Histone modifications regulate chromatin remodeling and gene expression in development and diseases. DOT1L, the sole histone H3K79 methyltransferase, is essential for embryonic development. Here, we report that DOT1L regulates male fertility in mouse. DOT1L associates with MLLT10 in testis. DOT1L and MLLT10 localize to the sex chromatin in meiotic and post-meiotic germ cells in an inter-dependent manner. Loss of either DOT1L or MLLT10 leads to reduced testis weight, decreased sperm count and male subfertility. H3K79me2 is abundant in elongating spermatids, which undergo the dramatic histone-to-protamine transition. Both DOT1L and MLLT10 are essential for H3K79me2 modification in germ cells. Strikingly, histones are substantially retained in epididymal sperm from either DOT1L- or MLLT10-deficient mice. These results demonstrate that H3K79 methylation promotes histone replacement during spermiogenesis.

Spermatogenesis is a complex developmental process that produces sperm. It consists of three phases: mitotic, meiotic and haploid. During the mitotic phase, spermatogonial stem cells renew to sustain life-long production of sperm, then differentiate and enter meiosis. During meiosis, homologous chromosomes pair and undergo meiotic recombination. Meiotic germ cells undergo one round of DNA replication but two successive rounds of cell divisions, resulting in haploid round spermatids. During the haploid phase (i.e. spermiogenesis), round spermatids undergo dramatic structural changes to become spermatozoa, including nuclear elongation and condensation, histone-to-protamine replacement, formation of flagella and shedding of the cytoplasm.

Dramatic chromatin remodeling occurs during spermiogenesis. Most histones in spermatids are replaced by sperm-specific transition proteins and then by sperm-specific protamines (Rousseaux et al., 2008). Protamines are highly basic proteins of lower molecular weight than histones. As a result, the histone-to-protamine replacement causes much more compact chromatin in sperm. Global histone acetylation precedes histone replacement (Meistrich et al., 1992; Oliva and Mezquita, 1982). GCN5, a histone acetyltransferase, acetylates histones in spermatids (Luense et al., 2019). BRDT, a testis-specific bromodomain protein, binds to acetylated histones to induce reorganization of chromatin when ectopically expressed in somatic cells (Pivot-Pajot et al., 2003). Genetic studies have shown that both GCN5 and BRDT play a role in the histone-to-protamine replacement during spermiogenesis (Gaucher et al., 2012; Luense et al., 2019; Shang et al., 2007). Therefore, histone acetylation enhances chromatin accessibility and facilitates nucleosome eviction during spermiogenesis.

Like histone acetylation, histone methylations also play important roles in chromatin remodeling and gene expression. DOT1L is the sole methyltransferase for histone H3 at lysine 79 (H3K79) and is responsible for mono-, di and tri-methylation (Feng et al., 2002; Lacoste et al., 2002; Ng et al., 2002; van Leeuwen et al., 2002). Dot1, the yeast orthologue of DOT1L, methylates H3K79 in the context of nucleosomes and chromatin (Lacoste et al., 2002; van Leeuwen et al., 2002). In addition, ubiquitylation of H2B at lysine 120 directly stimulates intranucleosomal methylation of H3K79 by human DOT1L (McGinty et al., 2008). In budding yeast, Dot1 functions in telomere silencing and in meiotic checkpoint control (San-Segundo and Roeder, 2000). In mouse, DOT1L is essential for embryonic development and early erythropoiesis (Feng et al., 2010; Jones et al., 2008). In mouse embryonic stem (ES) cells, DOT1L is important for establishment of heterochromatin structures at telomeres, centromeres and pericentromeres (Jones et al., 2008; Malla et al., 2021 preprint). The phosphorylated C-terminal domain of RNA polymerase II recruits DOT1L, which leads to H3K79 methylations in actively transcribed genes (Kim et al., 2012). Several DOT1L-associated proteins have been identified in cancer cells (Vlaming and van Leeuwen, 2016). In MLL leukemia, DOT1L interacts with AF9, MLLT10 (i.e. AF10) and ENL. MLLT10 regulates the H3K79 methyltransferase activity in leukemic cells. The fusion of MLL-MLLT10 leads to malignant transformation of myeloid progenitor cells and upregulation of HoxA9 in leukemia (Deshpande et al., 2014; Okada et al., 2005). Therefore, DOT1L plays important roles in development and leukemic transformation.

Mouse DOT1L is essential for self-renewal of spermatogonial stem cells (Lin et al., 2022). Inhibition of DOT1L leads to downregulation of HoxC cluster transcription factors in spermatogonial stem cells (Lin et al., 2022). In contrast, oocyte-specific inactivation shows that DOT1L is dispensable for oogenesis and that maternal DOT1L is not required for mouse development (Liao and Szabo, 2020). In mouse spermatocytes, DOT1L and H3K79 methylation exhibit dynamic localization patterns (Ontoso et al., 2014). Intriguingly, both DOT1L and H3K79me2 are abundant in diplotene spermatocytes; however, DOT1L accumulates in the XY body but H3K79me2 mark is excluded from the XY body, suggesting that DOT1L may play a role in male meiosis in mouse (Ontoso et al., 2014). Here, we report that DOT1L forms a complex with MLLT10 in mouse testis. DOT1L and MLLT10 localize to the XY chromatin in late meiotic spermatocytes and early round spermatids in an inter-dependent manner. Conditional inactivation of either Dot1l or Mllt10 results in depletion of H3K79me2 in germ cells, reduced sperm count and decreased male fertility. We also find that H3K79me2 is abundant in round and elongating spermatids. Unexpectedly, our study shows that histones are retained in Dot1l-deficient or Mllt10-deficient epididymal sperm, suggesting that H3K79 methylation promotes histone-to-protamine replacement during spermiogenesis.

DOT1L interacts with MLLT10 in testis

To enable systematic identification of DOT1L-associated proteins in testis, we generated Dot1l-3×FLAG mice using the CRISPR/Cas9 genome editing approach (Fig. S1). The adult homozygous FLAG-tagged mice were fertile, showing that the C-terminal FLAG tagging did not affect the DOT1L function. For immunoprecipitation (IP) with anti-FLAG antibody, protein extracts were prepared from wild-type (no FLAG tag) and Dot1l-3×FLAG testes at postnatal day 26, which contain spermatogonia, spermatocytes and round spermatids. Immunoprecipitated proteins were identified by mass spectrometry. As expected, DOT1L had the highest number of peptides identified in the DOT1L-3×FLAG testis IP (Fig. 1A). In addition to DOT1L, MLLT10 and MLLT6 ranked second and third, respectively, in the number of peptides in the DOT1L-3×FLAG testis IP but had no peptides in the wild-type testis IP (Fig. 1A). MLLT10 is a known DOT1L-binding protein in leukemic cells (Deshpande et al., 2014; Okada et al., 2005). MLLT6 (also known as AF17) competes with MLLT3 (also known as AF9) for binding to DOT1L in epithelial cells (Reisenauer et al., 2009). However, none of these MLLT proteins has been studied in germ cells. To validate the DOT1L-MLLT10 interaction, we first performed co-IP in testis with the anti-FLAG antibody followed by western blotting analysis (Fig. 1B). As expected, the DOT1L-3×FLAG was immunoprecipitated with the anti-FLAG antibody. MLLT10 was co-immunoprecipitated only in the DOT1L-3×FLAG testis IP, demonstrating that MLLT10 is associated with DOT1L in testis (Fig. 1B). Reciprocal co-IP confirmed the DOT1L-MLLT10 association in testis (Fig. 1C).

Fig. 1.

DOT1L is associated with MLLT10 in mouse testis. (A) Identification of DOT1L-associated proteins in testis by co-immunoprecipitation (co-IP) and mass spectrometry. Proteins listed have the highest number of peptides identified by mass spectrometry in the complexes immunoprecipitated with anti-FLAG monoclonal antibody. (B) Co-IP of MLLT10 with DOT1L-3×FLAG in 2-month-old testes. (C) Reciprocal co-IP of DOT1L-3×FLAG with MLLT10 in 2-month-old testes. Input was 1% of the protein extract used for IP (B,C). The experiments were replicated twice. (D) Western blot analysis of DOT1L, MLLT10 and H3K79me2 in a panel of adult mouse tissues. Asterisk indicates a non-specific band. (E) Western blot analysis of DOT1L, MLLT10 and H3K79me2 in mouse testes at various ages. The first appearance of types of germ cells in postnatal testes is shown. ACTB serves as a loading control. Validation of primary antibodies is shown in Fig. S7. Images of full-length western blots in B-D were shown in Fig. S8.

Fig. 1.

DOT1L is associated with MLLT10 in mouse testis. (A) Identification of DOT1L-associated proteins in testis by co-immunoprecipitation (co-IP) and mass spectrometry. Proteins listed have the highest number of peptides identified by mass spectrometry in the complexes immunoprecipitated with anti-FLAG monoclonal antibody. (B) Co-IP of MLLT10 with DOT1L-3×FLAG in 2-month-old testes. (C) Reciprocal co-IP of DOT1L-3×FLAG with MLLT10 in 2-month-old testes. Input was 1% of the protein extract used for IP (B,C). The experiments were replicated twice. (D) Western blot analysis of DOT1L, MLLT10 and H3K79me2 in a panel of adult mouse tissues. Asterisk indicates a non-specific band. (E) Western blot analysis of DOT1L, MLLT10 and H3K79me2 in mouse testes at various ages. The first appearance of types of germ cells in postnatal testes is shown. ACTB serves as a loading control. Validation of primary antibodies is shown in Fig. S7. Images of full-length western blots in B-D were shown in Fig. S8.

We next examined the expression of DOT1L and MLLT10 in mouse tissues (Fig. 1D). Surprisingly, DOT1L was abundant in testis but not detected in other somatic tissues that were examined. Likewise, MLLT10 was abundant in testis and present at a low level in brain, but not in other tissues. In contrast, H3K79me2 was present in all tissues. We next investigated their expression in postnatal developing testes (Fig. 1E). In the first wave of spermatogenesis after birth, different germ cells appear at defined ages (Fig. 1E). DOT1L was present at low levels at postnatal days 7, 15 and 21, but its abundance increased at postnatal day 28, when elongated spermatids first appeared. The abundance of MLLT10 was sharply increased at postnatal day 21, when round spermatids first appeared. These results suggest that DOT1L and MLLT10 may play an important role in germ cells of late stages during spermatogenesis.

Accumulation of DOT1L and MLLT10 at the XY chromatin in meiotic and post-meiotic germ cells

We investigated the expression and subcellular localization of DOT1L and MLLT10 by immunofluorescence of testis sections (Fig. 2A). Consistent with our western blot data in juvenile testes (Fig. 1E), DOT1L and MLLT10 were expressed in late spermatocytes and early spermatids. DOT1L was abundant in the nuclei of diplotene spermatocytes, metaphase spermatocytes, secondary spermatocytes, round spermatids and step 9 elongating spermatids (Fig. 2B). DOT1L was not detected in spermatids at step 11 and beyond. The MLLT10 expression pattern mostly overlapped with DOT1L, except that MLLT10 was also abundant in mid- to late-pachytene spermatocytes (Fig. 2B). H3K79me2 was present from late pachytene spermatocytes through step 11 elongating spermatids. Notably, the H3K79me2 signals increased in intensity in elongating spermatids (step 9-11) and then disappeared abruptly in condensing spermatids (step 12) (Fig. 2B,C). Sex chromosomes form the XY body and are γH2AX positive in pachytene and diplotene spermatocytes. Strikingly, although DOT1L did not accumulate in the XY body in pachytene spermatocytes, it accumulated in the XY body in diplotene spermatocytes. DOT1L was also concentrated in a peri-chromocenter area in round spermatids, which had a characteristic intermediate DAPI staining (weaker than the chromocenter) and was previously shown to be the sex chromosome (Namekawa et al., 2006). MLLT10 began to accumulate on the XY body in late pachytene cells and had the same localization pattern on sex chromosomes in diplotene cells and round spermatids as DOT1L. In metaphase I spermatocytes, DOT1L and MLLT10 localized to one pair of chromosomes, which were likely to be sex chromosomes. Nuclear spread analysis confirmed the localization pattern of DOT1L and MLLT10 in spermatocytes (Fig. S2A,B). In secondary spermatocytes, DOT1L and MLLT10 also occupied one subnuclear region, which likely corresponds to sex chromosomes (Fig. 2B). Unlike DOT1L and MLLT10, H3K79me2 was excluded from the XY body in diplotene cells (Fig. 2B, Fig. S2C). However, like DOT1L and MLLT10, H3K79me2 also had increased signals in the peri-chromocenter region (sex chromosome) in round spermatids. The largely overlapping localization patterns of DOT1L and MLLT10 in male germ cells were consistent with the DOT1L-MLLT10 interaction in testis. These results suggest a possible role for DOT1L and MLLT10 in meiosis and spermiogenesis.

Fig. 2.

Accumulation of DOT1L and MLLT10 at the sex chromatin in late prophase I spermatocytes through early round spermatids. (A,B) Immunofluorescence analysis of DOT1L, MLLT10 and H3K79me2 in wild-type adult testes. Stages of the seminiferous tubules are shown. Enlarged views of the outlined cells are shown in B. Scale bars: 20 μm in A; 10 μm in B. (C) Summary of dynamic expression and localization of DOT1L, MLLT10 and H3K79me2 in postnatal male germ cells. Lep, leptotene; Zyg, zygotene; E-Pa, early pachytene; M-Pa, mid-pachytene; L-Pa, late pachytene; Dip, diplotene; MI, metaphase I spermatocytes; SS, secondary spermatocytes; RS, round spermatids; ES, elongated spermatids; CS, condensing spermatids. Arabic numerals indicate the step of spermatids. X/Y indicates X or Y chromosome. Roman numerals indicate the stage of the tubule.

Fig. 2.

Accumulation of DOT1L and MLLT10 at the sex chromatin in late prophase I spermatocytes through early round spermatids. (A,B) Immunofluorescence analysis of DOT1L, MLLT10 and H3K79me2 in wild-type adult testes. Stages of the seminiferous tubules are shown. Enlarged views of the outlined cells are shown in B. Scale bars: 20 μm in A; 10 μm in B. (C) Summary of dynamic expression and localization of DOT1L, MLLT10 and H3K79me2 in postnatal male germ cells. Lep, leptotene; Zyg, zygotene; E-Pa, early pachytene; M-Pa, mid-pachytene; L-Pa, late pachytene; Dip, diplotene; MI, metaphase I spermatocytes; SS, secondary spermatocytes; RS, round spermatids; ES, elongated spermatids; CS, condensing spermatids. Arabic numerals indicate the step of spermatids. X/Y indicates X or Y chromosome. Roman numerals indicate the stage of the tubule.

Loss of DOT1L causes apoptosis in metaphase I spermatocytes and reduces sperm production

We have previously demonstrated that DOT1L is essential for self-renewal of spermatogonial stem cells by inactivating Dot1l using the Ddx4-Cre (Lin et al., 2022). To circumvent the loss of spermatogonial stem cells, we inactivated Dot1l using the Stra8-Cre, which is expressed in differentiating spermatogonia before meiotic entry (Lin et al., 2017). We generated Dot1lfl/−Stra8-Cre conditional knockout (referred to as Dot1lcKO) mice. We found that the adult Dot1lcKO testis was smaller (Fig. 3A). The testis weight of adult Dot1lcKO males was significantly reduced (Fig. 3B). The sperm count of Dot1lcKO males was sharply decreased (Fig. 3C). Mating tests showed that, out of six Dot1lcKO males tested, two males were infertile and the remaining four males produced fewer and smaller litters (Fig. 3D,E). Western blot analysis revealed that DOT1L was absent in Dot1lcKO testes (Fig. 3F). H3K79me2 was substantially lower in Dot1lcKO testes than in Dot1lfl/+ testes but not absent (Fig. 3F). Immunofluorescence of testis sections showed that the focal localization of DOT1L in metaphase spermatocytes was lost in Dot1lcKO testes (Fig. S3A). H3K79me2 was absent in germ cells but still present in somatic cells in Dot1lcKO testes, as expected (Fig. S3B). Histological analysis of Dot1lcKO testes revealed no spermatogenic arrest (Fig. 3G). In addition, Dot1lcKO testes lacked defects in self-renewal of spermatogonial stem cells. This was expected, because Stra8-Cre was not expressed in spermatogonial stem cells. On closer examination by histology, we observed an increase in apoptotic spermatocytes in stage XII tubules from Dot1lcKO testes (Fig. 3G). Indeed, TUNEL assays showed a significant increase in the number of apoptotic spermatocytes in stage XII tubules from Dot1lcKO testes (Fig. 3H). Therefore, the increased apoptosis in spermatocytes at stage XII could be at least partially responsible for reduced sperm production in Dot1lcKO males.

Fig. 3.

Inactivation of Dot1l in germ cells leads to increased apoptosis in metaphase spermatocytes and reduced sperm production. (A) Images of testes from 2-month-old Dot1lfl/− and Dot1lfl/−Stra8-Cre (Dot1lcKO) mice. (B,C) Reduced testis weight (B) and reduced sperm count (C) in Dot1lcKO males compared with control males (Dot1lfl/+, Dot1lfl/− or Dot1lfl/+Stra8-Cre). N, number of 2- to 3-month-old males. Data are mean±s.d. A paired Student's t-test was used to test for significance. (D) Reduced number of litters sired by Dot1lcKO males. Each 8-week-old male was housed with two wild-type females for 3 months. (E) Reduced size of litters sired by Dot1lcKO males in comparison with control (Dot1lfl/+ and Dot1lfl/−) males. Each 8-week-old male was housed with two wild-type females for 3 months. Data are mean±s.d. Unpaired Student's t-test was used to test for significance. (F) Western blot analysis of DOT1L and H3K79me2 in 2-month-old testes. SYCP3 is a meiosis-specific protein. ACTB serves as a loading control. Asterisks indicate a non-specific band. (G) Histology (Hematoxylin and Eosin staining) of 2-month-old testes. Stage XII tubules are shown. Diakinesis spermatocytes, black arrowheads; metaphase spermatocytes, white arrowheads; apoptotic cells, red arrowheads. Scale bar: 50 μm. (H) TUNEL analysis of testes from 2-month-old mice. White arrowheads indicate metaphase spermatocytes. Red arrowheads indicate TUNEL-positive spermatocytes. Scale bar: 50 μm. The plot on the left shows the number of TUNEL-positive cells per stage XII tubule cross-section. N, number of mice; n, number of tubule cross sections analyzed. ****P<0.0001 (Mann–Whitney U-test). Data are mean±s.d.

Fig. 3.

Inactivation of Dot1l in germ cells leads to increased apoptosis in metaphase spermatocytes and reduced sperm production. (A) Images of testes from 2-month-old Dot1lfl/− and Dot1lfl/−Stra8-Cre (Dot1lcKO) mice. (B,C) Reduced testis weight (B) and reduced sperm count (C) in Dot1lcKO males compared with control males (Dot1lfl/+, Dot1lfl/− or Dot1lfl/+Stra8-Cre). N, number of 2- to 3-month-old males. Data are mean±s.d. A paired Student's t-test was used to test for significance. (D) Reduced number of litters sired by Dot1lcKO males. Each 8-week-old male was housed with two wild-type females for 3 months. (E) Reduced size of litters sired by Dot1lcKO males in comparison with control (Dot1lfl/+ and Dot1lfl/−) males. Each 8-week-old male was housed with two wild-type females for 3 months. Data are mean±s.d. Unpaired Student's t-test was used to test for significance. (F) Western blot analysis of DOT1L and H3K79me2 in 2-month-old testes. SYCP3 is a meiosis-specific protein. ACTB serves as a loading control. Asterisks indicate a non-specific band. (G) Histology (Hematoxylin and Eosin staining) of 2-month-old testes. Stage XII tubules are shown. Diakinesis spermatocytes, black arrowheads; metaphase spermatocytes, white arrowheads; apoptotic cells, red arrowheads. Scale bar: 50 μm. (H) TUNEL analysis of testes from 2-month-old mice. White arrowheads indicate metaphase spermatocytes. Red arrowheads indicate TUNEL-positive spermatocytes. Scale bar: 50 μm. The plot on the left shows the number of TUNEL-positive cells per stage XII tubule cross-section. N, number of mice; n, number of tubule cross sections analyzed. ****P<0.0001 (Mann–Whitney U-test). Data are mean±s.d.

We examined meiotic progression in Dot1lcKO testes. The composition of prophase I spermatocytes was comparable between Dot1lcKO and control testes (Fig. S4A). We did not observe defects in chromosomal synapsis in Dot1lcKO spermatocytes. RPA, a single-strand DNA-binding heterotrimer (RPA1, RPA2 and RPA3), localizes to DSBs (Shi et al., 2019). The number of RPA2 foci was similar between Dot1lcKO and control spermatocytes (Fig. S4B). We next examined the XY body. SCML2 localizes to the XY body in spermatocytes (Hasegawa et al., 2015; Luo et al., 2015). SCML2 still localized to the XY body in both pachytene and diplotene spermatocytes from Dot1lcKO testes (Fig. S5A). H3K9me2, a silencing histone modification, accumulated at the XY body in both Dot1lcKO and control spermatocytes (Fig. S5B). As in control spermatocytes, H3K4me3 and RNA polymerase II were excluded from the XY body in Dot1lcKO pachytene and diplotene spermatocytes, showing that meiotic sex chromatin inactivation (MSCI) remained intact (Fig. S5C,D). These analyses showed that meiotic progression was apparently normal in Dot1lcKO testes.

Germ cell-specific deletion of Mllt10 causes reduced sperm production and male subfertility

Global deletion of Mllt10 causes embryonic lethality (Deshpande et al., 2014). To study its role in germ cells, we generated Mllt10fl/−Ddx4-Cre conditional knockout (referred to as Mllt10cKO) mice. Ddx4-Cre begins to be expressed in germ cells at embryonic day 15 (E15) (Gallardo et al., 2007). Therefore, Mllt10 was expected to be deleted in all germ cells, including spermatogonia, in postnatal Mllt10cKO testis. We found that adult Mllt10cKO testis was slightly smaller than Mllt10fl/− testis (Fig. 4A). Western blot analysis showed the absence of MLLT10 in Mllt10cKO testis (Fig. 4B). Immunofluorescence showed that MLLT10 signals were absent in Mllt10cKO testis (Fig. S3C). H3K79me2 was absent in germ cells but still present in somatic cells in Mllt10cKO testis, demonstrating that MLLT10 is required for the methyltransferase activity of DOT1L in germ cells (Fig. S3D). The testis weight of Mllt10cKO mice was lower than that of control (Fig. 4C). The sperm count of Mllt10cKO males was reduced by 50% (Fig. 4D). Mating tests showed that although the number of litters was the same, the size of litters sired by Mllt10cKO males was significantly lower (Fig. 4E,F). In conclusion, loss of MLLT10 results in subfertility in males.

Fig. 4.

Inactivation of Mllt10 in germ cells reduces sperm production and fertility. (A) Image of testes from 2-month-old Mllt10fl/− and Mllt10fl/−Ddx4-Cre (Mllt10cKO ) mice. (B) Western blot analysis of MLLT10 in 2-month-old testes. ACTB serves as a loading control. (C,D) Comparison of testis weight (C) and sperm count (D) in 8-week-old Mllt10cKO and control males (Mllt10fl/+, Mllt10fl/− or Mllt10fl/+Ddx4-Cre). N, number of males. (E,F) Comparison of litter number (E) and litter size (F) between 8-week-old Mllt10cKO and control (Mllt10fl/+ and Mllt10fl/−) males. N, number of male mice. Each 8-week-old male was housed with two wild-type females for 2 months. An unpaired Student's t-test was used to test for significance. (G) Histology (Hematoxylin and Eosin staining) of 2-month-old testes. Tubules at stages VII-VIII are shown. Pa, pachytene; RS, round spermatid; ES, elongated spermatid. Scale bar: 50 μm. (H) TUNEL analysis of testes from 2-month-old mice. White arrowheads indicate metaphase cells. Red arrowheads indicate TUNEL-positive cells. Scale bar: 50 μm. (I) Count of TUNEL-positive cells per tubule cross-section at all stages (left) or only stage XII (right). N, number of mice; n, number of tubule cross-sections. Data are mean±s.d. NS, non-significant (Mann–Whitney U-test).

Fig. 4.

Inactivation of Mllt10 in germ cells reduces sperm production and fertility. (A) Image of testes from 2-month-old Mllt10fl/− and Mllt10fl/−Ddx4-Cre (Mllt10cKO ) mice. (B) Western blot analysis of MLLT10 in 2-month-old testes. ACTB serves as a loading control. (C,D) Comparison of testis weight (C) and sperm count (D) in 8-week-old Mllt10cKO and control males (Mllt10fl/+, Mllt10fl/− or Mllt10fl/+Ddx4-Cre). N, number of males. (E,F) Comparison of litter number (E) and litter size (F) between 8-week-old Mllt10cKO and control (Mllt10fl/+ and Mllt10fl/−) males. N, number of male mice. Each 8-week-old male was housed with two wild-type females for 2 months. An unpaired Student's t-test was used to test for significance. (G) Histology (Hematoxylin and Eosin staining) of 2-month-old testes. Tubules at stages VII-VIII are shown. Pa, pachytene; RS, round spermatid; ES, elongated spermatid. Scale bar: 50 μm. (H) TUNEL analysis of testes from 2-month-old mice. White arrowheads indicate metaphase cells. Red arrowheads indicate TUNEL-positive cells. Scale bar: 50 μm. (I) Count of TUNEL-positive cells per tubule cross-section at all stages (left) or only stage XII (right). N, number of mice; n, number of tubule cross-sections. Data are mean±s.d. NS, non-significant (Mann–Whitney U-test).

Histological analysis of adult Mllt10cKO testes did not reveal spermatogenic block or an apparent loss of germ cells (Fig. 4G). Germ cell-specific deletion of Dot1l using the Ddx4-Cre causes a failure in self-renewal of spermatogonial stem cells, resulting in Sertoli cell-only tubules in testes as early as postnatal day 40 (Lin et al., 2022). However, Sertoli cell-only tubules were not observed in Mllt10cKO testes up to the age of 4 months, showing that MLLT10, unlike DOT1L, is dispensable for self-renewal of spermatogonial stem cells. Furthermore, there was no increase in apoptosis of spermatocytes in stage XII tubules or other tubules from Mllt10cKO testes (Fig. 4H,I). We monitored chromosomal synapsis, XY body formation and meiotic DSB formation by surface nuclear spread analysis of spermatocytes (Fig. S6). The composition of prophase I spermatocytes was similar between Mllt10fl/− and Mllt10cKO testes (Fig. S6A). The XY body was formed in Mllt10cKO pachytene and diplotene cells, evidenced by the localization of γH2AX and H3K9me2 (Fig. S6C,D). Chromosomal synapsis and formation of meiotic DSBs appeared to be normal in Mllt10cKO pachytene cells (Fig. S6B,E). These analyses show that MLLT10 is dispensable for meiosis.

Inter-dependent localization of DOT1L and MLLT10 at the sex chromatin from diplotene spermatocytes to round spermatids

DOT1L and MLLT10 form a complex in testis (Fig. 1). We sought to address whether their abundance is dependent on each other by western blotting analysis in knockout testes. MLLT10 was readily detected in Dot1lcKO testes but at a lower abundance than in Dot1lfl/+ testes (Fig. 5A). The DOT1L abundance was much lower in Mllt10cKO testes than in Mllt10fl/+ testes (Fig. 5B). The H3K79me2 levels were reduced in both Dot1lcKO testes and Mllt10cKO testes (Fig. 5A,B). These data show that DOT1L and MLLT10 stabilize each other in testes and that MLLT10 also plays a role in facilitating DOT1L in H3K79 methylation in germ cells.

Fig. 5.

Interdependent localization of DOT1L and MLLT10 at the sex chromosomes from diplotene spermatocytes to round spermatids. (A) Western blot analysis of 2-month-old Dot1lfl/+, Dot1lfl/− and Dot1lcKO testes. (B) Western blot analysis of 2-month-old Mllt10fl/+, Mllt10fl/− and Mllt10cKO testes. Asterisk indicates a non-specific band. (C) Immunofluorescence analysis of MLLT10 in staged seminiferous tubules from 2-month-old Dot1lfl/+ and Dot1lcKO testes. White arrowheads indicate accumulation of MLLT10 at the sex chromatin in Dot1lfl/+ germ cells. Red arrowheads indicate loss of accumulation of MLLT10 at the sex chromatin (γH2AX-positive) in Dot1lcKO diplotene spermatocytes. (D) Immunofluorescence analysis of DOT1L in staged seminiferous tubules from 2-month-old Mllt10fl/+ and Mllt10cKO testes. White arrowheads indicate accumulation of DOT1L at the sex chromatin in Mllt10fl/+ germ cells; red arrowheads indicate loss of accumulation of DOT1L at the sex chromatin in Mllt10cKO diplotene spermatocytes. (C,D) Le, leptotene; Zy, zygotene; Pa, pachytene; Di, diplotene; M, metaphase; SS, secondary spermatocytes; RS, round spermatids. Enlarged view of boxed cells in the tubules are shown in the right columns. Scale bars: 10 μm.

Fig. 5.

Interdependent localization of DOT1L and MLLT10 at the sex chromosomes from diplotene spermatocytes to round spermatids. (A) Western blot analysis of 2-month-old Dot1lfl/+, Dot1lfl/− and Dot1lcKO testes. (B) Western blot analysis of 2-month-old Mllt10fl/+, Mllt10fl/− and Mllt10cKO testes. Asterisk indicates a non-specific band. (C) Immunofluorescence analysis of MLLT10 in staged seminiferous tubules from 2-month-old Dot1lfl/+ and Dot1lcKO testes. White arrowheads indicate accumulation of MLLT10 at the sex chromatin in Dot1lfl/+ germ cells. Red arrowheads indicate loss of accumulation of MLLT10 at the sex chromatin (γH2AX-positive) in Dot1lcKO diplotene spermatocytes. (D) Immunofluorescence analysis of DOT1L in staged seminiferous tubules from 2-month-old Mllt10fl/+ and Mllt10cKO testes. White arrowheads indicate accumulation of DOT1L at the sex chromatin in Mllt10fl/+ germ cells; red arrowheads indicate loss of accumulation of DOT1L at the sex chromatin in Mllt10cKO diplotene spermatocytes. (C,D) Le, leptotene; Zy, zygotene; Pa, pachytene; Di, diplotene; M, metaphase; SS, secondary spermatocytes; RS, round spermatids. Enlarged view of boxed cells in the tubules are shown in the right columns. Scale bars: 10 μm.

We next examined whether the subcellular localization of MLLT10 was affected in Dot1lcKO testes (Fig. 5C). MLLT10 accumulated on the sex chromosomes (XY body) in diplotene cells and formed one large blob (presumably sex chromosomes) in wild-type metaphase spermatocytes, secondary spermatocytes and round spermatids (Figs 2 and 5C). Notably, MLLT10 failed to localize to sex chromosomes in spermatocytes and round spermatids from Dot1lcKO testes (Fig. 5C). DOT1L had a similar accumulation pattern on the sex chromosomes in wild-type spermatocytes and round spermatids, but failed to localize to the sex chromosomes from Mllt10cKO testes (Fig. 5D). Therefore, these results demonstrate the inter-dependent localization of DOT1L and MLLT10 at the sex chromosomes from diplotene spermatocytes to round spermatids.

Histone retention in epididymal sperm from Dot1lcKO and Mllt10cKO mice

While performing sperm count, we noticed abnormal morphology in sperm from the Dot1lcKO cauda epididymis (Fig. 6A,B). We observed three types of sperm in the Dot1lcKO males. Although morphologically normal sperm (type 1) were present, some sperm (type 2) had head foldback onto the flagella and a minority of sperm had coiled flagella (type 3) (Fig. 6A,B). The normal sperm (type 1) and head foldback sperm (type 2) each accounted for about 50% of the sperm (Fig. 6C). The type 2 sperm appeared to have a round head morphology under bright field illumination. However, DAPI staining revealed that type 2 sperm had nuclear morphology similar to normal sperm (Fig. 6A,B). Type 3 sperm also had apparently typical nuclear morphology. Under a dissecting microscope, all three types of sperm from the Dot1lcKO males appeared motile.

Fig. 6.

Histone retention in epididymal sperm from Dot1lcKO and Mllt10cKO mice. (A) Morphology of cauda epididymal sperm from 4-month-old Dot1lfl/+ (control) and Dot1lcKO males. Type 1, normal; type 2, sperm head foldback; type 3, coiled flagellum; BF, bright field. Scale bars: 25 µm. (B) Enlarged view of three types of sperm from Dot1lcKO males. (C) Quantification of three types of cauda sperm from 4-month-old Dot1lfl/+ and Dot1lcKO males. N, number of males; n, number of sperm counted per male. An unpaired Student's t-test was used to test for significance. (D) Western blot analysis of histone H3 in cauda epididymal sperm from 4-month-old Dot1lfl/+ and Dot1lcKO males. Acetyl-tubulin serves as a loading control. The experiment was replicated three times. (E) Western blot analysis of histone H3 in cauda epididymal sperm from 3-month-old control and Mllt10cKO males. The experiment was replicated twice.

Fig. 6.

Histone retention in epididymal sperm from Dot1lcKO and Mllt10cKO mice. (A) Morphology of cauda epididymal sperm from 4-month-old Dot1lfl/+ (control) and Dot1lcKO males. Type 1, normal; type 2, sperm head foldback; type 3, coiled flagellum; BF, bright field. Scale bars: 25 µm. (B) Enlarged view of three types of sperm from Dot1lcKO males. (C) Quantification of three types of cauda sperm from 4-month-old Dot1lfl/+ and Dot1lcKO males. N, number of males; n, number of sperm counted per male. An unpaired Student's t-test was used to test for significance. (D) Western blot analysis of histone H3 in cauda epididymal sperm from 4-month-old Dot1lfl/+ and Dot1lcKO males. Acetyl-tubulin serves as a loading control. The experiment was replicated three times. (E) Western blot analysis of histone H3 in cauda epididymal sperm from 3-month-old control and Mllt10cKO males. The experiment was replicated twice.

Most histones are replaced by protamines in sperm. H3K79me2 reached the highest abundance in elongating spermatids (step 9-11), in which histone-to-protamine replacement occurs (Fig. 2). Therefore, we sought to address whether histone-to-protamine exchange was affected in the Dot1lcKO testis. Western blot analysis showed that histone H3 was substantially retained in epididymal sperm from the Dot1lcKO males in comparison with the Dot1lfl/+ (control) males (Fig. 6D). In addition, the abundance of histone H3 increased in epididymal sperm from Mllt10cKO males (Fig. 6E). These results demonstrate that DOT1L and MLLT10 promote histone replacement during spermiogenesis.

Dramatic chromatin reorganization occurs during spermiogenesis. A major event is the replacement of most histones by protamines in elongating spermatids. Histone acetylation precedes the histone-to-protamine exchange in spermatids (Meistrich et al., 1992; Oliva and Mezquita, 1982). GCN5, a histone acetyltransferase, and BRDT, an acetylated histone reader, function in histone exchange in late spermatids (Gaucher et al., 2012; Luense et al., 2019; Shang et al., 2007). BRD4, another acetylated histone reader, localizes as a ring structure under the acrosome and has been postulated to facilitate histone removal (Bryant et al., 2015). The bromodomain in BRDT and BRD4 recognizes acetylated histones. In this study, we found that H3K79me2 reached the highest abundance in elongating spermatids from step 9 to 11. Histone H3 was retained in epididymal sperm from either DOT1L- or MLLT10-deficient mice. Our result is consistent with the histone retention in spermatids from DOT1L conditional knockout testes as reported by Malla et al. (2023). Therefore, in addition to histone acetylation, histone H3K79 methylation also plays an important role in histone removal in spermatids. Histone acetylation and histone H3K79 methylation are associated with active gene transcription and thus are features of open chromatin. In spermatids, the open chromatin state induced by histone acetylation and histone methylation possibly facilitates nucleosome eviction and thus histone removal.

Two proteins were previously reported to play a role in histone removal: CHD5 and RNF8. CHD5, a chromatin remodeler, plays a multi-faceted role in histone removal in spermatids (Li et al., 2014). CHD5 binds to unmodified histone 3 (H3) with its PHD fingers (Oliver et al., 2012). Loss of CHD5 compromises H4 acetylation and perturbs the expression of transition proteins and protamines. The role of RNF8, a ubiquitin E3 ligase, in histone removal in spermatids is contradictory. In one study, it was claimed that RNF8-dependent histone ubiquitylation induces H4K16 acetylation, resulting in nucleosome removal (Lu et al., 2010). However, another study concluded that RNF8 is required for neither H4K16 acetylation nor histone-to-protamine exchange (Abe et al., 2021). Here, we report that the DOT1L-MLLT10 complex is important, highlighting the multi-factorial regulation of this complex process.

We have previously reported that DOT1L is essential for self-renewal of spermatogonial stem cells (Lin et al., 2022). In this study, we find that DOT1L plays additional roles during male meiosis and spermiogenesis. In Dot1l-deficient testis, the increased apoptosis in metaphase I spermatocytes is presumably caused by activation of the spindle assembly checkpoint, which is usually triggered by defects in chromosome segregation. However, chromosome synapsis and meiotic recombination appeared to be unaffected in Dot1l-deficient testis. In C. elegans, Dot-1.1 plays a role in the regulation of chromosome synapsis and meiotic recombination, but is not essential for either meiotic process (Lascarez-Lagunas et al., 2020). In budding yeast, Dot1 functions in the meiotic checkpoint control (San-Segundo and Roeder, 2000). Such a role for Dot-1.1 in meiotic checkpoint in C. elegans is not as clear-cut as in yeast (Lascarez-Lagunas et al., 2020). Although the methyltransferase activity of DOT1L is evolutionarily conserved, the role of DOT1L and its orthologues during meiosis differs between the single cell eukaryotes (yeast) and metazoans.

DOT1L interacts with MLLT10 in leukemic cells (Deshpande et al., 2014; Okada et al., 2005). Direct fusion of either DOT1L or MLLT10 to MLL (mixed lineage leukemia) leads to leukemic transformation (Okada et al., 2005). The methyltransferase activity of DOT1L depends on its binding partner MLLT10 in leukemic cells (Deshpande et al., 2014). We find that DOT1L and MLLT10 form a complex in testis. Although DOT1L is essential for self-renewal of spermatogonial stem cells (Lin et al., 2022), the current study did not identify a role for MLLT10 in self-renewal of spermatogonial stem cells, suggesting that the requirement of DOT1L in spermatogonial stem cells is independent of MLLT10. However, the localization of DOT1L and MLLT10 on the sex chromatin in spermatocytes and spermatids is inter-dependent. Importantly, MLLT10 is required for H3K79me2 in spermatocytes and spermatids. Furthermore, loss of either DOT1L or MLLT10 leads to histone retention in epididymal sperm. Therefore, like in leukemic transformation, the interaction between DOT1L and MLLT10 is essential for H3K79 methylation and histone removal during spermiogenesis.

Generation of Dot1l and Mllt10 conditional knockout mice

Dot1l floxed (Dot1lfl) mice and Mllt10 floxed (Mllt10fl) mice were generated previously (Bernt et al., 2011; Deshpande et al., 2014). To inactivate Dot1l or Mllt10 specifically in germ cells, floxed mice were crossed with the Cre mouse strains Ddx4-Cre (stock number 006954, Jackson Laboratory) and Stra8-Cre (Gallardo et al., 2007; Lin et al., 2017). PCR genotyping primers were as follows: the Dot1l floxed allele (418 bp), p2 (5′ CCCAAAAGGGTCTTTTCACA 3′) and p3 (5′ ATGGGATTTCATGGAAGCAA 3′); the Dot1l deletion allele (620 bp), p1 (5′ CTCACAGTCACATACTACCTCTGAC 3′) and p3 (5′ ATGGGATTTCATGGAAGCAA 3′); Ddx4-Cre (240 bp), Vasa-Cre-1 (5′ CACGTGCAGCCGTTTAAGCCGCGT 3′) and Vasa-Cre-2 (5′ TTCCCATTCTAAACAACACCCTGAA 3′); the Mllt10 floxed allele (602 bp), Flox-F (5′ TGCTCGGATCAAAGCTTCC 3′) and Flox-R (5′ TCTGTCTCTGTCCCTCACAAC 3′); the Mllt10 deletion allele (250 bp), 1a (5′ CACAGCCTACTTCAAAGAAC 3′) and 4a (5′ ATTAGAGTCCATCCCACTTC 3′); and Stra8-Cre (400 bp), F (5′ ACTCCAAGCACTGGGCAGAA 3′) and R2 (5′ CGTTTACGTCGCCGTCCAG 3′). The Institutional Animal Care and Use Committee (IACUC) of the University of Pennsylvania approved all procedures.

Generation of Dot1l-3×FLAG knock-in mice

A guide RNA mapping to the last coding exon of the Dot1l gene was designed (Fig. S1). DNA oligos for sgRNA contained overhangs for ligation into the BbsI site in the px330 vector (Addgene 127875). sgRNA was generated through in vitro transcription with T7 RNA polymerase. The ssDNA repair template contained the 3×FLAG-coding sequence (Fig. S1). The sgRNA (50 ng/μl), CAS9 protein (100 ng/μl) and template ssDNA (100 ng/μl) were mixed according to a protocol described previously (Yang et al., 2014). This mixture was microinjected into mouse zygotes to produce founder mice at the PennVet Transgenic Core facility. Injected zygotes were cultured in vitro until the two-cell stage and were then transferred into oviducts of 0.5 day post-coitum pseudopregnant foster mothers. Around 19.5 days post coitum, recipient mothers delivered pups. Two positive founders were verified by Sanger sequencing and bred with DBA2 mice for germline transmission, which was successful. The Dot1l-3×FLAG mice were bred to homozygosity.

Sperm count and mating test

For sperm count, one cauda epididymis from each mouse was harvested and placed in a tissue dish with 1×PBS. The cauda was minced into small pieces with scissors, fixed in paraformaldehyde and counted on the Hausser Bright-Line hematocytometer. For each mating test, one 8-week-old control or conditional knockout male was housed with two 8-week-old wild-type females for 3 months. The litter number and size during this period were recorded.

Immunoprecipitation, co-immunoprecipitation and mass spectrometry

Protein extracts prepared from 100 mg of 26-day-old wild-type and Dot1lFlag/Flag testes were used for immunoprecipitation with a murine IgG1 monoclonal anti-FLAG antibody covalently conjugated to agarose (A2220, Sigma). Proteins were eluted with FLAG peptides. Eluted proteins were run to 1 cm away from the loading well on an 8% SDS-PAGE gel and stained with Coomassie Brilliant Blue G-250 dye (Sigma). The stained gel was cut and sent for protein identification by mass spectrometry at the Wistar Proteomics Core Facility (Table S1). The peptide maps of DOT1L and MLLT10 are shown in Fig. S9.

For co-immunoprecipitation experiments followed by western blotting, 100 mg of 26-day or adult testes (wild type or Dot1lFlag/Flag) were homogenized in 1 ml lysis buffer (50 mM Tris, 150 mM NaCl, 5 mM MgCl2, 1 mM DTT, 0.5% deoxycholate and 1% Triton) with a cocktail of proteinase inhibitors. The lysate was precleared by incubating with agarose beads at 4°C for 2 h. Pre-cleared lysate was used for immunoprecipitation with anti-FLAG or anti-MLLT10 antibodies followed by western blotting with anti-DOT1L, anti-FLAG or anti-MLLT10 antibodies.

Histological, immunofluorescence and TUNEL analyses

For histological analysis, testes were fixed in Bouin's solution, dehydrated through a gradient of ethanol, embedded with paraffin wax and sectioned. Sections were deparaffinized in xylene, rehydrated through a gradient of ethanol, and stained with Hematoxylin and Eosin. Sections were imaged on a Leica DM5500B microscope by DFC450 digital color camera (Leica Microsystems). For immunofluorescence including TUNEL analyses, testes were fixed in 4% paraformaldehyde (in 1×PBS) overnight at 4°C, dehydrated in 30% sucrose (in 1×PBS) overnight, embedded in optimal cutting temperature compound (6502, Thermo Scientific) and sectioned on a cryostat. Alternatively, testes were fixed in a fixative solution (30% formaldehyde, 15% ethanol and 5% glacial acetic acid) overnight. Fixed testes were dehydrated through a gradient of ethanol, embedded in paraffin wax and sectioned on a microtome. Paraffin wax-embedded slides were deparaffinized in xylene, rehydrated through a gradient of ethanol. Heat-induced epitope retrieval was performed for the rehydrated slides by incubation in the epitope retrieval buffer (1 mM EDTA buffer and 0.05% Tween-20 at pH 8.0) at 95°C for 20 min, excluding TUNEL assays. Epitope retrieval for TUNEL assays was performed by incubation with proteinase K (10 µg/ml) at room temperature for 10 min. Subsequently, slides were incubated with 0.5% Triton X-100 in 1×PBS at room temperature for 10 min. Surface nuclear spread analysis of spermatocytes has been described previously (Peters et al., 1997). In brief, seminiferous tubules were extracted from testes and incubated for 45 min to 1 h in the hypotonic buffer (50 mM sucrose, 30 mM Tris, 5 mM EDTA, 17 mM trisodium citrate dihydrate, 0.5 mM DTT and 1 mM phenylmethylsulfonyl fluoride). After hypotonic treatment, seminiferous tubules were minced into cell suspension in 100 mM sucrose and were then spread on a slide covered with paraformaldehyde solution containing Triton X-100 in a wet chamber overnight.

The slides were blocked with 10% goat serum in PBST at 37°C for 1 h followed by primary antibody incubation at 37°C overnight. The primary antibodies used for immunofluorescence were as follows: DOT1L (1:50, UP2565) (Lin et al., 2022), MLLT10 (1:200, HPA005747, Sigma), H3K79me2 (1:100, ab3594, Abcam), SYCP3 (1:500, ab97672, Abcam), SYCP2 (1:500) (Yang et al., 2006), SYCP1 (1:100, ab15090, Abcam), γH2AX (1:500, 16-202A, clone JBW301, Millipore), RPA2 (1:50; UP2436) (Shi et al., 2019), SCML2 (1:200; UP2323) (Luo et al., 2015), H3K9me2 (1:500, ab1220, Abcam), H3K4me3 (1:500, 39159, Active Motif) and POLII (1:50, sc-899, Santa-Cruz Biotech). Secondary antibodies were FITC- or Texas red-conjugated (1:200, Vector Laboratories, FI-1000 or TI-1000). TUNEL assays were performed with a TUNEL kit (TUNEL Enzyme, 11767305001, Roche) and the TUNEL Label kit (11767291910, Roche) at a ratio of 1:10. Mounting medium with DAPI (H-1200, Vectashield) was used for nuclear DNA counterstaining and fluorochrome preservation. Fluorescence images were captured with an ORCA Flash4.0 digital monochrome camera (Hamamatsu Photonics) on a Leica DM5500B microscope.

Protein extraction and western blot analysis

Sperm protein extract preparation and western blot analysis have been described previously (Luense et al., 2019; Zhou et al., 2012). Cauda epididymal sperm were collected from each male. Briefly, caudal epididymides were cut several times and incubated in 1×PBS solution at 37°C for 10 min to allow sperm to swim out. Sperm concentration was determined by counting with a hemocytometer. Sperm were collected by centrifugation at 800 g for 5 min at room temperature. The sperm pellet was resuspended in 100 μl of lysis buffer [20 mM Tris-HCl (pH 7.5), 1 mM MgCl2, 1 mM CaCl2, 137 mM NaCl, 10% glycerol, 1% NP-40, 300 nM TSA, benzonase (12.5 U/ml) and 1× protease inhibitors] and incubated at 4°C for 1 h. Alternatively, sperm were homogenized in 100 μl SDS-EDTA solution (1% SDS, 24 mM EDTA and 75 mM NaCl at pH 6.0) followed by centrifugation at 5000 g for 30 min at room temperature. An equal volume of the SDS-PAGE sample buffer [62.5 mM Tris (pH 6.8), 3% SDS, 10% glycerol, 5% β-mercaptoethanol and 0.02% Bromophenol Blue) was added to the saved supernatant,. The sample was heated at 95°C for 10 min and was used for western blot analysis.

Total protein extract from testis or cultured cells was prepared by homogenization in five volumes of protein extraction buffer [62.5 mM Tris-HCl (pH 6.8), 3% SDS, 10% glycerol and 5% 2-mercaptoethanol) and boiled at 95°C for 15 min followed by centrifugation at 12,000 g for 5 min. Samples were run in an SDS-PAGE gel and then transferred to the PVDF membrane. The membrane blot was blocked in 5% non-fat milk (in 1×PBST) at room temperature for 1 h. The primary antibodies used for western blot were as follows: FLAG (1:5000, F1804, Sigma), MLLT10 (1:1000, HPA005747, Sigma), DOT1L (1:1000, ab64077, Abcam), H3K79me2 (1:1000, ab3594, Abcam), ACTB (1:5000, A5441, Sigma), SYCP3 (1:500, 23024-1-AP, Proteintech), acetylated tubulin (1:2000, T7451, Sigma) and histone H3 (1:2000, 9715S, Cell Signaling Technology). Secondary antibodies were HRP-conjugated (1:5000, Cell Signaling Technology, 7076S or 7074S). ECL western blotting substrate (32106, ThermoFisher Scientific) was used for development.

We thank Fang Yang and Zhenlong Kang for comments on the manuscript. We thank Hsin Yao Tang and Thomas Beer at the Wistar Proteomics Core for mass spectrometry.

Author contributions

Conceptualization: H.L., M.L., P.J.W.; Methodology: H.L., I.G.C., N.A.L.; Validation: H.L.; Formal analysis: H.L., I.G.C., M.L., P.J.W.; Investigation: H.L., I.G.C.; Resources: K.M.B., A.J.D.; Writing - original draft: H.L., P.J.W.; Writing - review & editing: H.L., I.G.C., K.M.B., A.J.D., M.L., P.J.W.; Visualization: H.L., I.G.C.; Supervision: M.L., P.J.W.; Project administration: P.J.W.; Funding acquisition: H.L., M.L., P.J.W.

Funding

This study was supported by the National Institutes of Health/Eunice Kennedy Shriver National Institute of Child Health and Human Development (HD069592 and HD068157 to P.J.W.), the National Natural Science Foundation of China (32270905 to M.L), and a China Scholarship Council fellowship (to H.L.). Deposited in PMC for release after 12 months.

Data availability

All the data were provided in the manuscript and the supplementary information.

Abe
,
H.
,
Meduri
,
R.
,
Li
,
Z.
,
Andreassen
,
P. R.
and
Namekawa
,
S. H.
(
2021
).
RNF8 is not required for histone-to-protamine exchange in spermiogenesisdagger
.
Biol. Reprod.
105
,
1154
-
1159
.
Bernt
,
K. M.
,
Zhu
,
N.
,
Sinha
,
A. U.
,
Vempati
,
S.
,
Faber
,
J.
,
Krivtsov
,
A. V.
,
Feng
,
Z.
,
Punt
,
N.
,
Daigle
,
A.
,
Bullinger
,
L.
et al. 
(
2011
).
MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L
.
Cancer Cell
20
,
66
-
78
.
Bryant
,
J. M.
,
Donahue
,
G.
,
Wang
,
X.
,
Meyer-Ficca
,
M.
,
Luense
,
L. J.
,
Weller
,
A. H.
,
Bartolomei
,
M. S.
,
Blobel
,
G. A.
,
Meyer
,
R. G.
,
Garcia
,
B. A.
et al. 
(
2015
).
Characterization of BRD4 during mammalian postmeiotic sperm development
.
Mol. Cell. Biol.
35
,
1433
-
1448
.
Deshpande
,
A. J.
,
Deshpande
,
A.
,
Sinha
,
A. U.
,
Chen
,
L.
,
Chang
,
J.
,
Cihan
,
A.
,
Fazio
,
M.
,
Chen
,
C. W.
,
Zhu
,
N.
,
Koche
,
R.
et al. 
(
2014
).
AF10 regulates progressive H3K79 methylation and HOX gene expression in diverse AML subtypes
.
Cancer Cell
26
,
896
-
908
.
Feng
,
Q.
,
Wang
,
H.
,
Ng
,
H. H.
,
Erdjument-Bromage
,
H.
,
Tempst
,
P.
,
Struhl
,
K.
and
Zhang
,
Y.
(
2002
).
Methylation of H3-lysine 79 is mediated by a new family of HMTases without a SET domain
.
Curr. Biol.
12
,
1052
-
1058
.
Feng
,
Y.
,
Yang
,
Y.
,
Ortega
,
M. M.
,
Copeland
,
J. N.
,
Zhang
,
M.
,
Jacob
,
J. B.
,
Fields
,
T. A.
,
Vivian
,
J. L.
and
Fields
,
P. E.
(
2010
).
Early mammalian erythropoiesis requires the Dot1L methyltransferase
.
Blood
116
,
4483
-
4491
.
Gallardo
,
T.
,
Shirley
,
L.
,
John
,
G. B.
and
Castrillon
,
D. H.
(
2007
).
Generation of a germ cell-specific mouse transgenic Cre line, Vasa-Cre
.
Genesis
45
,
413
-
417
.
Gaucher
,
J.
,
Boussouar
,
F.
,
Montellier
,
E.
,
Curtet
,
S.
,
Buchou
,
T.
,
Bertrand
,
S.
,
Hery
,
P.
,
Jounier
,
S.
,
Depaux
,
A.
,
Vitte
,
A. L.
et al. 
(
2012
).
Bromodomain-dependent stage-specific male genome programming by Brdt
.
EMBO J.
31
,
3809
-
3820
.
Hasegawa
,
K.
,
Sin
,
H. S.
,
Maezawa
,
S.
,
Broering
,
T. J.
,
Kartashov
,
A. V.
,
Alavattam
,
K. G.
,
Ichijima
,
Y.
,
Zhang
,
F.
,
Bacon
,
W. C.
,
Greis
,
K. D.
et al. 
(
2015
).
SCML2 establishes the male germline epigenome through regulation of histone H2A ubiquitination
.
Dev. Cell
32
,
574
-
588
.
Jones
,
B.
,
Su
,
H.
,
Bhat
,
A.
,
Lei
,
H.
,
Bajko
,
J.
,
Hevi
,
S.
,
Baltus
,
G. A.
,
Kadam
,
S.
,
Zhai
,
H.
,
Valdez
,
R.
et al. 
(
2008
).
The histone H3K79 methyltransferase Dot1L is essential for mammalian development and heterochromatin structure
.
PLoS Genet.
4
,
e1000190
.
Kim
,
S. K.
,
Jung
,
I.
,
Lee
,
H.
,
Kang
,
K.
,
Kim
,
M.
,
Jeong
,
K.
,
Kwon
,
C. S.
,
Han
,
Y. M.
,
Kim
,
Y. S.
,
Kim
,
D.
et al. 
(
2012
).
Human histone H3K79 methyltransferase DOT1L protein [corrected] binds actively transcribing RNA polymerase II to regulate gene expression
.
J. Biol. Chem.
287
,
39698
-
39709
.
Lacoste
,
N.
,
Utley
,
R. T.
,
Hunter
,
J. M.
,
Poirier
,
G. G.
and
Cote
,
J.
(
2002
).
Disruptor of telomeric silencing-1 is a chromatin-specific histone H3 methyltransferase
.
J. Biol. Chem.
277
,
30421
-
30424
.
Lascarez-Lagunas
,
L. I.
,
Herruzo
,
E.
,
Grishok
,
A.
,
San-Segundo
,
P. A.
and
Colaiacovo
,
M. P.
(
2020
).
DOT-1.1-dependent H3K79 methylation promotes normal meiotic progression and meiotic checkpoint function in C. elegans
.
PLoS Genet.
16
,
e1009171
.
Li
,
W.
,
Wu
,
J.
,
Kim
,
S. Y.
,
Zhao
,
M.
,
Hearn
,
S. A.
,
Zhang
,
M. Q.
,
Meistrich
,
M. L.
and
Mills
,
A. A.
(
2014
).
Chd5 orchestrates chromatin remodelling during sperm development
.
Nat. Commun.
5
,
3812
.
Liao
,
J.
and
Szabo
,
P. E.
(
2020
).
Maternal DOT1L is dispensable for mouse development
.
Sci. Rep.
10
,
20636
.
Lin
,
Z.
,
Hsu
,
P. J.
,
Xing
,
X.
,
Fang
,
J.
,
Lu
,
Z.
,
Zou
,
Q.
,
Zhang
,
K. J.
,
Zhang
,
X.
,
Zhou
,
Y.
,
Zhang
,
T.
et al. 
(
2017
).
Mettl3-/Mettl14-mediated mRNA N(6)-methyladenosine modulates murine spermatogenesis
.
Cell Res.
27
,
1216
-
1230
.
Lin
,
H.
,
Cheng
,
K.
,
Kubota
,
H.
,
Lan
,
Y.
,
Riedel
,
S. S.
,
Kakiuchi
,
K.
,
Sasaki
,
K.
,
Bernt
,
K. M.
,
Bartolomei
,
M. S.
,
Luo
,
M.
et al. 
(
2022
).
Histone methyltransferase DOT1L is essential for self-renewal of germline stem cells
.
Genes Dev.
36
,
752
-
763
.
Lu
,
L.-Y.
,
Wu
,
J.
,
Ye
,
L.
,
Gavrilina
,
G. B.
,
Saunders
,
T. L.
and
Yu
,
X.
(
2010
).
RNF8-dependent histone modifications regulate nucleosome removal during spermatogenesis
.
Dev. Cell
18
,
371
-
384
.
Luense
,
L. J.
,
Donahue
,
G.
,
Lin-Shiao
,
E.
,
Rangel
,
R.
,
Weller
,
A. H.
,
Bartolomei
,
M. S.
and
Berger
,
S. L.
(
2019
).
Gcn5-mediated histone acetylation governs nucleosome dynamics in spermiogenesis
.
Dev. Cell
51
,
745
-
758.e746
.
Luo
,
M.
,
Zhou
,
J.
,
Leu
,
N. A.
,
Abreu
,
C. M.
,
Wang
,
J.
,
Anguera
,
M. C.
,
De Rooij
,
D. G.
,
Jasin
,
M.
and
Wang
,
P. J.
(
2015
).
Polycomb protein SCML2 associates with USP7 and counteracts histone H2A ubiquitination in the XY chromatin during male meiosis
.
PLoS Genet.
11
,
e1004954
.
Malla
,
A. B.
,
Yu
,
H.
,
Kadimi
,
S.
,
Lam
,
T.
,
Cox
,
A. L.
,
Smith
,
Z. D.
and
Lesch
,
B. J.
(
2021
).
DOT1L bridges transcription and heterochromatin formation at pericentromeres
.
bioRxiv
Malla
,
A. B.
,
Rainsford
,
S. R.
,
Smith
,
Z. D.
and
Lesch
,
B. J.
(
2023
).
DOT1L promotes spermatid differentiation by regulating expression of genes required for histone-to-protamine replacement
.
Development
150
,
dev201497
.
Mcginty
,
R. K.
,
Kim
,
J.
,
Chatterjee
,
C.
,
Roeder
,
R. G.
and
Muir
,
T. W.
(
2008
).
Chemically ubiquitylated histone H2B stimulates hDot1L-mediated intranucleosomal methylation
.
Nature
453
,
812
-
816
.
Meistrich
,
M. L.
,
Trostle-Weige
,
P. K.
,
Lin
,
R.
,
Bhatnagar
,
Y. M.
and
Allis
,
C. D.
(
1992
).
Highly acetylated H4 is associated with histone displacement in rat spermatids
.
Mol. Reprod. Dev.
31
,
170
-
181
.
Namekawa
,
S. H.
,
Park
,
P. J.
,
Zhang
,
L. F.
,
Shima
,
J. E.
,
Mccarrey
,
J. R.
,
Griswold
,
M. D.
and
Lee
,
J. T.
(
2006
).
Postmeiotic sex chromatin in the male germline of mice
.
Curr. Biol.
16
,
660
-
667
.
Ng
,
H. H.
,
Feng
,
Q.
,
Wang
,
H.
,
Erdjument-Bromage
,
H.
,
Tempst
,
P.
,
Zhang
,
Y.
and
Struhl
,
K.
(
2002
).
Lysine methylation within the globular domain of histone H3 by Dot1 is important for telomeric silencing and Sir protein association
.
Genes Dev.
16
,
1518
-
1527
.
Okada
,
Y.
,
Feng
,
Q.
,
Lin
,
Y.
,
Jiang
,
Q.
,
Li
,
Y.
,
Coffield
,
V. M.
,
Su
,
L.
,
Xu
,
G.
and
Zhang
,
Y.
(
2005
).
hDOT1L links histone methylation to leukemogenesis
.
Cell
121
,
167
-
178
.
Oliva
,
R.
and
Mezquita
,
C.
(
1982
).
Histone H4 hyperacetylation and rapid turnover of its acetyl groups in transcriptionally inactive rooster testis spermatids
.
Nucleic Acids Res.
10
,
8049
-
8059
.
Oliver
,
S. S.
,
Musselman
,
C. A.
,
Srinivasan
,
R.
,
Svaren
,
J. P.
,
Kutateladze
,
T. G.
and
Denu
,
J. M.
(
2012
).
Multivalent recognition of histone tails by the PHD fingers of CHD5
.
Biochemistry
51
,
6534
-
6544
.
Ontoso
,
D.
,
Kauppi
,
L.
,
Keeney
,
S.
and
San-Segundo
,
P. A.
(
2014
).
Dynamics of DOT1L localization and H3K79 methylation during meiotic prophase I in mouse spermatocytes
.
Chromosoma
123
,
147
-
164
.
Peters
,
A. H.
,
Plug
,
A. W.
,
Van Vugt
,
M. J.
and
De Boer
,
P.
(
1997
).
A drying-down technique for the spreading of mammalian meiocytes from the male and female germline
.
Chromosome Res.
5
,
66
-
68
.
Pivot-Pajot
,
C.
,
Caron
,
C.
,
Govin
,
J.
,
Vion
,
A.
,
Rousseaux
,
S.
and
Khochbin
,
S.
(
2003
).
Acetylation-dependent chromatin reorganization by BRDT, a testis-specific bromodomain-containing protein
.
Mol. Cell. Biol.
23
,
5354
-
5365
.
Reisenauer
,
M. R.
,
Anderson
,
M.
,
Huang
,
L.
,
Zhang
,
Z.
,
Zhou
,
Q.
,
Kone
,
B. C.
,
Morris
,
A. P.
,
Lesage
,
G. D.
,
Dryer
,
S. E.
and
Zhang
,
W.
(
2009
).
AF17 competes with AF9 for binding to Dot1a to up-regulate transcription of epithelial Na+ channel alpha
.
J. Biol. Chem.
284
,
35659
-
35669
.
Rousseaux
,
S.
,
Reynoird
,
N.
,
Escoffier
,
E.
,
Thevenon
,
J.
,
Caron
,
C.
and
Khochbin
,
S.
(
2008
).
Epigenetic reprogramming of the male genome during gametogenesis and in the zygote
.
Reprod. Biomed. Online
16
,
492
-
503
.
San-Segundo
,
P. A.
and
Roeder
,
G. S.
(
2000
).
Role for the silencing protein Dot1 in meiotic checkpoint control
.
Mol. Biol. Cell
11
,
3601
-
3615
.
Shang
,
E.
,
Nickerson
,
H. D.
,
Wen
,
D.
,
Wang
,
X.
and
Wolgemuth
,
D. J.
(
2007
).
The first bromodomain of Brdt, a testis-specific member of the BET sub-family of double-bromodomain-containing proteins, is essential for male germ cell differentiation
.
Development
134
,
3507
-
3515
.
Shi
,
B.
,
Xue
,
J.
,
Yin
,
H.
,
Guo
,
R.
,
Luo
,
M.
,
Ye
,
L.
,
Shi
,
Q.
,
Huang
,
X.
,
Liu
,
M.
,
Sha
,
J.
et al. 
(
2019
).
Dual functions for the ssDNA-binding protein RPA in meiotic recombination
.
PLoS Genet.
15
,
e1007952
.
Van Leeuwen
,
F.
,
Gafken
,
P. R.
and
Gottschling
,
D. E.
(
2002
).
Dot1p modulates silencing in yeast by methylation of the nucleosome core
.
Cell
109
,
745
-
756
.
Vlaming
,
H.
and
Van Leeuwen
,
F.
(
2016
).
The upstreams and downstreams of H3K79 methylation by DOT1L
.
Chromosoma
125
,
593
-
605
.
Yang
,
F.
,
De La Fuente
,
R.
,
Leu
,
N. A.
,
Baumann
,
C.
,
Mclaughlin
,
K. J.
and
Wang
,
P. J.
(
2006
).
Mouse SYCP2 is required for synaptonemal complex assembly and chromosomal synapsis during male meiosis
.
J. Cell Biol.
173
,
497
-
507
.
Yang
,
H.
,
Wang
,
H.
and
Jaenisch
,
R.
(
2014
).
Generating genetically modified mice using CRISPR/Cas-mediated genome engineering
.
Nat. Protoc.
9
,
1956
-
1968
.
Zhou
,
J.
,
Yang
,
F.
,
Leu
,
N. A.
and
Wang
,
P. J.
(
2012
).
MNS1 is essential for spermiogenesis and motile ciliary functions in mice
.
PLoS Genet.
8
,
e1002516
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information