Skip to content
BY 4.0 license Open Access Published by De Gruyter Open Access April 20, 2022

The role of passage numbers of donor cells in the development of Arabian Oryx – Cow interspecific somatic cell nuclear transfer embryos

  • Aiman A. Ammari EMAIL logo , Muath G. ALghadi , Ahmad R. ALhimaidi and Ramzi A. Amran
From the journal Open Chemistry

Abstract

The cloning between different animals known as interspecific somatic cell nuclear transfer (iSCNT) was carried out for endangered species. The iSCNT has been characterized by a poor success rate due to several factors that influence the formation of the SCNT in various cytoplasms. The cell cycle of the transferred somatic cell, the passage number of the cultured somatic cell, the mitochondria oocytes, and their capabilities are among these factors. This study investigates the role of the passage number of the Arabian Oryx somatic cell culture when transplanted to an enucleated domestic cow oocyte and embryo development in vitro. The fibroblast somatic cell of the Arabian Oryx was cultured for several passage lanes (3–13). The optimal passage cell number was found to be 10–13 Oryx cell lines that progressed to various cell stages up to the blastula stage. There was some variation between the different passage numbers of the oryx cell line. The 3–9 cell line did not show a good developmental stage. These could be attributed to several factors that control the iSCNT as stated by several investigators. More investigation is needed to clarify the role of factors that affect the success rate for the iSCNT.

1 Introduction

Enucleated, metaphase II sheep oocytes fused with 8- or 16-cell embryonic blastomeres were employed in the first report of embryonic nuclear transfer in mammals [1,2]. Following that, other attempts to clone cattle and other animals were made. Numerous examples of effective nuclear transfer utilizing donor cells have been published for sheep [3,4,5], cows [6,7], rabbits [8,9], pigs [10], mice [11], monkeys [12], goats [13,14], and camel [15,16]. At the morula and blastocyst stages, donor cells were also revealed to be totipotent and reprogrammable by cytoplasmic factors in cow [6] and rabbits [8]. American researchers successfully injected the somatic cells of threatened animals (Bos gaurus) into an enucleated oocyte, obtained embryo at blastula stage and transferred to a recipient’s cow [17]. In addition, Saikhun et al. [18] also succeeded to inject buffalo somatic cells into an enucleated oocyte from a species cow (Bos indicus) [19]. Furthermore, Ikumi et al. [20] succeeded in transferring a somatic cell from a whale into an in vitro cloned embryo.

The endangered grey wolf was successfully cloned from somatic cells obtained from the dead carcass [21]. In wild goats, somatic cells were transferred into the oocyte of local goats [22]. Although the use of intra- or interspecies somatic cell nuclear transfer (SCNT) has resulted in the generation of cloned offspring in a variety of mammalian species, the efficiency of somatic cell cloning remains often very or extremely low. Therefore, extensive investigations are needed to identify the determinants of SCNT-mediated cloning, which can be responsible for ameliorating the efficiency of this modern-assisted reproductive technology. Among these determinants, the most important role is played by: (1) the source of nuclear donor somatic cells (NDSCs) [23,24,25,26], (2) the abilities of nuclear recipient oocytes to be artificially activated [26,27,28], and (3) molecular interactions between mitochondrial and nuclear DNA molecules in SCNT-derived embryos [29,30,31,32]. It is also worth noting about the remarkable role of: (4) the abilities of donor cell nuclear genes to epigenomically reprogram their transcriptional activities in somatic cell-cloned embryos [33,34,35] and (5) the molecular quality and survival rates of the in vitro cultured NDSCs and SCNT-derived embryos [36,37,38,39]. Taking the above-indicated determinants affecting the efficiency of somatic cell cloning, the research is required to be undertaken to improve the epigenetic reprogrammability (i.e., dedifferentiability) of donor cell nuclei in interspecies SCNT-derived embryos that have been reconstructed with somatic cells stemming from endangered or extinct mammalian species. The technique of transferring somatic cells from threatened wild species of mammals such as the Arabian Oryx is essential in their preservation. Also, the cytoplasm of cow oocytes supports the development of embryos resulting from SCNT of different types of animals [40]. Cow and Arabian Oryx are both members of the family Bovidae. The cloning of Arabian Oryx via the injection of the enucleated oocyte from cow somatic cells is realistic.

The Arabian Oryx is the smallest member of the Oryx genus and is found across the Arabian Peninsula. They vanished from the wild in the early 1970s, but zoos and private custodians preserved them. The Arabian Oryx was categorized as endangered by the International Union for Conservation of Nature in 1986 [41]. In addition, it was the first mammal to be reclassified as vulnerable after being declared extinct in the wild in 2011 [42]. This study aims to produce cloned Arabian Oryx embryos by transferring different passages of fibroblast somatic cells from Arabian Oryx into enucleated cow oocytes.

2 Materials and methods

2.1 Chemicals

Both chemicals, media and hormones, were obtained from Sigma-Aldrich Corp. (St. Louis, MO, USA), unless otherwise specified.

2.2 Ovaries, Oocyte, and maturation

Ovaries were obtained from a local slaughterhouse in Riyadh and transported in 0.9% Sodium Chloride to the laboratory during 1–2 h. The oocytes were extracted from the ovarian follicles using a 19-gauge needle connected to syringe containing 0.5 mL of tissue culture medium (Caisson Lab. Inc., Smithfield, USA). Cumulus oocyte complexes (n 706) with more than three layers of cumulus cells and homogenous cytoplasm (Figure 1a) were matured in TCM-199 at 39°C and 5% CO2 in atmospheric air at high humidity for 24 h as mentioned in our previous research [43].

Figure 1 
                  (a) Immature oocyte, (b) mature oocyte, (c) oocyte with FPB after 24 h (the arrow = 1st polar body) and oocyte cutting, (d) the nucleus was extracted and stained, and then examined under fluorescence microscopy, (e) donor cells from Arabian Oryx, (f) injection somatic cell, (g) 2 cell stage, (h) = 4 cell stage, (i) = 8 cell stage, (j) = 16 cell stage, (k) morula stage, and (l) blastocyst stage.
Figure 1

(a) Immature oocyte, (b) mature oocyte, (c) oocyte with FPB after 24 h (the arrow = 1st polar body) and oocyte cutting, (d) the nucleus was extracted and stained, and then examined under fluorescence microscopy, (e) donor cells from Arabian Oryx, (f) injection somatic cell, (g) 2 cell stage, (h) = 4 cell stage, (i) = 8 cell stage, (j) = 16 cell stage, (k) morula stage, and (l) blastocyst stage.

2.3 Zona dissection with micropipettes

Cumulus-enclosed oocytes were placed on a 35 mm Petri plate with hyaluronidase enzyme (300 IU/1 mL) after in vitro maturation period. Cumulus cells were manually removed from oocyte by using a glass Pasteur pipette. Then, for zona dissection, only the oocytes with an extruded first polar body (FPB) would be utilized. The directing of the oocyte was controlled by the cutting pipette. When the polar body is at 12 o’clock, an oocyte position with the holding pipette, a slot or slit in the zona pellucida (ZP) was made with the cutting pipette on the opposite side of oocyte (Figure 1c) [4].

2.4 Oocyte enucleation

Cutting pipettes were used to softly spin the oocyte to extract a little part of cytoplasm contained in the plasma membrane immediately beneath the FPB of oocytes outside the ZP. Enucleation was ascertained by separating the resultant from the oocyte and cultured primarily in TCM-199. Earle’s salt is mixed with 10% fetal bovine serum and 20 μg/2 mL Hoechst for 30 m (Sigma B2261). The ooplasm was then subjected to UV light to check for the existence of a metaphase II stage [4].

2.5 Preparation of fibroblast donor somatic cells from Arabian Oryx

Oryx ears were utilized to acquire fibroblast donor cells, which have a normal diploid karyotype, and were used to transduce cell nuclei by lanes 3–13. The fibroblast cells were cultured in Dulbecco’s modified minimal basic 0.5 μg/mL Penicillin streptomycin sigma and 10% fetal bovine serum at 39°C and 5% (CO2) in atmospheric air at high humidity. The cells were grown to 80% confluence and NT cells were separated by 0.25% trypsin-EDTA solution (Gibco 25200) (Figure 1e) [4].

2.6 Analysis of cell cycle by flow cytometry

Flow cytometry was used to determine the cell cycle of Oryx cells. Cells were extracted by centrifugation, washed once with PBS after being treated with three passages, and incubated for 6–8 h. The cells were then fixed in chilly 70% alcohol for 30 m. After that, the cell suspensions were incubated with RNase for 30 min in the dark. By using a Beckman Coulter flow cytometer, 10,000 events of PI-stained cells were recorded in the FL4 Log channel using a 675 nm band-pass filter (Cytomics FC 500, Beckman Coulter, Fullerton, CA, USA). CXP capture and analysis software was used to compute the cell cycle stages and analyze the data (Figure 2).

Figure 2 
                  The different passage cells to make sure from their presence in the G0 stage.
Figure 2

The different passage cells to make sure from their presence in the G0 stage.

2.7 Nuclear transfer, fusion, and activation

One somatic cell from an oryx fibroblast was inserted by polar body dissection pipette into the perivitelline space of every enucleated cows’ oocytes. Immediately following NT at room temperature, the cell fusion was done via a single alternating current pulse of 0.2 kV/cm for one second followed by a single DC pulse of 2.5 kV/cm for 50 s. After fusing, 5 μM ionomycin was employed to activate selected fused SCNT at room temperature for 5 min. Then, they were cultured in SOF medium supplemented with 2 mg/500 μL BSA, 20 μL/2 mL MEM (50×), 20 μL/2 mL MEM (100×), 50 μg/mL Penicillin streptomycin, 5 μg/500 μL Cycloheximide using a 35 mm Petri dish coated by mineral oil that has been tried on embryos, for 6 h at 39°C with 5% CO2. All steps of this protocol follow the method described in a previous study (Figure 1f) [4].

2.8 In vitro culture of iSCNT embryos

After 6 h of activation, iSCNT embryos were cultured for 42 h in SOF medium supplemented with 2 mg/500 L BSA., 20 μL/2 ml MEM (50×), 20 μL/2 mL MEM (100×) 50, μg/2 mL Penicillin streptomycin using Petri dishes 35 mm coated by mineral oil at 39°C for 6 h with mixture gas 90, 5, 5% of N2, CO2, O2, respectively (Figure 1g–l) [4,43].

2.9 Statistical analysis

All results were recorded and analyzed using the Mine Tab INSTAT program. The Blastocyst and cleavage rates were analyzed by using the Chi-square test.

3 Results

In this study, the collected oocytes (n 706) injected with different passage lanes 3–13 of donor cells showed different developmental rates of iSCNT Oryx – Cow embryos. From passage 3 up to 9, was no considerable rate of development, which illustrates the collected oocyte numbers, with enucleation rate, fusion rate, embryo cleavage rate, and blastula development rate. The rate of the different passage 10 to 13 was 80, 91.2, 66.3, and 84%, respectively. The fusion rate of the different passage 10 to 13 was 43, 61.4, 71, and 70%, respectively (Table 1).

Table 1

The role of passage numbers of donor cells in the development of Arabian Oryx – Cow iSCNT embryos

Passage number No. of oocytes Enucleation rate% Fusion rate% Cleavage rate% Blastocyst rate%
Passage 10 190 125 (80) 35 (43) (8) 14% (5) 62%*
Passage 11 140 114 (91.2) 70 (61.4) (25) 57%* (0) 0%
Passage 12 271 112 (66.3) 80 (71.4) (8) 16% (1) 12%
Passage 13 105 142 (84) 100 (70.4) (17) 48%* (0) 0%

*Significant difference at p < 0.01 compared to the others without star at the same type of passage or columns.

In the passage from 10 to 13, the cleavage rate of the iSCNT embryos injection with the different passage was 14, 57, 16, and 48%, respectively. The cleavage rate of the iSCNT in passages 11 and 13 indicated a considerable increase (p < 0.01) than passage lines 10 and 12 (Table 1).

While passage line 10 produced a better blastula rate compared to the other passage lines, it was significantly higher (p < 0.01) than passage lines 11, 12, and 13 (Table 1). In general, the total embryos at the blastocyst stage were five embryos (62%) from passage 10 and one embryo (12%) from passage 12, while no embryos developed up to the blastocyst stage from passages 11 and 13 (Table 1).

4 Discussion

The failure of cloning has been related to several variables, one of which is a greater rate of oocyte degeneration. This might be owing to the kind of somatic cell employed since most of the donor cells were from skin or cumulus cells, which are not in the G0 stage, or the electrical current used to fuse the somatic cell with the oocytes, which could damage the ova’s cytoplasm. In addition, due to the long term of the fibroblast cell, the rate of development of the SCNT may be affected by in vitro culture [45].

This work demonstrated the ability of the cytoplasm of domestic cows to reprogram adult Arabian Oryx fibroblast nuclei. Interspecies somatic cell nuclear transfer embryos have the potential to develop to the blastocyst. The development rate of iSCNT rebuilt Arabian Oryx–Cow embryos can be obtained at a low rate as per Li et al. [45]. SCNT efficiency was substantially lower when embryonic germ (EG) cells with lower passage numbers (3–5) were employed as donor cells than when fetal fibroblast donor cells were used. Higher passage EG cells (numbers 9–12) produced similar blastocyst rates, implying that higher passage EG cells are better suited for SCNT. This agrees with our result regarding the use of passes more than 10. The SCNT development was dramatically reduced when lower passage (numbers 3–5) porcine EG cells were used as donor cells, but higher passage (numbers 9–12) EG cells produced a greater blastula rate. However, these results disagree with Jin et al. [44] which they got better-cloned embryo development from the first passage cells compared to 4–6 passages.

There are several factors reported about early and late passages [45,46,47], which affect the proliferation of donor somatic cell culture, such as the abnormalities in chromosomes, DNA damage, and DNA methylation. The bulk of iSCNT research has focused on establishing whether an oocyte can reprogram a foreign species’ nucleus and support the initial cleavage divisions. Fewer studies, on the other side, have looked at the variables that cause embryonic failure in less taxonomically various species during the preimplantation stages [48]. Two of the key studied challenges in iSCNT include failure to reprogram the somatic cell and incompatibility between the nucleus and the cytoplasm [49,50]. Incompatibilities between the nucleus and the cytoplasm have been found to cause harmful cellular processes as an outcome of species variation in studies on and nuclear hybrid embryos. Larger species’ divergence lengths, according to some theories, may hinder communication between the nucleus and cytoplasm components such as the mitochondrial genome [51,52].

Many studies have shown the molecular that may cause the failure of the development of the embryos it produces by iSCNT [53,54,55]. In our research, we studied two species known to produce related species (Bovidae species) through interspecies cloning, which indicates some similarities between the two species.

Furthermore, the timing of DNA replication appears to vary among SCNT embryos [56]. In addition, the cytoplasm of donated oocytes for iSCNT is from different species which have different regulations for DNA demethylation of somatic cells [57,58].

The findings of this study revealed information about some of the factors that contribute to the development of Arabian Oryx – Cow iSCNT embryos in certain passages and another passage failure in others. The reduced rate of cleavage and blastocyst formation in iSCNT might be attributed to mitochondrial function, which causes embryo development failure during the early stages of division in embryos [53,59]. The results of our study showed the best cleavage and blastula rate obtained from passages 10 and 12. There may be other reasons that are recommended for more investigation such as gene expression of the Cow–Oryx iSCNT.

5 Conclusion

Our current investigations have proved the occurrence of intergroup variability in the in vitro developmental outcomes of interspecies (oryx → bovine) cloned embryos generated using somatic cell lines at different passage numbers (between 10th and 13th passage). In contrast, the use of somatic cell lines between passages 3 and 9 has resulted in remarkable alleviations in the in vitro developmental capabilities of oryx → bovine iSCNT-derived embryos to reach the blastocyst stage. The intergroup differences between the extracorporeal developmental competencies of oryx → bovine cloned embryos might be evoked by a variety of factors that affected the iSCNT efficiency. Among them, one of the most important factors seems to be molecular cooperation between nuclear and mitochondrial genomes in interspecies cloned embryos [32,39,60]. The successful developmental rates of such embryos appear to be arising from epigenomic reprogrammability of foreign (xenogeneic) somatic cell nuclear genomes in iSCNT-derived embryos [61,62,63,64,65,66,67]. For those reasons, additional intensive studies are needed to clarify how the high impact of the abovementioned molecular factors is exerted on the ex vivo development of oryx → bovine iSCNT-derived embryos.

Acknowledgment

The Project for funding this work number (RSP-2021/232) at King Saud University, Riyadh, Saudi Arabia.

  1. Funding information: The authors sincerely acknowledge the Researchers Supporting Project for funding this work number (RSP-2021/232) at King Saud University, Riyadh, Saudi Arabia.

  2. Author contributions: Conceptualization, A.A.A. and M.G.A.; methodology, A.A.A. and R.A.A; validation, A.R.A., A.A.A.; formal analysis, A.A.A.; investigation, A.A.A. and M.G.A.; data curation, A.R.A.; writing – original draft preparation, A.A.A. and A.R.A.; writing – review and editing, A.A.A., A.R.A.; visualization, A.R.A., A.A.A., and M.Q.A.; supervision, A.R.A., and M.Q.A. All authors have read and agreed to the published version of the manuscript.

  3. Conflict of interest: The authors declare that they have no conflict of interest.

  4. Ethical approval: All of the experiments were conducted according to the Guidelines for the Institutional Animal Care and Use Committee of the Zoology Department, College of Science at King Saud University.

  5. Data availability statement: The authors confirm that the data supporting the findings of this study are available within the article.

References

[1] Willadsen SM. The developmental capacity of blastomeres from 4-and 8-cell sheep embryos. Embryo Exp Morph. 1981;65:165–72.10.1242/dev.65.1.165Search in Google Scholar

[2] Willadsen SM. Nuclear transplantation in sheep embryos. Nature. 1998;320:63–5.10.1038/320063a0Search in Google Scholar

[3] Alhimaidi A, Ghadi Al, Iwamoto M, Almutary D, Alfuraje M, Alzeer M, et al. The enhancement of Scripted in the development of cloned sheep embryos. Int J Sci Res. 2014;3:538–41.Search in Google Scholar

[4] Al Ghadi MQ, Alhimaidi AR, Iwamoto D, Al Mutary MG, Ammari AA, Saeki KO, et al. The in vitro development of cloned sheep embryos treated with Scriptaid and Trichostatin (A). Saudi J Biol Sci. 2020;27:2280–6.10.1016/j.sjbs.2020.04.039Search in Google Scholar

[5] Li H, Chen S, Piao S, An T, Wang C. Production of artificial synthetic spidroin gene 4S-transgenic cloned sheep embryos using somatic cell nuclear transfer. Anim Biotechnol. 2021;32:616–26.10.1080/10495398.2020.1737098Search in Google Scholar

[6] Bondioli KR, Westhusin ME, Looney CR. Production of identical bovine offspring by nuclear transfer. Theriogenology. 1990;33:165–74.10.1016/0093-691X(90)90607-USearch in Google Scholar

[7] Singina GN, Sergiev PV, Lopukhov AV, Rubtsova MP, Taradajnic NP, Ravin NV, et al. Production of a cloned offspring and crispr/cas9 genome editing of embryonic fibroblasts in cattle. Doklady Biochem Biophys. 2021;496:48–51.10.1134/S1607672921010099Search in Google Scholar PubMed PubMed Central

[8] Collas P, Balise JJ, Robl JM. Influence of cell cycle stage of the donor nucleus on development of nuclear transplant rabbit embryos. Biol Reprod. 1992;46:492–500.10.1095/biolreprod46.3.492Search in Google Scholar PubMed

[9] Qu P, Shen C, Du Y, Qin H, Luo S, Fu S, et al. Melatonin protects rabbit somatic cell nuclear transfer (SCNT) embryos from electrofusion damage. Sci Rep. 2020;10:1–9.10.1038/s41598-020-59161-6Search in Google Scholar PubMed PubMed Central

[10] Prather RS, Sims MM, First NL. Nuclear transplantation in early pig embryos. Biol Reprod. 1989;41:414–8.10.1095/biolreprod41.3.414Search in Google Scholar PubMed

[11] Kamimura S, Inoue K, Mizutani E, Kim JM, Inoue H, Ogonuki N, et al. Improved development of mouse somatic cell nuclear transfer embryos by chlamydocin analogues, class I and IIa histone deacetylase inhibitors. Biol Reprod. 2021;105:543–53.10.1093/biolre/ioab096Search in Google Scholar PubMed PubMed Central

[12] Meng L, Ely JJ, Stouffer RL, Wolf DP. Rhesus monkeys produced by nuclear transfer. Biol Reprod. 1997;57:454–9.10.1095/biolreprod57.2.454Search in Google Scholar

[13] Hajian M, Jafarpour F, Aghamiri SM, Varnosfaderani SR, Esfahani MHN. Effects of ovary storage temperature and embryo vitrification on somatic cell nuclear transfer outcomes in goats. Reprod Fertil Dev. 2020;32:419–24.10.1071/RD18529Search in Google Scholar

[14] Skrzyszowska M, Samiec M. Generating cloned goats by somatic cell nuclear transfer – molecular determinants and application to transgenics and biomedicine. Int J Mol Sci. 2021;22(14):7490.10.3390/ijms22147490Search in Google Scholar

[15] Wani NA, Wernery U, Hassan FAH, Wernery R, Skidmore JA. Production of the first cloned camel by somatic cell nuclear transfer. Biol Reprod. 2021;82:373–9.10.1095/biolreprod.109.081083Search in Google Scholar

[16] Wani NA, Kumar KP, Hong S, Umer MA. Telomere length in dromedary camels (Camelus dromedarius) produced by somatic cell nuclear transfer (SCNT) and their age-matched naturally produced counterparts. Theriogenology. 2022;177:151–6.10.1016/j.theriogenology.2021.10.012Search in Google Scholar

[17] Lanza RP, Cibelli JB, Diaz F, Moraes CT, Farin PW, Farin CE, et al. Cloning of an endangered species (Bos gaurus) using interspecies nuclear transfer. Cloning. 2000;2:79–90.10.1089/152045500436104Search in Google Scholar

[18] Saikhun Pavasuthipaisit K, Jaruansuwan M, Kitiyanant Y. Xenonuclear transplantation of buffalo (Bubalus bubalis) fetal and adult somatic cell nuclei into bovine (Bos indicus) oocyte cytoplasm and their subsequent development. Theriogenology. 2002;57:1829–37.10.1016/S0093-691X(02)00667-2Search in Google Scholar

[19] Lu F, Shi D, Wei J, Yang S, Wei Y. Development of embryos reconstructed by interspecies nuclear transfer of adult fibroblasts between buffalo (Bubalus bubalis) and cattle (Bos indicus). Theriogenology. 2005;64:1309–19.10.1016/j.theriogenology.2005.03.005Search in Google Scholar PubMed

[20] Ikumi S, Sawai K, Takeuchi Y, Iwayama H, Ishikawa H, Ohsumi S, et al. Interspecies Somatic Cell Nuclear Transfer for in Vitro Production of Antarctic Minke Whale (Balaenoptera bonaerensis) Embryos. Cloning Stem Cell. 2004;6:284–93.10.1089/clo.2004.6.284Search in Google Scholar PubMed

[21] Kim J:. Chemosensitization prevents tolerance of Aspergillus fumigatus to antimycotic drugs. Biochem Biophys Res Commun. 2008;372:71–266.10.1016/j.bbrc.2008.05.030Search in Google Scholar PubMed

[22] Gray R, Dobson R. Extinct ibex is resurrected by cloning. Telegraph. 2009;71:1026–34.Search in Google Scholar

[23] Azuma R, Hatanaka Y, Shin SW, Murai H, Miyashita M, Anzai M, et al. Developmental competence of interspecies cloned embryos produced using cells from large Japanese field mice (Apodemus speciosus) and oocytes from laboratory mice (Mus musculus domesticus). J Reprod Dev. 2020;66(3):255–63.10.1262/jrd.2019-167Search in Google Scholar PubMed PubMed Central

[24] Podstawski P, Samiec M, Skrzyszowska M, Szmatoła T, Semik-Gurgul E, Ropka-Molik K. The induced expression of BPV E4 gene in equine adult dermal fibroblast cells as a potential model of skin sarcoid-like Neoplasia. Int J Mol Sci. 2022;23(4):1970.10.3390/ijms23041970Search in Google Scholar PubMed PubMed Central

[25] Wiater J, Samiec M, Wartalski K, Smorąg Z, Jura J, Słomski R, et al. Characterization of Mono- and Bi-transgenic pig-derived epidermal keratinocytes expressing human FUT2 and GLA Genes – In vitro studies. Int J Mol Sci. 2021;22(18):9683.10.3390/ijms22189683Search in Google Scholar PubMed PubMed Central

[26] Wang C, Swanson WF, Herrick JR, Lee K, Machaty Z. Analysis of cat oocyte activation methods for the generation of feline disease models by nuclear transfer. Reprod Biol Endocrinol. 2009;7:148.10.1186/1477-7827-7-148Search in Google Scholar PubMed PubMed Central

[27] Samiec M, Skrzyszowska M, Lipiński D. Pseudophysiological transcomplementary activation of reconstructed oocytes as a highly efficient method used for producing nuclear-transferred pig embryos originating from transgenic foetal fibroblast cells. Pol J Vet Sci. 2012;15(3):509–16.10.2478/v10181-012-0078-3Search in Google Scholar PubMed

[28] Oh HJ, Lee BC, Kim MK. Optimal treatment of 6-dimethylaminopurine enhances the in vivo development of canine embryos by rapid initiation of DNA synthesis. Int J Mol Sci. 2021;22(14):7757.10.3390/ijms22147757Search in Google Scholar PubMed PubMed Central

[29] Srirattana K, St John JC. Transmission of dysfunctional mitochondrial DNA and its implications for mammalian reproduction. Adv Anat Embryol Cell Biol. 2019;231:75–103.10.1007/102_2018_3Search in Google Scholar PubMed

[30] Samiec M. The role of mitochondrial genome (mtDNA) in somatic and embryo cloning of mammals. A Rev J Anim Feed Sci. 2005;14(2):213–33.10.22358/jafs/67008/2005Search in Google Scholar

[31] Yin T, Wang J, Xiang H, Pinkert CA, Li Q, Zhao X. Dynamic characteristics of the mitochondrial genome in SCNT pigs. Biol Chem. 2019;400(5):613–23.10.1515/hsz-2018-0273Search in Google Scholar PubMed

[32] Samiec M, Skrzyszowska M. Extranuclear inheritance of mitochondrial genome and epigenetic reprogrammability of chromosomal telomeres in somatic cell cloning of mammals. Int J Mol Sci. 2021;22(6):3099.10.3390/ijms22063099Search in Google Scholar PubMed PubMed Central

[33] Su G, Wang L, Gao G, Wu S, Yang L, Wu M, et al. C23 gene regulates the nucleolin structure and biosynthesis of ribosomes in bovine intraspecific and interspecific somatic cell nuclear transfer embryos. FASEB J. 2021;35(11):e21993.10.1096/fj.202100737RRSearch in Google Scholar PubMed

[34] Wiater J, Samiec M, Skrzyszowska M, Lipiński D. Trichostatin A-assisted epigenomic modulation affects the expression profiles of not only recombinant human α1,2-fucosyltransferase and α-galactosidase a enzymes but also Galα1 → 3Gal epitopes in porcine Bi-transgenic adult cutaneous fibroblast cells. Int J Mol Sci. 2021;22(3):1386.10.3390/ijms22031386Search in Google Scholar PubMed PubMed Central

[35] Kim MJ, Oh HJ, Choi YB, Lee S, Setyawan EMN, Lee SH, et al. Suberoylanilide hydroxamic acid during in vitro culture improves development of dog-pig interspecies cloned embryos but not dog cloned embryos. J Reprod Dev. 2018;64(3):277–82.10.1262/jrd.2017-112Search in Google Scholar PubMed PubMed Central

[36] Samiec M, Skrzyszowska M, Opiela J. Creation of cloned pig embryos using contact-inhibited or serum-starved fibroblast cells analysed intra vitam for apoptosis occurrence. Ann Anim Sci. 2013;13(2):275–93.10.2478/aoas-2013-0009Search in Google Scholar

[37] Alsalim H, Jafarpour F, Ghazvini Zadegan F, Nasr-Esfahani MH, Niasari-Naslaji A. Epigenotoxic effect of dimethyl sulfoxide on buffalo somatic cells and buffalo-bovine interspecies somatic cell nuclear transfer embryos. Cell J. 2019;20(4):544–51.Search in Google Scholar

[38] Gorczyca G, Wartalski K, Wiater J, Samiec M, Tabarowski Z, Duda M. Anabolic steroids-driven regulation of porcine ovarian putative stem cells favors the onset of their neoplastic transformation. Int J Mol Sci. 2021;22(21):11800.10.3390/ijms222111800Search in Google Scholar PubMed PubMed Central

[39] Gupta MK, Das ZC, Heo YT, Joo JY, Chung HJ, Song H, et al. Transgenic chicken, mice, cattle, and pig embryos by somatic cell nuclear transfer into pig oocytes. Cell Reprogram. 2013;15(4):322–8.10.1089/cell.2012.0074Search in Google Scholar PubMed PubMed Central

[40] Damiani R, Haimov S. A high-resolution dual Doppler technique for fixed multi-antenna airborne radar. IEEE Trans Geosci Remote Sens. 2006;42:3475–89.10.1109/TGRS.2006.881745Search in Google Scholar

[41] IUCN. Arabian Oryx (Oryx leucoryx). IUCN Red List of Threatened Species; 2008. WWW.iucnredlist.org SSC Antelope Specialist Group.Search in Google Scholar

[42] IUCN. Arabian Oryx (Oryx leucoryx). IUCN Red List of Threatened Species; 2012. WWW.iucnredlist.org SSC Antelope Specialist Group.Search in Google Scholar

[43] Al Mutary MG, Al Ghadi MQ, Ammari AA, Al Himadi AR, Al Jolimeed A,H, Arafah M, et al. Effect of different concentrations of resveratrol on the quality and in vitro fertilizing ability of ram semen stored at 5° C for upto 168h. Theriogenology. 2020;152:139146.10.1016/j.theriogenology.2020.05.001Search in Google Scholar PubMed

[44] Jin Y, Zhang M, Ju X, Liang S, Xiong Q, Zhao L, et al. Factors influencing the somatic cell nuclear transfer efficiency in pigs. Front Agric Sci Eng. 2019;6:73–80.10.15302/J-FASE-2018220Search in Google Scholar

[45] Li J, Gao Y, Petkov S, Purup S, Hyttel P, Callesen H. Passage number of porcine embryonic germ cells affects epigenetic status and blastocyst rate following somatic cell nuclear transfer. Anim Reprod Sci. 2014;147:39–46.10.1016/j.anireprosci.2014.03.012Search in Google Scholar PubMed

[46] Noer A, Sørensen AL, Boquest AC, Collas P. Stable CpG hypomethylation of adipogenic promoters in freshly isolated, cultured, and differentiated mesenchymal stem cells from adipose tissue. Mol Biol Cell. 2006;17:3543–56.10.1091/mbc.e06-04-0322Search in Google Scholar PubMed PubMed Central

[47] Wagner M, Schmelz K, Dörken B, Tamm I. Epigenetic and genetic analysis of the survivin promoter in acute myeloid leukemia. Leukemia Res. 2008;32:1054–60.10.1016/j.leukres.2007.11.013Search in Google Scholar PubMed

[48] Mastromonaco GF, Favetta LA, Smith LC, Filion F, king WA. The Influence of nuclear content on developmental competence of gaur x cattle hybrid in vitro fertilized and somatic cell nuclear transfer embryos. Biol Reprod. 2007;76:514–23.10.1095/biolreprod.106.058040Search in Google Scholar PubMed

[49] Wang K, Out HH, Chen Y, Lee Y, Latham K, Cibelli JB. Reprogrammed transcriptome in rhesus-bovine interspecies somatic cell nuclear transfer embryos. PLoS ONE. 2001;6:19–22.10.1371/journal.pone.0022197Search in Google Scholar PubMed PubMed Central

[50] Lagutina I, Zakhartchenko V, Fulka H, Colleoni S, Wolf E, Fulka J, et al. Formation of nucleoli in interspecies nuclear transfer embryos derived from bovine, porcine, and rabbit oocytes and nuclear donor cells of various species. Reproduction. 2011;141:453.10.1530/REP-10-0266Search in Google Scholar PubMed

[51] Kenyon L, Moraes CT. Expanding the functional human mitochondrial DNA database by the establishment of primate xenomitochondrial cybrids. Proc Natl Acad Sci. 1997;94:9131–5.10.1073/pnas.94.17.9131Search in Google Scholar PubMed PubMed Central

[52] Narbonne P, Simpson DE, Gurdon JB. Deficient induction response in a Xenopus nucleocytoplasmic hybrid. PLoS Biol. 2011;9:100–19.10.1371/journal.pbio.1001197Search in Google Scholar PubMed PubMed Central

[53] Lagutina I, Fulka H, Lazzari G, Galli C. Interspecies somatic cell nuclear transfer: advancements and problems. Cell Reprogram. 2013;15:374–84.10.1089/cell.2013.0036Search in Google Scholar PubMed PubMed Central

[54] Amarnath D, Choi I, Moawad AR, Wakayama T, Campbell KHS. Nuclear-cytoplasmic incompatibility and inefficient development of pig-mouse cytoplasmic hybrid embryos. Reproduction. 2011;142:295–307.10.1530/REP-11-0044Search in Google Scholar PubMed

[55] Yan J, Kelly R, Peters A, Fulka H, Dickinson A, Mitchell DA, et al. Interspecies somatic cell nuclear transfer is dependent on compatible mitochondrial DNA and reprogramming factors. PLoS One. 2011;6:48–50.Search in Google Scholar

[56] Shago M, Yi Z. Somatic cell nuclear transfer reprogramming: mechanisms and applications. Cell Stem Cell Rev. 2018;23:471–85.10.1016/j.stem.2018.06.018Search in Google Scholar PubMed PubMed Central

[57] Beaujean N, Taylor JE, Mcgarry M, Gardner JO, Wilmut I, Loi P, et al. The effect of interspecific oocytes on demethylation of sperm DNA. Proc Natl Acad Sci. 2004;101:7636–40.10.1073/pnas.0400730101Search in Google Scholar PubMed PubMed Central

[58] Chen T, Zhang YL, Yang Liu SZ, Schatten H, Chen DY, Sun QY. The DNA methylation events in normal and cloned rabbit embryos. FEBS Lett. 2004;578:69–72.10.1016/j.febslet.2004.10.073Search in Google Scholar PubMed

[59] González Grajales LA, Favetta LA, King WA, Mastromonaco GF. Developmental competence of 8–16-cell stage bison embryos produced by interspecies somatic cell nuclear transfer. Reprod Fertil Dev. 2016;28:1360–8.10.1071/RD14376Search in Google Scholar PubMed

[60] Son YB, Jeong YI, Hwang KC, Jeong YW, Hwang WS. Mitochondrial metabolism assessment of Lycaon-dog fetuses in interspecies somatic cell nuclear transfer. Theriogenology. 2021 Apr 15;165:18–27.10.1016/j.theriogenology.2021.01.010Search in Google Scholar PubMed

[61] Hua S, Lu C, Song Y, Li R, Liu X, Quan F, et al. High levels of mitochondrial heteroplasmy modify the development of ovine-bovine interspecies nuclear-transferred embryos. Reprod Fertil Dev. 2012;24(3):501–9.10.1071/RD11091Search in Google Scholar PubMed

[62] No JG, Hur TY, Zhao M, Lee S, Choi MK, Nam YS, et al. Scriptaid improves the reprogramming of donor cells and enhances canine-porcine interspecies embryo development. Reprod Biol. 2018;18(1):18–26.10.1016/j.repbio.2017.11.001Search in Google Scholar PubMed

[63] Saini M, Selokar NL, Raja AK, Sahare AA, Singla SK, Chauhan MS, et al. Effect of donor cell type on developmental competence, quality, gene expression, and epigenetic status of interspecies cloned embryos produced using cells from wild buffalo and oocytes from domestic buffalo. Theriogenology. 2015;84(1):101–8.e1.10.1016/j.theriogenology.2015.02.018Search in Google Scholar PubMed

[64] Samiec M, Skrzyszowska M. Can reprogramming of overall epigenetic memory and specific parental genomic imprinting memory within donor cell-inherited nuclear genome be a major hindrance for the somatic cell cloning of mammals? – A review. Ann Anim Sci. 2018a;18(3):623–38.10.2478/aoas-2018-0015Search in Google Scholar

[65] Samiec M, Skrzyszowska M. Intrinsic and extrinsic molecular determinants or modulators for epigenetic remodeling and reprogramming of somatic cell-derived genome in mammalian nuclear-transferred oocytes and resultant embryos. Pol J Vet Sci. 2018b;21(1):217–27.Search in Google Scholar

[66] Zuo Y, Gao Y, Su G, Bai C, Wei Z, Liu K, et al. Irregular transcriptome reprogramming probably causes the developmental failure of embryos produced by interspecies somatic cell nuclear transfer between the Przewalski’s gazelle and the bovine. BMC Genomics. 2014;15(1):1113.10.1186/1471-2164-15-1113Search in Google Scholar PubMed PubMed Central

[67] Wittayarat M, Sato Y, Do LT, Morita Y, Chatdarong K, Techakumphu M, et al. Histone deacetylase inhibitor improves the development and acetylation levels of cat-cow interspecies cloned embryos. Cell Reprogram. 2013;15(4):301–8.10.1089/cell.2012.0094Search in Google Scholar PubMed PubMed Central

Received: 2022-01-28
Revised: 2022-03-28
Accepted: 2022-03-31
Published Online: 2022-04-20

© 2022 Aiman A. Ammari et al., published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 28.3.2024 from https://www.degruyter.com/document/doi/10.1515/chem-2022-0153/html
Scroll to top button