• No results found

Heat Shock Factor 5 Is Essential for Spermatogenesis in Zebrafish

N/A
N/A
Protected

Academic year: 2021

Share "Heat Shock Factor 5 Is Essential for Spermatogenesis in Zebrafish"

Copied!
16
0
0

Loading.... (view fulltext now)

Full text

(1)

http://www.diva-portal.org

This is the published version of a paper published in Cell reports.

Citation for the original published paper (version of record):

Saju, J M., Hossain, M S., Liew, W C., Pradhan, A., Thevasagayam, N M. et al. (2018)

Heat Shock Factor 5 Is Essential for Spermatogenesis in Zebrafish

Cell reports, 25(12): 3252-3261

https://doi.org/10.1016/j.celrep.2018.11.090

Access to the published version may require subscription.

N.B. When citing this work, cite the original published paper.

Permanent link to this version:

(2)

Report

Heat Shock Factor 5 Is Essential for

Spermatogenesis in Zebrafish

Graphical Abstract

Highlights

d

hsf5 is predominantly expressed in testis

d

Hsf5 loss of function causes infertility in zebrafish males, but

not in females

d

The

hsf5

/

mutant testes start showing defects from

meiosis-I during spermatogenesis

d

The mutants have low sperm count with structural defects in

their spermatozoa

Authors

Jolly M. Saju,

Mohammad Sorowar Hossain,

Woei Chang Liew, ..., Amit Anand,

Per-Erik Olsson, La´szlo´ Orba´n

Correspondence

amit@tll.org.sg (A.A.),

per-erik.olsson@oru.se (P.-E.O.),

orban@georgikon.hu (L.O.)

In Brief

Saju et al. show that heat shock factor 5

(

hsf5) is predominantly expressed in

testis. Loss of Hsf5 leads to a drastic

reduction in sperm counts and severe

morphological abnormalities in

spermatozoa, leading to infertility. Hsf5

appears to be required for proper

progression of meiosis-I during

spermatogenesis.

Saju et al., 2018, Cell Reports25, 3252–3261 December 18, 2018ª 2018 The Author(s).

(3)

Cell Reports

Report

Heat Shock Factor 5 Is Essential

for Spermatogenesis in Zebrafish

Jolly M. Saju,1Mohammad Sorowar Hossain,1,2,7Woei Chang Liew,1Ajay Pradhan,3Natascha May Thevasagayam,1 Lydia Shun En Tan,1Amit Anand,4,*Per-Erik Olsson,3,*and La´szlo´ Orba´n1,5,6,8,*

1Reproductive Genomics Group, Temasek Life Sciences Laboratory, Singapore, Singapore 2Department of Biological Sciences, National University of Singapore, Singapore, Singapore

3Biology, The Life Science Center, School of Science and Technology, O¨ rebro University, O¨rebro, Sweden 4Bioimaging and Biocomputing, Temasek Life Sciences Laboratory, Singapore, Singapore

5Frontline Fish Genomics Research Group, Department of Animal Sciences, Georgikon Faculty, University of Pannonia, Keszthely, Hungary 6Centre for Comparative Genomics, Murdoch University, Murdoch, Australia

7Present address: Biomedical Research Foundation, Dhaka, Bangladesh 8Lead Contact

*Correspondence:amit@tll.org.sg(A.A.),per-erik.olsson@oru.se(P.-E.O.),orban@georgikon.hu(L.O.)

https://doi.org/10.1016/j.celrep.2018.11.090

SUMMARY

Heat shock factors (Hsfs) are transcription factors

that regulate responses to heat shock and other

envi-ronmental stimuli. Four heat shock factors (Hsf1-4)

have been characterized from vertebrates to date. In

addition to stress response, they also play important

roles in development and gametogenesis. Here, we

study the fifth member of heat shock factor family,

Hsf5, using zebrafish as a model organism. Mutant

hsf5

/

males, generated by CRISPR/Cas9

tech-nique, were infertile with drastically reduced sperm

count, increased sperm head size, and abnormal tail

architecture, whereas females remained fertile. We

show that Hsf5 is required for progression through

meiotic prophase 1 during spermatogenesis as

suggested by the accumulation of cells in the

lepto-tene and zygolepto-tene-pachylepto-tene stages and increased

apoptosis in post-meiotic cells.

hsf5

/

mutants

show gonadal misregulation of a substantial number

of genes with roles in cell cycle, apoptosis, protein

modifications, and signal transduction, indicating an

important role of Hsf5 in early stages of

spermato-genesis.

INTRODUCTION

Heat shock factors (Hsfs) are a family of transcription factors involved in differentiation, development, reproduction, and stress-induced adaptation by regulating temperature-controlled heat shock protein (hsp) genes and other non-hsp genes as well (reviews:A˚kerfelt et al., 2010; Gomez-Pastor et al., 2018). Four members of the heat shock factor family have been character-ized from vertebrates prior to this study (reviews:Fujimoto and Nakai, 2010; Gomez-Pastor et al., 2018). Multiple heat shock factors in plants and vertebrates appear to mediate a wide array of responses to versatile forms of physiological and environ-mental stimuli (Dayalan Naidu and Dinkova-Kostova, 2017;

Park and Seo, 2015). Some heat shock factors have an important role in male gonad development and spermatogenesis.

Heat-responsive heat shock factors are ubiquitously expressed across many tissues during thermal stress; however, Hsf1 and Hsf2 are also known to play specific roles in gametogenesis during non-stress conditions (review:Abane and Mezger, 2010). Sperm production in Hsf1-knockout mouse testes was decreased ( Sal-mand et al., 2008), but the animals still exhibited normal reproduc-tive ability (Salmand et al., 2008; Zhang et al., 2002) and spermato-genesis (Izu et al., 2004). On the other hand, overexpression of

Hsf1 led to defective spermatogenesis (Nakai et al., 2000). Hsf2 showed enhanced expression in mouse testis between day 14 to 21 postnatal development (Fiorenza et al., 1995) and localized to the nuclei of spermatocytes in a stage-specific manner (Alastalo et al., 1998; Sarge et al., 1994). Disruption of Hsf2 expression re-sulted in several negative phenotypes, including reduced testis size and sperm count (Kallio et al., 2002; Wang et al., 2003). Similar to Hsf1-knockout, Hsf2 null mutant male mice were fertile (Wang et al., 2003). However, disruption of both Hsf1 and Hsf2 resulted in male sterility due to severe defects in spermatogenesis (Wang et al., 2004).

Zebrafish (Danio rerio, Cyprinidae) is an important vertebrate model organism for development, genetics, and reproduction (see e.g., Dahm and Geisler, 2006). Prior to this study, three heat shock factors—Hsf1, Hsf2 and Hsf4—have been isolated and characterized from zebrafish (Swan et al., 2012; Yeh et al., 2006). Array-based transcriptomic studies performed in our lab earlier on adult zebrafish gonads identified heat shock factor 5 (hsf5), the fifth, hitherto uncharacterized member of the heat shock factor family. Although the expression of Hsf5 was shown to be testis enhanced earlier in several mammalian species (see e.g.,Chalmel et al., 2012; Kogo et al., 2010), its detailed func-tional characterization has not been performed to date. RESULTS

Identification and Cloning of Hsf5, an Uncharacterized Heat Shock Factor

We identified hsf5 initially as an expressed sequence tag (EST) with enhanced expression in adult zebrafish testis (Li et al.,

(4)

2004). Mapping of the full-length cDNA to the latest genome assembly (GRCz11; released in May 2017; for primers see

Table S1) revealed a genomic locus that consists of six exons spanning a region of over 20 kb on Chromosome 5 (3,094,980– 3,118,313 bp). Bioinformatic analysis predicted a protein of 47 kDa and with an isoelectric point of pH 7.34 with a helix-turn-helix DNA binding domain (DBD) in the N-terminal region (between the 14thand 119thamino acids; AA in short), which is

the most conserved functional domain in heat shock factors across vertebrates (Figure 1A; Table S2). The DBD showed 37%–39% identity with those of other zebrafish heat shock factors (Table S3). However, the zebrafish protein lacked any other domains, such as Neuregulin, heptad repeats A/B (HR-A/B) and C (HR-C) described from other heat shock factors (Figure 1A).

Homology and phylogenetic analysis grouped the protein together with its orthologs in several vertebrate species as a different branch, away from previously described heat shock fac-tors that formed four separate clusters (Hsf1–4;Figure 1B). These data confirm that this is the fifth member of the vertebrate heat shock factor family and is classified as heat shock factor 5 (Hsf5). We examined experimentally whether hsf5 expression is altered upon heat shock as observed for many other heat shock response proteins. The expression level of hsf5 did not increase upon heat shock in adult testis, whereas transcript levels of

hsp70 showed a significant upregulation (Figures S1A and S1B). hsf5 Gene Shows Sexually Dimorphic Expression The hsf5 transcript was maternally deposited in zebrafish oo-cytes and the first sign of its zygotic expression was observed Figure 1. Characterization of Zebrafish Heat Shock Factor 5 and Generation ofhsf5 Mutants Using CRISPR/Cas9-Based Strategy (A) Schematic representation of four members of zebrafish heat shock factor (Hsf) protein family for comparing the relative location of specific domains. For Hsf5, two transcript variants (hsf5_tv1 and hsf5_tv2) are shown. HR, heptad repeat.

(B) Phylogenetic analysis of vertebrate heat shock factors showed that all orthologs of zebrafish Hsf5 are located on a separate branch (in bold rectangle) clearly apart from other heat shock factor family members. The tree was constructed with the neighbor-joining method based on the amino acid sequences; we per-formed a 10003 bootstrapping for a robust comparison. hs, Homo sapiens; mm, Mus musculus; gg, Gallus; xt, Xenopus tropicalis; om, Oncorhynchus mykiss; and dr, Danio rerio (GenBank IDs shown inTable S2).

(C) Schematic representation of three mutant lines generated by CRISPR/Cas9 method: Hsf5sg40

has a deletion of five bases, and it introduces a stop codon after 121 amino acids (AA). Hsf5sg41

has a deletion of 7 bases, whereas Hsf5sg42

has an addition of 25 bases, introducing a stop codon after 114 AA and 131 AA, respectively. All three mutations are frameshift type resulting in a truncated protein.

(D) Western blot confirmed loss of the C-terminal regions in the mutants. Adult testes from Hsf5sg40

and Hsf5sg41

and WT were used. Actin was used as an internal control; n = 2 biological replicates in each group.

(E) Blue Native PAGE analysis using total testicular lysates showed a band of 242 kDa (arrow), suggesting that Hsf5 is present either in oligomeric state or as complexed with other proteins; n = 4 biological replicates.

(5)

at the mid-blastula transition stage (Figure S1C). In adults, the

hsf5 expression level was significantly higher in the testis than

in other organs, including the ovary (Figure S1D).

Testis-enhanced expression pattern of hsf5 was established during gonadal development. In comparison to two early testic-ular markers, amh and nr5a1a, and ovarian marker cyp19a1a, which showed upregulated expression in respective gonads from 30 days post-fertilization (dpf) (Hossain, 2010; Wang and Orban, 2007), hsf5 expression remained low in developing go-nads from both sexes until 35 dpf (Figures S1E–S1H). Expression levels of hsf5 increased substantially in testis, but not in other or-gans, from 35 dpf (Figure S1H). This suggests that Hsf5 may function in downstream processes of testis formation and/or maturation.

Cloning of the full-length hsf5 revealed the presence of two transcript variants, hsf5_tv1 and hsf5_tv2 (Figures 1A,S2A, and S2B). The shorter variant (hsf5_tv2) lacked the third exon and the resulting protein contained 335 AAs compared to 416 AAs in hsf5_tv1, yet the DBD remained the same for both isoforms (Figures 1A andS2A). RT-PCR showed that the shorter transcript variant was expressed in testes—albeit at much lower levels compared to that of longer one—and could not be detected in ovaries (Figure S2B).

Generation of Hsf5 Mutants in Zebrafish Using CRISPR/ Cas9

We generated loss-of-function mutants by CRISPR/Cas9 tech-nology targeting a region in exon2 of hsf5 gene (Figure S3A; seeSTAR Methodsfor details).

Mapping of the mutation sites of F0 founder males and fe-males showed different rates of mutation (17%–54%; for typical examples, seeFigure S3B). Three mutant lines were selected for downstream analysis and were named as Hsf5sg40, Hsf5sg41and Hsf5sg42. Each line carried a different mutation, as follows: a 5 bp or 7 bp deletion or a 25 bp insertion, respectively, at the ex-pected site in exon #2 coding for DBD (Figure S3C). At the pro-tein level, mutations introduced premature stop codons at the terminal region of DBD (AA14–AA119) at AA121, AA114, and AA131 in Hsf5sg40, Hsf5sg41and Hsf5sg42mutants, respectively,

generating severely truncated proteins (Figure 1C). We specu-late that if the truncated Hsf5 is expressed in mutants, the func-tion of protein will be drastically affected. From here onward, we refer to all these mutant alleles collectively as hsf5/.

Homozygous hsf5/ mutants carrying a 7 bp deletion (labelled as Hsf5sg41inFigure 1C) were used for western blot, histology, microscopy, and RNA sequencing (RNA-seq) studies. An antibody generated against the C-terminal region of Hsf5 de-tected protein of predicted size from wild-type (WT) testicular protein extract, while this antibody could not detect any expres-sion in Hsf5sg40(or Hsf5sg41) testicular extracts with western blot-ting (Figure 1D; for validation of the anti-Hsf5 antibody, see Fig-ures S2C–S2F). We also examined the status of Hsf5 protein in testis with blue native polyacrylamide gel electrophoresis using total testicular protein extracts. We observed a band around 242 kDa, indicating the existence of Hsf5 as an oligomer or as a part of a big complex with its binding partners (Figure 1E).

To study the potential role of Hsf5 in heat shock response, we examined the expression of hsp70 upon heat treatment on the

hsf5/mutants. Following heat exposure, the expression level of hsp70 transcript in hsf5/mutants was comparable to that of WT, suggesting that Hsf5 function may not be critical for heat shock response (Figure S1A).

The hsf5/mutant males could not produce viable offspring when crossed with WT, homozygous (hsf5/) and heterozygous (hsf5+/) females, although heterozygous males produced viable offspring, whereas hsf5/mutant females were fertile. The em-bryos resulting from hsf5/mutant males and WT females ex-hibited lethality before the age of 1 dpf (data not shown). In addi-tion, a significant decrease in the gonadosomatic index (GSI) was found in the homozygous mutants, suggesting the role of Hsf5 in the maintenance of some gonadal cells and fertility (Figure S3I).

Hsf5 Protein Is Predominantly Observed in Spermatocytes

We examined Hsf5 localization in the testis, where immunostain-ing with anti-Hsf5 antibody revealed abundant Hsf5 presence (Figure 2A). Spermatogenesis is a multi-step process during which diploid germ cells (spermatogonia) are converted into haploid spermatozoa. The main cell types of this transition are spermatogonia (2n), spermatocytes (2n), spermatids (n) and spermatozoa (n) (Leal et al., 2009). Hsf5 signal was predomi-nantly observed in primary spermatocytes (Figure 2B). Compar-ison of Hsf5 localization with that of Sycp3, a known primary spermatocyte marker, shows that Hsf5 expression was abun-dant in primary spermatocytes, while its expression was comparatively lower in spermatogonia, spermatids and sperma-tozoa. Unlike Hsf5, Sycp3 did not show expression in other germ cells (Figures 2C and 2D).

The specificity of anti-Hsf5 antibody was strengthened by the absence of significant signals in the hsf5/testis (Figures 2E andS4A). In agreement with qPCR results, Hsf5 signals upon im-munostaining and western blotting in ovaries were much lower than those in testes (Figures S1D, S2C, and S2F). Similarly, Hsf5 signals in ovaries also seem to be restricted to the germ cells. We confirmed germline-specific signals of Hsf5 by comparing its distribution with germ cell marker Ddx4 (Figures 2G–2I andS4B).

A closer examination of Hsf5 signals in testis revealed that the bulk was outside of the nucleus, whereas the rest was localized as foci in the nucleus (Figures 2J and 2K).

Hsf5 Is Required for Proper Spermatogenesis and Fertility in Males

These results prompted us to perform a histological comparison of WT and hsf5/mutant testes. Expectedly, well-developed lumina filled with spermatozoa could be observed in WT, whereas hsf5/mutants exhibited a drastic loss of spermato-zoa (Figure 3A). A quantitative comparison of testicular cell types revealed a significant increase in the number of primary spermatocytes and a drastic reduction of spermatozoa in

hsf5/ mutants compared to WT, whereas the number of spermatogonia was comparable (Figure 3C). Spermatogonia are the largest germ cells with a large nucleus and poorly condensed chromatin; primary spermatocytes are characterized by the coarse chromatin strands and bouquet configuration

(6)

chromosomes. Spermatids and spermatozoa have dark round nuclei with minimal or no cytoplasm (Figure 3B). Analysis of

hsf5/semen smear using haemocytometer showed a drastic reduction of sperm count (Figure 3D) and decreased sperm motility (data not shown).

We examined the defects in hsf5/spermatozoa at greater resolution, using scanning electron microscopy. We found that

hsf5/spermatozoa had significantly enlarged heads in com-parison to WT and appeared to cluster together with crenate arrangement of cytoplasmic membrane (Figure 3E). Most of the hsf5/spermatozoa appeared to lack flagellum or had a short, kinked one in comparison to WT (Figure 3E).

We examined the cytoskeleton of hsf5/spermatozoa with transmission electron microscopy, which revealed irregular shape

Figure 2. Hsf5 Protein Showed Similar Local-ization Pattern in Spermatocytes to a Known Spermatocyte Marker Sycp3

(A and B) Representative images show immuno-staining with anti-Hsf5 antibodies on adult WT testis sections (B is a zoomed-in image of A).

(C and D) Adjacent sections were immunostained with anti-Sycp3 antibodies (D is a zoomed-in image of C), and nuclei were counterstained with DAPI. Strong Hsf5 and Sycp3 signals were detected in spermatocytes, while weak Hsf5 signals were seen in spermatogonia, spermatids and spermatozoa. (E) Immunostaining with anti-Hsf5 antibody did not show significant signals in hsf5/testis. (F) Immunostaining suggests low levels of Hsf5 in WT ovary.

(G–I) Representative images show immunostaining of adult WT ovaries from tg(ddx4:ddx4-egfp) trans-genic zebrafish with Ddx4 (green; G) and anti-Hsf5 (red; H) antibodies, respectively, and merged images showing signals from both antibodies (I). Hsf5 localization pattern follows that of Ddx4 and does not stain the gonadal somatic cells. White ar-rows indicate the outer layer of oocytes.

(J) Localization of Hsf5 protein (green) as granule-like structures in the nucleus of primary spermato-cytes; orthogonal views are presented to emphasize the presence of foci in the nucleus (red).

(K) Maximum-intensity Z-projection of images. For (A)–(K), n = 3 biological replicates in each group. Scale bars: 50mm in (A), (C), (G), (H), and (I); 30 mm in (B) and (D); 400mm in (E); 100 mm in (F); and 5 mm in (G), (H), and (K). See alsoFigure S4.

and disruption of cytoplasmic membrane at various regions around the nucleus along with intense vacuolization in most sperm heads (91%; n = 103; Figure 3F). The cross-section of flagellar axoneme of most

hsf5/mutant spermatozoa showed se-vere deformity in the arrangement of micro-tubules. Instead of a typical ‘‘9+2’’ pattern for microtubule arrangement (Loreng and Smith, 2017), the majority of hsf5/ sper-matozoa showed severe structural defects with partial or complete loss of central crotubules and/or irregular arrangement of peripheral duplet mi-crotubules (Figure 3G and 3I). Longitudinal sections through the flagella also revealed a lack of central doublet microtubules and irregular arrangement of central and peripheral microtubules in most of the hsf5/mutant sperm cells (Figure 3H).

Severe reduction in sperm count as well as frequent defects in sperm shape and structure in hsf5/mutants suggest that Hsf5 is important for spermatogenesis and fertility in males.

Spermatogenesis in hsf5/Mutant Testes Appears to

Be Compromised during Meiotic Division

Increase in the number of primary spermatocytes and severe reduction of sperm count in hsf5/ prompted us to examine primary spermatocytes at different stages of meiosis.

(7)

We performed immunostaining with anti-Sycp3 to compare the progression of hsf5/mutant and WT primary spermatocytes (Ozaki et al., 2011) through prophase-1.

hsf5/ testes showed a marked increase in the clusters of Sycp3-positive cells, confirming accumulation of primary spermatocytes in comparison to WT, which was suggested Figure 3. Histological and Electron Microscopic High-Resolution Analysis of Testes and Spermatozoa Revealed Severe Reduction of Sperm Count and Defective Shape of Spermatozoa inhsf5/Mutant

(A) H&E staining of testicular sections showed severe loss of spermatozoa in hsf5/testis lumen, while abundant spermatozoa filled the lumen of WT. (B) Representative germ cell stages: spermatogonia (SPG), primary spermatocyte (PS) and spermatozoa (SZ).

(C) Segmentation-based quantification of different cell types using histology images reflected accumulation of primary spermatocyte (PS) and drastic reduction of spermatozoa (SZ) in hsf5/testis in comparison to WT.

(D) Haemocytometer-based sperm counting reflected severe reduction in the number of spermatozoa in hsf5/compared to WT.

(E) Comparison of WT (top left and right) and hsf5/(bottom left and right) spermatozoa with scanning electron microscope showed clumping of spermatozoa (bottom left) with enlarged head and defective and shorter flagellum (bottom right) in the latter.

(F) Comparison of WT and hsf5/sperm heads using transmission electron microscopy (TEM) showed the latter with dislocated cytoplasmic membrane (blue arrows) and intense vacuolization.

(G) Representative magnified TEM images of cross section through the central region of a WT and hsf5/sperm flagella showed severe defects in the orga-nization of axoneme in the latter.

(H) Representative magnified TEM images of longitudinal section of WT and hsf5/sperm flagella showed severe defects in microtubules of the mutant. Mi-crotubules in WT sperm flagella are marked by white arrows.

(I) Quantitation of defects in the head and microtubule arrangements in axonemes of WT and hsf5/spermatozoa.

For (C) and (D), error bars indicate± SD; n = 8 biological replicates for (C) and 4 for (D); difference between groups was statistically examined with unpaired two-tailed Student’s t-test; *p < 0.05; ***p < 0.001. For (E)–(I), n = 4 biological replicates in each group. Scale bars: 20mm in (A); 10 mm in (B) and (E, left); 2 mm in (E, right) and (F); 0.2mm in (G); and 0.5 mm in (H).

(8)

by histological analysis (Figures 3A, 4A, and 4B). Detailed analysis of the localization of Sycp3 allowed us to differentiate between different stages of meiotic prophase in primary sper-matocytes, namely, pre-leptotene, leptotene and zygotene-pachytene. At the pre-leptotene stage, Sycp3 appeared as a small particle at one side of the cell (Figure 4E), in agreement with previously published reports (Saito et al., 2011). At the leptotene stage, Sycp3 staining seemed to highlight the bou-quet-shaped chromosomal arrangement (Figure 4F), whereas at the zygotene-pachytene stage, Sycp3 localization ap-peared reticulate, staining condensed chromosomes ( Fig-ure 4G). hsf5/mutant testes showed a lower number of cells at the pre-leptotene stage and significantly higher ones in the subsequent leptotene and zygotene-pachytene stages in comparison to WT (Figure 4H). In addition, the size of the cells at the above stages was also greater in hsf5/testes than WT (Figure S5), which might have resulted from defects in cell division.

Reduction in the number of post-meiotic cells and their aber-rant shape pointed to potential involvement of programmed cell death. Indeed, fluorescence-based TUNEL assay showed an increased number of apoptotic cells among germ cells in mu-tants than WT (Figures 4C and 4D).

Accumulation of the cells during meiotic leptotene and zygo-tene-pachytene stages and reduction of cells in post-meiotic stages suggest that Hsf5 is required for proper progression dur-ing meiotic prophase-I in spermatogenesis.

Hsf5 Has a Potentially Important Role in Regulating Cell Cycle and Apoptosis

Comparative transcriptome analyses of adult testes from the

hsf5/ mutant and WT individuals using RNA sequencing (RNA-seq) indicated significant differences (Figure 5A). Twelve percent (1,533/12,772) of the genes tested were differentially expressed in the mutant, suggesting that Hsf5 loss leads to a substantial change in the testicular transcriptome landscape (Figure 5B). Among the differentially expressed genes (DEGs) in hsf5/ testis, those showing downregulation were more abundant than upregulated ones (Table S4). The expression of a few hsp genes, such as hsp90b1, hsp70.1 and hsp90aa1, were marginally differentially expressed in hsf5/ mutants (Table S4), whereas others remained unchanged.

Gene ontology (GO) analysis revealed that 416 DEGs were classified under biological processes. Among all the categories, ‘‘protein modification’’ function was most abundant, followed by ‘‘negative regulation of biological process’’ and ‘‘apoptosis or Figure 4. hsf5/Testes Showed Higher Number of Primary Spermatocytes and Apoptotic Cells Compared to Wild-Type

(A and B) Representative images showing immunostaining of WT (A) and hsf5/testis sections (B) with anti-Sycp3 antibody. More spermatocytes were accumulated in the mutant. For (A) and (B), n = 4 biological replicates in each group.

(C and D) TUNEL staining of the wild-type (C) and hsf5/(D) testis, showed a higher number of apoptotic cells in the latter. n = 3 biological replicates. (E–G) High-resolution confocal images of anti-Sycp3 immunostaining in WT testis showed spermatocytes at different stages of meiotic prophase-1. Sycp3 appears as small particles at one side of the cell in pre-leptotene stage (E), bouquet-shaped chromosomal arrangement in leptotene stage (F), and in reticulate manner at zygotene-pachytene stage (G).

(H) Relative proportion of cells in the above three stages quantified from Sycp3-stained WT and hsf5/testes. Cell counts at preleptotene stage were signif-icantly lower, whereas those at the leptotene and zygotene-pachytene stages were higher in the mutant compared to WT. For (E)–(H), n = 5 biological replicates in each group.

Scale bars: 100mm in (A)–(D) and 10 mm in (E)–(G). Difference between WT and hsf5/was examined with Kolmogorov-Smirnov test, **p < 0.01. The error bars

(9)

regulation of cell death’’ (Figure 5C). Analysis of the biological function of upregulated and downregulated genes separately showed similar enrichment patterns. Upregulated genes showed enrichment of ‘‘negative regulation of cellular/biological pro-cess,’’ ‘‘cell death,’’ and ‘‘cell cycle’’ (Figure 5D; Table S5). Downregulated genes showed enrichment of ‘‘cell adhesion,’’ ‘‘cell cycle,’’ and ’’nervous system.’’ Such an increased expres-sion of apoptotic genes and negative regulators correlated with an increased level of apoptosis and failure of the cell cycle regu-lation in hsf5/mutants (Figure 4D).

Misregulation of genes involved in cell cycle and apoptosis likely explain the increase in number of primary spermatocytes and apoptosis during spermatogenesis. Among these genes, we interestingly observed upregulation of tp53, tp63, and other members of the tp53 pathway (Table S4). In addition to explaining defects in cell division, this also suggested major defects in the G1/S phase, along with misregulation of several of G1/S transition checkpoint proteins in hsf5/mutant testis (Tables S4andS5). Downregulated expression of important genes responsible for male sexual differentiation, e.g., amh, ddx, and fshr, also pointed to a function of Hsf5 in these processes.

Analysis of molecular function and localization of protein prod-ucts of DEGs revealed that 474 of them have been assigned mo-lecular function. GO analysis revealed that ‘‘catalytic activity,’’ ‘‘ion binding,’’ and ‘‘nucleotide binding’’ were the most abundant functions (Figure 5E). This was in agreement with maximum enrichment of genes with protein modification as their biological function.

Overall, GO analysis of testicular transcriptomes together with developmental defects in hsf5/testis (Figures 3and4) strongly

suggest that loss of Hsf5 results in misregulation of genes involved in signal transduction, apoptosis, and cell cycle regula-tion that likely monitor the compleregula-tion of early phases of meiosis during spermatogenesis.

DISCUSSION

Here, we describe a hitherto uncharacterized heat shock fac-tor, Hsf5 from zebrafish, which is required for spermatogen-esis. We speculate that Hsf5 might not play a significantly important role in heat shock response (Figure S1A), albeit we cannot rule out a modulatory function under certain stress conditions, as shown earlier for mammalian HSF2 (Jaeger et al., 2016; Ostling et al., 2007). The increased expression level of mammalian Hsf5 orthologs in testis during meiosis strongly suggests a conserved testis-specific function of this heat shock factor in the whole vertebrate clade (Chalmel et al., 2012). Although the hsf5 ortholog, as well as hsf1 and

hsf2, are transiently expressed in spermatocytes and

sperma-tids in mouse, only a combined loss of Hsf1 and Hsf2 leads to severe gonadal phenotype, including infertility due to abnormal sperm shape and reduced sperm numbers (Ji et al., 2012; Wang et al., 2004). Loss of zebrafish Hsf5 alone results in a similar phenotype, indicating its dedicated function in spermatogenesis. While Hsf5 is required for progression through meiotic prophase-1, we speculate that its loss could result in polyploidy (Revay et al., 2009) or defects in chromo-some packaging (Guthauser et al., 2011), both known to cause infertility. The increased size of sperm heads (Figure 3F) could be an indication of such problems.

Figure 5. The Transcriptome ofhsf5/Zebrafish Gonad Is Substantially Different from that of Wild-Type (A) Principal-component analysis (PCA) plot demonstrating the clustering of mutants further away from WT.

(B) MA plot representing the differentially expressed genes (DEGs; in red).

(C) Enrichment of biological processes among DEGs in hsf5/mutant testis, as shown by GO analysis.

(D) Representation of enrichment of biological function separately for genes with upregulated and downregulated expression level. (E) Enrichment of genes under molecular function among DEGs in hsf5/mutant testis, as shown by GO analysis.

(10)

The expression level of zebrafish hsf5 was the highest in the primary spermatocytes, and its localization was predominantly observed at cytoplasmic bridges (Figures 2A, 2B, 2J, and 2K). In mice and Drosophila, these structures connect germ cells and function as channels to transport gene products (Braun et al., 1989). Hsf5 localization as speckles in the nucleus of pri-mary spermatocytes suggests a potential role in gene regulation during meiosis-1 (Figures 2J and 2K). Accumulation of primary spermatocytes at the leptotene and zygotene-pachytene stages of hsf5/mutants along with increased size of these cells ( Fig-ures 3,4, andS5) point towards defects in cell cycle. A lower number of cells in the pre-leptotene stage in hsf5/ mutant might be the result of feedback from accumulated cells at later stages to retard entry of mature spermatogonia into a non-pro-gressive meiotic stage. These phenotypes underscore the vital role of Hsf5 for progression through meiotic prophase-1.

Misregulation of genes from different functional categories may explain our observations. The most important indicator of failure of cell cycle control was misregulation of genes involving monitoring G1/S transition, including tp53, tp63, and other pro-teins of the p53 pathway. Both tp53 and tp63 were upregulated along with G1/S regulators cdk5, rap3, and top2b. These pro-teins are involved in DNA surveillance, and their misregulation in-dicates failure of proper cell division in mutants and that molec-ular function of Hsf5 is likely to ensure proper progression through prophase-1.

Genes with biological functions ‘‘molecular function,’’ ‘‘cata-lytic activity,’’ and ‘‘ion binding’’ were predominant among DEGs in hsf5/mutant testis. The majority of their protein prod-ucts were serine - threonine kinases, phosphatases and cyto-skeleton-component binding enzymes, which are important reg-ulators of cell cycle and signal transduction (Figures 5C and 5E;

Table S5).

Cell cycle failure might have triggered increased apoptosis, which is reflected in the enrichment of apoptotic genes, including those in the p53 pathway, among DEGs (Figure 5C). Curiously, we also observed upregulation of many apoptosis inhibitory genes, such as faima, ppp1r13ba, clrn, and ppm1k, in the

hsf5/ mutant testis (Table S4). Upregulated expression of these genes might be required to support accumulated primary spermatocytes in the hsf5/mutant testis (Figures 3,4B, and

S5). In addition to apoptosis, other factors (e.g., upregulated autophagy and ubiquitination) might also contribute to the desta-bilization of spermatogenetic processes, leading to the degrada-tion of cells.

We could not attribute downregulation of important testicular development genes, like amh, ddx4 and fshr, as a direct conse-quence of Hsf5 loss of function. Severe developmental abnor-mality in testicular cell types in hsf5/ mutant testis could be a reason for their misregulation. Interestingly, rho kinase

rock2, misregulated in hsf5/testis, was shown to be required for proper localization of germplasm RNAs (Miranda-Rodrı´guez et al., 2017).

Differences in the cellular composition between WT and

hsf5/mutant gonads are likely to contribute substantially to the gene expression landscapes and DEGs. As our data on DEGs are from full gonads, they cannot be broken down to cell-type levels. Cell-type-specific gene expression data would

be necessary to differentiate between DEGs resulting directly from Hsf5 loss of function and those that are caused indirectly by the changes in the proportion of cell types.

We describe the functional characterization of heat shock factor 5, with bona fide function in germ cell development and a critical role in meiotic progression in zebrafish males. Future analysis of its interacting partners and the effect of its loss on transcription, splicing, and the epigenetic state of genes in germ cells might provide more information about additional functions.

STAR+METHODS

Detailed methods are provided in the online version of this paper and include the following:

d KEY RESOURCES TABLE

d CONTACT FOR REAGENT AND RESOURCE SHARING d EXPERIMENTAL MODEL AND SUBJECT DETAILS

B Zebrafish strain

d METHOD DETAILS

B Identification of hsf5 and its two isoforms B Bioinformatic analysis of Hsf5

B Sampling and isolation of nucleic acids B Gene expression analyses

B Characterization of anti-Hsf5 antibody B Immunostaining and data analysis B Generation and screening of mutants B Propagation and genotyping of mutants B Histological and EM analysis of gonads B Sperm quantification and TUNEL assay B RNA sequencing and transcriptome analysis

d QUANTIFICATION AND STATISTICAL ANALYSIS d DATA AND SOFTWARE AVAILABILITY

SUPPLEMENTAL INFORMATION

Supplemental Information includes five figures and five tables and can be found with this article online athttps://doi.org/10.1016/j.celrep.2018.11.090.

ACKNOWLEDGMENTS

This research was supported by the National Research Foundation, Prime Minister’s Office, Singapore under its Competitive Research Programme (award NRF-CRP7-2010-001 to L.O.), the National Research, Development and Innovation Office of Hungary through its Frontline Research Grant (award KKP 126764 to L.O.), internal research grants from Temasek Life Sciences Laboratory (to L.O.), and the Swedish Research Council, Knowledge Founda-tion, Sweden, and O¨ rebro University (to P.-E.O.). The authors thank Kellee Siegfried, Toshihiro Kawasaki, Haiwei Song, Bharath SR, Xuhua Tang, Sza-bolcs Nagy, and Andra´s Orosz for their advice and Terence Goh for his help with the graphical abstract. The tg(ddx4:ddx4-EGFP) zebrafish line and the anti-Sycp3 antibody were kind gifts from Lisbeth Olsen and Noriyoshi Sakai, respectively.

AUTHOR CONTRIBUTIONS

L.O., J.M.S., and M.S.H. conceived the study; J.M.S., A.A., P.-E.O., and L.O. designed the experiments; J.M.S. generated the mutants; J.M.S., A.A., M.S.H., A.P., W.C.L., N.M.T., and L.S.E.T. performed the experiments and/ or analyzed the data: J.M.S., M.S.H., A.P., W.C.L., N.M.T., L.S.E.T., A.A.,

(11)

P.-E.O., and L.O. discussed and interpreted the data; and J.M.S., A.A., W.C.L., P.-E.O., and L.O. wrote and corrected the manuscript.

DECLARATION OF INTERESTS

The authors declare no competing interests. Received: February 7, 2018

Revised: August 24, 2018 Accepted: November 26, 2018 Published: December 18, 2018

REFERENCES

Abane, R., and Mezger, V. (2010). Roles of heat shock factors in gametogen-esis and development. FEBS J. 277, 4150–4172.

A˚kerfelt, M., Morimoto, R.I., and Sistonen, L. (2010). Heat shock factors: inte-grators of cell stress, development and lifespan. Nat. Rev. Mol. Cell Biol. 11, 545–555.

Alastalo, T.P., Lo¨nnstro¨m, M., Leppa¨, S., Kaarniranta, K., Pelto-Huikko, M., Sistonen, L., and Parvinen, M. (1998). Stage-specific expression and cellular localization of the heat shock factor 2 isoforms in the rat seminiferous epithe-lium. Exp. Cell Res. 240, 16–27.

Anders, S., Pyl, P.T., and Huber, W. (2015). HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169.

Baek, M., Park, T., Heo, L., Park, C., and Seok, C. (2017). GalaxyHomomer: a web server for protein homo-oligomer structure prediction from a monomer sequence or structure. Nucleic Acids Res. 45, W320–W324.

Benson, D.A., Cavanaugh, M., Clark, K., Karsch-Mizrachi, I., Ostell, J., Pruitt, K.D., and Sayers, E.W. (2018). GenBank. Nucleic Acids Res. 46, D41–D47.

Braun, R.E., Behringer, R.R., Peschon, J.J., Brinster, R.L., and Palmiter, R.D. (1989). Genetically haploid spermatids are phenotypically diploid. Nature 337, 373–376.

Chalmel, F., Lardenois, A., Evrard, B., Mathieu, R., Feig, C., Demougin, P., Gat-tiker, A., Schulze, W., Je´gou, B., Kirchhoff, C., and Primig, M. (2012). Global human tissue profiling and protein network analysis reveals distinct levels of transcriptional germline-specificity and identifies target genes for male infer-tility. Hum. Reprod. 27, 3233–3248.

Dahm, R., and Geisler, R. (2006). Learning from small fry: the zebrafish as a ge-netic model organism for aquaculture fish species. Mar. Biotechnol. (NY) 8, 329–345.

Dayalan Naidu, S., and Dinkova-Kostova, A.T. (2017). Regulation of the mammalian heat shock factor 1. FEBS J. 284, 1606–1627.

Dobin, A., Davis, C.A., Schlesinger, F., Drenkow, J., Zaleski, C., Jha, S., Batut, P., Chaisson, M., and Gingeras, T.R. (2013). STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21.

Fiorenza, M.T., Farkas, T., Dissing, M., Kolding, D., and Zimarino, V. (1995). Complex expression of murine heat shock transcription factors. Nucleic Acids Res. 23, 467–474.

Fujimoto, M., and Nakai, A. (2010). The heat shock factor family and adaptation to proteotoxic stress. FEBS J. 277, 4112–4125.

Gomez-Pastor, R., Burchfiel, E.T., and Thiele, D.J. (2018). Regulation of heat shock transcription factors and their roles in physiology and disease. Nat. Rev. Mol. Cell Biol. 19, 4–19.

Guthauser, B., Albert, M., Ferfouri, F., Ray, P.F., Rabiey, G., Selva, J., and Vi-alard, F. (2011). Inverse correlation between chromatin condensation and sperm head size in a case of enlarged sperm heads. Reprod. Biomed. Online

23, 711–716.

Hossain, M.S. (2010). Molecular analyses of gonad differentiation and function in zebrafish. PhD thesis (The National University of Singapore).

Izu, H., Inouye, S., Fujimoto, M., Shiraishi, K., Naito, K., and Nakai, A. (2004). Heat shock transcription factor 1 is involved in quality-control mechanisms in male germ cells. Biol. Reprod. 70, 18–24.

Jaeger, A.M., Pemble, C.W., 4th, Sistonen, L., and Thiele, D.J. (2016). Struc-tures of HSF2 reveal mechanisms for differential regulation of human heat-shock factors. Nat. Struct. Mol. Biol. 23, 147–154.

Ji, Z.-L., Duan, Y.-G., Mou, L.-S., Allam, J.-P., Haidl, G., and Cai, Z.-M. (2012). Association of heat shock proteins, heat shock factors and male infertility. Asian Pac. J. Reprod. 1, 76–84.

Kallio, M., Chang, Y., Manuel, M., Alastalo, T.P., Rallu, M., Gitton, Y., Pirkkala, L., Loones, M.T., Paslaru, L., Larney, S., et al. (2002). Brain abnormalities, defective meiotic chromosome synapsis and female subfertility in HSF2 null mice. EMBO J. 21, 2591–2601.

Kogo, H., Kowa-Sugiyama, H., Yamada, K., Bolor, H., Tsutsumi, M., Ohye, T., Inagaki, H., Taniguchi, M., Toda, T., and Kurahashi, H. (2010). Screening of genes involved in chromosome segregation during meiosis I: toward the iden-tification of genes responsible for infertility in humans. J. Hum. Genet. 55, 293–299.

Krøvel, A.V., and Olsen, L.C. (2002). Expression of a vas:EGFP transgene in primordial germ cells of the zebrafish. Mech. Dev. 116, 141–150.

Leal, M.C., Cardoso, E.R., No´brega, R.H., Batlouni, S.R., Bogerd, J., Franc¸a, L.R., and Schulz, R.W. (2009). Histological and stereological evaluation of ze-brafish (Danio rerio) spermatogenesis with an emphasis on spermatogonial generations. Biol. Reprod. 81, 177–187.

Li, Y., Chia, J.M., Bartfai, R., Christoffels, A., Yue, G.H., Ding, K., Ho, M.Y., Hill, J.A., Stupka, E., and Orban, L. (2004). Comparative analysis of the testis and ovary transcriptomes in zebrafish by combining experimental and computa-tional tools. Comp. Funct. Genomics 5, 403–418.

Loreng, T.D., and Smith, E.F. (2017). The central apparatus of cilia and eukary-otic flagella. Cold Spring Harb. Perspect. Biol. 9, a028118.

Love, M.I., Huber, W., and Anders, S. (2014). Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550.

Miranda-Rodrı´guez, J.R., Salas-Vidal, E., Lomelı´, H., Zurita, M., and Schnabel, D. (2017). RhoA/ROCK pathway activity is essential for the correct localization of the germ plasm mRNAs in zebrafish embryos. Dev. Biol. 421, 27–42.

Nakai, A., Suzuki, M., and Tanabe, M. (2000). Arrest of spermatogenesis in mice expressing an active heat shock transcription factor 1. EMBO J. 19, 1545–1554.

Ostling, P., Bjo¨rk, J.K., Roos-Mattjus, P., Mezger, V., and Sistonen, L. (2007). Heat shock factor 2 (HSF2) contributes to inducible expression of hsp genes through interplay with HSF1. J. Biol. Chem. 282, 7077–7086.

Ozaki, Y., Saito, K., Shinya, M., Kawasaki, T., and Sakai, N. (2011). Evaluation of Sycp3, Plzf and Cyclin B3 expression and suitability as spermatogonia and spermatocyte markers in zebrafish. Gene Expr. Patterns 11, 309–315.

Park, C.J., and Seo, Y.S. (2015). Heat shock proteins: A review of the molecular chaperones for plant immunity. Plant Pathol. J. 31, 323–333.

Ran, F.A., Hsu, P.D., Wright, J., Agarwala, V., Scott, D.A., and Zhang, F. (2013). Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281– 2308.

Revay, T., Nagy, S., Kopp, C., Flyckt, A., Rens, W., Rath, D., Hidas, A., Kovacs, A., Johannisson, A., Rodriguez-Martinez, H., and Andersson, M. (2009). Mac-rocephaly in bull spermatozoa is associated with nuclear vacuoles, diploidy and alteration of chromatin condensation. Cytogenet. Genome Res. 126, 202–209.

Robinson, M.D., and Oshlack, A. (2010). A scale normalization method for dif-ferential expression analysis of RNA-seq data. Genome Biol. 11, R25.

Saito, K., Siegfried, K.R., N€usslein-Volhard, C., and Sakai, N. (2011). Isolation and cytogenetic characterization of zebrafish meiotic prophase I mutants. Dev. Dyn. 240, 1779–1792.

Salmand, P.A., Jungas, T., Fernandez, M., Conter, A., and Christians, E.S. (2008). Mouse heat-shock factor 1 (HSF1) is involved in testicular response to genotoxic stress induced by doxorubicin. Biol. Reprod. 79, 1092–1101.

Sarge, K.D., Park-Sarge, O.-K., Kirby, J.D., Mayo, K.E., and Morimoto, R.I. (1994). Expression of heat shock factor 2 in mouse testis: potential role as a regulator of heat-shock protein gene expression during spermatogenesis. Biol. Reprod. 50, 1334–1343.

(12)

Sievers, F., Wilm, A., Dineen, D., Gibson, T.J., Karplus, K., Li, W., Lopez, R., McWilliam, H., Remmert, M., So¨ding, J., et al. (2011). Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega. Mol. Syst. Biol. 7, 539.

Sreenivasan, R., Cai, M., Bartfai, R., Wang, X., Christoffels, A., and Orban, L. (2008). Transcriptomic analyses reveal novel genes with sexually dimorphic expression in the zebrafish gonad and brain. PLoS ONE 3, e1791.

Swan, C.L., Evans, T.G., Sylvain, N., and Krone, P.H. (2012). Zebrafish HSF4: a novel protein that shares features of both HSF1 and HSF4 of mammals. Cell Stress Chaperones 17, 623–637.

Tamura, K., Peterson, D., Peterson, N., Stecher, G., Nei, M., and Kumar, S. (2011). MEGA5: molecular evolutionary genetics analysis using maximum like-lihood, evolutionary distance, and maximum parsimony methods. Mol. Biol. Evol. 28, 2731–2739.

van der Ven, L., and Wester, P. (2017). Histology and Histopathology Atlas of the Zebrafish V2.01,http://zfin.org/hh_atlas/.

Wang, X.G., and Orban, L. (2007). Anti-M€ullerian hormone and 11 beta-hy-droxylase show reciprocal expression to that of aromatase in the transforming gonad of zebrafish males. Dev. Dyn. 236, 1329–1338.

Wang, G., Zhang, J., Moskophidis, D., and Mivechi, N.F. (2003). Targeted disruption of the heat shock transcription factor (hsf)-2 gene results in increased embryonic lethality, neuronal defects, and reduced spermatogen-esis. Genesis 36, 48–61.

Wang, G., Ying, Z., Jin, X., Tu, N., Zhang, Y., Phillips, M., Moskophidis, D., and Mivechi, N.F. (2004). Essential requirement for both hsf1 and hsf2 transcrip-tional activity in spermatogenesis and male fertility. Genesis 38, 66–80.

Wang, X.G., Bartfai, R., Sleptsova-Freidrich, I., and Orban, L. (2007). The timing and extent of ‘juvenile ovary’ phase are highly variable during zebrafish testis differentiation. J. Fish Biol. 70, 33–44.

Westerfield, M. (1995). The zebrafish book. A guide for the laboratory use of zebrafish (Danio rerio) (Eugene: University of Oregon Press).

Wittig, I., Braun, H.-P., and Scha¨gger, H. (2006). Blue native PAGE. Nat. Pro-toc. 1, 418–428.

Yeh, F.L., Hsu, L.Y., Lin, B.A., Chen, C.F., Li, I.C., Tsai, S.H., and Hsu, T. (2006). Cloning of zebrafish (Danio rerio) heat shock factor 2 (HSF2) and similar pat-terns of HSF2 and HSF1 mRNA expression in brain tissues. Biochimie 88, 1983–1988.

Zhang, Y., Huang, L., Zhang, J., Moskophidis, D., and Mivechi, N.F. (2002). Targeted disruption of hsf1 leads to lack of thermotolerance and defines tis-sue-specific regulation for stress-inducible Hsp molecular chaperones. J. Cell. Biochem. 86, 376–393.

Zhao, S., and Fernald, R.D. (2005). Comprehensive algorithm for quantitative real-time polymerase chain reaction. J. Comput. Biol. 12, 1047–1064.

(13)

STAR+METHODS

KEY RESOURCES TABLE

CONTACT FOR REAGENT AND RESOURCE SHARING

Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, La´szlo´ Orba´n (orban@georgikon.hu).

EXPERIMENTAL MODEL AND SUBJECT DETAILS Zebrafish strain

This study and all procedures were approved by Temasek Life Sciences Laboratory Institutional Animal Care and Use Committee (approval ID: TLL(F)-10-002) for experiments carried out at Temasek Life Sciences Laboratory (License for Animal Research Facility No. VR016) and O¨ rebro University by Linko¨pings djurfo¨rso¨ksetiska na¨mnd (Linbko¨pings Animal Care and Use Committee, Approval ID: 32-10) for experiments carried out at O¨ rebro University (License for Animal Research Facility: No. 5.2.18-2863/13).

REAGENT or RESOURCE SOURCE IDENTIFIER Antibodies

Rabbit anti-Hsf5 This study N/A

Chicken anti-Ddx4 Abcam Cat. # ab13970; RRID: AB_300798 Rabbit anti-Sycp3 Saito et al., 2011 N/A

Rabbit anti-fluorescein ThermoFisher Scientific Cat. # A889; RRID: AB_221561 Alexa fluor 488 anti-rabbit ThermoFisher Scientific Cat. # A32731; RRID: AB_2633280 Alexa fluor 555 anti-rabbit ThermoFisher Scientific Cat. #A27039; RRID: AB_2536100 Alexa fluor 488 anti-chicken ThermoFisher Scientific Cat. # A11039; RRID: AB_2534096 Critical Commercial Assays

TruSeq RNA Library Preparation Illumina Cat. #20020598

In situ cell- death detection kit fluorescein V17 Roche Diagnostics Cat. # 1684795910

Deposited Data

RNA seq This study SRP124146 Experimental Models: Organisms/Strains

Danio rerio, AB strain Orban lab, Fish facility N/A

Tg(ddx4:ddx4-EGFP) Krøvel and Olsen, 2002 N/A

Danio rerio, Hsf5sg40 This study N/A

Danio rerio, Hsf5sg41 This study N/A

Danio rerio, Hsf5sg42 This study N/A

Oligonucleotides

SeeTable S1 This study N/A

gRNA target 50TACAATCCCAACTTCAGACGAGG 30 ToolGen Inc (Seoul,Korea) N/A Recombinant DNA

pGEM-T Easy Vector Promega Cat.# A1360 Software and Algorithms

ImageJ NIH https://fiji.sc/

CLUSTAL OMEGA Sievers et al., 2011 N/A MEGA Version 5 Tamura et al,. 2011 N/A Galaxy Homomer Baek et al., 2017 N/A Imaris www.bitplane.com N/A GeneMapper Applied Biosystem N/A bioconductoR Robinson and Oshlack, 2010 N/A

(14)

Zebrafish (Danio rerio, AB strain) were raised, maintained and crossed according to the standard protocol (Westerfield, 1995). Gonadal samples of different developmental stages were collected from a tg(ddx4:ddx4-egfp) transgenic zebrafish line (Krøvel and Olsen, 2002). Fish were reared in AHAB recirculation systems (Aquatic Habitats, Apopka, FL, USA) at ambient temperature (26-28C).

METHOD DETAILS

Identification of hsf5 and its two isoforms

The consensus hsf5 sequence (1,581bp) was derived from an expressed sequence tag (EST) showing testis-enhanced expression from our adult zebrafish testis cDNA library (Li et al., 2004; Sreenivasan et al., 2008) and seven other zebrafish testis-derived ESTs from GenBank (Benson et al., 2018). The consensus sequence was confirmed by reverse transcription PCR (RT-PCR) using total RNA extracted from adult testis (for primers sequences please seeTable S1). To obtain the complete hsf5 cDNA sequence, rapid ampli-fication of cDNA ends (RACE) were performed using RLM-RACE kit (Ambion) leading to the identiampli-fication of a full-length 1.7 kb tran-script (FJ969446, NM_001089476). The coding region of zebrafish hsf5 cDNA was amplified from various organs and different devel-opmental stages and sequenced for verification. Expression of hsf5 transcript variants was analyzed by RT-PCR using the primer pair hsf5_FL (seeTable S1for primer sequences) that amplified both hsf5_tv1 and hsf5_tv2. eef1a1l1 was used as the reference gene. Bioinformatic analysis of Hsf5

The Hsf5 protein was analyzed using the following softwares: Conserved Domain Database (CDD) and SMART software (both avail-able at:https://expasy.org/). The full-length amino acid sequence of heat shock factor orthologs were retrieved from GenBank (Table S2). Sequences were aligned by CLUSTAL OMEGA software (Sievers et al., 2011). Estimation of molecular phylogeny was carried out by neighbor-joining method with Poisson correction model as implemented in MEGA Version 5 (Tamura et al., 2011). Confidence in the phylogeny was assessed by bootstrap re-sampling of the data (1000). We used ExPASy (Expert Protein Analysis System,https:// expasy.org/), a proteomics server to analyze protein sequences and structures and PrepCalc (PrepCalc.com) to calculate the mo-lecular weight and physiochemical properties of Hsf5. Momo-lecular weight was calculated as 47254.22 g/mol (47kDa). This protein has an isoelectric point of pH 7.34 carrying a net charge at pH 7.14 and it was estimated to be soluble in water. Analysis Hsf5 amino-acid sequence using Galaxy homomer (Baek et al., 2017) predicted the oligomeric state of Hsf5 as a dimer in all 5 structural models using a similarity-based approach.

Sampling and isolation of nucleic acids

For the analysis of early gene expression, RNA was extracted from pooled zebrafish embryos (30 individuals/pool). In order to inves-tigate the expression of hsf5 during gonad development, samples from the isolated gonads (20, 25, 30, 35 and 40 dpf) were collected from a total of six tg(ddx4:ddx4-egfp) individuals for each time point. Previously, our lab showed that those transgenic individuals that did not seem to show visually detectable Egfp signal during their early gonadal development (20-24 dpf) became exclusively males (Wang et al., 2007). Therefore, we considered such individuals as presumptive males (four samples), whereas individuals with strong Egfp signal were considered as presumptive females (four samples). For spatial analysis of hsf5 expression profiles in adult zebrafish, samples from nine different organs (testis, ovary, kidney, liver, brain, gut, gill, skin and eye) were collected from three individuals. For heat shock experiment, two groups were formed from adult male siblings. The first group was heat-treated for one hour at 37C, whereas the second group was kept at ambient temperature (26-28C). Total RNAs were extracted using RNeasy RNA extraction kit (QIAGEN) according to the manufacturer’s instructions.

Gene expression analyses

Expression analyses of hsf5 during early embryonic development, gonad development and in adult tissues were performed by using real-time quantitative PCR (qPCR) using the iCycler iQ Real-time Detection system and SYBR Green chemistry (Bio-Rad). Samples were assayed in triplicate and each experiment had at least three biological replicates. To normalize the expression, several house-keeping genes (including actb1, rpl13 and eef1a1l1) were assayed. In the present study, we used eef1a1l1 for early developmental stages and adult tissues, while rpl13 was used as a reference for the developing gonads. The efficiency of each reaction was calcu-lated using PCR miner (Zhao and Fernald, 2005). The relative gene expression level was determined using the delta-CT method and is presented as a log2 of relative quantity (RQ) of the target gene.

Characterization of anti-Hsf5 antibody

An antibody was raised against the C-terminal region (amino acids 403-416) of the zebrafish Hsf5 protein by Agrisear AB (Va¨nna¨s, Sweden). This region is not conserved among the tested Hsf5 orthologs in vertebrates. The specificity of the antibody to recognize native Hsf5 protein was demonstrated through ELISA assays, Blue Native PAGE, western blot and immunohistochemistry on zebra-fish gonads. For ELISA 96 well plates were coated with 4mg/ml of peptide. Rabbit immune serum, pre-immune serum and purified anti-Hsf5 antibody were diluted and incubated for 1 hr at room temperature. Rabbit secondary antibody at 1:8000 dilution was used and EC-Blue substrate system (Medicago AB) was used for reaction development. The plate was read in spectrophotometer at 650 nm. Western blot was performed on whole testis tissue lysate samples for WT and mutants. Following SDS-PAGE, proteins

(15)

were transferred onto PVDF blotting membrane and blocked with 3% non-fat milk for 2 hours at room temperature. The membrane was incubated with anti-Hsf5 antibody at a dilution of 1:5000 overnight at 4C, washed multiple times and incubated with anti-rabbit secondary antibody (P0448, DAKO) at a dilution of 1:6000 for 2 hours at room temperature. Following washing, the membrane was incubated with Clarity Western ECL Substrate (Bio-Rad) and imaged using ChemiDoc Touch Gel Imaging System (BIo-Rad). Testis tissue for Blue Native PAGE was prepared as described previously (Wittig et al., 2006). Immunoblotting conditions were kept same as for western blots.

Immunostaining and data analysis

For immunohistochemical (IHC) analyses, testes and ovaries from adult zebrafish (90 dpf) were isolated and fixed in 4% paraformal-dehyde in PBS pH 7.4 (Sigma) for 2 hours (hr) at room temperature and washed three times with PBS and equilibrated in graded sucrose series. Samples were frozen in tissue freezing media (Leica Biosystems), and 5-7mm sections were cut using a cryotome (Leica Biosystems). After blocking in 3% BSA, the sections were then incubated either with anti-Hsf5, anti-Sycp3 antibody or with anti-Hsf5 and anti-Ddx4 for 16 hr at

4C at 1:1000 (anti-Hsf5 and anti-Ddx4) and 1:400 (anti-Sycp3) dilutions, in PBS containing 1% BSA and 0.1% Triton X. Alexa Flour 488 anti-rabbit / Alexa Flour 555 anti-rabbit / Alexa Flour 488 anti-chicken (Invitrogen) at 1:1,000 dilutions were used as a secondary antibody to incubate for 2 hr at room temperature followed by DAPI (Calbiochem) staining for 5 mins. The image was captured using a Leica SP8 Confocal Laser Scanning Biological Microscope. Randomly chosen images stained with Sycp3 from five biological rep-licates were used for counting different cell types in meiotic prophase stages. Spermatocytes at different phases of meiosis were identified by nuclear characteristics, such as size, chromosome condensation, and meiotic figures of the chromosomes using anti-Sycp3 antibody staining. Nuclei were stained by DAPI. Sycp3-stained WT and hsf5/mutant testis images were acquired using Lecia SP8 confocal. We used these data to count cells at different stages of meiosis-1 in five WT and mutant testis, using segmen-tation in Imaris software (www.bitplane.com).

Generation and screening of mutants

Custom-designed guide RNA (gRNA) targeting the DNA Binding Domain at second exon of hsf5 and recombinant Cas9 protein (Streptococcus pyogenes) were manufactured by ToolGen Inc (Seoul, Korea). gRNA was designed using CRISPR design tool (http://zlab.bio/guide-design-resources) and off-target analysis using RGEN Tools by Seoul University (http://www.rgenome.net/ cas-offinder). gRNA and Cas9 protein were co-injected into 350 one-cell stage zebrafish embryos in three separate experiments. Each embryo was injected with 2 nl of solution containing 12.5 ng/ml of sgRNA and 300 ng/ml of Cas9 protein. Injected embryos were grown to 60 dpf for fin clipping. A total of 57 F0 founder individuals at three months post-fertilization (mpf) were screened by fluorescent genotyping and 16 of them by T7E1 assay (for typical results seeFigures S3B and S3E–S3G). The genomic region sur-rounding the CRISPR target site was PCR-amplified using primer pair ‘hsf5a’ (seeTable S1for primer sequences) and cloned into a pGEMT Easy vector (Promega) and sequenced. T7E1 assay was carried out using T7E1 assay kit (NEB) following the manufacturer’s recommended protocol. Relative proportion of mutation was estimated by the formula of Indel % = 100 x (1-O(1-fcut)) as described in (Ran et al., 2013).

Propagation and genotyping of mutants

F0 mosaic founders were raised to sexual maturity and crossed with wild-type (WT) partners to generate F1. The resulting embryos were grown to three mpf, genomic DNA was isolated from tail fin biopsies and subjected to genotyping to identify the mutants. Heterozygous mutants from F1 were crossed to generate F2 for subsequent analyses. For genotyping, the forward primer of ‘hsf5b’ primer pair (seeTable S1for primer sequences) was labelled with FAM fluorescent dye on the 50end and PCR was performed in a 25 ul volume to amplify a 267 bp fragment spanning the targeted region of the hsf5 locus. PCR products were mixed with internal size standard, GeneScan 500 Rox (Applied Biosystems) and subjected to capillary electrophoresis using 3730xl DNA analyzer (Applied Biosystem). The genotypes of mutants were analyzed using GeneMapper software version 5.0 (Applied Biosystem). For fertility tests, twelve WT male and twelve WT female zebrafish were selected as potential partners for pairwise crossing. The total number of eggs produced and the survival rate at 24 hours post-fertilization (hpf) was recorded for three to six rounds of breeding. After that, six and three WT females with consistent production of good quality eggs were selected to be paired with hsf5/and

hsf5+/ male partners, respectively. Eight WT males were paired with hsf5/females and the transparent eggs from all were collected separately after three hpf to determine the survival rate at 24 hpf.

Histological and EM analysis of gonads

Male zebrafish were euthanized with ethyl 3-aminobenzoate methanesulfonate (Sigma) and their testicular tissues were fixed in 4% formaldehyde at 4C overnight. After dehydration, samples were embedded in plastic resin (Leica). Serial cross-sections of 2mm were cut by a microtome (Leica), dried on slides at 42C overnight, stained with hematoxylin and eosin, and then mounted in Per-mount (Fisher-Brand) and imaged with phase contrast microscope with a 63x and 100x oil objective lens. Ten sections each from eight WT and hsf5/mutant testes were chosen randomly and scored using high-resolution light microscope (Leica) at a magnifi-cation of3 100. Staging of germ cells was performed as described previously (Leal et al., 2009), using collection of images explaining the histology and toxicological pathology of the zebrafish (van der Ven and Wester, 2017) as a reference using ImageJ thresholding.

(16)

For Scanning Electron Microscopy (SEM), three WT and mutant sperm specimens each were fixed using 2.5% glutaraldehyde, de-hydrated in ascending graded ethanol and dropped onto a round glass slide and dried using a silica gel drier. After the surface was treated with electric conduction, the specimens were observed under a Jeol JSM-6360LV scanning electron microscope (Joel). For Transmission Electron Microscopy (TEM), samples (n = 4 individuals) were fixed with 2.5% glutaraldehyde in 0.1 M phosphate buffer (pH 7.2), washed 3X in 0.1 M phosphate buffer (pH 7.2) for 15 minutes each dehydrated in graded series of alcohol (in water) for 15 mi-nutes each, dried at critical point, mounted on specimen stub with silver paste, sputter coated with gold and imaged under a Jeol JEM-1230 Transmission Electron Microscope (Jeol).

Sperm quantification and TUNEL assay

Sperm counts were determined by haemocytometer. Sperm suspension stock from four WT and hsf5/mutant each were diluted 5,000 times before counting with the haemocytometer under light microscope at 20X magnification.

TUNEL staining was performed on mutant and WT testis fixed in 4% paraformaldehyde, embedded in paraffin. Cross sections of 5mm were rehydrated and a commercial in situ cell- death detection kit fluorescein (Roche Diagnostics) was used for labelling the slides. To preserve the tissue morphology while showing the TUNEL labelling, we used signal amplification incorporating secondary detection. The sections were then incubated with anti-fluorescein antibody (Invitrogen) for 16 hr at 4C at 1:400 dilution in PBS con-taining 1% BSA and 0.1% Triton X. Alexa Flour 488 anti-rabbit (Invitrogen) at 1:1000 dilutions were used as a secondary antibody for 2 hr at RT followed by DAPI (Calbiochem) staining for 5 mins. The image was captured using SP8 gSTED Confocal Laser Scanning Biological Microscope (Leica). Gonado-somatic indices (GSI) were calculated for WT and hsf5/mutant. We compared eight bio-logical replicates and examined the significance of the difference between the mutant and WT, using Student’s t-test.

RNA sequencing and transcriptome analysis

RNA was extracted from the intact testes of five adult hsf5/and WT siblings (at five mpf of age) using an Ambion RNAqueous-micro kit (Thermo Fisher Scientific). Sequencing libraries were constructed using TruSeq RNA Library Prep Kit v2 (Illumina) following the manufacturer’s instructions and sequenced on NextSeq 500 (Illumina). Mapping was performed on GRCz10 genome assembly using STAR v2.5.3a, using the soft-clipping option (Dobin et al., 2013). The bam files produced by STAR were then used as input for HTSeq-count (Anders et al., 2015), using GRCz10 gtf file from Ensembl (www.ensembl.org), to count the number of reads mapping to each gene, in intersection-nonempty mode. The files having counts for each gene thus produced by HTSeq-count were used for analyzing differentially expressed genes using the DESeq2 statistical package from bioconductoR (Love et al., 2014). To reduce the noise created by less significant variations, we also used log2 fold shrinkage. We selected the genes which have both p-value and false discovery rate less than 0.05, as differentially expressed (DEGs). Principal component analysis was done using the DESeq2 utility. The MA plot for the DEGs was drawn from the DESeq2 results. DEGs produced from the DESeq2 were subjected to further analysis using the GOSeq package, available on bioconductoR (Robinson and Oshlack, 2010). We only looked at the biological process of those DEGs, for which the p-value was less than 0.05.

QUANTIFICATION AND STATISTICAL ANALYSIS

For qPCR data analysis, mean± standard deviation was calculated. Statistical differences in relative mRNA expression between experimental groups were assessed by 2 tailed Student’s t-test. Differences were considered statistically significant at * p < 0.05; ** p < 0.01; and *** p < 0.001. We used Kolmogorov-Smirnov test to determine the significance in the differences between the cell number and area at meiotic prophase-I. The number of replicates for each experiment is detailed in figure caption forFigures 3,

4,S1,S3, andS5and corresponding methods sections. DATA AND SOFTWARE AVAILABILITY

References

Related documents

Further optimization of crystallization conditions and making new constructs with shorter linker between MBP and the target protein can help in getting better crystals that can be

3.4 SDS-PAGE analysis of purified fractions of MBP-HSF1-232 fusion protein after amylose affinity chromatography. The first lane is the marker, second one is the crude extract and

Stöden omfattar statliga lån och kreditgarantier; anstånd med skatter och avgifter; tillfälligt sänkta arbetsgivaravgifter under pandemins första fas; ökat statligt ansvar

Interestingly, germ cells from both the 1- and 7-year-old samples consisted of only undifferentiated spermatogonia (corresponding to States 0–1) (Figures 2A, 2D, and 2E) that

To evaluate the contribution of different factors (expression levels, duplication status [i.e., paralog or single-copy gene], evolu- tionary time [duplication or speciation age])

The overall aim of the studies performed within the frame of the present thesis was to examine the expression of four heat shock proteins (HSPs) in exercised human skeletal muscle

(PAPER IV (54)): This paper provides evidence for further links between protein oxidation and Hsps by showing that induction of the heat shock regulon in response to

But nor would it make any sense producing a forgery in the first place, as a modern copy painted in a modern style would be much higher valued than any worned-out original, which