Drosophila stocks
Fly stocks were obtained from the Bloomington Drosophila Stock Center (BDSC) at Indiana University, unless indicated otherwise. Flies were kept on standard fly food at 25 °C.
To analyze chromatin loading, we used fly strains coexpressing EGFP-tagged condensin subunits and His2Av-mRFP1. Lines for the analysis of SMC2 (w*; P [w+, His2Av-mRFP1]II.1; M [w+, gSMC2h-EGFP]ZH-96E) and Cap-D2 (w*; M [w+, EGFP-Cap-D2]ZH-51D/CyO; P [w+, His2Av-mRFP1]III.1/TM3,Sb) were described previously (Herzog et al. 2013). Lines expressing C-terminally fused variants of Barren and Cap-G were generated using a CRISPR/Cas9-based approach (see below). The resulting Cap-G-FE and Barren-FE transgenes were then combined with a chromosome carrying a transgene allowing expression of His2Av fused with mRFP1 (Schuh et al. 2007).
For RNAi-mediated knockdown of Condensin mRNA, we used fly strains expressing either short hairpin RNAs (shRNAs), as in the case of SMC2 (y1sc* v1; P [TRiP.HMS00360]attP2) and Cap-G (w*; P [w+, UAS-CapG-RNAi 20.2]ZH-96E), or a long hairpin RNA (lhRNA) as in the case of Barren (P [KK101679]VIE-260B). The Cap-G shRNA expressing construct was generated by cloning a double-stranded oligonucleotide corresponding to nucleotides 1148-1168 of the Cap-G reading frame, flanked by NheI and EcoRI sites, into the plasmid pWalium20, which is identical to pValium20 (Ni et al. 2011) except that it contains white+ as selectable marker instead of vermilion+. The construct was injected into y1, w1, M [vas-int]ZH2A; M[3xP3-RFP, attP’]ZH96E embryos to establish transgenic lines.
For expression of UAS-transgenes, we used bam-GAL4-VP16 (Chen and McKearin 2003), ey-GAL4 (Hazelett et al. 1998), and maternal α4tub-GAL4-VP16 (Micklem et al. 1997).
For the construction of fly stocks expressing siRNA-resistant variants, siRNA target sites of SMC2 (CAAAACAAGTTCCTCATCAA) and Cap-G (GGCAGTGTCTTAGCGAATATC) were mutated based on a PCR-mediated approach. A first fragment comprising the SMC2 genomic sequence up to the region encoding the mutated siRNA target site was PCR-amplified using the primers KS39 (5′-GCGGTTAATTAAACGTTAAAATAATTGAATGAAGC-3′) and KS42 (5′-CCATTAATCAGAAATTTATTTTTGCCTCCGACAACCAC-3′). A second fragment corresponding to a region spanning the mutated target site and the downstream sequence of the target site was PCR-amplified using the primers KS40 (5′-ATAAACGCGTATGACGCAGCTCGATCTCTGAGGTC-3′) and KS41 (5′-GGCAAAAATAAATTTCTGATTAATGGCAAGCTGGTGC-3′). The two PCR-generated DNA fragments partially overlap in the region encoding the mutated siRNA recognition site. After purification (PCR purification kit, Thermo Scientific), the two PCR products were pooled and served as template for a final PCR using the flanking primers KS39 and KS40. The final PCR product was used to replace the native sequence in the plasmid pattB-SMC2h-EGFP (Herzog et al. 2013). Target site mutation of Cap-G was carried out analogous to that described above for SMC2 but with primer pairs IH01 (5′-ATATCCTAGGGGCTGAGGAGGGCAATGAG-3′)/IH02 (5′-CCAGGTACTCGGACAGGCATTGCCAATATAACAACAGC-3′), IH03 (5′-ATTGGCAATGCCTGTCCGAGTACCTGGAGACGGAAGCG-3′)/IH04 (5′-ATCACTAAGTGAAAGTTAATTAAGTTAG-3′), and IH01/IH04 for the final amplification. As template, the plasmid pattB-Cap-GFL-EGFP (Herzog et al. 2013) was used. For target site mutation of Barren, 104 silent mutations were introduced in the 530 bp spanning recognition site by gene synthesis and this region was replaced within the plasmid pattB-barren-EGFP. pattB-barren-EGFP contains a 6.8 kb genomic fragment encompassing barren as well as 1247 bp and 3219 bp of genomic sequences upstream and downstream of the barren reading frame, respectively. The EGFP reading frame was fused to the 3′-terminus of the barren reading frame. This genomic barren-EGFP transgene fully rescues the lethality associated with the barrL305/Df(2 L)Exel7077 transheterozygous mutant situation (data not shown). Transgenic lines of the siRNA-resistant transgenes were generated by φC31 integrase-mediated germline transformation via injection of the plasmids pattB-Cap-G-siRNAres, pattB-SMC2h-siRNAres, and pattB-barren-siRNAres into y1w* M [vas-int. Dm]ZH-2A; M[3xP3-RFP.attP’]ZH-68E embryos.
For deGradFP dependent destruction of Barren-FE in the male germ line, we generated w*; Barren-FE/Barren-FE; P [w+,UASP-NSlmb-vhh-GFP4]III.1/ P [w+, bam-GAL4-VP16] or w*; Barren-FE/CyO; P [w+,UASP-NSlmb-vhh-GFP4]III.1/ P [w+, bam-GAL4-VP16] males by standard crossing schemes. As controls, we also generated w*; Barren-FE/Barren-FE; P [w+,UASP-NSlmb-vhh-GFP4]III.1/TM3, Sb or w*; Barren-FE/CyO; P [w+,UASP-NSlmb-vhh-GFP4]III.1/ TM3, Sb flies, which do not carry a Gal4 driver. The w*; P [w+, UASP-NSlmb-vhh-GFP4]III.1 transgene was described previously (Urban et al. 2014).
CRISPR/Cas9-mediated genome engineering
gRNA design
To generate variants of Barren and Cap-G carrying a C-terminal EGFP-fusion expressed from the endogenous loci, we employed the CRISPR/Cas9-induced HDR pathway to insert the coding sequence for EGFP downstream of their respective reading frames within the genome. Target sites for Cas9 were chosen in a way that the double-strand breaks (DSBs) were generated in close proximity to the designated fusion site, i.e., the translational termination codon, and with a low risk of potential off-target effects. To identify optimal target sequences and assess specificity of the CRISPR targets, we used the CRISPR Optimal Target Finder algorithm at http://tools.flycrispr.molbio.wisc.edu/targetFinder/ (Gratz et al. 2014). In order to supply gRNAs from a plasmid DNA source, designated target site sequences (Barren: 5′-GCTAATTCCGCAGGAGGACTTGG-3′ ➔ cleavage 36 nt upstream of the translational stop codon within exon 3 of Barren; Cap-G: 5′-GAAGCGCGTGACGCGGGCAGTGG-3′ ➔ cleavage 48 nt upstream of the translational stop codon within exon 6 of Cap-G) were synthesized as a pair of short complementary oligonucleotides and cloned into the pU6-BbsI-gRNA vector backbone (Gratz et al. 2014) according to the instructions provided on flyCRISPR.
HDR template cloning
The HDR templates were assembled in the plasmid pSLfa1180fa (Horn and Wimmer 2000) and contained the EGFP-encoding sequence preceded by a removable FRT-SV40-3xP3-FRT (or initially FRT-3xP3-FRT) expression cassette. These regions were flanked by appropriate homologous sequences (at least 1 kb homology arms upstream and downstream of the cleavage site) for efficient HDR-mediated repair. To avoid cleavage of the HDR templates by Cas9, silent point mutations were introduced into the protospacer regions and the PAM sites. The details of the cloning strategy are available upon request.
Microinjection and screening
HDR templates and the gRNA-encoding plasmids were co-injected into transgenic embryos expressing Cas9 under control of the nos-promotor (Port et al. 2014). Six micrograms of gRNA- and 6 μg HDR template-encoding plasmids were co-precipitated and dissolved in 20 μl of injection buffer (0.1 mM NaP, 5 mM KCl; pH 6.8) prior to injection. To isolate integration events, individual adult males developing from injected embryos were outcrossed to females of the balancer stock w*; Sco/CyO, P [ry+, ftz lacZ]. Each F1 brood was scored for green eye fluorescence due to expression of EGFP under control of the eye-specific 3xP3-promotor, indicating integration events. Individual recombined chromosomes were isolated and the resulting balanced lines are referred to as Barren-FSV3FE/CyO, P [ry+, ftz lacZ] or w*; Cap-G-FSV3FE/CyO, P [ry+, ftz lacZ] or w*; Cap-G-F3FE/CyO, P [ry+, ftz lacZ] (the latter lacking the SV40 terminator sequence). The correct integration at the desired locus was confirmed by PCR analyses.
To remove the promotor-cassette, and at the same time generate a translational fusion between Barren or Cap-G and EGFP, Flp mRNA was injected into embryos derived from parents with the genotypes w*; Barren-FSV3FE/CyO, P [ry+, ftz lacZ] or w*; Cap-G-FSV3FE/CyO, P [ry+, ftz lacZ] or w*; Cap-G-F3FE/CyO, P [ry+, ftz lacZ]. Single-injected, non-CyO males were outcrossed to w*; Sco/CyO, P [ry+, ftz lacZ] virgin females and the F1 generation was screened for the loss of green eye fluorescence.
Flp mRNA production
The Flp-recombinase encoding sequence was amplified with the oligodeoxynucleotides SH381 (5′-CGATCATAATACGACTCACTATAGGGGTCACAACATGGGCCCAAAAAAGAAAAGA-3′) and SH382 (5′-ATGGCGCGCCTTATATGCGTC-3′) from the plasmid pAS1834 (generously provided by Olaf Stemmann). The PCR product served as template for Flp mRNA synthesis by in vitro transcription and subsequent polyadenylation using the mMessage mMachine R T7 Ultra Kit (Thermo Fisher, Invitrogen) according to the manufacturers’ recommendations.
Culture of isolated spermatid cysts
To determine the chromatin association of the EGFP-fused condensin subunits during male meiosis, pupal testes were dissected at approximately 1 day after puparium formation in Shields and Sang M3 insect culture medium (Sigma) supplemented with 10% fetal bovine serum (Sigma) as well as 100 U/ml penicillin and 100 μg/ml streptomycin. The dissected pupal testes were transferred into culture dishes (ibidi μ-Dish; article no. 81158) and teared open with thin needles. Spermatid cysts were gently squeezed out of the testes. For microscopy, isolated cysts were transferred into sterile glass-bottom culture dishes (ibidi 8-well μ-Slide; article no. 80826) with fresh medium using glass Pasteur pipettes. To avoid floating of the cysts, a drop of 1% methyl cellulose was added to each well.
Cysts were staged according to their morphology and the presence of three chromatin territories indicating prophase of meiosis I. For analysis of progression through meiosis II, appropriate cysts were identified by the size and the number of the nuclei within the cysts. Multi-stack confocal images were acquired every 3 min using a Leica Confocal TCS SP5 system (Carl Zeiss,Germany) equipped with a × 40/1.25 oil-immersion objective, a 488-nm Ar laser, and a 561-nm DPSS561 laser for the excitation of EGFP and mRFP1, respectively.
Male fertility test
To assess male fertility, 2–4-day-old single males were crossed to three 5–12-day-old virgin wild-type females. Ten single males were analyzed per genotype. Crosses were maintained at 25 °C on standard medium supplemented with yeast paste. After 4 days, males were discarded and females were flipped into fresh vials and maintained for a second period of 4 days. Females were then removed and all vials were further incubated at 25 °C. Progeny was counted over a period of 10 days starting with the first day of eclosion. For statistical analysis, unpaired Student’s t tests (www.graphpad.com) were performed.
Analysis of seminal vesicles and early embryos
To analyze sperm content within seminal vesicles, males were collected shortly after eclosion, restricted from females and maintained at 25 °C. After 10 days, seminal vesicles were dissected and fixed at room temperature for 20 min in a mixture of 300 μl heptane and 150 μl fixation solution (1× PBS, 0.5% Nonidet NP 40 and 2% para-formaldehyde). Fixed seminal vesicles were then washed twice with PBS and treated with Hoechst 33258 (1 μg/ml in PBS) to stain DNA. Confocal microscopy was employed to determine the focal plane showing the maximum extent of the seminal vesicles. ImageJ (Schneider et al. 2012) was used to calculate the area of the seminal vesicles within these focal planes.
For analysis of early embryonic development, 0–3-h-old embryos were collected on apple-juice agar plates and dechorionized. Embryos were fixed in a 1:1 mixture of n-heptane and methanol for 5 min at room temperature, washed with PBST (PBS plus 0.1% Triton X-100) and treated with Hoechst 33258 (1 μg/ml in PBS) to stain DNA. Prior to mounting, embryos were washed three times in PBS for 5 min each.
Nondisjunction analysis
For the analysis of nondisjunction rates of the 4th chromosome after knockdown of condensin subunits in the male germ line, we adopted and modified a previously published assay (Hartl et al. 2008b). Twenty males (2–3 days old) were crossed to 30 virgin females carrying the compound chromosome C(4) RM, ci1, eyR. Because in these females the 4th chromosomes are attached, the eggs either carry the compound C(4) RM, ci ey chromosome (diplo-4), or no 4th chromosome (nullo-4). Nullo-4 eggs fertilized by normal haploid sperm create nullo-4/+ progeny, while the fertilization of C(4) RM, ci1, eyR eggs with haploid sperm creates C(4) RM, ci1, eyR/+ progeny. Both classes are viable and appear normal with respect to ci and ey according to wild-type alleles on the paternal 4th chromosome. In the case of 4th chromosome missegregation events during male meiosis, exceptional classes of progeny arise, one of which can be phenotypically detected. This is, when nullo-4 sperm fertilize diplo-4 oocytes. In this case, progeny exhibit the ci and ey mutant phenotype due to carrying exclusively the mutant alleles present on the compound chromosome, which are not complemented by wild type alleles provided by the father. The additional exceptional classes go undetected with this assay because they are either lethal (0/0) or appear phenotypically wild-type (0/++ and C(4) RM, ci1, eyR/++).
To quantify sex chromosome nondisjunction rates, 20 males (2–3 days old) were crossed to 30 virgin wild-type females. In addition to the transgenes needed for downregulation of condensin subunits, males were bearing an Y chromosome (Dp(1;Y)BsYy+) carrying two X translocations with a dominant allele of Bar (BS) and the wild-type allele of yellow, respectively. The y+ allele was dispensable for the evaluation of the nondisjunction test. Offspring that arises from sperm bearing the normal sex chromosome content, either one X or one Y, corresponds to the genotypes XX and X BsYy+, respectively. In this case, all female flies have a wild-type eye phenotype, whereas all males have reduced eyes due to the BS allele. If exceptional classes of sperm are created that are XY, or lack either sex chromosome entirely, then BS females (XX BsYy+) and males with wild-type eyes (X0 males) arise, respectively, among the offspring. Diplo-X sperm will result in a lethal Triplo-X combination after fertilization.
To specifically analyze X chromosome nondisjunction during meiosis II, 20 males (2–3 days old) were crossed to 30 virgin females of the genotype C(1) RM, y2su (wa)1wa/0 (BDSC stock no. 700) carrying a compound X-chromosome. Regular female progeny from these crosses inherits the compound X chromosome from the mothers and a Y chromosome from the fathers. These females are phenotypically characterized by wild-type eye color and yellow pigmentation of the cuticle. If X-chromosome nondisjunction occurs during meiosis II in the fathers, sperm containing two X-chromosomes can fertilize eggs without a gonosome resulting in female progeny that can be phenotypically distinguished from their siblings. These exceptional females are yellow+, and carry a w− -allele on their X-chromosomes from the father. In the case of Cap-G-RNAi, all exceptional progeny harbor one mini-white+ allele due to the presence of either the bam-GAL4 or the UAS-Cap-G-RNAi transgene, which result in orange eyes. In the case of Barren-RNAi the progeny receives none, one, or two copies of a mini-white+ marked transgene, since bam-GAL4 and UAS-Barren-RNAi reside on different chromosomes. Thus, the progeny has white, orange or red eyes, respectively.
Squashed testes preparations and immunofluorescence staining
3–4 pairs of testes were dissected from young males of the desired genotype and placed into a drop of PBS (137 mM NaCl; 2.7 mM KCl; 10 mM Na2HPO4; 1.8 mM KH2PO4; pH 7.4) on a poly-L-lysine-coated microscopy slide. A siliconized cover slip was placed on the samples, covered with 4 layers of tissue, and testes were gently squashed by applying some pressure manually. After snap-freezing in liquid nitrogen, the cover slip was immediately removed using a clean scalpel. The slides were then transferred into a Coplin jar filled with ice-cold 95% ethanol, and dehydrated at − 20 °C for at least 10 min. Samples were treated with 4% formaldehyde in PBS for 10 min at room temperature to fix the testes. After fixation, slides were washed two times with PBST (PBS/0.3% Triton X-100) and once with PBT (PBS/0.1% Tween 20) 15 min each. For the blocking step, slides were immersed in PBT/1% BSA for 30 min. Slides were removed from the jar; 60 μl of primary antibody solution was applied to the squashed testes, protected with a cover slip, and incubated in a dark, moist chamber at 4 °C overnight. After washing four times in PBT/1% BSA for 15 min each, secondary goat antibodies conjugated with Alexa 488 or Cy3 were applied for 2 h analogous to the primary antibody treatment. All antibodies were diluted in PBT/1% BSA. Following additional washes with PBT/1% BSA, DNA was stained with Hoechst 33258 (1 μg/ml). Finally, samples were washed four times in PBS and mounted with Fluoromount G (Southern Biotech).
Fluorescent in situ hybridization (FISH)
A labeled X-chromosome-specific probe was prepared and used in FISH analyses as described previously (Urban et al. 2014) with some modifications. Testes were dissected from 1 to 2-day old males and incubated in a droplet of 0.5% sodium citrate on a microscopy slide coated with poly-L-lysine for 10 min. The sodium citrate solution was carefully removed, and testes were coated with a 45% acetic acid/2% para-formaldehyde solution for 3 min to fix the sample. Following a squashing step as described above, testes were dehydrated sequentially with ice-cold 70% and 100% ethanol. After air-drying, testes were sequentially incubated with 2× SSCT (0.3 M sodium chloride; 30 mM sodium citrate; 0.1% (v/v) Tween 20), 2× SSCT-25% formamide, and 2× SSCT-50% formamide for 10 min each, followed by incubation in fresh 2× SSCT-50% formamide for 3 h at 37 °C. The testes were then coated with 36 μl of hybridization buffer (20% dextrane sulfate, 15% formamide in 2× SSCT) supplemented with 100 ng of fluorescently labeled probe and protected with a cover slip. Probe and chromosomal DNA were denatured at 95 °C for 5 min, and the hybridization reaction was carried out overnight at 37 °C in a humid chamber. After hybridization, slides were washed three times with pre-warmed (37 °C) 2× SSCT-50% formamide for 1 h each, then once with 2× SSCT-25% formamide, and once with 2× SSCT for 10 min/wash. The samples were rinsed with PBS and DNA was stained with Hoechst 33258 (1 μg/ml in PBS). Finally, the testes were washed once in PBS and mounted in Fluoromount G Medium (Southern Biotech).
Immunoblotting
For immunoblotting experiments, tissues were dissected in PBS and homogenized in sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) sample buffer. Protein samples were then separated on Tris-glycine-based polyacrylamide gels and blotted onto nitrocellulose membranes. For detection of bound antibodies on immunoblots, the horseradish peroxidase-based system from p.j.k was used according to the manufacturer’s recommendations.
Antibodies
Antibodies against Drosophila SMC2, Drosophila Cap-G, Drosophila Cid, and EGFP have been described previously (Herzog et al. 2013; Jäger et al. 2005). An antibody against Barren was raised in rabbits using bacterially expressed full-length protein as antigen. The antiserum was affinity purified using standard procedures. A mouse monoclonal antibody directed against α-tubulin was obtained from Sigma. For immunoblotting, rabbit antibodies were used at a 1∶3000 dilution and the anti-α-tubulin antibody at a 1:20,000 dilution. For immunofluorescence analyses, the anti-Cid antibody and the anti-α-tubulin antibody were diluted 1:500 and 1:8000, respectively. Secondary antibodies conjugated with fluorophores (Molecular probes) or horseradish peroxidase (Jackson laboratories) were obtained commercially.