κb. by sirtuin 1 contributes to renoprotection in unilateral ureteral

04_polarity. The formation of synaptic vesicles

Supplemental Materials. STK16 regulates actin dynamics to control Golgi organization and cell cycle

Effects of essential and non-essential amino acids on in-vitro maturation, fertilization and development of immature bovine oocytes

Dako IT S ABOUT TIME. Interpretation Guide. Agilent Pathology Solutions. ALK, ROS1 and RET IQFISH probes (Dako Omnis) MET IQFISH probe (Dako Omnis)

A549 and A549-fLuc cells were maintained in high glucose Dulbecco modified

Incidence of Chromosomal Abnormalities from a Morphologically Normal Cohort of Embryos in Poor- Prognosis Patients

General Biology. Overview: The Key Roles of Cell Division. Unicellular organisms

Effects of Preservation of Porcine Oocytes by Dibutyryl Cyclic AMP on in vitro Maturation, Fertilization and Development

RayBio Annexin V-FITC Apoptosis Detection Kit

Rejuvenation of Gamete Cells; Past, Present and Future

Scanning Electron Microscopic Observations on the Sperm Penetration through the Zona Pellucida of Mouse Oocytes Fertilized in vitro

(a) Significant biological processes (upper panel) and disease biomarkers (lower panel)

Maturation and Freezing of Bovine Oocytes

STUDIES OF THE HUMAN UNFERTILIZED TUBAL OVUM*t

Cell cycle co-ordination in embryo cloning by nuclear transfer

Supplementary tables and figure legends

Name: Xueming Zhao. Professional Title: Professor. Animal embryo biotechnology, mainly including in vitro maturation (IVM), in vitro fertilization

MHM : A Unique and Improved IVF Handling Media

Is the mouse a good model for the human with respect to the development of the preimplantation embryo in vitro?

Multi-Parameter Apoptosis Assay Kit

The Cell Cycle and How Cells Divide

Supplementary Figure 1. Properties of various IZUMO1 monoclonal antibodies and behavior of SPACA6. (a) (b) (c) (d) (e) (f) (g) .

FDA Compliant Patented & Patent Pending

The Cytotoxic Effect of Cryoprotective Agents on in vitro Fertilization Rates of Mammalian Oocytes

The Cell Cycle CHAPTER 12

In vitro production of equine embryos and genetic testing

Fig. 1. A zona-free hamster oocyte penetrated by several guinea pig spermatozoa.

SHORT COMMUNICATION CHICAGO, ILLINOIS. Visualization of Chromosomes in Single Human Blastomeres

Development: is the growth of an individual organism from a simple to a more complex or mature level. A slow process of progressive change

Toxic effects of oxygen on human embryo development

The storage of cow eggs at room temperature and at low temperatures

but it still needs a bit of work

SUPPORTING ONLINE MATERIAL

Type of file: PDF Title of file for HTML: Supplementary Information Description: Supplementary Figures

Do small differences in oxygen concentration influence oocyte and embryo developmental competence?

Effect of addition of exogenous growth factor on in vitro development of preimplantation stage buffalo embryos

Ahtiainen et al., http :// /cgi /content /full /jcb /DC1

Development of normal mice from metaphase I oocytes fertilized with primary spermatocytes

Toxic Effect of Cryoprotectants on Embryo Development in a Murine Model

Organizing the cytoplasm during oocyte maturation. Why investigate oocyte maturation? Oocyte maturation: from symmetry to asymmetry

Transcription:

J. Mamm. Ova Res. Vol. 23, 128 134, 2006 128 Original Distributions of Mitochondria and the Cytoskeleton in Hamster Embryos Developed In Vivo and In Vitro Hiroyuki Suzuki 1 *, Manabu Satoh 1 ** and Katsuya Kabashima 1 1 Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Aomori 036-8561, Japan Abstract: To clarify the causes of low viability of hamster embryos following in vitro culture, the present study compared the distributions of mitochondria and the cytoskeleton in embryos grown in vivo and in vitro. Hamster 2- and 4-cell embryos were characterized by perinuclear clustering of mitochondria, the degree of which was almost the same for in vivo and in vitro embryos. In the cell cortex and cell-to-cell contact region, however, microfilaments were located less densely in in vitro embryos than in in vivo ones. The nucleus moved towards the apex of the blastomere at the late 8-cell stage, when embryos begin the process of compaction. The density of mitochondria seemed to increase in the cell-to-cell contact region during this cellular rearrangement. Mitochondria were concentrated at the perinuclear region of in-vivo 8-cell embryos, whereas they were diffused into the subcortical region of in-vitro 8-cell embryos. Such a diffusion pattern of mitochondrial distribution was also noted in the morulae and blastocysts grown in vitro. These results show that both mitochondrial translocation and cytoskeletal reorganization did not proceed normally in the hamster embryos cultured in vitro, probably resulting in decreased viability of these embryos. Key words: Cytoskeleton, Hamster Embryo, Mitochondria Introduction The ability to support in vitro development of preimplantation embryos is basic for recent animal biotechnology. However, hamster embryos are very Received: February 20, 2006 Accepted: July 13, 2006 *To whom correspondence should be addressed. e-mail: suzuki@cc.hirosaki-u.ac.jp **Present address: IVF Osaka Clinic, Higashi-Osaka, Osaka 577-0013, Japan sensitive to culture milieus, which may seriously affect their viability after embryo transfer (see review [1]). In particular, the energy substrate requirements for hamster embryo development in vitro are markedly different from those of mouse embryos, and mitochondrial metabolism during preimplantation development must still be resolved. Translocation of active mitochondria, being associated with energy production, has some functional correlation with successful preimplantation development [1 5]. Under unsuitable culture conditions, mitochondria are distributed inappropriately, resulting in early embryonic failure [2 5]. Maternal age also influences the loss of mitochondrial activity, leading to lower embryonic development and pregnancy rates in humans [6]. Our previous study revealed that redistribution of the cytoskeleton in hamster embryos occurs during the process of embryonic compaction and is accompanied by an outward migration of blastomere nuclei [7]. Furthermore, it has been reported that the cytoskeletal elements are involved in the mechanism of mitochondrial translocation in mouse [3, 8], hamster [4], and pig embryos [9]. The present study was undertaken to compare the mitochondrial and cytoskeletal distributions between in vivo and in vitro embryos in the hamster. Understanding the relationship between the organelle-cytoskeletal network and the subsequent development of early embryos will contribute to improvement of embryo culture systems for the hamster. Materials and Methods Collection and culture of embryos Golden hamsters (Mesocricetus auratus), 10 12 weeks old, were superovulated as described previously [7]. Embryos at the 2-cell to blastocyst stages were

Suzuki, et al. 129 collected from oviducts and/or uteri with hamster embryo culture medium-3 supplemented with 1.0 mm hypotaurine (HECM-3ht) [10] equilibrated with 10% CO 2, 5% O 2, and 85% N 2 at 37.5 C. Two-cell embryos were recovered at 5:00 6:00 p.m. on day 2 of pregnancy, 4-cell embryos were recovered at 1:00 2:00 a.m. on day 3, early 8-cell embryos were recovered at 11:00 12:00 a.m. on day 3, late 8-cell embryos were recovered at 4:00 6:00 p.m. on day 3, and morulae and blastocysts were recovered at 0:00 1:00 a.m. on day 4 [7, 11]. Two-cell embryos were cultured in HECM-3ht in a humidified atmosphere of 10% CO 2, 5% O 2, and 85% N 2 at 37.5 C, and their development was checked at the time points mentioned above. In some cases, the culture period was extended to obtain embryos at the stage expected. Two-cell embryos cultured for 5 h were referred to as in vitro 2-cells. The experimental design was approved by the Ethical Committee for Experimentation with Animals, Hirosaki University. Fluorescence observations Active mitochondria were stained with rhodamine 123 (Rh123, 10 µg/ml, Sigma, St. Louis, MO, USA) for 15 min in HECM-3ht, as described previously [12], washed 3 times in HECM-3ht, mounted onto slide glasses, and imaged immediately after labeling. To assess the nuclear configuration and distribution of microtubules and microfilaments, the eggs were processed as reported previously [7]. After fixation in microtubule stabilization buffer, the samples were exposed overnight to primary monoclonal anti-β tubulin antibody (1:200; Sigma) at 5 C and incubated with fluorescein isothiocyanate (FITC)-conjugated secondary antibody (1:200; Sigma) at 37 C for 2 h. After rinsing, the samples were stained with rhodamine-phalloidin (1:1000; Molecular Probes, Eugene, OR, USA) for microfilaments for 1 h and then stained for DNA with Hoechst 33342 (10 µg/ml) in mounting medium containing PBS and glycerol (1:1). Fluorescence imaging The samples were viewed under a fluorescent microscope (BX-FLA, Olympus, Tokyo, Japan). A U- MNIBA filter set (Olympus) was used for Rh123 and FITC, a U-MWIB set (Olympus) was used for rhodamine, and a U-MWU set (Olympus) was used for Hoechst. A cooled CCD video system (ImagePoint, Photometrics Ltd., Tucson, AZ, USA) was used to obtain images on a computer, and color adjustment was performed using the IPLab-Spectrum P software (Signal Analytics Corporation, Vienna, VA, USA). Quantification of fluorescence intensity To compare mitochondrial intensity among the specific stages, digital images were obtained at a constant exposure of 0.1 sec. The images of embryos stained with Rh123 (10 17 eggs for each stage) were analyzed by quantifying the average pixel intensities of the cytoplasm, according to a previous study [12]. The distribution density of microfilaments and microtubules was assessed by their fluorescence intensity. Briefly, embryos showing pixel intensity of more than 90% of the average intensity of in vivo embryos were referred to as having dense distribution of microfilaments and microtubules. As reported previously [7], blastomeres showing nuclear migration, the nuclei of which were located in the apical cytoplasm, were also recorded. Statistical analyses The mean mitochondrial intensities at different stages in vivo and in vitro were analyzed by ANOVA. Proportional data were analyzed by Chi-square test or Fisher s exact probability test. Results Distribution of active mitochondria and the cytoskeleton A total of 421 embryos were analyzed for the distributions of their mitochondria (68 in vivo and 72 in vitro embryos) and cytoskeletons (135 in vivo and 146 in vitro embryos). In blastomeres of the 2-4-cell embryos grown in vivo, most mitochondria were accumulated in the perinuclear region, while few mitochondria were noted in the cell cortex (Figs. 1a, b). A similar distribution pattern was noted in in vitro embryos at the corresponding stages (Figs. 1a, b ), but microfilament staining was decreased in the cell cortex and cell-to-cell contact region of the in vitro embryos (Figs. 2a, b vs. 2a, b). Surprisingly, the mitochondria of 2-cell embryos cultured for 5 hours had significantly decreased fluorescence intensity, approximately 10%, compared to the 2-cell embryos just after recovery (Table 1). A similar rate of decline was noted in the fluorescence intensity of in vitro early- and late-8-cell embryos compared with in vivo ones, but no significant difference was found at the 4-cell stage between in vivo and in vitro embryos (Table 1). In the early 8-cell embryos grown in vivo, the cell cortex and perinuclear region showed intense mitochondrial staining (Figs. 1c-1, c-2). In in vitro embryos, however, mitochondria had extended into the

130 J. Mamm. Ova Res. Vol. 23, 2006 Fig. 1. Fluorescence micrographs showing the distribution of mitochondrial clustering in hamster embryos developed in vivo (a f) and in vitro (a f ). The bar in (a) represents 50 µm for all micrographs. Two-cell embryos (a, a ); 4-cell embryos (b,b ); 3 blastomeres from an early 8- cell embryo in vivo (c-1); the other 5 blastomeres from the same embryo (c-2); 4 blastomeres from an early 8-cell embryo in vitro (c -1); the other 4 blastomeres from the same embryo (c - 2); 4 blastomeres from a late 8-cell embryo in vivo (d-1); the other 4 blastomeres from the same embryo (d-2); 4 blastomeres from a late 8-cell embryo in vitro (d -1); the other 4 blastomeres from the same embryo (d -2); morulae (e, e ); blastocysts (f, f ). Two- and fourcell embryos are characterized by complete perinuclear clustering of active mitochondria with a thin cortical ring both in vivo (a, b) and in vitro (a, b ). In in vivo 8-cell embryos (c, d), the cortex became strongly stained with Rh123, and perinuclear clustering is still observed (arrows), whereas perinuclear mitochondria are dispersing into the middle layer in in vitro embryos, resulting in a wider distribution of mitochondria (c, d ). In vitro embryos display partial migration into the subcortical region (c, d, e ; arrowheads). The mitochondrial localization of morulae and blastocysts became difficult to evaluate because the signals overlapped each other due to increasing cell numbers. However, fluorescence intensity seems to decline slightly in in vitro embryos (e, f ) compared to in vivo ones (e, f). subcortical (intermediate) region at various degrees (Figs. 1c f ), showing a diffusion pattern for mitochondrial distribution after the 8-cell stage. In the late 8-cell stages onward, the nuclei of the blastomeres were located in the apex of the cells (Figs. 2d, d ), as reported previously [7]. In association with the outward migration of the nucleus, mitochondrial clusters also moved outwards (Figs. 1d, d ). But the percentage of

Suzuki, et al. 131 Fig. 2. Fluorescence micrographs showing the distribution of the nucleus and cytoskeleton of hamster embryos developed in vivo (a f) and in vitro (a f ). The bar in (a) represents 50 µm for all micrographs. Microfilaments are red, microtubules are green, and nuclei are blue; yellow shows the distribution of microfilaments and microtubules. Two-cell embryos (a, a ); 4-cell embryos (b, b ); 4 blastomeres from an early 8-cell embryo in vivo (c-1); the other 4 blastomeres from the same embryo (c-2); 3 blastomeres from an early 8-cell embryo in vitro (c -1); the other 5 blastomeres from the same embryo (c -2); 4 blastomeres from a late 8-cell embryo in vivo (d- 1); the other 4 blastomeres from the same embryo (d-2); 4 blastomeres from a late 8-cell embryo in vitro (d -1); the other 4 blastomeres from the same embryo (d -2); morulae (e, e ); blastocysts (f, f ). The nuclei of individual cells were located in the center region of the cytoplasm of 2- cell (a, a ) and 4-cell (b, b ) embryos and in most cells of the early 8-cell embryos (c, c ). As reported previously [7], outward migration of the nucleus of the blastomere is observed in the late 8-cell embryos (arrows). Such features are also noted in the outer cells of morulae (e, e ) and blastocysts (f, f ). Decreased intensity in microfilaments and/or microtubules is frequently observed in the in vitro embryos from the 8-cell stage onwards (c, d, e, f ; arrowheads). blastomeres showing nuclear migration was significantly higher in in vivo embryos than in in vitro embryos (94%, n=33 vs. 57%, n=37; P<0.01). After the 8-cell stage, the proportion of embryos showing a dense distribution of microfilaments or microtubules decreased more significantly in vitro than in vivo (Table 2). In morulae and blastocysts under the conventional fluorescent microscope used in this study, it became difficult to identify the distribution of mitochondria and the cytoskeleton within each blastomere because the

132 J. Mamm. Ova Res. Vol. 23, 2006 Table 1. Fluorescence intensity of mitochondria in the hamster embryos* Developmental In vivo In vitro Stage No. of embryos Mean ± SEM No. of embryos Mean ± SEM 2-cell 12 193.8 ± 4.8 A 10** 173.1 ± 5.0 B 4-cell 11 173.9 ± 2.4 13 171.1 ± 4.5 Early 8-cell 13 199.3 ± 8.7 a 12 177.9 ± 4.9 b Late 8-cell 11 208.5 ± 3.1 A 13 187.7 ± 3.1 B *The morula and blastocyst stages were not evaluated because the signals overlapped. **The 2-cell embryos were cultured for 5 h after recovery. Values with different superscripts are significantly different between in vivo and in vitro embryos (p<0.05 between a and b; p<0.01 between A and B). Table 2. Percentage of embryos showing a dense distribution of the cytoskeleton* Developmental In vivo In vitro Stage No. of embryos MFs MTs No. of embryos MFs MTs 2-cell 24 92 96 21 81 90 4-cell 26 85 92 33 88 79 Early 8-cell 32 84 a 100 A 35 57 b 69 B Late 8-cell 33 94 A 97 A 37 49 B 30 B *The morula and blastocyst stages were not evaluated because the signals overlapped. Embryos showing pixel intensity of more than 90% of the average intensity of in vivo embryos were referred to as having dense distribution for microfilaments (MFs) and microtubules (MTs). Values with different superscripts are significantly different between in vivo and in vitro embryos (p<0.05 between a and b; p<0.01 between A and B). Table 3. Developmental schedule of hamster embryos grown in vivo and cultured in HECM-3ht Developmental In vivo In vitro stage Hours PEA* Hours PEA* No. (%) of embryos developed** 4-cell 43 44 44 149 (100) Early 8-cell 54 55 55 146 (98.0) Late 8-cell 60 61 61 144 (96.6) Morula 64 65 68 131 (87.9) Blastocyst 68 69 72 56 (37.6) *Post-egg activation [11]. **Culture was initiated from 2-cell embryos. Times given for developmental stages are when approximately 50% of the embryos were at the stage shown. fluorescence signals overlapped each other. At these stages, therefore, only the outer cells of the embryos, which were not overlapped, were able to be evaluated. Perinuclear clustering of mitochondria became obscure at the morula or later stages (Figs. 1e, f). Fluorescence observations for different components within each blastomere revealed that microtubules were found around the nuclei, and the distribution of the mitochondria was somewhat correlated with that of the microtubules. Furthermore, some active mitochondria were found in the cell-to-cell contact region of in vivo embryos (Figs. 1c-2, 1d-1, d-2), where microfilaments were located in a high density, especially around the time of compaction (Figs. 2c-1, 2d-1, d-2). Decreased intensity of the cytoskeleton and diffused distribution of mitochondria were more frequently noted in in vitro embryos (Figs. 1c f, 2c f ) than in vivo ones (Figs. 1c f, 2c f). Developmental schedule of hamster embryos Table 3 shows the timing of development of the hamster embryos grown in vivo and in vitro. The time scale was presented as hours post-egg activation (PEA) and was based on the estimated time after sperm penetration according to Bavister et al. [11]. A total of 149 embryos recovered at 2-cell stage were cultured.

Suzuki, et al. 133 All embryos (100%) developed to the 4-cell stage after 7 8 h of culture, namely 43 44 h PEA, to the early 8- cell stage 54 55 h PEA, and to the late 8-cell stage 60 61 h PEA. Thus, embryo development to the 8-cell stage was very similar between in vivo and in vitro embryos. More than 95% of embryos reached the late 8-cell stage. However, in vitro development was retarded around compaction; in vivo embryos developed to the morula stage 64 65 h PEA, whereas in vitro embryos developed to this stage 68 h PEA. Afterward, 87.9% of the embryos reached the morula or later stages and 37.6% developed into blastocysts. Once embryos reached the morula stage, no more delays were noted in formation of the blastocoele. Therefore, embryonic development was delayed at least 4 hours preferentially from the late 8-cell to morula stages under the culture condition used in this study. Discussion Our previous study showed that mitochondria in the ooplasm intensified their activities progressively from the germinal vesicle stage to the first metaphase stage and reorganized dramatically during maturation and fertilization in hamster oocytes [12]. The present study clearly demonstrated that the mitochondrial localization of embryonic cells changed after the 8-cell stage. At the late 8-cell stage, hamster embryos were characterized by concentration of mitochondria in the cell-to-cell contact region and perinuclear region in the in vivo embryos, but this was not clear in the in vitro embryos. Such heterogeneity in mitochondrial distribution may be associated with energy production/utilization for compaction. In vitro culture may change mitochondrial localization in the cytoplasm of hamster embryos, especially after the 8-cell stage, where the distributional density of microfilaments and microtubules is reduced. Furthermore, the percentage of blastomeres showing nuclear migration significantly decreased in in vitro embryos compared to in vivo ones (57% vs. 94%, P<0.01). This observation suggests that the nucleus migrates from the central cytoplasm to the apical cytoplasm around the late 8-cell stage [7], but some failure occurred in the mechanism of nuclear migration in vitro. Of particular interest was the observation that in-vitro embryonic development was delayed from the late 8-cell to morula stages, namely around the time of compaction. This suggests that the process of compaction may require increased energy production in a particular region of each blastomere and that culture conditions that disrupt energy production in the embryo may retard the compaction process. Previous studies have shown that changes in culture conditions can alter the organization or structure of mitochondria in the oocytes or embryos of hamsters [4, 13 15], mice [2, 3], and cattle [16, 17]. It has been reported that exposure of hamster 2-cell embryos to suboptimal culture conditions (addition of glucose and phosphate, which disrupts development) causes dispersion of mitochondria away from the nuclei [5, 13, 14]. The observations of the present study revealed that a similar dispersion of mitochondria was noted from the 8-cell stage onwards for hamster embryos cultured in HECM- 3ht. Translocation of mitochondria in oocytes/embryos is associated with reorganization of the cytoskeleton. However, experiments in which the cytoskeleton was disrupted with inhibitors have presented confusing results. Some researchers suggest that microfilaments play a role in the translocation of mitochondria [4], whereas others indicate that microtubules play a role in the translocation [8, 9]. In our preliminary observations of hamster 2-cell embryos, a treatment of cytochalasin D (a microfilament disruptor) caused mitochondria to disappear from the cortical cytoplasm, and a treatment of nocodazole (a microtubule disruptor) caused the breakup of perinuclear clustering of mitochondria. Therefore, both microfilaments and microtubules may be mutually related to spatial organization of the mitochondria in the cytoplasm of embryos. Further studies are needed to understand how mitochondrial reorganization is controlled during early embryonic development. In conclusion, a combination of translocation of mitochondria and reorganization of the cytoskeletal networks may be involved in the developmental program of cytoplasmic alterations, and this type of organelle-cytoskeletal network was not maintained normally under the culture conditions used in this study. These findings may offer some explanation for low viability of hamster embryos cultured in vitro. Acknowledgements The authors thank the staff of the Gene Research Center at Hirosaki University for use of the image analyzing system. References 1) Bavister, B.D. (2000): Interactions between embryos and the culture milieu. Theriogenology, 53: 619 626.

134 J. Mamm. Ova Res. Vol. 23, 2006 2) Muggleton-Harris, A.L. and Brown, J.J.G. (1988): Cytoplasmic factors influence mitochondrial reorganization and resumption of cleavage during culture of early mouse embryos. Hum. Reprod., 3: 1020 1028. 3) Tokura, T., Noda, Y., Goto, Y. and Mori, T. (1993): Sequential observation of mitochondrial distribution in mouse oocytes and embryos. J. Assisted Reprod. Gen., 10: 417 426. 4) Barnett, D.K., Kimura, J. and Bavister, B.D. (1996): Translocation of active mitochondria during hamster preimplantation embryo development studied by confocal laser microscopy. Dev. Dyn., 205: 64 72. 5) Barnett, D.K., Clayton, M.K., Kimura, J. and Bavister, B.D. (1997): Glucose and phosphate toxicity in hamster preimplantation embryos involves disruption of cellular organization, including distribution of active mitochondria. Mol. Reprod. Dev., 48: 227 237. 6) Wilding, M., Dale, B. and Placido, G.D. (2001): Mitochondrial aggregation patterns and activity in human oocytes and preimplantation embryos. Hum. Reprod., 16: 909 917. 7) Suzuki, H., Azuma, T., Koyama, H. and Yang, X. (1999): Development of cellular polarity of hamster embryos during compaction. Biol. Reprod., 61: 521 526. 8) Van Blerkom, J. (1991): Microtubule mediation of cytoplasmic and nuclear maturation during the early stages of resumed meiosis in cultured mouse oocytes. Proc. Natl. Acad. Sci. USA. 88: 5031 5035. 9) Sun, Q.Y., Wu, G.M., Lai, L., Park, K.W., Cabot, R., Cheong, H.T., Day, B.N., Prather, R.S. and Schatten, H. (2001): Translocation of active mitochondria during pig oocyte maturation, fertilization and early embryo development in vitro. Reproduction, 122: 155 163. 10) Barnett, D.K. and Bavister, B.D. (1992): Hypotaurine requirement for in vitro development of golden hamster one-cell embryos into morulae and blastocysts and production of term offspring from in vitro-fertilized ova. Biol. Reprod., 47: 297 304. 11) Bavister, B.D., Leibfried, M.L. and Lieberman, G. (1983): Development of preimplantation embryos of the golden hamster in a defined culture medium. Biol. Reprod., 28: 235 247. 12) Suzuki, H., Satoh, M. and Toyokawa, K. (2005): Changes in distribution of active mitochondria during oocyte maturation and fertilization in the hamster. J. Mamm. Ova. Res., 22: 163 169. 13) Lane, M. and Bavister, B.D. (1998): Calcium homeostasis in early hamster preimplantation embryos. Biol. Reprod., 59: 1000 1007. 14) Squirrell, J.M., Lane, M. and Bavister, B.D. (2001): Altering intercellular ph disrupts development and cellular organization in preimplantation hamster embryos. Biol. Reprod., 64: 1845 1854. 15) Ludwig, T.E., Squirrell, J.M. and Bavister, B.D. (2001): Relationship between development, metabolism, and mitochondrial organization in 2-cell hamster embryos in the presence of low levels of phosphate. Biol. Reprod., 65: 1648 1654. 16) Krisher, R.L. and Bavister, B.D. (1998): Responses of oocytes and embryos to the culture environment. Theriogenology, 49: 103 114. 17) Abe, H., Otoi, T., Tachikawa, S., Yamashita, S., Satoh, T. and Hoshi, H. (1999): Fine structure of bovine morulae and blastocysts in vivo and in vitro. Anat. Embryol., 199: 519 527.