reproduction · reproduction research unequal distribution of 16s mtrrna at the 2-cell stage...

17
REPRODUCTION RESEARCH Unequal distribution of 16S mtrRNA at the 2-cell stage regulates cell lineage allocations in mouse embryos Zhuxia Zheng 1,2 , Hongmei Li 3 , Qinfen Zhang 3 , Lele Yang 1 and Huayu Qi 1 1 Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, A303, 190 Kaiyuan Boulevard, Science City, Huangpu District, Guangzhou, Guangdong 510630, China, 2 School of Life Science, University of Science and Technology of China, Hefei 230026, China and 3 State Key Lab for Bio-control, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China Correspondence should be addressed to H Qi; Email: [email protected] Abstract Cell lineage determination during early embryogenesis has profound effects on adult animal development. Pre-patterning of embryos, such as that of Drosophila and Caenorhabditis elegans, is driven by asymmetrically localized maternal or zygotic factors, including mRNA species and RNA binding proteins. However, it is not clear how mammalian early embryogenesis is regulated and what the early cell fate determinants are. Here we show that, in mouse, mitochondrial ribosomal RNAs (mtrRNAs) are differentially distributed between 2-cell sister blastomeres. This distribution pattern is not related to the overall quantity or activity of mitochondria which appears equal between 2-cell sister blastomeres. Like in lower species, 16S mtrRNA is found to localize in the cytoplasm outside of mitochondria in mouse 2-cell embryos. Alterations of 16S mtrRNA levels in one of the 2-cell sister blastomere via microinjection of either sense or anti- sense RNAs drive its progeny into different cell lineages in blastocyst. These results indicate that mtrRNAs are differentially distributed among embryonic cells at the beginning of embryogenesis in mouse and they are functionally involved in the regulation of cell lineage allocations in blastocyst, suggesting an underlying molecular mechanism that regulates pre-implantation embryogenesis in mouse. Reproduction (2016) 151 351–367 Introduction Embryogenesis following fertilization lays down the foundation for future animal development. During early embryogenesis, descendent of the totipotent zygote (the 1-cell embryo that formed by sperm–egg fusion) gradually lose their developmental potential and acquire specific cell fate. Understanding the molecular inner works that underlie the regulation of cell fate determina- tion during early embryogenesis has been one of the central issues in developmental biology (Johnson 2009, Rossant & Tam 2009, St Johnston & Ahringer 2010, Nance 2014, Du et al. 2015). In lower species, such as Drosophila and Caenorhab- ditis elegans, embryogenesis is believed being pre- patterned. Unfertilized eggs and zygotes are highly polarized cells with distinctive cellular geometry, containing predisposed cell fate determinants that are asymmetrically localized in the cell, including various RNA species and RNA binding proteins (Johnstone & Lasko 2001, St Johnston & Ahringer 2010). Differentially segregated cell fate determinants can drive the inherited cells into specific cell lineages, such as the germ line cells (Johnstone & Lasko 2001, Kimble & Crittenden 2007). The early cellular polarity of eggs or zygotes can be translated into axes of developing embryos, such as the animal–vegetal and anterior–posterior axes, which are strongly correlated with the development of body axes in adult animals. Although the underpinning molecular mechanisms of these correlations remain to be fully elucidated, cell lineage formation during early embryogenesis in lower species is profoundly influenced by pre-existing intrinsic factors, mostly inherited from the maternal source. In mammals, the establishment of distinguishable cell lineages following fertilization is thought to occur first at the blastocyst stage during pre-implantation embryo- genesis (Rossant & Tam 2009). Mammalian blastocyst initially contains two cell-lineages: the inner cell mass (ICM) and the trophectoderm (TE). With development, ICM cells are further differentiated into epiblast (Epi) that will give rise to the embryo proper and primitive endoderm (PrE) that together with the TE will generate extra-embryonic tissues, including placenta. It is postu- lated that, in mouse, the blastocyst is bi-laterally symmetric and can be divided into embryonic half q 2016 Society for Reproduction and Fertility DOI: 10.1530/REP-15-0301 ISSN 1470–1626 (paper) 1741–7899 (online) Online version via www.reproduction-online.org Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PM via free access

Upload: others

Post on 24-Jan-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

  • R

    EPRODUCTIONRESEARCH

    Unequal distribution of 16S mtrRNA at the 2-cell stageregulates cell lineage allocations in mouse embryos

    Zhuxia Zheng1,2, Hongmei Li3, Qinfen Zhang3, Lele Yang1 and Huayu Qi1

    1Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and RegenerativeMedicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes ofBiomedicine and Health, Chinese Academy of Sciences, A303, 190 Kaiyuan Boulevard, Science City, HuangpuDistrict, Guangzhou, Guangdong 510630, China, 2School of Life Science, University of Science and Technology ofChina, Hefei 230026, China and 3State Key Lab for Bio-control, School of Life Sciences, Sun Yat-sen University,Guangzhou 510275, China

    Correspondence should be addressed to H Qi; Email: [email protected]

    Abstract

    Cell lineage determination during early embryogenesis has profound effects on adult animal development. Pre-patterning of embryos,

    such as that of Drosophila and Caenorhabditis elegans, is driven by asymmetrically localized maternal or zygotic factors, including

    mRNA species and RNA binding proteins. However, it is not clear how mammalian early embryogenesis is regulated and what the early

    cell fate determinants are. Here we show that, in mouse, mitochondrial ribosomal RNAs (mtrRNAs) are differentially distributed between

    2-cell sister blastomeres. This distribution pattern is not related to the overall quantity or activity of mitochondria which appears equal

    between 2-cell sister blastomeres. Like in lower species, 16S mtrRNA is found to localize in the cytoplasm outside of mitochondria in

    mouse 2-cell embryos. Alterations of 16S mtrRNA levels in one of the 2-cell sister blastomere via microinjection of either sense or anti-

    sense RNAs drive its progeny into different cell lineages in blastocyst. These results indicate that mtrRNAs are differentially distributed

    among embryonic cells at the beginning of embryogenesis in mouse and they are functionally involved in the regulation of cell lineage

    allocations in blastocyst, suggesting an underlying molecular mechanism that regulates pre-implantation embryogenesis in mouse.

    Reproduction (2016) 151 351–367

    Introduction

    Embryogenesis following fertilization lays down thefoundation for future animal development. Duringearly embryogenesis, descendent of the totipotent zygote(the 1-cell embryo that formed by sperm–egg fusion)gradually lose their developmental potential and acquirespecific cell fate. Understanding the molecular innerworks that underlie the regulation of cell fate determina-tion during early embryogenesis has been one of thecentral issues in developmental biology (Johnson 2009,Rossant & Tam 2009, St Johnston & Ahringer 2010,Nance 2014, Du et al. 2015).

    In lower species, such as Drosophila and Caenorhab-ditis elegans, embryogenesis is believed being pre-patterned. Unfertilized eggs and zygotes are highlypolarized cells with distinctive cellular geometry,containing predisposed cell fate determinants that areasymmetrically localized in the cell, including variousRNA species and RNA binding proteins (Johnstone &Lasko 2001, St Johnston & Ahringer 2010). Differentiallysegregated cell fate determinants can drive the inheritedcells into specific cell lineages, such as the germ line

    q 2016 Society for Reproduction and Fertility

    ISSN 1470–1626 (paper) 1741–7899 (online)

    cells (Johnstone & Lasko 2001, Kimble & Crittenden2007). The early cellular polarity of eggs or zygotes canbe translated into axes of developing embryos, such asthe animal–vegetal and anterior–posterior axes, whichare strongly correlated with the development of bodyaxes in adult animals. Although the underpinningmolecular mechanisms of these correlations remain tobe fully elucidated, cell lineage formation during earlyembryogenesis in lower species is profoundly influencedby pre-existing intrinsic factors, mostly inherited fromthe maternal source.

    In mammals, the establishment of distinguishable celllineages following fertilization is thought to occur firstat the blastocyst stage during pre-implantation embryo-genesis (Rossant & Tam 2009). Mammalian blastocystinitially contains two cell-lineages: the inner cell mass(ICM) and the trophectoderm (TE). With development,ICM cells are further differentiated into epiblast (Epi) thatwill give rise to the embryo proper and primitiveendoderm (PrE) that together with the TE will generateextra-embryonic tissues, including placenta. It is postu-lated that, in mouse, the blastocyst is bi-laterallysymmetric and can be divided into embryonic half

    DOI: 10.1530/REP-15-0301

    Online version via www.reproduction-online.org

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

    http://dx.doi.org/10.1530/REP-15-0301

  • 352 Z Zheng and others

    (including ICM and polar TE) and abembryonic half(containing mural TE) along animal–vegetal (A–V) axis(Gardner 1997). How this bi-lateral symmetry is relatedto the development of body axes in adult animals iscurrently not understood. It is also not clear when andhow different cell lineages and the axes of blastocystare generated during early mammalian embryogenesis.Although unfertilized mouse eggs have an apparentpolarity, drastic cellular changes following fertilizationmake it uncertain whether the maternal polarity isrelevant to the cell lineage determination and axisformation in blastocyst (Whitaker 2006, Li & Albertini2013). Previous research showed that embryonic cellsdo not gain obvious morphological and moleculardifferences until after 8-cell stage (Ducibella & Anderson1975, Johnson & Ziomek 1981, Dard et al. 2004,Guo et al. 2010). Other lines of evidences also suggestedthat cell lineage specification occurs only at much laterstages and embryonic axis formation may be influencedby extrinsic cues, such as the geometry of the egg’sextracellular coat, Zona pellucida (ZP), rather thanintrinsic factors (Motosugi et al. 2005, Kurotaki et al.2007, Tabansky et al. 2013). Data from clonal analysesand transplantation experiments in mouse, however,suggested that cells within early embryos up to16–32-cell stages could have similar developmentalpotential (Fujimori et al. 2003, Tarkowski et al. 2010).One explanation for these seemingly contradictoryobservations is that early embryonic cells possessdevelopmental plasticity that allows them to adaptdifferent developmental conditions brought by bothintrinsic and extrinsic stimuli (Rossant & Tam 2009,Zernicka-Goetz et al. 2009).

    Recent research in mouse have provided direct linksbetween cell-lineage allocations in the blastocyst andearly embryonic events following fertilization (Gardner2001, Piotrowska & Zernicka-Goetz 2001, Piotrowskaet al. 2001), indicating the existence of early cell fatedeterminants in mammals. CDX2, the TE specific markerin the blastocyst, was found to express heterogeneouslyamong blastomeres of 8-cell embryos when cells arestarting to be allocated into either inner or outer layers(Niwa et al. 2005, Jedrusik et al. 2008). It was shown thatboth epigenetic modifiers (arginine methyltransferasePRMT4 and demethylase PRDM14) and histone argininemethylation (H3R26me) patterns are differentiallyexpressed in mouse 4-cell blastomeres (Torres-Padillaet al. 2007, Burton et al. 2013). The chromatin bindingdynamics of POU5F1/OCT4 has also been shown todiffer among embryonic cells at 4-cell stage (Plachtaet al. 2011). These earlier molecular asymmetries arefunctionally relevant to cell lineage allocations and axisformation in the blastocyst, reminiscent of the early cellfate determinants for mammalian embryogenesis.However, it remains to be determined whether the4-cell stage is the earliest embryonic time when cellsgain molecular variations.

    Reproduction (2016) 151 351–367

    First found in Drosophila, one of the maternal cell fatedeterminants that influence the formation of germ cellsin invertebrates is the posterior mitochondrial ribosomalRNAs (mtrRNAs) that are localized outside of theorganelle (Kobayashi et al. 1993, Iida & Kobayashi1998). Examination of mtrRNAs in other species,including Xenopus and Sea urchin, also showed thatthey are localized asymmetrically and outside ofmitochondria in early embryos, suggesting their possiblefunctions as early cell fate determinants in these species(Kobayashi et al. 1998, Ogawa et al. 1999). Investigationof mtrRNAs in mouse pre-implantation embryossuggested that they are localized asymmetrically in thecytoplasm of MII oocytes toward the animal pole wherethe first polar body emits. Similar to that in lower species,16S mtrRNA was also found in the cytoplasm outsideof mitochondria in MII oocytes (Ninomiya & Ichinose2007). However, it was suggested that mtrRNAs wereabsent in mouse 2-cell embryos and no difference wasfound in their distribution among blastomeres of pre-implantation embryos. Their functional roles duringcell lineage specification in mouse have yet to bedetermined. In the present study, we re-examined theexpression and distribution patterns of mtrRNAs inmouse pre-implantation embryos. Results showed thatmtrRNAs are constantly expressed during pre-implan-tation embryogenesis. Both small and large mtrRNAs areunequally distributed between 2-cell sister blastomeres,suggesting a molecular variation between cells at thebeginning of embryogenesis in mouse. Further functionalanalyses suggested that alterations of levels of largesubunit 16S mtrRNA in 2-cell blastomeres functionallyinfluence cell lineage allocations in the blastocyst.

    Materials and methods

    Animal handling and mouse embryos

    CD1 mice were used for embryo isolations for most of theexperiments, except that when testing the hormonal effects onembryos, C57BL/6 mice were also used. Adult female mice (1.5-to 2-month of age) were superovulated by i.p. injection of 10 IUof pregnant mare serum gonadotropin (PMSG) per mouse,followed by 10 IU of human chorionic gonadotropin (HCG)48 h later. Injected female mice were placed with adult malemice overnight and checked for copulation plugs the followingmorning. Animals were sacrificed by cervical dislocationfollowing CO2 anesthetization. Embryos at different stages wereisolated in pre-warmed M2 media (Sigma M7167) and washed inKSOM media (Specialty Media, MR-020P-D). Unfertilized eggswere isolated from females without mating 16–18 h post HCGinjection. Zygotes, early 2-cell, middle 2-cell, late 2-cell, 4-cell,8-cell, morula and blastocyst embryos were collected 24–26 h,31–32 h, 45–46 h, 52–53 h, 56–60 h, 67–68 h, 77–78 h and90–92 h post HCG injection. As controls, CD1 mice that werenaturally mated and superovulated C57BL/6 mice were also usedfor the isolation of embryos. To isolate single blastomeres from2-cell and 4-cell embryos, embryos were placed in Tyrode’s

    www.reproduction-online.org

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

  • 16S mtrRNA regulates cell lineage allocations 353

    solution to remove ZP and blastomeres were washed andseparated by pipetting in PBS/PVP-40 (4 mg/ml). Separatedblastmeres were then transferred into PCR tubes. All animalhusbandryand usage were carried out according to the guidelinesof IACUC and approved by the Animal Care and Use Committeeof Guangzhou Institutes of Biomedicine and Health, ChineseAcademy of Sciences (Permit number: 2008023).

    cDNA cloning and real time quantitative RT-PCR

    cDNAs of 12S and 16S mtrRNA were cloned from mousemitochondria genome extracted from CD1 mouse ovary usingMini Plasmid Extraction kit (Tiangen, Beijing, China). 12S mtrRNAcDNA was amplified using RT-PCR with a 5 0-primer containingT7 promoter sequence for in vitro transcription. 16S mtrRNAcDNA was cloned into pCIneo expression vector (Promega)between SalI/NotI sites under the T7 promoter. PlasmidpEGFP-N2 containing EGFP coding sequence was used forPCR and in vitro transcription of Egfp mRNA.

    For the examination of 16S mtrRNA and Actin expression inmouse pre-implantation embryos, 50 embryos at each pre-implantation stage were collected from superovulated CD1mice. Embryos of each group were lysed in 10 ml lysis buffer(DEPC-H2O containing 0.1% Triton X-100, 0.1 M DTTand 1 mlRNase OUT Recombinant RNase inhibitor, 5000 U, Invitrogen).Same amount of in vitro transcribed Egfp mRNA was added toeach sample as internal control. The RT reaction was carriedout with an adapter–oligo–dT24 primer at 50 8C for 1.5 h. Realtime quantitative PCR was done with SYBR Green Mix (Takara,Otsu Shiga, Japan) in optical 96-well reaction plates on a CFX96Realtime System (Bio-Rad). PCRs were performed in triplicateand signals obtained were normalized against that of Egfp usingthe following equation: 2(CtegfpKCtgeneX).

    Single-cell quantitative RT-PCR, expression of mitochondrialgenes and the expression of 16S mtrRNA following micro-injection were examined in the similar fashion usingendogenous Actin as control. PCRs were performed intriplicate and signals obtained were normalized against thatof Actin using the following equation: 2(CtactinKCtgeneX).Relative gene expression levels were calculated using Excelsoftware. Primers used for cDNA cloning and qRT-PCR aresummarized in Supplementary Table S1, see section onsupplementary data given at the end of this article. Detailedexperimental procedures are provided in SupplementaryMaterials and Methods.

    Whole-mount in situ hybridization of mouse embryos

    Briefly, digoxigenin (DIG)-labeled RNA probes were transcribedin vitro using T7 RNA polymerase (Takara) and DIG RNA LabelingMix (Roche), and purified and stored in DEPC-H2O at K80 8C.Collected embryos were fixed and stained with DIG-labeled cRNAantisense or sense probes (1 mg/ml in pre-hybridization solution).They were then washed and incubated with anti-DIG alkalinephosphatase (AP) conjugates (Roche) at 1:2000 dilution in 1%bovine serum albumin (BSA)/1! Phosphate Buffered Salinecontaining 0.1% Tween20 (PBST) for 2 h. They were furtherwashed before transferred into Staining buffer containingBCIP/NBT solution. Stained embryos were then mounted onto

    www.reproduction-online.org

    glass slides in 50% glycerol/PBS and examined with an invertedmicroscope (Olympus IX71). ZP surrounding embryos weredissolved during hybridization. All procedures were carried outat room temperature unless indicated. Detailed experimentalprocedures are provided in Supplementary Materials and Methods.

    Whole mount in situ hybridization (ISH) was carried outfor the following genes: Actin, 12S mtrRNA, 16S mtrRNA,cytochrome b (Cytb), NADH dehydrogenase subunit 2 (Nd2),cytochrome c-1 (Cyc1), glyceraldehyde-3-phosphate dehydro-genase (Gapdh), hypoxanthine guanine phosphoribosyl trans-ferase (Hprt), high mobility group box3 (Hmgb3), TATA boxbinding protein (TBP)-associated factor 9 (Taf9) and immediateearly response 5 (Ier5). Primers used for amplifying probes aresummarized in Supplementary Table S1.

    ISH at electron microscopic level

    ISH at the electron microscopic level (ISH-EM) was carried outaccording to the procedures described before (Yamashita et al.2009), with some modifications. Briefly, isolated embryoswere first fixed and hybridized with 16S mtrRNA long cRNAprobes as described above. They were then processed forthin (100 nm) sections cut with an Ultramicrotome (LeicaInstruments, Wetzlar, Germany) and mounted onto nickel grids.Grids were subsequently stained with primary mouse mono-clonal anti-DIG antibody (1:200, Sigma) and secondary goatanti-mouse IgG conjugated with 12-nm colloidal gold (1:20,Jackson ImmunoResearch, West Grove, PA, USA). Stainedsamples were examined using an electron microscope (TecnaiG2 Spirit, FEI). Average numbers of colloidal gold particles onsections of stained embryos were counted and calculated foraverage from ten frames of electron micrographs at 16 800!magnification. All procedures were carried out at roomtemperature unless indicated. Transmission electronmicroscopy of thin (100 nm) embryo sections without ISH wasalso carried out according to the method described previously(Zhang et al. 2004). Detailed experimental procedures areprovided in Supplementary Materials and Methods.

    Fluorescent ISH and immunofluorescent staining ofmouse embryos

    Stellaris oligonuleiotide probe sets (20-mers) that cover theentire lengths of 12S mtrRNA and 16S mtrRNA weresynthesized and tagged with Quasar 570 Dye and Quasar670 Dye (Biosearch Technologies, Petaluma, CA, USA)respectively. Collected mouse embryos were fixed and stainedwith 125 nM Stellaris probes, either singly or together. Stainedembryos were then mounted onto glass slides.

    For immunostaining of embryos with antibodies, embryoswere fixed with 4% paraformaldehyde (PFA)/PBST (1! PBScontaining 0.1% Triton X-100), for 15 min. After washing twicewith PBST, they were blocked in 2% BSA/PBST for 2 h andincubated with primary antibodies for 2 h. Fluorescently taggedsecondary antibodies were then used to stain the embryosin 1% BSA/PBST for another 2 h. Nuclei were stained with40, 6-diamidino-2-phenylindole dihydrochloride (DAPI) (0.5 mg/ml)for 3 min. Stained embryos were washed three times withPBST and mounted onto glass slides in 50% glycerol/PBS.

    Reproduction (2016) 151 351–367

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

    http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1

  • 354 Z Zheng and others

    Live embryos were stained with MitoTracker Red (200 mM,Molecular Probes) for detecting mitochondria content andtetramethyl rhodamine methyl ester (TMRM, 25 mM, MolecularProbes) for detecting mitochondrial membrane potential, withor without pre-treatment of protonophore carbonyl cyanidep-trifluoromethoxyphenyl-hydrazone (FCCP). Hoechst 33342(10 mg/ml) was used for nuclei staining for 30 min. In somecases, embryos stained with MitoTracker Red were furtherprocessed for fluorescent ISH (FISH) of mtrRNAs using Stellarisprobes as described above. Embryos microinjected with 16SmtrRNA sense or anti-sense RNAs, together with Egfp mRNA,were also stained with TMRM in the same way 8 h post-injection.Stained embryos were examined with Confocal Laser ScanningMicroscopy (CLSM, LSM 710 NLO, Carl Zeiss, Jena, Germany).

    Primary antibodies used were: rabbit polyclonal anti-E-cadherin (Abcam, 1:50); mouse monoclonal anti-CDX2(Abcam, ab115595, 1:50); rabbit polyclonal anti-NANOG(Abcam, ab80892, 1:100) and TRITC-Phalloidin (Sigma,1:5000). Respective secondary antibodies used were:goat-anti-rabbit FITC conjugate (Molecular Probes, 1:1000);Goat-anti-mouse Alexa Fluor568 (Molecular Probes,1:1000) or goat-anti-rabbit Alexa Fluor568 (Molecular Probes,1:1000). Detailed experimental procedures are provided inSupplementary Materials and Methods.

    RNA microinjection of 2-cell embryos and cellallocation analysis

    mRNAs of Egfp were transcribed in vitro using mMessagemMachine SP6/T7 Ultra Kit with Capping analogous (Ambion)from linearized pCS2 vector containing EGFP open readingframe (ORF) with human serum albumin 5 0 and 3 0-UTRs.Full-length 16S mtrRNA sense or antisense RNAs weretranscribed in vitro using T7 promoter with the same kitwithout Capping analogous. Transcribed RNAs were purifiedusing RNeasy Mini Kit (Qiagen), re-suspended in DEPC-H2Oand stored at K80 8C. Ten fragments (100–200 nt in length) ofanti-sense RNAs, spanning the entire 16S mtrRNA sequencewere prepared and mixed together for microinjection. Singleblastomere of a 2-cell embryo was microinjected with eitherEgfp mRNA (0.8 mg/ml) alone, or together with 16S mtrRNAsense (1.5 mg/ml, 2:1) or anti-sense RNAs (1.5 mg/ml, 2:1)respectively.

    Injected embryos were cultured in vitro in KSOM for 46–48 htill blastocyst stage in a humidified incubator containing 5%CO2, 37 8C. Embryos were then fixed in 4% PFA/PBST andstained with DAPI (0.5 mg/ml). Each embryo containing EGFP-labeled (EGFPC) cells was scanned at 2-mm intervals usingCLSM. Scanned images from each embryo were composedtogether and examined at three dimensions using ZEN 2010software (Carl Zeiss). Total number of cells and positions ofindividual cells within an embryo were determined by theirnuclear DAPI staining, in relation to the surrounding cells.Using ortho module of the software, cells that were located onthe surface of the embryo along either one of the three axes(X, Yand Z) were designated as TE cells, whereas cells that arelocated on the inside of the embryo and surrounded by othercells along all three axes were assigned as ICM cells. Positionsof EGFPC cells were determined by superimposing the green

    Reproduction (2016) 151 351–367

    fluorescence over DAPI staining. Total number of cells witheither EGFP signal or CDX2 staining or both were counted inthe same way. Alternatively, distributions of EGFPC progenycells in either embryonic, abembryonic or both sides withinblastocysts were examined using 3-D confocal images.Embryos were grouped into three different patterns accordingto the allocations of EGFPC cells within them. Pattern 1contains EGFPC cells in the embryonic side only; pattern 2contains EGFPC cells in both embryonic and abembryonicsides and pattern 3 contains EGFPC cells in the abembryonicside only. Total numbers of cells with or without EGFP ineither ICM or TE and patterns of embryos followingmicroinjection were counted and calculated using Excelsoftware. Cell numbers surveyed were summarized inSupplementary Tables, see section on supplementary datagiven at the end of this article.

    Statistical analyses

    Signal intensities of whole-mount ISH were measured againstbackground using outlined single blastomere from lightmicrographs using Image-Pro 6 software. Fluorescent signals,including that of FISH, were quantified from entire stacks ofconfocal sections for each sister blastomere using ImageJsoftware. Ratios of signal intensities (RZOD1/OD2, OD1OOD2, OD: optical density measured for each sister blastomere)between 2-cell sister blastomeres were then calculated usingExcel. Statistical significances of signal variances betweenexperimental and control groups were assessed using non-parametric Mann–Whitney test for independent samples.Alternatively, significance of variation was also examinedusing z-test for two samples’ means. One-way ANOVA andpost-hoc Turkey test were performed to assess the significanceof differences when there were more than two groups of datain the experiment using QIMacros software. c2 test wasperformed for assessing the significance of changes ofembryonic patterns following microinjection. Significancewas set as P!0.05. Data are presented as meanGS.D.

    Results

    Expression of mtrRNAs in mouse pre-implantationembryos

    The expression of 16S mtrRNA in mouse oocytes andpre-implantation embryos following fertilization wasfirst examined using quantitative RT-PCR. In order tohave an internal control with constant quantity, sameamount of in vitro transcribed Egfp mRNA was added toeach RNA sample extracted from embryos at variousstages before RT reaction. Quantitative RT-PCR was thenused to examine the expression of 16S mtrRNA. It wasfound that levels of 16S mtrRNA remained constant inMII oocytes, zygotes and 2-cell embryos and increaseddramatically after 4-cell stage, similar to the expressionpattern of Actin control (Fig. 1D). No apparent decreaseof 16S mtrRNA was found in 2-cell embryos.

    To confirm the expression of mtrRNAs in pre-implantation embryos, ISH of both small (12S) and

    www.reproduction-online.org

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

    http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1

  • A

    B Early 2-cell Mid 2-cell Late 2-cellZygoteMII oocyte

    3-cell 4-cell 8-cell Morula Blastocyst

    16S

    mtr

    RN

    A

    20 µm

    MII oocyte Early 2-cell Mid 2-cell Late 2-cell

    4-cell

    12S

    mtr

    RN

    A

    Zygote

    BlastocystMorula8-cell3-cell

    20 µm

    20 µm

    Act

    in

    MII oocyte Early 2-cell Mid 2-cell Late 2-cellZygote

    4-cell BlastocystMorula8-cell3-cell

    C

    D E 2-cell 4-cellZygote

    10 µm12S mtrRNA/16S mtrRNA02

    4

    6

    8

    10

    12

    14

    16

    NO RT Oocyte 1-cell 2-cell 4-cell 8-cell Morula Blastocyst

    qRT-

    PC

    R o

    f 16S

    mtr

    RN

    A a

    nd A

    ctin

    16S mtrRNA

    Actin

    *

    *

    *

    16S mtrRNA regulates cell lineage allocations 355

    www.reproduction-online.org Reproduction (2016) 151 351–367

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

  • 356 Z Zheng and others

    large (16S) mtrRNAs was applied using long cRNAprobes containing DIG labeled uridine, coupled withanti-DIG-AP conjugates. Strong signals of both 12S and16S mtrRNAs were seen in embryos at all pre-implantation stages (Fig. 1A and B). In consistence withprevious report, mtrRNAs were found to concentrate inthe area toward the animal pole where the first polarbody emits in MII oocytes (Ninomiya & Ichinose 2007),whereas they appeared to distribute in the cytoplasm ofzygotes surrounding pro-nuclei. Following compactionafter 8-cell stage, higher levels of mtrRNAs were foundin inner cells of morula and blastocysts, comparingwith the outer cells of embryos. In contrast, Actin controlappeared to be evenly distributed in all embryonic cells(Fig. 1C). Using short oligonucleotide probes (20-mers)encompassing the entire lengths of mtrRNAs, FISH wasfurther performed to determine the expression ofmtrRNAs in mouse embryos. These Stellaris oligonucleo-tide probes were tagged with either Quasar 570 orQuasar 670 Dye for 12S and 16S mtrRNAs respectively.Similarly, signals of mtrRNAs were seen in pre-implantation embryos at all stages, including 2-celland 4-cell embryos (Fig. 1E). These results indicate thatmtrRNAs are continuously expressed in mouse embryosacross all pre-implantation stages.

    mtrRNAs are differentially distributed between mouse2-cell blastomeres

    Although no clear asymmetric distribution of mtrRNAswas found in zygotes, one of the 2-cell sister blastomeresin some embryos appeared to contain more mtrRNAsthan the other (Fig. 1A and B, late 2-cells). To betterquantify and define the differences of mtrRNAs betweensister blastomeres, 2-cell embryos were separated intoearly, middle and late stages and hybridized withDIG-labeled long cRNA probes (Fig. 2A). The opticaldensities of ISH signals in sister blastomeres were thenmeasured in pair for each 2-cell embryo outlined fromlight micrographs. Ratios of signal intensity betweentwo sister blastomeres (the higher vs the lower one,RZOD1/OD2, OD1OOD2) were calculated as anindicator for differences of RNA levels. Average ratioswere obtained from groups of embryos at each 2-cellstage for each RNA species examined (Fig. 1B and C,Supplementary Table S2). It was found that the ratios

    Figure 1 Expression of mitochondrial ribosomal RNAs in mouse pre-implanembryos at different stages. (B) In situ hybridization of 16S mtrRNA in mousembryos at different stages. MII oocytes were also included. Stars indicate thbetween late 2-cell blastomeres in (A) and (B). (D) Quantitative RT-PCR of 16remained at constant level before 2-cell stage and greatly increased afterwaof Actin also increased after 2-cell stage (red dashed line). Same amount ofMaterials and methods). PCR without RT was included as negative control.reaction. (E) Fluorescent in situ hybridization (FISH) of mouse pre-implantaprobes tagged with Quasar 570 (for 12S mtrRNA) and Quasar 670 dye (for 16green. Shown are merged confocal images.

    Reproduction (2016) 151 351–367

    of Actin mRNA signals were around 1 between sisterblastomeres at all three 2-cell stages, suggesting an equalamount of Actin mRNA between sister blastomeres.In contrast, ratios of signals were much higher for both12S mtrRNA (RZ1.4G0.2, nZ24, late stage) and16S mtrRNA (RZ1.7G0.6, nZ30, late stage) between2-cell sister blastomeres (Fig. 2B and C). The signaldifferences of 12S and 16S mtrRNAs appeared to besignificantly varied when their ratios were comparedwith those of Actin control at the same stage (P!0.05,Mann–Whitney test). Furthermore, variations of mtrRNAlevels between sister blastomeres increased with thedevelopment of 2-cell embryos, comparing to that ofActin (P!0.05, one-way ANOVA, aZ0.05) (Supple-mentary Table S3). The difference of 16S mtrRNAbetween 2-cell sister blastomeres could reach up tothreefold in some of the embryos examined (Supple-mentary Table S2).

    To confirm that the relative quantification of ISHsignals indeed reflects different levels of RNA speciesbetween 2-cell sister blastomeres, additional controlexperiments were performed on house-keeping genes(Gapdh and Hprt) and genes that have been shown tohighly express at 2-cell stage (Hmgb3, Taf9 and Ier5)(Zeng et al. 2004). Unlike mtrRNAs, ratios of ISH signalsfor all genes examined were similar to that of Actin(Rz1.1–1.2, nZ29–52), indicating that their mRNAs areevenly distributed between 2-cell sister blastomeres(Supplementary Figure S1A and B, see section onsupplementary data given at the end of this article).This also suggested that relative comparison of ISHsignals could reflect the differences of RNA levelsin-between cells and the mtrRNAs are indeed distributedunevenly between sister blastomeres of 2-cell embryos.Although the differences of mtrRNAs between 2-cellsister blastomeres can be as high as threefold in someembryos, the average differences of their signals betweensister blastomeres are about 10–50% when their ratios ofintensities were subtracted with those of Actin controls atthe same stage (13G18% for 12S mtrRNA and 58G67%for16S mtrRNA at late 2-cell stage respectively). Thesedifferences (less than twofold) and large variationsamong samples could be difficult to reveal using PCRmethod due to the exponential amplification effect(Weaver et al. 2010). Nevertheless, single-cell quan-titative RT-PCR of dissected blastomeres suggested similar

    tation embryos. (A) In situ hybridization of 12S mtrRNA in mousee embryos at different stages. (C) In situ hybridization of Actin in mousee position of the first polar body. Note the difference in signal intensitiesS mtrRNA in pre-implantation embryos. The expression of 16S mtrRNA

    rds (black dashed line, three independent experiments). The expressionEgfp mRNA was added to each sample and used as internal control (see50 embryos at each stage were used for cell lysate preparation and RTtion embryos. Embryos were hybridized with Stellaris oligonucleotideS mtrRNA) simultaneously. 16S mtrRNA signals were pseudo-colored in

    www.reproduction-online.org

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

    http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1

  • Early 2-cell Mid 2-cell Late 2-cell

    16S

    mtr

    RN

    A s

    ense

    Act

    in12

    S m

    trR

    NA

    BA

    16S

    mtr

    RN

    A

    10 µm

    OD1

    OD2

    R = OD1/OD2, OD1 > OD2

    C

    16S mtrRNA

    Actin

    Early (n=29, 30)

    Middle (n=38, 27)

    Late (n=30, 25)

    Ratio of intensity between sister blastomeres

    2-cell stage

    1.4 ± 0.3 1.9 ± 0.7 1.7 ± 0.6

    1.2 ± 0.2 1.3 ± 0.2 1.2 ± 0.1

    0.0

    0.5

    1.0

    1.5

    2.0

    2.5

    3.0

    Early-2C Mid-2C Late-2C

    Rat

    io o

    f int

    ensi

    ty b

    etw

    een

    sist

    er b

    last

    omer

    es (

    high

    /low

    )

    Actin 16S mtrRNA

    P=3.082×10–6

    P=0.006

    P=1.427×10–5

    12S mtrRNA

    Actin

    Early (n=29, 22)

    Middle (n=49, 49)

    Late (n=24, 19)

    Ratio of intensity between sister blastomeres

    2-cell stage

    1.2 ± 0.2 1.3 ± 0.2 1.4 ± 0.2

    1.1 ± 0.1 1.2 ± 0.1 1.2 ± 0.1

    0.0

    0.5

    1.0

    1.5

    2.0

    2.5

    3.0

    Early-2C Mid-2C Late-2C

    Rat

    io o

    f int

    ensi

    ty b

    etw

    een

    sist

    er b

    last

    omer

    es (

    high

    /low

    )

    Actin 12S mtrRNA

    P=0.018P=0.003 P=0.003

    F

    D

    20 µm12S mtrRNA

    12S mtrRNA

    DAPI

    n=20 n=23

    Ratio of intensity between sister blastomeres

    Mid 2-cells

    1.12 ± 0.09 1.22 ± 0.19

    1.06 ± 0.04 1.08 ± 0.05

    16S mtrRNA

    DAPI

    1.0

    1.1

    1.2

    1.3

    1.4

    1.5

    DAPI 12S mtrRNA DAPI 16S mtrRNA

    Rat

    io o

    f int

    ensi

    ty b

    etw

    een

    sist

    er b

    last

    omer

    es (

    high

    /low

    ) P=0.0003

    P=0.048

    20 µm16S mtrRNA

    E

    Figure 2 Distribution of mitochondrial ribosomal RNAs between mouse 2-cell sister blastomeres. (A) In situ hybridization of mouse 2-cell embryos.Mouse 2-cell embryos were separated into early, middle and late stages and hybridized with long cRNA probes for 12S and 16S mtrRNAs respectively.Actin was used ascontrol. Nosignals above backgroundwere detectedwhen embryoswere hybridizedwith16S mtrRNA sense probes. (B and C) Ratiosofin situ hybridization signal intensities for Actin and mtrRNAs between 2-cell sister blastomeres (RZOD1/OD2, OD1OOD2). Embryonic stages, numberof embryos measured and average ratios of signal intensities are shown in tables (upper panels). Bar graphs are displayed in lower panels. Significances ofthe differences between control and experimental groups were assessed using Mann–Whitney test for two independent samples. (D and E) Fluorescentin situ hybridization (FISH) of 12S and 16S mtrRNAs in mouse 2-cell embryos using Stellaris oligonucleotide probes (20-mers). Probes were tagged withQuasar 570 and Quasar 670 dyes for 12S and 16S mtrRNAs respectively. 16S mtrRNA signal was pseudo-colored as green. (F) Quantification of FISHsignals between 2-cell sister blastomeres. Comparing to DAPI staining, both mtrRNAs appeared higher in one blastomere than the other. Number ofembryos measured and the average ratios of signal intensity are shown in table (upper panel). Bar graph is shown in lower panel. Significances of thedifferences between control and experimental groups were assessed using Mann–Whitney test. Error bars: S.D.

    16S mtrRNA regulates cell lineage allocations 357

    high variations and differences of mtrRNAs between sisterblastomeres (Supplementary Figure S1G). We furtherquantified mtrRNA levels more directly using thefluorescent signals following FISH of mtrRNAs. Both

    www.reproduction-online.org

    12S and 16S mtrRNAs could be readily seen in thecytoplasm of blastomeres when 2-cells were hybridizedwith short fluorescent probes (Fig. 2D and E). Quantifi-cation of the fluorescent signals in paired sister

    Reproduction (2016) 151 351–367

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

    http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1

  • A 16S mtrRNA

    Actin 16S sense

    12S mtrRNA

    C

    B

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2

    Rat

    io o

    f int

    ensi

    ty o

    f sis

    ter

    blas

    tom

    eres

    Actin 12S mtrRNAP=6.57×10–5 P=1.07×10–6

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2

    Blastomere-1 Blastomere-2 Blastomere-3 Blastomere-4

    Blastomere-1 Blastomere-2 Blastomere-3 Blastomere-4

    Rat

    io o

    f int

    ensi

    ty o

    f sis

    ter

    blas

    tom

    eres

    Actin 16S mtrRNAP=9.55×10–6 P=3.10×10–6

    Figure 3 Distribution of mtrRNAs among sister blastomeres of mouse4-cell embryos. (A) In situ hybridization of mouse 4-cell embryos. Sisterblastomeres of 4-cell embryos were separated manually by micro-manipulation before being hybridized together with indicated probes.Shown are representative light micrographs. Images of in situhybridization with 16S mtrRNA sense probe from a separateexperiment was included as control. (B) Ratios of in situ hybridizationsignal intensities for Actin and 12S mtrRNA among 4-cell sisterblastomeres. Comparing to the sister blastomere with the highest signalintensity (set as 1), ratios of Actin and 12S mtrRNA signals for the twolowest blastomeres are 0.93G0.02, 0.90G0.03 (nZ16) and 0.82G0.11, 0.73G0.1 (nZ22), respectively. (C) Ratios of in situ hybridizationsignal intensities for Actin and 16S mtrRNA among 4-cell sisterblastomeres. Comparing with the sister blastomere with the highestsignal intensity (set as 1), ratios of Actin and 16S mtrRNA signals for thetwo lowest blastomeres are 0.95G0.03, 0.92G0.04 (nZ15) and0.83G0.07, 0.75G0.07 (nZ16) respectively. Significances of thedifferences between control and experimental groups were assessedusing Mann–Whitney test (see also Supplementary Tables S4 and S5).Error bars: S.D.

    358 Z Zheng and others

    blastomeres showed that 2-cell embryos containeddifferential levels of mtrRNAs in sister blastomeres(about 10–20% at the examined mid-2-cell stage),comparing with the equally distributed nuclear DAPIsignals (nZ20, 23; PZ0.048, 0.0003 for 12S and 16SmtrRNAs respectively, Mann–Whitney test) (Fig. 2F).

    It has been suggested that hormone treatment duringsuperovulation or different mouse strains would influ-ence the quality of embryos obtained, therefore castingenvironmental effects on development (Sanfins et al.2003). To eliminate the possibility that these artificialtreatments will deviate the distribution of mtrRNAsbetween 2-cell sister blastomeres, mid-2-cell embryosisolated from CD1 mice that were naturally mated orfrom C57BL/6 mice that were superovulated washybridized with DIG-labeled long cRNA probes.Comparing with the Actin controls that were processedat the same time, expression of 16S mtrRNA was foundto be more differential between 2-cell sister blastomeresfrom these mice. The average ratios of ISH signalintensity for 16S mtrRNA were significantly morevariable (1.4G0.3 for CD1 mice, nZ23; 1.4G0.4 forC57BL/6, nZ20) comparing with those of Actin(P!0.05, z-test for two samples’ means) (SupplementaryFigure S1C, D, E and F). This suggested that the unevendistribution of 16S mtrRNA between 2-cell sisterblastomeres was not affected by hormone injection ordifferent strains of mice. Taken together, these resultsindicated that mtrRNAs are differentially distributedbetween mouse 2-cell sister blastomeres.

    To examine whether the asymmetric distribution ofmtrRNAs would persist with further cell divisions, levelsof mtrRNAs among blastomeres in 4-cell embryos wereexamined using ISH. To better visualize 4-cell sisterblastomeres, they were first manually separated intosingle cells from each embryo (Fig. 3A). Groups of foursister blastomeres of the same embryo were thenhybridized with the same long cRNA probes together.Signals of mtrRNAs in individual blastomeres wereimaged and measured as described above. Relativesignal ratios were compared among sister blastomereswhen setting the blastomere with the highest intensityas 1. It was found that 4-cell sister blastomeres containeddescending amounts of mtrRNAs, with two of themcontaining w 20% less of mtrRNAs than the other two(Fig. 3B and C). The differences of 12S and 16S mtrRNAsamong sister blastomeres were significantly varied(P!0.05, nZ22 and 16 respectively), comparing withthose of Actin (PZ0.438, nZ15, one-way ANOVA,aZ0.05) (Supplementary Tables S4 and S5). The highvariations and differences of mtrRNA levels among 4-cellsister blastomeres were also seen using single cellquantitative RT-PCR (Supplementary Figure S1H).These results suggested that the asymmetric distributionof mtrRNAs could be propagated with further celldivisions. Recent studies showed that mouse 4-cellblastomeres contain differentially expressed epigenetic

    Reproduction (2016) 151 351–367 www.reproduction-online.org

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

    http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1

  • C

    A

    MitoTracker DAPI Merge20 µm 20 µm 20 µm

    16S mtrRNA regulates cell lineage allocations 359

    modifiers that influence the destination of respectiveprogeny cells in blastocyst (Torres-Padilla et al. 2007,Burton et al. 2013). Although it is not clear at this stagewhether the differentially distributed mtrRNAs in mouse2-cell embryos are related to the differential geneexpression at later stages, the results nevertheless suggestthat molecular asymmetry occurs in mouse 2-cellembryos, earlier than previously found embryonic stage.

    20 µm

    TMRM Hoechst Merge

    Hoechst MergeTMRM (w. FCCP)

    D

    B

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2

    1.4

    Rat

    io o

    f int

    ensi

    ty(h

    igh/

    low

    )

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2

    1.4

    TMRM Hoechst

    MitoTracker DAPI

    Rat

    io o

    f int

    ensi

    ty(h

    igh/

    low

    )

    Figure 4 Mouse 2-cell embryos contain overall equal amount andactivity of mitochondria. (A) MitoTracker Red staining of mouse 2-cellembryos. Nuclei were stained with DAPI. Shown are representativeconfocal images. (B) Ratio of signal intensities of MitoTracker Redbetween 2-cell sister blastomeres. Similar ratios of mitochondria(RZ1.09G0.09) and DAPI (RZ1.09G0.07, nZ30) indicate equalamounts of mitochondria between sister blastomeres. (C) TMRMstaining of live 2-cell embryos. As control, FCCP was used to un-couplemitochondrial membrane potential before TMRM staining (lowerpanels). Nuclei were stained with Hoechst 33342. Shown arerepresentative confocal images. (D) Ratio of TMRM signal intensitiesbetween 2-cell blastomeres. Similar ratios of TMRM (RZ1.12G0.08)and Hoechst (RZ1.08G0.12, nZ40) indicate the equal mitochondrialactivity between sister blastomeres.

    Mouse 2-cell embryos contain cytoplasmic 16S mtrRNAoutside of mitochondria

    Since mtrRNAs are essential components of mito-chondrial ribosomes, whether the differential local-ization of mtrRNAs reflects differences in mitochondriadistribution or mitochondrial activity between 2-cellsister blastomeres was examined next. Whole-mountimmunofluorescent staining of embryos with Mito-Tracker Red showed overall even distribution ofmitochondria in the cytoplasm of 2-cell blastomeres.Quantification of MitoTracker Red signals in paired sisterblastomeres of 2-cell embryos from confocal micro-graphs gave rise to the ratio of 1.1G0.08 between sisterblastomeres, similar to that of nuclear DAPI staining(RZ1.1G0.07, nZ30), indicating that 2-cell sisterblastomeres contain similar amounts of mitochondria(Fig. 4A and B). Interestingly, in contrary to apparenthigher levels of mtrRNAs in inner cells of morula andblastocyst (Fig. 1A and B), fluorescent signals ofMitoTracker Red in the inner cells appeared weakerthan those of outer cells (Supplementary Figure S2A, seesection on supplementary data given at the end of thisarticle), suggesting a probable higher mitochondriacontents in cells at the outer layer of morula andblastocysts. Next, the membrane potential of mito-chondria was examined using TMRM staining of live2-cell embryos. Comparing to embryos treated withFCCP (the membrane potential un-coupler), liveembryos were readily stained by the dye (Fig. 4C). Littledifference in the intensity of TMRM staining was foundbetween 2-cell sister blastomeres (RZ1.1G0.08,nZ40), similar to that of nuclear DNA staining(RZ1.08G0.12, Fig. 4D), suggesting that 2-cellblastomeres also have similar mitochondrial activity.Thus, neither the quantity nor the activity of mito-chondria is differentially distributed between sisterblastomeres of 2-cell embryos.

    One possibility for un-evenly distributed mtrRNAsseen in 2-cell embryos is the incomplete hybridizationof long anti-sense cRNA probes due to their difficultyto penetrate mitochondrial membranes, as previouslysuggested (Ninomiya & Ichinose 2007). To test this, theexpression of two mitochondria encoded genes (Nd2and Cytb) and a nuclear gene (Cyc1) was analyzed usingrespective long cRNA probes. Quantitative RT-PCR firstshowed that both Nd2 and Cytb were expressed at levelshigher than that of 16S mtrRNA at most pre-implantation

    www.reproduction-online.org Reproduction (2016) 151 351–367

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

    http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1

  • 360 Z Zheng and others

    embryonic stages, whereas Cyc1 was expressed at muchlower level (Fig. 5A). However, both mRNAs of Nd2 andCytb that are known to be inside of mitochondria werenot detected by ISH, whereas Cyc1 mRNA was readilyseen in morula and blastocysts using the same method(Fig. 5B). This suggested that long cRNA probes was able

    A

    C

    D

    Mitochondria

    16S mtrRNA

    500 nm

    B

    16S mtrRNA MitoTracker Merge

    MII

    oocy

    te

    20 µm

    12S m

    Nd2

    Mor

    ula

    20 µm

    0.0

    0.5

    1.0

    1.5

    2.0

    2.5

    3.0

    3.5

    4.0

    Oocyte 1-cell 2-cell 4-cell 8-cell Morula Blastocyst

    Gen

    e ex

    pres

    sion

    vs

    16S

    mtr

    RN

    A 16S mtrRNA Cytb Nd2 Cyc1

    Figure 5 Distribution of cytoplasmic 16S mtrRNA outside of mitochondria inShown are average expression levels relative to that of 16S mtrRNA from thremouse embryos. Collected embryos (from 1-cell to blastocyst) were hybridizencoded Cytb and Nd2 were not detectable in cytoplasm despite their higheat morula and blastocyst stages. Anti-sense and sense probes for mtrRNAs wSignals of 16S mtrRNA (detected by fluorescent oligonucleotide probes) weMitoTracker Red) from MII oocytes to blastocysts. Inner cells of morula andsignals. (D) In situ hybridization of 16S mtrRNA at electron microscopic levoutside of mitochondria (arrows), as revealed by anti-DIG-colloidal gold se(right panel). Average numbers of gold particles per frame were calculatedSignificance of the difference was assessed using Mann–Whitney test. Error

    Reproduction (2016) 151 351–367

    to detect relatively small amount of mRNAs in thecytoplasm of cells during ISH, but may indeed bedifficult to localize RNA species within mitochondria.It also suggested that the differentially distributedmtrRNAs detected by long cRNA probes were probablylocalized outside of the organelle. Since small

    500 nm

    Mitochondria

    16S mtrRNA sense

    16.7

    5.30

    5

    10

    15

    20

    25

    16S mtrRNA Sense

    No.

    Gol

    d/fr

    ame

    P=0.002

    16S mtrRNA MitoTracker Merge2-

    cell

    20 µm

    50 µm

    trRNA 16S mtrRNA

    16S mtrRNA sense

    Cytb

    Cyc1

    Bla

    stoc

    yst

    20 µm

    mouse 2-cell embryos. (A) Quantitative RT-PCR of mitochondrial genes.e independent experiments. (B) In situ hybridization of pre-implantationed with long cRNA probes for indicated genes. Signals for mitochondriar levels of expression (A), whereas nuclear encoded Cyc1 could be seenere used as controls. (C) Co-staining of 16S mtrRNA and mitochondria.re found to partially co-localize with mitochondria (detected byblastocysts contain higher levels of 16S mtrRNA than MitoTracker Redel (ISH-EM). 16S mtrRNA (arrow heads) was detected in the cytoplasmcondary antibodies. Sense probe was used as negative controlfrom ten frames of electron micrographs in each group (Inset, nZ10).bars: S.D.

    www.reproduction-online.org

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

  • 16S mtrRNA regulates cell lineage allocations 361

    oligonucleotide probes could better penetrate mito-chondria, FISH signals of mtrRNAs may represent bothcytoplasmic and mitochondrial fractions of mtrRNAs.Using fluorescently tagged oligonucleotide FISH probes,together with MitoTracker Red, the subcellular local-ization of 16S mtrRNA in mouse embryos was furtherexamined. In control experiments, fluorescent signals of16S mtrRNA were found to completely overlap with thatof MitoTracker Red in mouse embryonic fibroblast (MEF)and 293T cells (Supplementary Figure S2B). However, inmouse MII oocytes and pre-implantation embryos,signals of 16S mtrRNA were found to only partiallyoverlap with MitoTracker Red staining, suggesting that16S mtrRNA was also present in the cytoplasm outside ofmitochondria in mouse female germ cells and embryos(Fig. 5C). Green fluorescent signals of 16S mtrRNA thatdid not overlap with MitoTracker Red were found in thecytoplasm of MII oocytes toward animal pole and moreconcentrated in inner cells in morula and blastocysts,suggesting that 16S mtrRNA at these regions was likelyoutside of mitochondria, consistent with ISH resultsusing long cRNA probes (Fig. 1).

    Mitochondrial rRNAs have been found outside ofmitochondria in early embryos of Drosophila, Sea urchinand Xenopus (Kobayashi et al. 1993, 1998, Ogawa et al.1999). In mouse, it has been suggested thatMII oocytesalsocontain mtrRNAs in the cytoplasm (Ninomiya & Ichinose2007). To further determine the sub-cellular localizationsof 16S mtrRNA in relation to mitochondria, 2-cell embryoswere examined at electron microscopic level followingISH. In control experiment, transmission electronmicroscopy of thin sections (100 nm) of 2-cell embryosshowed that mitochondria appeared to be spherical inshape with clear cristae (Supplementary Figure S2C).Following ISH, mitochondria were less well maintainedand the internal cristae of mitochondria were sometimeslost due to repeated sample treatments. However, theoverall morphology of mitochondria remained identifiable(Supplementary Figure S2D). Using anti-DIG antibodyconjugated with 12-nm colloidal gold particles, it wasfound that 16S mtrRNA was distributed in the cytoplasmof 2-cell blastomeres outside of mitochondria (Fig. 5D).The average number of gold particles on thin sections of2-cell embryos was significantly higher when anti-senseprobes of 16S mtrRNA were used (16.7G7.8), comparingwith that of sense control (5.3G2.7, PZ0.002, nZ10,Mann–Whitney test) (Fig. 5D, inset). Collectively, theseresults indicate that mouse 2-cell blastomeres contain 16SmtrRNA in the cytoplasm outside of mitochondria.

    16S mtrRNA in 2-cell embryos regulates cell allocationsin blastocyst

    First found in Drosophila, 16S mtrRNA was shown todrive the germ cell formation during early embryogen-esis (Iida & Kobayashi 1998). To find out whether thedifferentially expressed 16S mtrRNA in mouse 2-cell

    www.reproduction-online.org

    embryos could have functional roles in cell-lineageallocations during pre-implantation embryogenesis,levels of 16S mtrRNA were altered in one sisterblastomere of 2-cell embryos using microinjection ofeither sense or antisense RNAs. In vitro transcribed EgfpmRNA was used as control or co-injected into the sameblastomere in order to trace the fate of its descendentinto blastocysts (Fig. 6A). Injected embryos weredeveloped till blastocyst stage in vitro and scanned at2-mm interval using confocal microscopy. Confocalsections of each individual blastocyst were compiledinto single 3-D images, from which the number andlocations of cells were analyzed (Supplementary Movies1 and 2, see section on supplementary data given at theend of this article). In the three dimensional space, cellsthat are located on the outer-most layer on either axis ofthe three dimensions (X, Yand Z) were considered as theTE cells, whereas cells that are surrounded by others onall three axes were counted as the ICM cells (Fig. 6B).Cell positions were determined by the nuclear DAPIstaining and green fluorescence was used to identifythe EGFP-labeled (EGFPC) cells. For the ease of analysesof 3D cell allocations, embryos containing 40–45 cells(46–48 h in vitro culture following microinjection),corresponding to early blastocysts were used in mostcases. At this stage, ICM and TE cells are segregated,whereas epiblast and primitive endoderm cells of theICM are yet to be differentiated. To confirm the cellpositioning method, embryos were co-stained with cell–cell junctional E-Cadherin or plasma membrane markerTRITC-Phalloidin in some cases so that cell boundarieswere clearly defined. It was found that cells in both ICMand TE could be similarly positioned with or without cellmembrane labeling (Supplementary Figure S3). Thus formost of the cell allocation analyses, embryos withoutcell membrane labeling were used. Total numbers ofEGFPC cells that were allocated into either ICM or TEwere then counted in order to assess the effects ofchanging 16S mtrRNA levels on cell lineage allocations.

    Quantitative RT-PCR showed that anti-sense mtrRNAdecreased 16S mtrRNA level in injected blastomeres,while sense RNA increased it by about 60% (Supple-mentary Figure S4A, see section on supplementary datagiven at the end of this article). The decrease of 16SmtrRNA following anti-sense microinjection was alsosupported by the decreased fluorescent signals ininjected blastomeres when compared to the non-injected ones using FISH (Supplementary Figure S4B).Examination of Egfp mRNA-injected embryos firstshowed that EGFPC cells derived from microinjectedblastomere occupied blastocyst in both ICM and TEwithout affecting the overall development of embryosin vitro. All groups of blastocysts contained similarnumbers of cells when cultured for the same length oftime in vitro (Supplementary Table S6). There wereabout 34% of EGFPC cells in ICM and 66% of EGFPC

    cells in TE in Egfp-injected embryos (Fig. 6F). Comparing

    Reproduction (2016) 151 351–367

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

    http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1

  • 362 Z Zheng and others

    with the Egfp-injected control, increasing 16S mtrRNA(sense injection) increased the number of EGFPC

    descendent in ICM (to about 49%) (PZ0.005, nZ18),while reduced 16S mtrRNA level (anti-sense injection)caused an increase in the number of EGFPC cells in TE(to about 78%) (PZ0.0001, nZ19, Mann–Whitney test)(Fig. 6F and Supplementary Table S6). These changes ofcell allocations of EGFPC cells following microinjectionappeared statistically significant when they werecompared together across different conditions (P!0.001,one-way ANOVA, aZ0.05) (Supplementary Table S7).

    The cell allocation effects of altered 16S mtrRNAlevels were further examined by cell lineage specificgene expression. Using antibodies against cell-lineagemarkers NANOG (pluripotency marker) and CDX2(TE marker), in vitro cultured blastocysts were immu-nostained following microinjection. It was found thatpatterns of marker gene expression in either ICM or TEcell lineages were not changed with respective allo-cations in blastocyst following either sense or anti-sense

    B

    D

    34%49%

    22%

    66%51%

    78%

    0.00.10.20.30.40.50.60.70.80.91.0

    Egfp 16S mtrRNA+Egfp

    16S mtrRNAanti-sense+

    Egfp

    Allo

    catio

    ns o

    f GF

    P+

    cells

    in b

    last

    ocys

    t

    ICM

    TE

    P=0.001P=0.0005

    P=0.001P=0.0006

    A

    Y

    X

    Z

    Y

    X

    Z

    ICMTE

    In vitro culture

    Imaging analysisMicroinjection

    46–48 h

    C

    GF

    0

    10

    20

    30

    40

    50

    60

    Tota

    l

    Tota

    l

    Tota

    l

    Cdx

    2+

    Cdx

    2+

    Cdx

    2+

    Cdx

    2+E

    GF

    P+

    Cdx

    2+E

    GF

    P+

    Non-injected

    16Ssense

    16S anti-sense

    Non-injected

    16Ssense

    16S anti-sense

    16Ssense

    16S anti-sense

    Ave

    rage

    num

    ber

    of c

    ells P=0.744

    P=0.007

    P=0.0002

    Non-injected

    NANOG

    Non-injected

    CDX2

    X

    YZ

    X

    Y

    10 µm

    10 µm 10 µm

    X

    Y

    +

    +

    + +

    +

    +Z

    X

    Y

    Z ICM cell Z TE cell

    16S mtrRNA+Egfp

    NANOG

    16S mtrRNA+Egfp

    CDX2

    16S anti-sense+Egfp

    NANOG

    16S anti-sense+Egfp

    CDX2

    Merge

    Merge

    Merge

    Merge

    Merge

    20 µmMerge

    E

    10 µm

    10 µm

    10 µm

    Reproduction (2016) 151 351–367

    injection, comparing to the non-injected control group(Fig. 6C, D and E). NANOG was primarily found in ICMcells with occasional staining in TE cells, whereas CDX2was exclusively localized in TE cells at the outer layer inall groups. Since NANOG is a pluripotency marker butnot a strict ICM specific marker at the early blastocyststage and CDX2 is an exclusive TE lineage marker,changes of CDX2 expressing (CDX2C) cells in micro-injected embryos were further analyzed. It was foundthat, while overall development of the embryosremained unaltered (embryos contained similar numberof cells during similar period of culturing time in allgroups), EGFPC cells contained higher fraction ofCDX2C cells (16.7G5.0, 69.5G13.3% of total EGFPC

    cells) when 16S anti-sense was injected, comparing withsense injected embryos (9.4G4.2, 42.6G16% of totalEGFPC cells, PZ0.0002, nZ18–19, Mann–Whitneytest) (Fig. 6G, Supplementary Tables S8 and S9). Theseresults suggested that changes of 16S mtrRNA did notchange the marker gene expression prior to cell lineageallocations and levels of 16S mtrRNA in 2-cellblastomeres could deviate the segregation of ICM andTE cell lineages in blastocyst. Neither anti-sense norsense 16S mtrRNA brought changes to mitochondrial

    Figure 6 Influences of 16S mtrRNA in 2-cell blastomeres on cellallocations in mouse blastocyst. (A) Schematic drawing of 2-cellembryo microinjection. In vitro transcribed Egfp mRNA was injectedinto one blastomere at random, either singly or together with 16SmtrRNA. Embryos were cultured in vitro for 46–48 h till blastocyst stageand EGFPC descendent cells were traced for their locations in eitherinner cell mass (ICM) or trophectoderm (TE) within blastocyst.(B) Representative confocal images showing locations of EGFPC ICMand TE cells. Blastocysts cultured in vitro following microinjectionwere scanned under a confocal microscope at 2-mm interval. EGFPC

    descendent cells were traced for their locations as marked by DAPIstaining using 3-D compository images. Cells that are surrounded byother cells along three axes (X, Y and Z) are denoted as ICM cells (leftpanel) and cells that are located on the outside along either one of thethree axes are denoted as TE cells (right panel). Shown are centersections of a scanned blastocyst with indicated ICM and TE cells onthree dimensions. (C, D and E) Immunostaining of NANOG and CDX2in mouse blastocysts with or without microinjection. Embryos werecultured in vitro from 2-cell stage, when one blastomere wasmicroinjected with indicated RNA species. Similar to the control (C),NANOG was found to primarily express in ICM cells, with occasionalstaining in TE cells (D), whereas CDX2 was found to express exclusivelyin TE cells in all groups (E). Cell nuclei were stained with DAPI.Representative confocal images of immunostaining of NANOG orCDX2 are shown on the left and merged images with either DAPI orEGFP signals are shown on the right in each panel. (F) 16S mtrRNAinfluences cell allocations in blastocyst. Comparing to Egfp mRNAinjected controls, 16S mtrRNA sense or anti-sense increased EGFPC

    cells’ occupation in either ICM or TE, respectively (nZ18–19; see alsoSupplementary Tables S6 and S7). (G) While total number of cells werenot changed in blastocysts with or without microinjection, 16S mtrRNAanti-sense increased CDXC cells in EGFPC fraction (nZ18–20; see alsoSupplementary Tables S8). Pair-wised comparison was assessed forsignificance using Mann–Whitney test, while significance ofdifferences among three groups were assessed using one-way ANOVAand post-hoc Turkey analyses (see also Supplementary Table S9).

    www.reproduction-online.org

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

    http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1

  • EGFP

    DAPI

    Merge 50 µm 50 µm

    n=101 n=144 n=127

    Pattern-1(Embryonic)

    Pattern-2(Mixed)

    Pattern-3(Ab-embryonic)

    Em

    A

    B

    C

    Ab

    15.3%

    68.1%

    16.7%

    16S mtrRNA anti-sense+Egfp

    31.7%

    42.6%

    25.7%

    Egfp

    46.5%40.9%

    12.6%

    16S mtrRNA+EgfpPattern 1Pattern 2Pattern 3

    Figure 7 Influences of 16S mtrRNA in 2-cell blastomeres on celllineages of blastocyst. (A) Schematic drawing of categorization ofblastocysts containing differentially distributed EGFPC progeny cells.Pattern 1: EGFPC descendent found only at embryonic side (left panel);pattern 2: EGFPC descendent found at both embryonic andabembryonic sides (mixed, middle panel); pattern 3: EGFPC descen-dent found only at abembryonic side (right panel). (B) Representativeconfocal micrographs of blastocysts developed from microinjected2-cell embryos, corresponding to three patterns depicted in (A).Cell nuclei were stained with DAPI. Dotted lines show the embryonic-abembryonic boundary. (C) Changes of embryonic patterning inblastocyst following 2-cell blastomere microinjection with 16SmtrRNA or anti-sense RNAs. One sister blastomere of 2-cell embryowas microinjected with indicated RNAs. Comparing to theEgfp-injected control, 16S mtrRNA anti-sense decreased descendentallocation in embryonic cell lineage (pattern 1, green) and increasedtheir mixed cell allocations (pattern 2, light green), while 16S mtrRNAincreased EGFPC cells in embryonic cell lineage and decreased theirabembryonic allocation (pattern 3, orange). Numbers of embryos

    16S mtrRNA regulates cell lineage allocations 363

    activity between 2-cell blastomeres (SupplementaryFigure S4C and D), suggesting that the extra-mito-chondrial fraction of the mtrRNA may be accountablefor the cell lineage allocation effects.

    Since bi-laterally symmetric mouse blastocysts can bedivided into embryonic half (containing ICM andpolar TE) and abembryonic half (containing mural TE)along the A–V axis, cell allocation effects of 16S mtrRNAalong the A–V axis following microinjection was furtheranalyzed. Based on the distribution of EGFPC descen-dent in either embryonic or abembryonic allocations inblastocysts, embryos were separated into three differentcategories (Fig. 7A and B). Among 101 Egfp-injectedembryos, 31.7% of embryos contained EGFPC progeniesonly at embryonic side (pattern 1, embryonic) and25.7% of embryos contained EGFPC cells only atabembryonic side (pattern 3, abembryonic). The remain-ing 42.6% of embryos contained EGFPC cells in bothcell allocations (pattern 2, mixed). However, whensingle 2-cell sister blastomeres were co-injected withanti-sense 16S mtrRNA and Egfp mRNA, blastocystscontaining EGFPC progenies in embryonic half reducedfrom 31.7% to 15.3%, whereas blastocysts containingEGFPC cells in the mixed cell allocations increased to68.1% (nZ144). Including the remaining 16.7%embryos with EGFPC cells located only at abembryonichalf, a total of 84.8% embryos contained progenies frominjected blastomeres at abembryonic allocations(comparing to the total of 68.3% in the control group).The dramatic increase of embryos with mixed cellallocations is probably caused by the randomness atchoosing the 2-cell sister blastomeres for microinjection.In contrast, when in vitro transcribed 16S mtrRNAwas co-injected with Egfp mRNA into single 2-cellblastomeres, blastocysts containing EGFPC descendentallocated to embryonic cell-lineage increased to 46.5%,whereas blastocysts with EGFPC abembryonic cellsdecreased to 12.6% (nZ127) (Fig. 7C). Statisticalanalysis suggested that these changes of embryos indifferent patterns were significantly relevant to themicroinjection of either sense or anti-sense 16S mtrRNAsamong experimental groups (c2Z39.18, P!0.001, c2

    test, Supplementary Table S10). Taken together, theseresults suggested that the increase of 16S mtrRNA in2-cell blastomeres facilitates their progeny cells toallocate into pluripotent embryonic cell lineage,whereas reduction of 16S mtrRNA deviates the descen-dent into abembryonic allocation.

    analyzed are indicated below pie charts.

    Discussion

    It has been reported that the total number of mito-chondria and the mitochondrial DNA remain largelyunchanged in pre-implantation mouse embryos (Piko &Taylor 1987). Since embryonic mitochondria are mostlyinherited from the maternal source (Cree et al. 2008,Mishra & Chan 2014), the number of mitochondria in

    www.reproduction-online.org

    embryonic cells gradually decreases with cell divisionuntil post-implantation stage when cell proliferationaccelerates. However, mtrRNAs are actively transcribedduring pre-implantation development (Piko & Chase1973, Piko & Taylor 1987). Consistent with this,quantitative RT-PCR showed that the level of 16S

    Reproduction (2016) 151 351–367

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

    http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1

  • 364 Z Zheng and others

    mtrRNA is maintained constant from MII oocytes to2-cell embryos and increased dramatically after 4-cellstage. This suggests that mitochondrial genome isprobably expressed continuously in the absence ofDNA replication, separating its gene expressionregulation away from that of nuclear genome (Leeet al. 2014).

    In unfertilized eggs, mtrRNAs appeared to localizemore towards the animal pole where the first polar bodyemits. Uneven distribution of mtrRNAs was alsoobserved throughout early embryos at different stagesfrom 2-cell and 4-cell (higher in half of the blastomeres)to morula and blastocyst (higher in inner cells). It is notclear at this stage whether the maternal polarity is relatedto the molecular asymmetry observed in early embryos.Several studies have shown that LEPTIN and STAT3 areasymmetrically localized in unfertilized eggs and earlyembryos in mouse, accumulating at the animal poletoward the first polar body (Antczak & Van Blerkom1997, Schulz & Roberts 2011). This is similar to that ofmtrRNAs in unfertilized eggs (Fig. 1A and B). A largemulti-protein complex (subcortical maternal complex(SCMC)) was also found to localize in the subcorticalregions of mouse eggs and early embryos. Geneticdepletion of the SCMC components in mouse causeddevelopmental retardation at 2-cell stage (Li et al. 2008).During compaction, these polarized factors accumulatedifferentially in outer cells vs inner cells, generatingpolarity gradient within the embryo that may be linkedto the cell-lineage determination and axis formationin blastocyst. However, none of them was found todifferentiate in-between sister blastomeres followingfertilization when embryogenesis starts. Recent researchsuggested that differentially localized epigenetic factorsappearing at very early stage (4-cell stage) might regulatecell lineage determination in blastocysts (Torres-Padillaet al. 2007, Plachta et al. 2011, Burton et al. 2013).However, how these early molecular asymmetries aregenerated and whether there are even earlier cell fatedeterminants in mouse pre-implantation embryosremain elusive. Results presented here indicate thatmouse 2-cell sister blastomeres contain quantitativelydifferent mtrRNAs, suggesting that the molecular asym-metry may occur at the very beginning of mouseembryogenesis. Whether the differential mtrRNAs in2-cell blastomeres are functionally related to themolecular asymmetries that occur during the develop-ment of early embryos requires further exploration.

    Although the origin of differentially distributedmtrRNAs remains to be determined, their differencesbetween sister blastomeres appear to be more evident atlate 2-cell stage. One possibility is that the partitionedmaternal mtrRNAs are differentially retained or degradedwith the development of embryos. The transcriptionactivity of mitochondrial genome could also differbetween sister blastomeres. Alternatively, since thedifferential distribution of mtrRNAs were detected

    Reproduction (2016) 151 351–367

    mainly outside of the organelle, mitochondria in sisterblastomeres could export differential amount ofmtrRNAs into the cytoplasm. It will be interesting tofind out whether the post-transcriptional modificationsof RNAs and mitochondrial activities contribute to thedifferential distribution of mitochondrial small and largeribosomal RNAs between 2-cell sister blastomeres.Although it is known that embryonic mitochondria aremainly inherited from the maternal source, it remainspossible that sperm entry or the paternal cellularcomponents could contribute to the regulation ofembryogenesis beyond fertilization and activation ofzygote. Previous sperm labeling and tracing experimentin mouse showed that sperm tail enters the egg duringfertilization and remains in one of the 2-cell blastomeres(Jefferson & Williams 2012). The experiment showed thatsperm tail remained largely intact at 2-cell stage. Giventhat sperm mid-piece contains mitochondrial sheathwith rather compacted configuration, this is differentfrom what’s seen with unevenly distributed mtrRNAs,which have diffused localization patterns. Research inDrosophila showed that sperm mitochondria aredestroyed through autophagic and endocytic effectsfollowing fertilization (Politi et al. 2014). Whetherpaternal mitochondria are also eliminated in the samemanner in mouse remains to be determined. Previousstudies suggested that the mode of the first cell divisionsis relevant to the progenies’ cell lineage allocations inblastocysts (Piotrowska et al. 2001, Piotrowska-Nitsche& Zernicka-Goetz 2005). Sequential meridional–equitorial divisions of 2-cell blastomeres showed morepredictable fate of the descendent in the blastocyst (theblastomere that divide meridionally first tend to allocateinto the embryonic hemisphere) than 2-cell blastomeresthat are divided in the same directions (meridional–meridional or equatorial–equatorial divisions). Perhapsthe non-uniform dividing orientations could help togenerate molecular gradients among progenies whichare necessary preludes for cells’ further differentiation.In this regard, it will be interesting to find out whetherthe levels of mtrRNAs are relevant to the regulation ofthe first cell divisions and cell lineage allocations inearly embryos.

    How are mitochondria encoded mtrRNAs localizedin the cytoplasm outside of the organelle? AlthoughmtrRNAs outside of mitochondria have been found invarious species, including Drosophila, Sea urchin,Xenopus and mouse (Kobayashi et al. 1993, 1998,Ogawa et al. 1999, Ninomiya & Ichinose 2007), it is notclear what molecular mechanisms that govern theircytoplasmic distribution. It was suggested that variousRNA species encoded by mitochondrial genome couldbe exported into cytoplasm of human cells via unknownmechanisms (Attardi & Attardi 1968, Maniataki &Mourelatos 2005). One intriguing phenomenon is thatneither the number nor the membrane potential ofmitochondria has apparent differences between 2-cell

    www.reproduction-online.org

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

  • 16S mtrRNA regulates cell lineage allocations 365

    sister blastomeres. It is known that mouse pre-implan-tation embryos utilize pyruvate and lactate instead ofglucose to meet their energy and metabolic requirementsand maintain relatively dormant mitochondrial oxidativephosphorylation activity until blastocyst stage (Krisher &Prather 2012). Interestingly, inner cells of morula andblastocyst appear to contain higher levels of mtrRNAswhen long cRNA probes were used for hybridization,whereas outer cells showed higher contents of mito-chondria as indicated by TMRM staining (Fig. 1A and B,Supplementary Figure S2A). The possible higher levelsof cytoplasmic mtrRNAs in inner cells of late pre-implantation embryos are in parallel with the celllineage allocation effects of 16S mtrRNA. It is possiblethat the expression and distribution of mtrRNAs is relatedto unknown mitochondrial activities in early embryos.MtrRNAs are important components of mitochondrialribosomes for mitochondrial protein synthesis. Despitethe dormant oxidative phosphorylation activity ofmitochondria, mitochondrial protein synthesis remainsactive in early embryos (Piko & Chase 1973, Cascio &Wassarman 1981). One possible functional conse-quence of the cytoplasmic localization of mtrRNAs canbe the participation of mitochondrial ribosomes, whichare more close to prokaryotic ribosomes than to itseukaryotic cytoplasmic counterparts (Amunts et al.2014), in the synthesis of specific subsets of mRNAsthat may play crucial roles during pre-implantationembryogenesis (Amikura et al. 2001). Intriguingly, lateststudies showed that mitochondrial genome containsshort ORFs (sORFs) that reside within mtrRNA’s geneticloci, which encode short polypeptides that are translatedfollowing the export of mtrRNAs (Hashimoto et al. 2001,Lee et al. 2015). These polypeptides (the 24-aa longHumanin from 16S mtrRNA and the 16-aa long MOTS-cfrom 12S mtrRNA) are biologically functional and elicitcritical roles in regulating insulin sensitivity andmaintaining metabolic homeostasis in neuronal andskeletal muscle cells (Hashimoto et al. 2001, Muzumdaret al. 2009, Lee et al. 2015). It will be interesting to fullyelucidate whether mtrRNAs regulate cell lineage allo-cations during early embryogenesis via RNA-mediatedor alternative mechanisms and how mitochondrialgenome participates in the process.

    The mechanisms of cell fate determination regulatedby early embryonic events during embryogenesis havebeen under intensive investigation (Rossant & Tam2009). It is known that blastomeres in early embryospossess developmental plasticity during experimentalmanipulations. Although molecular alterations withinisolated embryonic cells have not been examined, theadaptive nature of embryos suggests that mammalianpre-implantation embryogenesis is a subject of morecomplicated regulation than previously anticipated(Zernicka-Goetz et al. 2009). During microinjectionexperiments, altered 16S mtrRNA levels influenced celllineage allocations of descendent in the blastocyst.

    www.reproduction-online.org

    Although it is not clear whether effects brought byalterations of mtrRNAs will elicit in a stoichiometric ordose-dependent manner, statistical analyses showed thataltered 16S mtrRNA levels brought significant changesto cell lineage allocations of descendent. Specific celllineage markers (such as NANOG and CDX2), however,maintained constant within respective positions ofembryonic cells, even when progeny cells of injectedblastomeres were deviated to either ICM or TE. Thissuggests that 16S mtrRNA may participate in theregulation of cell allocations prior to cell lineage specificgene expression (Nance 2014, Rayon et al. 2014).Recent genome-wide gene expression analyses onearly embryonic cells in both mouse and C. elegansindicated a bi-facet model of cell lineage determination,in which an earlier stochastic heterogeneous gene-expression stage when cells appear non-distinguishableis followed by a late reinforced homogeneous gene-expression stage when cells gain respective molecularsignatures (Ohnishi et al. 2014, Du et al. 2015). Thesesuggested that perhaps earlier events preluding moredefinitive cell-lineage determination pathways areregulated by post-transcriptional mechanisms, involvingvarious RNA species and RNA binding proteins. Thecorrelation between mtrRNAs and cell lineage allo-cations in the blastocyst indicates that differentiallyexpressed molecules, such as mtrRNAs, are functionallyinvolved in the regulation of cell fate determinationat the very beginning of animal development. Futureresearch is required to elucidate the molecularmechanisms that govern the partitioning of mtrRNAsand how they regulate the cell lineage allocation duringmouse pre-implantation embryogenesis.

    Supplementary data

    This is linked to the online version of the paper at http://dx.doi.org/10.1530/REP-15-0301.

    Declaration of interest

    The authors declare that there is no conflict of interest thatcould be perceived as prejudicing the impartiality of theresearch reported.

    Funding

    This work was supported by the Key Technologies R&DProgram, Ministry of Science and Technology, China(2010CB945402, 2007CB947504), the National NatureScience Foundation of China (30871403, 81561138001), andpartly supported by the Chinese Academy of Sciences.

    Acknowledgements

    We would like to thank Xingguo Liu and Xiaodong Shu forsharing reagents; Hongwen Pang for the help with using ZEN

    Reproduction (2016) 151 351–367

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

    http://www.reproduction-online.org/cgi/content/full/REP-15-0301/DC1http://dx.doi.org/10.1530/REP-15-0301http://dx.doi.org/10.1530/REP-15-0301

  • 366 Z Zheng and others

    2010 software. We would like to thank David Albertini for thekind suggestion on the experiments of hormonal effects onmice. We would also like to thank the anonymous reviewers fortheir valuable comments.

    References

    Amikura R, Kashikawa M, Nakamura A & Kobayashi S 2001 Presenceof mitochondria-type ribosomes outside mitochondria in germ plasmof Drosophila embryos. PNAS 98 9133–9138. (doi:10.1073/pnas.171286998)

    Amunts A, Brown A, Bai XC, Llacer JL, Hussain T, Emsley P, Long F,Murshudov G, Scheres SH & Ramakrishnan V 2014 Structure of the yeastmitochondrial large ribosomal subunit. Science 343 1485–1489.(doi:10.1126/science.1249410)

    Antczak M & Van Blerkom J 1997 Oocyte influences on early development:the regulatory proteins leptin and STAT3 are polarized in mouse andhuman oocytes and differentially distributed within the cells of thepreimplantation stage embryo. Molecular Human Reproduction 31067–1086. (doi:10.1093/molehr/3.12.1067)

    Attardi G & Attardi B 1968 Mitochondrial origin of membrane-associatedheterogeneous RNA in HeLa cells. PNAS 61 261–268. (doi:10.1073/pnas.61.1.261)

    Burton A, Muller J, Tu S, Padilla-Longoria P, Guccione E & Torres-Padilla ME 2013 Single-cell profiling of epigenetic modifiers identifiesPRDM14 as an inducer of cell fate in the mammalian embryo. CellReports 5 687–701. (doi:10.1016/j.celrep.2013.09.044)

    Cascio SM & Wassarman PM 1981 Program of early development in themammal: synthesis of mitochondrial proteins during oogenesis and earlyembryogenesis in the mouse. Developmental Biology 83 166–172.(doi:10.1016/S0012-1606(81)80019-X)

    Cree LM, Samuels DC, de Sousa Lopes SC, Rajasimha HK, Wonnapinij P,Mann JR, Dahl HH & Chinnery PF 2008 A reduction of mitochondrialDNA molecules during embryogenesis explains the rapid segregation ofgenotypes. Nature Genetics 40 249–254. (doi:10.1038/ng.2007.63)

    Dard N, Louvet-Vallee S, Santa-Maria A & Maro B 2004 Phosphorylation ofezrin on threonine T567 plays a crucial role during compaction in themouse early embryo. Developmental Biology 271 87–97. (doi:10.1016/j.ydbio.2004.03.024)

    Du Z, Santella A, He F, Shah PK, Kamikawa Y & Bao Z 2015 The regulatorylandscape of lineage differentiation in a metazoan embryo. Develop-mental Cell 34 592–607. (doi:10.1016/j.devcel.2015.07.014)

    Ducibella T & Anderson E 1975 Cell shape and membrane changes in theeight-cell mouse embryo: pre-requisities for morphogenesis of theblastocyst. Developmental Biology 47 45–58. (doi:10.1016/0012-1606(75)90262-6)

    Fujimori T, Kurotaki Y, Miyazaki J & Nabeshima Y 2003 Analysis of celllineage in two- and four-cell mouse embryos. Development 1305113–5122. (doi:10.1242/dev.00725)

    Gardner RL 1997 The early blastocyst is bilaterally symmetrical and its axisof symmetry is aligned with the animal–vegetal axis of the zygote in themouse. Development 124 289–301.

    Gardner RL 2001 Specification of embryonic axes begins before cleavage innormal mouse development. Development 128 839–847.

    Guo G, Huss M, Tong GQ, Wang C, Li Sun L, Clarke ND & Robson P 2010Resolution of cell fate decisions revealed by single-cell gene expressionanalysis from zygote to blastocyst. Developmental Cell 18 675–685.(doi:10.1016/j.devcel.2010.02.012)

    Hashimoto Y, Niikura T, Tajima H, Yasukawa T, Sudo H, Ito Y, Kita Y,Kawasumi M, Kouyama K, Doyu M et al. 2001 A rescue factor abolishingneuronal cell death by a wide spectrum of familial Alzheimer’s diseasegenes and Ab. PNAS 98 6336–6341. (doi:10.1073/pnas.101133498)

    Iida T & Kobayashi S 1998 Essential role of mitochondrially encoded largerRNA for germ-line formation in Drosophila embryos. PNAS 9511274–11278. (doi:10.1073/pnas.95.19.11274)

    Jedrusik A, Parfitt DE, Guo G, Skamagki M, Grabarek JB, Johnson MH,Robson P & Zernicka-Goetz M 2008 Role of Cdx2 and cell polarity incell allocation and specification of trophectoderm and inner cell mass inthe mouse embryo. Genes and Development 22 2692–2706. (doi:10.1101/gad.486108)

    Reproduction (2016) 151 351–367

    Jefferson WN & Williams CJ 2012 Early mouse embryo asymmetry.Molecular Reproduction and Development 79 433. (doi:10.1002/mrd.22050)

    Johnson MH 2009 From mouse egg to mouse embryo: polarities, axes, andtissues. Annual Review of Cell and Developmental Biology 25 483–512.(doi:10.1146/annurev.cellbio.042308.113348)

    Johnson MH & Ziomek CA 1981 The foundation of two distinct cell lineageswithin the mouse morula. Cell 24 71–80. (doi:10.1016/0092-8674(81)90502-X)

    Johnstone O & Lasko P 2001 Translational regulation and RNA localizationin Drosophila oocytes and embryos. Annual Review of Genetics 35365–406. (doi:10.1146/annurev.genet.35.102401.090756)

    Kimble J & Crittenden SL 2007 Controls of germline stem cells, entry intomeiosis, and the sperm/oocyte decision in Caenorhabditis elegans.Annual Review of Cell and Developmental Biology 23 405–433. (doi:10.1146/annurev.cellbio.23.090506.123326)

    Kobayashi S, Amikura R & Okada M 1993 Presence of mitochondrial largeribosomal RNA outside mitochondria in germ plasm of Drosophilamelanogaster. Science 260 1521–1524. (doi:10.1126/science.7684857)

    Kobayashi S, Amikura R & Mukai M 1998 Localization of mitochondriallarge ribosomal RNA in germ plasm of Xenopus embryos. CurrentBiology 8 1117–1120. (doi:10.1016/S0960-9822(98)70466-X)

    Krisher RL & Prather RS 2012 A role for the Warburg effect inpreimplantation embryo development: metabolic modification tosupport rapid cell proliferation. Molecular Reproduction and Develop-ment 79 311–320. (doi:10.1002/mrd.22037)

    Kurotaki Y, Hatta K, Nakao K, Nabeshima Y & Fujimori T 2007 Blastocystaxis is specified independently of early cell lineage but aligns with the ZPshape. Science 316 719–723. (doi:10.1126/science.1138591)

    Lee MT, Bonneau AR & Giraldez AJ 2014 Zygotic genome activationduring the maternal-to-zygotic transition. Annual Review of Cell andDevelopmental Biology 30 581–613. (doi:10.1146/annurev-cellbio-100913-013027)

    Lee C, Zeng J, Drew BG, Sallam T, Martin-Montalvo A, Wan J, Kim SJ,Mehta H, Hevener AL, de Cabo R et al. 2015 The mitochondrial-derivedpeptide MOTS-c promotes metabolic homeostasis and reduces obesityand insulin resistance. Cell Metabolism 21 443–454. (doi:10.1016/j.cmet.2015.02.009)

    Li R & Albertini DF 2013 The road to maturation: somatic cell interactionand self-organization of the mammalian oocyte. Nature Reviews.Molecular Cell Biology 14 141–152. (doi:10.1038/nrm3531)

    Li L, Baibakov B & Dean J 2008 A subcortical maternal complex essentialfor preimplantation mouse embryogenesis. Developmental Cell 15416–425. (doi:10.1016/j.devcel.2008.07.010)

    Maniataki E & Mourelatos Z 2005 Human mitochondrial tRNAMet isexported to the cytoplasm and associates with the Argonaute 2 protein.RNA 11 849–852. (doi:10.1261/rna.2210805)

    Mishra P & Chan DC 2014 Mitochondrial dynamics and inheritance duringcell division, development and disease. Nature Reviews. Molecular CellBiology 15 634–646. (doi:10.1038/nrm3877)

    Motosugi N, Bauer T, Polanski Z, Solter D & Hiiragi T 2005 Polarity of themouse embryo is established at blastocyst and is not prepatterned. Genesand Development 19 1081–1092. (doi:10.1101/gad.1304805)

    Muzumdar RH, Huffman DM, Atzmon G, Buettner C, Cobb LJ, Fishman S,Budagov T, Cui L, Einstein FH, Poduval A et al. 2009 Humanin: a novelcentral regulator of peripheral insulin action. PLoS ONE 4 e6334.(doi:10.1371/journal.pone.0006334)

    Nance J 2014 Getting to know your neighbor: cell polarization in earlyembryos. Journal of Cell Biology 206 823–832. (doi:10.1083/jcb.201407064)

    Ninomiya Y & Ichinose S 2007 Subcellular distribution of mitochondrialribosomal RNA in the mouse oocyte and zygote. PLoS ONE 2 e1241.(doi:10.1371/journal.pone.0001241)

    Niwa H, Toyooka Y, Shimosato D, Strumpf D, Takahashi K, Yagi R &Rossant J 2005 Interaction between Oct3/4 and Cdx2 determinestrophectoderm differentiation. Cell 123 917–929. (doi:10.1016/j.cell.2005.08.040)

    Ogawa M, Amikura R, Akasaka K, Kinoshita T, Kobayashi S & Shimada H1999 Asymmetrical distribution of mitochondrial rRNA into smallmicromeres of Sea urchin embryos. Zoological Science 16 445–451.(doi:10.2108/zsj.16.445)

    www.reproduction-online.org

    Downloaded from Bioscientifica.com at 05/30/2021 09:05:08PMvia free access

    http://dx.doi.org/10.1073/pnas.171286998http://dx.doi.org/10.1073/pnas.171286998http://dx.doi.org/10.1126/science.1249410http://dx.doi.org/10.1093/molehr/3.12.1067http://dx.doi.org/10.1073/pnas.61.1.261http://dx.doi.org/10.1073/pnas.61.1.261http://dx.doi.org/10.1016/j.celrep.2013.09.044http://dx.doi.org/10.1016/S0012-1606(81)80019-Xhttp://dx.doi.org/10.1038/ng.2007.63http://dx.doi.org/10.1016/j.ydbio.2004.03.024http://dx.doi.org/10.1016/j.ydbio.2004.03.024http://dx.doi.org/10.1016/j.devcel.2015.07.014http://dx.doi.org/10.1016/0012-1606(75)90262-6http://dx.doi.org/10.1016/0012-1606(75)90262-6http://dx.doi.org/10.1242/dev.00725http://dx.doi.org/10.1016/j.devcel.2010.02.012http://dx.doi.org/10.1073/pnas.101133498http://dx.doi.org/10.1073/pnas.95.19.11274http://dx.doi.org/10.1101/gad.486108http://dx.doi.org/10.1101/gad.486108http://dx.doi.org/10.1002/mrd.22050http://dx.doi.org/10.1002/m