Neurogenesis is initiated by basic helix-loop-helix proneural proteins. Here, we show that Actin-related protein 6 (Arp6), a core component of the H2A.Z exchange complex SWR1, interacts with proneural proteins and is crucial for efficient onset of proneural protein target gene expression. Arp6 mutants exhibit reduced transcription in sensory organ precursors (SOPs) downstream of the proneural protein patterning event. This leads to retarded differentiation and division of SOPs and smaller sensory organs. These phenotypes are also observed in proneural gene hypomorphic mutants. Proneural protein expression is not reduced in Arp6 mutants. Enhanced proneural gene expression fails to rescue retarded differentiation in Arp6 mutants, suggesting that Arp6 acts downstream of or in parallel with proneural proteins. H2A.Z mutants display Arp6-like retardation in SOPs. Transcriptomic analyses demonstrate that loss of Arp6 and H2A.Z preferentially decreases expression of proneural protein-activated genes. H2A.Z enrichment in nucleosomes around the transcription start site before neurogenesis correlates highly with greater activation of proneural protein target genes by H2A.Z. We propose that upon proneural protein binding to E-box sites, H2A.Z incorporation around the transcription start site allows rapid and efficient activation of target genes, promoting rapid neural differentiation.

The basic helix-loop-helix (bHLH) proneural proteins are neural-inducing factors that confer neural fate to neuroepithelium or ectodermal cells. They promote neural fate determination, neural type specification and neural cell differentiation both in vivo and in vitro by transcriptional activation of a series of downstream genes (Bertrand et al., 2002; Wapinski et al., 2013). Nucleosome modification by histone acetyltransferase and demethylase and nucleosome relocation by the SWI/SNF remodeling complex are essential for regulation of proneural proteins to activate transcription in vertebrate cells (Koyano-Nakagawa et al., 1999; Lee et al., 2009; Lin et al., 2017; Seo et al., 2005).

Extensive studies in recent years have revealed that histone variant exchange alters the properties of the nucleosome core, thereby influencing chromatin-associated events, such as transcription, genome stability and DNA repair (reviewed by Martire and Banaszynski, 2020). The canonical H2A has two variants, H2A.Z and H2A.X, involved in nucleosome function and the DNA-damage response, respectively. H2A.Z typically constitutes 5-10% of the total H2A (Bonnet et al., 2019; Redon et al., 2002) and is incorporated into a subset of nucleosomes at nonrandom locations throughout the genome. H2A.Z is enriched within a few nucleosomes surrounding the transcriptional start site (TSS) (Barski et al., 2007; Guillemette et al., 2005; Mavrich et al., 2008; Whittle et al., 2008; Zilberman et al., 2008), although the extent of incorporation varies between different genes.

One of the significant functions of H2A.Z is transcriptional activation. H2A.Z incorporation levels largely correlate with gene expression in the metazoan (Mavrich et al., 2008; Whittle et al., 2008). It is also enriched in genes poised for activation in human cells (Hardy et al., 2009; Ku et al., 2012). Mechanistically, H2A.Z is required to recruit RNA polymerase II (Pol II) to the promoters (Adam et al., 2001). The height of the +1 nucleosome barrier to Pol II inversely correlates with enrichment of H2A.Z (Chen et al., 2019; Weber et al., 2014). Besides transcriptional activation, H2A.Z is implicated in transcriptional repression, gene silencing, and heterochromatin formation and spreading (reviewed by Giaimo et al., 2019).

H2A.Z is incorporated into nucleosomes by the highly conserved SWR1 complex. It partially unwraps DNA from the canonical histone core and catalyzes the replacement of an H2A-H2B dimer with an H2A.Z–H2B dimer (Hong et al., 2014; Willhoft et al., 2018). Actin-related protein 6 (Arp6) is a highly conserved component of the SWR1 complex. It physically interacts with the core ATPase of SWR1 and is essential for complex integrity and histone exchange activity (Scacchetti et al., 2020; Wu et al., 2005). Arp6 is a member of the actin superfamily (Muller et al., 2005; Poch and Winsor, 1997), and both Arp6 and nuclear actin are conserved members of the SWR1 complex in yeast and mammals (Mizuguchi et al., 2004; Ruhl et al., 2006).

Drosophila external sensory (ES) organ development is a classical model in which to study neurogenesis initiated by bHLH proneural proteins. Achaete (Ac) and Scute (Sc) were the first members of the highly conserved proneural protein family to be discovered (Villares and Cabrera, 1987). Both proteins are first expressed in proneural stripes to endow neural potential to the cells. Subsequently, high-level Ac and Sc are restricted to single sensory organ precursors (SOPs) to promote precursor determination, selection and differentiation by transcriptional activation of an array of downstream genes. Here, we report the crucial role of Arp6-mediated H2A.Z replacement for rapid SOP differentiation and division downstream of the proneural protein patterning event. We found that Arp6 is required for efficient transcription of Ac- and Sc-target genes in SOPs. Genetic analyses suggest that Arp6 functions downstream of or in parallel with proneural proteins to activate the SOP differentiation program. Transcriptomic analyses showed that both Arp6 and H2A.Z are preferentially required for proneural protein-induced transcription. Loss of H2A.Z did not reduce proneural protein binding to high-affinity E-box sites, suggesting that E-box binding is independent of H2A.Z enhancer incorporation. phyl is a proneural protein target gene that requires Arp6 and H2A.Z for its maximal expression. H2A.Z was incorporated into the nucleosome around the TSS of phyl in the absence of proneural proteins, and recruitment of proneural proteins to the E-box enhances H2A.Z incorporation through Arp6. Database analysis of the H2A.Z replacement at the +1 nucleosome preceding neurogenesis revealed a strong correlation between H2A.Z enrichment and efficient gene expression by proneural proteins. We propose that, in the absence of H2A.Z replacement at the +1 nucleosome, proneural proteins still bind to the cognate target sites with high affinity but initiate transcription at a slower rate, thus retarding precursor differentiation.

Arp6 associates with proneural proteins and is required for normal ES organ developmental timing

We have previously reported that nuclear actin physically interacts with Ac and Sc and promotes Ac-dependent gene expression (Hsiao et al., 2014). Nuclear actin is a component of several chromatin remodeling complexes, including the SWR1 complex (Cao et al., 2016; Klages-Mundt et al., 2018). Interestingly, the SWR1 complex component Arp6 was also listed as the candidate Ac-interacting protein in the same mass spectrometry analysis. To confirm the interaction between Arp6 and proneural proteins, we performed co-immunoprecipitation using the nuclear extract isolated from S2 cells. We showed that Arp6 was associated with Ac (lane 4 in Fig. 1A). The interaction between Arp6 and Ac was relatively specific as Ac either did not associate or barely associated with the other nuclear actin proteins Arp4, Arp5 and Arp8 (Fig. 1A) (reviewed by Klages-Mundt et al., 2018). We further tested the interaction between Arp6 and other proneural proteins. We found that Arp6 also interacted with Sc and Atonal (Ato) but failed to interact with the proneural protein heterodimeric partner Daughterless (Da) (Fig. 1B). Our results show that Arp6 physically interacts with proneural proteins Ac, Sc and Ato.

Fig. 1.

Arp6 is required for normal ES organ growth. (A) SDS-PAGE of anti-Flag co-immunoprecipitates from nuclear extracts of S2 cells transfected with plasmids encoding Myc-Ac and Flag-tagged Arp4, 5, 6, 8 or GFP. (B) SDS-PAGE of anti-Flag co-immunoprecipitates from nuclear extracts of S2 cells expressing Flag-Arp6 (or GFP) and Myc-tagged Ac, Ato, Da, Actin or Sc. Asterisks mark non-specific bands. (C) Diagrams of the Arp61 and Arp6D alleles generated by CRISPR-Cas9 and homologous-directed recombination (see Materials and Methods for details). Arp6 is encoded by a single exon. Black boxes represent the 5′ UTR and black triangles represent the 3′ UTR. White boxes are the protein-coding region. Red box indicates a 3xP3-RFP selectable marker cassette. Arp61 was generated by gRNA-1 and Arp6D was generated by gRNA-1 and gRNA-2. (D) RT-PCR analysis of Arp6 mRNA levels in whole larval extracts. Arp61 hemizygotes (lane 2) and Arp6D hemizygotes (lane 3) produced little and no Arp6 mRNA, respectively. (E) Numbers of viable progeny produced from the rescue experiment of Arp61 hemizygotes by Act5C-Gal4-driven Arp6 expression (see Materials and Methods). Exogenous expression of HA-Arp6 rescued the lethality of Arp61 mutants. (F,G) Adult nota with Arp61 (F) and Arp6D (G) mutant clones. Clones are marked by y and boundaries are marked by the dotted line. Red arrowheads indicate Arp6+/− macrochaetae, red arrows Arp6+/− macrochaetae, yellow arrowheads Arp6−/− macrochaetae and yellow arrows Arp6−/− microchaetae. (H) Rescue of small ES organ size in Arp61 MARCM clone by Arp6 expression via tubP-Gal4. (I) Quantification of shaft length. Graph shows the relative shaft length in homozygous versus heterozygous mutant tissue. The numbers of ES organs examined are listed at the bottom of each column. ****P<0.0001. ns, not significant. (J,K) Pupal nota with Arp61 clones. Pros-positive sheath cells (J) and Elav-positive neurons (K) were specified in the mutant clones. Scale bars: 50 µm. Clonal boundaries are marked by white lines, and clones are indicated by arrows. IB, immunoblot; IP, immunoprecipitation.

Fig. 1.

Arp6 is required for normal ES organ growth. (A) SDS-PAGE of anti-Flag co-immunoprecipitates from nuclear extracts of S2 cells transfected with plasmids encoding Myc-Ac and Flag-tagged Arp4, 5, 6, 8 or GFP. (B) SDS-PAGE of anti-Flag co-immunoprecipitates from nuclear extracts of S2 cells expressing Flag-Arp6 (or GFP) and Myc-tagged Ac, Ato, Da, Actin or Sc. Asterisks mark non-specific bands. (C) Diagrams of the Arp61 and Arp6D alleles generated by CRISPR-Cas9 and homologous-directed recombination (see Materials and Methods for details). Arp6 is encoded by a single exon. Black boxes represent the 5′ UTR and black triangles represent the 3′ UTR. White boxes are the protein-coding region. Red box indicates a 3xP3-RFP selectable marker cassette. Arp61 was generated by gRNA-1 and Arp6D was generated by gRNA-1 and gRNA-2. (D) RT-PCR analysis of Arp6 mRNA levels in whole larval extracts. Arp61 hemizygotes (lane 2) and Arp6D hemizygotes (lane 3) produced little and no Arp6 mRNA, respectively. (E) Numbers of viable progeny produced from the rescue experiment of Arp61 hemizygotes by Act5C-Gal4-driven Arp6 expression (see Materials and Methods). Exogenous expression of HA-Arp6 rescued the lethality of Arp61 mutants. (F,G) Adult nota with Arp61 (F) and Arp6D (G) mutant clones. Clones are marked by y and boundaries are marked by the dotted line. Red arrowheads indicate Arp6+/− macrochaetae, red arrows Arp6+/− macrochaetae, yellow arrowheads Arp6−/− macrochaetae and yellow arrows Arp6−/− microchaetae. (H) Rescue of small ES organ size in Arp61 MARCM clone by Arp6 expression via tubP-Gal4. (I) Quantification of shaft length. Graph shows the relative shaft length in homozygous versus heterozygous mutant tissue. The numbers of ES organs examined are listed at the bottom of each column. ****P<0.0001. ns, not significant. (J,K) Pupal nota with Arp61 clones. Pros-positive sheath cells (J) and Elav-positive neurons (K) were specified in the mutant clones. Scale bars: 50 µm. Clonal boundaries are marked by white lines, and clones are indicated by arrows. IB, immunoblot; IP, immunoprecipitation.

Close modal

To study the role of Arp6 in ac- and sc-dependent ES organ development, we generated a loss-of-function insertion allele (Arp61), a deletion allele (Arp6D) and a HA knock-in allele (Arp6HA) in which the 3XHA epitope was inserted at the end of the open reading frame by the CRISPR/Cas9 technique (see Materials and Methods and Fig. 1C and Fig. S2A). Compared with controls, Arp6 transcripts were greatly reduced in Arp61 hemizygous larvae and not detected in Arp6D hemizygotes (Fig. 1D). Arp6 mRNA level was moderately reduced by 20-40% in Arp6HA hemizygotes compared with the controls (Fig. S2B). Arp61 and Arp6D hemizygotes were lethal and most of them were arrested by early pupal stages. Arp6HA hemizygotes were semi-lethal and a few adults eclosed. The lethality of Arp61 could be rescued by the ubiquitous expression of Arp6 from a UAS-Arp6 transgene driven by Act5C-Gal4 (Fig. 1E), confirming that Arp61 is an Arp6 mutant allele.

To examine ES organ phenotypes, we generated Arp61 and Arp6D mutant clones on the notum. In both Arp61 and Arp6D clones, macrochaetae (yellow arrowheads) and microchaetae (yellow arrows) were reduced in size, exhibiting shorter and thinner shafts and smaller sockets, compared with non-clonal macrochaetae (red arrowheads) and microchaetae (red arrows) (Fig. 1F,G, and shaft length quantification in Fig. 1I). As a control, we also generated clones for yellow (y), which is used to mark the clone. The ES organs in the y clones showed no difference in shaft length compared with the y/+ heterozygous shaft (Fig. 1I). The reduction in ES organ size in Arp61 clones was completely rescued by ubiquitous expression of Arp6 driven by tubP-Gal4 (Fig. 1H), indicating that Arp6 is involved in normal ES organ development. The smaller ES organ size did not result from a general defect in cell growth because the clone size of Arp61 homozygous mutant was comparable to that of the homozygous wild-type twin spot (Fig. S1A). We found that the spacing between microchaetae was relatively unaffected in most clones, but was noticeably reduced in the large Arp6 mutant clones (Fig. S1B,C). The size of ES organs appeared normal in Arp6HA hemizygotes (Fig. S2C).

Although the sizes of shafts and sockets were smaller in Arp61 and Arp6D clones, most of them were present, indicating that the fate determination of outer support cells was largely normal. We also examined the inner daughter cells of the ES organs by dissecting notal tissue 24 h after puparium formation (APF). We found that the expression of Prospero (Pros) and Elav, which label sheath cells and neurons, respectively, were present in the Arp61 mutant clones (Fig. 1J,K). Careful examination of the numbers of Pros-positive and Elav-positive cells in Arp61 clones showed that loss of Arp6 caused little change in the number of sheath cells (3% reduction compared with heterozygous, n=30 in six clones) and a moderate reduction in the number of neurons (30% reduction compared with heterozygous, n=30 in seven clones). Together, these analyses indicate that the fate determination of ES organ daughter cells is mostly unaffected in Arp6 mutants.

Arp6 is required for timely SOP differentiation and division

We analyzed further how ES organ development is affected in Arp6 mutant clones by examining notal tissue dissected in a series of time points in pupal stages. We first examined the expression of the proneural protein Ac. We found that the pattern and level of Ac in proneural stripes were unaffected by the lack of Arp6 at 9 h APF (red arrows in Fig. 2A), suggesting that the initiation of Ac expression was normal. Whereas Ac-positive SOPs were isolated in non-clonal Arp61/+ cells at 10 h APF, Ac expression remained in clusters of cells in Arp61 clones (Fig. 2B). Single Ac-positive cells in Arp61 clones were observed at 12 h APF when the Ac level in Arp61/+ cells was diminished (Fig. 2C). Ac was undetected in both Arp61 clonal and Arp61/+ cells at 14 h APF (Fig. 2D). These results suggest a delay in the enrichment of Ac in SOP cells. We also examined Sc expression in a Sc-GFP knock-in line in which the C terminus of Sc is fused with the GFP (Chen et al., 2018). We found a similar delay in Sc expression in SOPs (Fig. 2E,F).

Fig. 2.

Arp6 is required for timely SOP development. (A-D′) Pupal nota with Arp61 clones. Proneural stripes and newly selected SOPs were labeled by anti-Ac staining (A-D). Differentiating SOPs were labeled by anti-Sens staining (A′-D′). Retardation in Arp61 mutant clones was observed in single SOP selection from proneural stripes (B), Ac downregulation (B,C) and Sens upregulation in differentiating SOPs (B′-D′). (E,F) Pupal nota of the knock-in sc-GFP with Arp61 clones. Sc-GFP restriction to single SOPs (E) and Sc-GFP downregulation in differentiating SOPs were retarded in mutant clones. (G-I) Divisions of SOPs and SOP lineage cells were retarded in Arp61 mutant clones. Hnt and Cut label the late SOPs and SOP lineage cells. All clonal boundaries are marked by yellow or white lines, and clones are indicated by arrows. Scale bars: 50 µm.

Fig. 2.

Arp6 is required for timely SOP development. (A-D′) Pupal nota with Arp61 clones. Proneural stripes and newly selected SOPs were labeled by anti-Ac staining (A-D). Differentiating SOPs were labeled by anti-Sens staining (A′-D′). Retardation in Arp61 mutant clones was observed in single SOP selection from proneural stripes (B), Ac downregulation (B,C) and Sens upregulation in differentiating SOPs (B′-D′). (E,F) Pupal nota of the knock-in sc-GFP with Arp61 clones. Sc-GFP restriction to single SOPs (E) and Sc-GFP downregulation in differentiating SOPs were retarded in mutant clones. (G-I) Divisions of SOPs and SOP lineage cells were retarded in Arp61 mutant clones. Hnt and Cut label the late SOPs and SOP lineage cells. All clonal boundaries are marked by yellow or white lines, and clones are indicated by arrows. Scale bars: 50 µm.

Close modal

The high-level proneural proteins in SOPs induce the expression of senseless (sens), which is a direct target of proneural proteins (Jafar-Nejad et al., 2003). Delays in Sens expression were also observed in the time series of notal tissues co-stained for Ac and Sens (Fig. 2A′-D′). Quantification of Sens levels in SOPs at 14 h APF confirmed that it was significantly decreased by 32% in Arp61 clones compared with the neighboring Arp61/+ cells (Fig. 2D′; Fig. S1D), indicating that Arp6 is required for maximal Sens expression. SOPs further divide into two daughter cells that could be labeled by Hindsight (Hnt; also known as Peb) and Cut proteins (Blochlinger et al., 1990; Pickup et al., 2002). Although Hnt and Cut expressions were detected in two adjacent daughter cells after SOP division in Arp61/+ cells, they were mostly localized in single, isolated cells in Arp61 clones (Fig. 2G,H). A delay in later cell division into three- to four-cell clusters was also observed (Fig. 2I). Taken together, the phenotypic analyses of Arp6 mutants demonstrate defects in the timely differentiation and division of SOP cells.

Hypomorphic ac sc mutant recapitulates Arp6 mutant phenotypes

In mutants lacking ac and sc activity, ES organs on the notum are completely absent (Campuzano et al., 1986; Dambly-Chaudiére and Ghysen, 1987). It is unclear whether partially lacking ac and sc would also lead to a delay in ES organ development. We tested this idea by examining ES organ phenotypes for the hypomorphic allele acsbm, a P-element insertion mutant affecting both ac and sc expression (Marcellini et al., 2005). In the acsbm clones at the adult nota, ES organs were smaller with a noticeable reduction in the shaft length compared with those in the neighboring heterozygous tissue (Fig. 3A,B). Like Arp61 mutant clones, upregulation of Sens was retarded in acsbm clones (Fig. 3C). Delays in cell division into two cells and further into three- to four-cell clusters or even into four to five-cell clusters were observed (Fig. 3D-F). Although retarded, sensory organ cell differentiation appeared normal because shaft and socket were found in every adult bristle in acsbm clones (Fig. 3A). Elav-positive cells were observed accompanying each Su(H)-positive socket cell at 28 h APF (Fig. 3G), indicating correct cell fate determination of neurons. These analyses together demonstrate that reduced proneural protein level retards SOP differentiation and division, but cell fate determination of daughter cells remain largely unaffected, as observed in Arp6 mutants.

Fig. 3.

Arp6 acts synergistically with proneural genes to promote SOP development. (A) Adult notum with an acsbm clone. ES organs were smaller in the clone, marked by y. (B) Quantification of shaft length. Graph shows the relative shaft length of acsbm homozygous versus acsbm heterozygous ES organs. The numbers of shafts examined are listed at the bottom of each column. ****P<0.0001. (C-G) Pupal nota with acsbm clones. Divisions of SOPs and SOP lineage cells were retarded in acsbm mutant clones (C-F). SOP lineage cells are labeled by anti-Sens (C,D), anti-Hnt (E), and anti-Cut (F) antibody staining. Elav-positive neurons (green) and Su(H)-positive socket cells (red) are present in acsbm clones (G). (H,I) Sens expression was partially reduced in Arp61 (H) clones and eliminated in acsbm Arp61 double-mutant clones (I). (J) Loss of ES organs in acsbm Arp61 double-mutant clone marked by y. (K,K′) Overexpression of ac failed to rescue Sens expression in Arp61 clones. Flag-ac was expressed in proneural stripes and SOPs by sca-Gal4. Clonal boundaries are marked by black or white lines, and clones are indicated by arrows. Scale bars: 50 µm.

Fig. 3.

Arp6 acts synergistically with proneural genes to promote SOP development. (A) Adult notum with an acsbm clone. ES organs were smaller in the clone, marked by y. (B) Quantification of shaft length. Graph shows the relative shaft length of acsbm homozygous versus acsbm heterozygous ES organs. The numbers of shafts examined are listed at the bottom of each column. ****P<0.0001. (C-G) Pupal nota with acsbm clones. Divisions of SOPs and SOP lineage cells were retarded in acsbm mutant clones (C-F). SOP lineage cells are labeled by anti-Sens (C,D), anti-Hnt (E), and anti-Cut (F) antibody staining. Elav-positive neurons (green) and Su(H)-positive socket cells (red) are present in acsbm clones (G). (H,I) Sens expression was partially reduced in Arp61 (H) clones and eliminated in acsbm Arp61 double-mutant clones (I). (J) Loss of ES organs in acsbm Arp61 double-mutant clone marked by y. (K,K′) Overexpression of ac failed to rescue Sens expression in Arp61 clones. Flag-ac was expressed in proneural stripes and SOPs by sca-Gal4. Clonal boundaries are marked by black or white lines, and clones are indicated by arrows. Scale bars: 50 µm.

Close modal

Next, we examined the functional interaction between proneural proteins and Arp6. Given that Ac and Sc levels were not reduced in Arp6 mutant clones, we tested whether Arp6 acts downstream or in parallel with proneural proteins in SOPs by rescue experiment. Overexpression of Ac by sca-Gal4 in a stripe pattern failed to restore the normal levels of Sens in SOPs of Arp61 clones (Fig. 3K,K′), indicating that the absence of Arp6 could not be replaced by enhanced Ac expression. We next examined the genetic interaction between Arp6 and ac sc by generating acsbm Arp61 double mutant clones. Whereas Sens was partially reduced in either acsbm or Arp61 mutants at 18 h APF (Fig. 3D,H), Sens expression was eliminated in the double-mutant clones (Fig. 3I) and the ES organs were almost absent in the double-mutant clone on adult notum (Fig. 3J). Together, these results support the suggestion that Arp6 functions downstream or in parallel with proneural proteins in SOPs to promote ES organ development.

Arp6 regulates proneural protein target gene transcription in SOPs

The retarded SOP differentiation and division in Arp6 mutant clones and the synergistic interaction with proneural genes prompted us to examine whether Arp6 regulates the proneural protein-activated transcriptional program. We first performed transcriptomic analysis to identify genes upregulated by proneural Ac-Da heterodimers. For comparison, we used the DNA-binding defective AcR29G mutant, in which arginine is replaced by glycine at the critical residue of the basic domain (Ma et al., 1994). We compared gene expression profiles of Ac-Da- and AcR29G-Da-transfected S2 cells. Among the 111 genes that were upregulated by at least 1.5-fold (log2 fold change>0.58) in two independent experiments, scabrous (sca) and phyllopod (phyl) were the top two upregulated genes (Table S1). Both sca and phyl are direct target genes of proneural proteins in SOPs (Mlodzik et al., 1990; Pi et al., 2004). We then performed quantitative RT-PCR to measure the levels of upregulation and showed that phyl expression was increased by more than 200-fold by Ac-Da heterodimers relative to AcR29G-Da, and sca was upregulated by 40-fold (Fig. 4A), confirming that phyl and sca are target genes of proneural proteins.

Fig. 4.

Arp6 regulates transcription of the Ac- and Sc-target genes phyl and sca in SOPs. (A) qRT-PCR analysis of Ac-Da-induced phyl and sca expression in S2 cells. Graph shows log2 fold changes in gene expression induced by Ac-Da relative to AcR29G-Da. n=4 biological repeats. (B,C) Pupal nota of phyl4.1-nGFP with Arp61 clones showing loss of Arp6 reduced phyl4.1-nGFP expression in proneural stripes (B) and SOPs (C). (D) Pupal notum of sca-lacZ with Arp61 clones. sca-lacZ expression was diminished in SOPs in mutant clones. Clonal boundaries are marked by white lines, and clones are indicated by arrows. Scale bars: 50 µm.

Fig. 4.

Arp6 regulates transcription of the Ac- and Sc-target genes phyl and sca in SOPs. (A) qRT-PCR analysis of Ac-Da-induced phyl and sca expression in S2 cells. Graph shows log2 fold changes in gene expression induced by Ac-Da relative to AcR29G-Da. n=4 biological repeats. (B,C) Pupal nota of phyl4.1-nGFP with Arp61 clones showing loss of Arp6 reduced phyl4.1-nGFP expression in proneural stripes (B) and SOPs (C). (D) Pupal notum of sca-lacZ with Arp61 clones. sca-lacZ expression was diminished in SOPs in mutant clones. Clonal boundaries are marked by white lines, and clones are indicated by arrows. Scale bars: 50 µm.

Close modal

Therefore, we examined whether the expression of phyl and sca could be modulated by Arp6 in SOPs. It has been shown that the 4.1 kb upstream regulatory region of phyl is sufficient to drive its expression in SOPs and rescues phyl mutant phenotypes when fused with phyl cDNA (Pi et al., 2004). The transcriptional reporter phyl4.1-nGFP was examined in Arp61 clones. At 12 h APF, phyl4.1-nGFP was expressed in strips of cells in Arp61/+ cells, and its expression was diminished in Arp61 clones (Fig. 4B). Reduced expression of phyl4.1-nGFP was also detected in single SOPs of Arp61 clones at 14 h APF (Fig. 4C). These results indicate that the Ac- and Sc-target gene phyl is regulated by Arp6 at the transcriptional level. We also examined whether sca transcription is regulated by Arp6 in SOPs. The enhancer trap sca-lacZ showed a markedly reduced expression in SOPs in the Arp61 clone compared with control Arp61/+ cells (Fig. 4D). Taken together, we conclude that Arp6 regulates the SOP genes phyl and sca at the transcriptional level.

H2A.Z is required for ES organ growth and SOP differentiation

Arp6 is a core component of the H2A.Z-exchange SWR1 complex, which is functionally conserved in yeast, fly and human (Ruhl et al., 2006; Scacchetti et al., 2020; Wu et al., 2005). In Arp61 clones, H2A.Z was eliminated from the developing notal cells (Fig. S3A). This could result from the degradation of free H2A.Z (Takahashi et al., 2017). Thus, we investigated whether loss of H2A.Z also impairs ES organ development. H2A.Z is encoded by a single copy of His2Av in the Drosophila genome. In His2Av810 null mutant clones, deformed bristles with tiny shafts and sockets were observed (Fig. 5A), demonstrating that H2A.Z is required for normal ES organ development. Although Ac expression in proneural stripes was largely unaffected (Fig. 5B), the progression of Ac proneural stripe to single SOP expression and diminish of Ac in SOPs were retarded in His2Av810 clones (Fig. 5C,D). Upregulation of Sens in SOPs, and SOP division to two daughter cells marked by Sens expression were also slowed down in His2Av810 clones (Fig. 5D′,E). In summary, His2Av810 clones, although presenting more severe phenotypes, recapitulate what was observed in Arp6 mutant clones. To evaluate further the functional interaction between Arp6 and H2A.Z in ES organ development, we examined the ES organ size by quantification of the shaft length in mutants partially defective for Arp6 and H2A.Z. H2A.Z was knocked down in the central region of the notum by RNA interference (RNAi) via pnr-Gal4. Compared with controls expressing RFP RNAi, knockdown of H2A.Z reduced the shaft length by 14% (Fig. S2D,E). In the heterozygous mutant carrying one copy of Arp6HA, knockdown of H2A.Z reduced the shaft length by a further 25% compared with the knockdown of RFP (Fig. S2D,E). The synergistic effect between Arp6 and H2A.Z to regulate shaft length suggests functional interaction in ES organ development.

Fig. 5.

H2A.Z is required for ES organ growth and timely SOP development. (A) Loss of H2A.Z in the His2Av810 mutant clone resulted in smaller ES organs, indicated by red arrows. (B-E) Pupal nota with His2Av810 clones. ac expression in proneural stripes was normal (B). However, SOP selection (C), Ac downregulation (D), Sens upregulation (D′), and division of SOP lineage cells (E) were severely affected in the mutant clones. (F-H′) Expression of Dom-B-GFP from the fosmid clone. (F-G′) Domino-B levels were initially comparable between Ac-positive proneural stripes (red in F′) and the neighboring epithelial cells, and between Sens-positive early SOPs (red in G′) and the neighboring epithelial cells. (H,H′) Higher Domino-B-GFP levels were observed in mature SOPs labeled by Hnt (red). Arrows indicate examples of Hnt-positive cells showing higher GFP intensity. Clonal boundaries are marked by yellow or white lines. Scale bars: 50 µm.

Fig. 5.

H2A.Z is required for ES organ growth and timely SOP development. (A) Loss of H2A.Z in the His2Av810 mutant clone resulted in smaller ES organs, indicated by red arrows. (B-E) Pupal nota with His2Av810 clones. ac expression in proneural stripes was normal (B). However, SOP selection (C), Ac downregulation (D), Sens upregulation (D′), and division of SOP lineage cells (E) were severely affected in the mutant clones. (F-H′) Expression of Dom-B-GFP from the fosmid clone. (F-G′) Domino-B levels were initially comparable between Ac-positive proneural stripes (red in F′) and the neighboring epithelial cells, and between Sens-positive early SOPs (red in G′) and the neighboring epithelial cells. (H,H′) Higher Domino-B-GFP levels were observed in mature SOPs labeled by Hnt (red). Arrows indicate examples of Hnt-positive cells showing higher GFP intensity. Clonal boundaries are marked by yellow or white lines. Scale bars: 50 µm.

Close modal

We next examined in more detail whether the Arp6-included SWR1 complex is regulated during SOP differentiation. In the Drosophila SWR1 complex, the catalytic core ATPase is encoded by the domino (dom)-B isoform (Scacchetti et al., 2020). We assessed Dom-B expression in a Dom-B-GFP protein trap line (Börner and Becker, 2016). The GFP protein was maintained at constant levels in proneural stripes and early Ac-positive SOPs compared with the epithelial cells (Fig. 5F-G′). Interestingly, the GFP level was elevated in the large, Hnt-positive SOPs (Fig. 5H,H′), suggesting that SWR1 activity might be upregulated in mature SOPs. Together, our data support that SWR1-dependent H2A.Z incorporation is required for timely SOP development downstream of the proneural patterning event.

Arp6 and H2A.Z is required for maximal gene expression activated by proneural proteins

Our analysis suggests that Arp6-mediated H2A.Z replacement might be required for the maximal expression of proneural protein target genes. We next assessed the requirement of Arp6 and H2A.Z for the Ac-Da heterodimer-dependent transcription in S2 cells by RNAi. In cells expressing Arp6 RNAi to reduce Arp6 expression by 93% (Fig. S3C), H2A.Z levels were reduced by 37% (Fig. S3B), indicating that H2A.Z incorporation is partially compromised by Arp6 knockdown in S2 cells. As shown in Fig. 4A, the Ac-Da heterodimer markedly induced phyl transcription in S2 cells. phyl expression was decreased by 24% (log2 fold change=−0.39) compared with control luciferase (Luc) knockdown (Fig. 6A). We also measured expression of the other target gene, sca, and found that the transcription induction was similarly compromised by Arp6 knockdown (Fig. 6A). Thus, the Ac-Da-induced maximal expression of phyl and sca requires the presence of Arp6.

Fig. 6.

H2A.Z is a crucial chromatin factor for Ac and Sc to promote gene expression. (A) Fold changes of Ac-Da-induced expression in S2 cells of Arp6 knockdown (KD) or H2A.Z KD versus control Luc KD. n=4 biological repeats. (B) Heatmap showing log2 fold change of gene expression. Top: 111 Ac-Da-induced genes. Bottom: 117 non-Ac-Da-induced genes. Left two columns: fold changes by Ac-Da versus AcR29G-Da heterodimers. Middle two columns: fold changes by Arp6 KD versus Luc KD. Right two columns: fold changes by H2A.Z KD versus Luc KD. n=2 biological repeats. (C) Scatter plot showing expression fold changes of 111 Ac-Da-induced (ADi) genes and 117 non-Ac-Da-induced (non-ADi) genes in Arp6 KD or H2A.Z KD cells. The mean is shown by the bar. (D) ChIP analyses of Ac-Da and AcR29G-Da bound to the E-box sites. The binding percentage (% input) is shown as the percentage bound by the anti-Flag antibody subtracted by that bound by IgG. n=2 biological repeats. (E,F) ChIP analyses showing the net DNA binding by Ac-Da versus AcR29G-Da (percentage of Ac-Da-bound subtracted by percentage of AcR29G-Da-bound) in Arp6 KD (E) and H2A.Z KD (F). n=2 for Arp6 KD and 3 for H2A.Z KD. (G) ChIP assay of Myc-H2A.Z enrichment around the +1 nucleosome region of phyl and ac by the anti-Myc antibody. ChIP signals were not detected in mock-treated cells. n=3 biological repeats. (H) ChIP analyses showing Myc-H2A.Z enrichment around the +1 nucleosome region of phyl in cells expressing Ac-Da or AcR29G-Da. Cells were also treated with RNAi to knockdown control Luc or Arp6 as indicated. n=3 biological repeats. (I,J) Scatter plots of expression fold changes in Arp6 KD (I) and H2A.Z KD (J) versus Luc KD. +, genes with H2A.Z replacement at +1 nucleosome. −, genes without H2A.Z replacement at the +1 nucleosome. (K) Arp6-induced H2A.Z replacement at the +1 nucleosome facilitates efficient transcription of proneural protein target genes. Left: The Arp6-including SWR1 complex promotes H2A.Z exchange at the +1 nucleosome of some proneural protein target genes before proneural protein expression. Right: In wild-type (WT) SOPs, proneural proteins initiate neurogenesis by binding to E-box sites and activating target gene transcription. Ac recruits Arp6 for more H2A.Z replacement at the +1 nucleosome, at least in the case of proneural protein target gene phyl. In SOPs defective for Arp6 or H2A.Z, proneural proteins still bind to the cognate target sites but initiate transcription at a slower rate because of reduced H2A.Z replacement at the +1 nucleosome. For all graphs, numbers at the bottom of columns indicate the number of measurements made. *P<0.05, **P<0.01, ****P<0.0001. ns, not significant.

Fig. 6.

H2A.Z is a crucial chromatin factor for Ac and Sc to promote gene expression. (A) Fold changes of Ac-Da-induced expression in S2 cells of Arp6 knockdown (KD) or H2A.Z KD versus control Luc KD. n=4 biological repeats. (B) Heatmap showing log2 fold change of gene expression. Top: 111 Ac-Da-induced genes. Bottom: 117 non-Ac-Da-induced genes. Left two columns: fold changes by Ac-Da versus AcR29G-Da heterodimers. Middle two columns: fold changes by Arp6 KD versus Luc KD. Right two columns: fold changes by H2A.Z KD versus Luc KD. n=2 biological repeats. (C) Scatter plot showing expression fold changes of 111 Ac-Da-induced (ADi) genes and 117 non-Ac-Da-induced (non-ADi) genes in Arp6 KD or H2A.Z KD cells. The mean is shown by the bar. (D) ChIP analyses of Ac-Da and AcR29G-Da bound to the E-box sites. The binding percentage (% input) is shown as the percentage bound by the anti-Flag antibody subtracted by that bound by IgG. n=2 biological repeats. (E,F) ChIP analyses showing the net DNA binding by Ac-Da versus AcR29G-Da (percentage of Ac-Da-bound subtracted by percentage of AcR29G-Da-bound) in Arp6 KD (E) and H2A.Z KD (F). n=2 for Arp6 KD and 3 for H2A.Z KD. (G) ChIP assay of Myc-H2A.Z enrichment around the +1 nucleosome region of phyl and ac by the anti-Myc antibody. ChIP signals were not detected in mock-treated cells. n=3 biological repeats. (H) ChIP analyses showing Myc-H2A.Z enrichment around the +1 nucleosome region of phyl in cells expressing Ac-Da or AcR29G-Da. Cells were also treated with RNAi to knockdown control Luc or Arp6 as indicated. n=3 biological repeats. (I,J) Scatter plots of expression fold changes in Arp6 KD (I) and H2A.Z KD (J) versus Luc KD. +, genes with H2A.Z replacement at +1 nucleosome. −, genes without H2A.Z replacement at the +1 nucleosome. (K) Arp6-induced H2A.Z replacement at the +1 nucleosome facilitates efficient transcription of proneural protein target genes. Left: The Arp6-including SWR1 complex promotes H2A.Z exchange at the +1 nucleosome of some proneural protein target genes before proneural protein expression. Right: In wild-type (WT) SOPs, proneural proteins initiate neurogenesis by binding to E-box sites and activating target gene transcription. Ac recruits Arp6 for more H2A.Z replacement at the +1 nucleosome, at least in the case of proneural protein target gene phyl. In SOPs defective for Arp6 or H2A.Z, proneural proteins still bind to the cognate target sites but initiate transcription at a slower rate because of reduced H2A.Z replacement at the +1 nucleosome. For all graphs, numbers at the bottom of columns indicate the number of measurements made. *P<0.05, **P<0.01, ****P<0.0001. ns, not significant.

Close modal

In cells knocked down for H2A.Z by approximately 90% (Fig. S3B), phyl expression was also decreased by 37% (Fig. S3D). Unexpectedly, examination of exogenously expressed Ac and Da levels revealed that they were much higher in H2A.Z-knockdown S2 cells compared with Luc-knockdown cells (Fig. S3E) but remained comparable between Arp6 knockdown and Luc knockdown cells (Fig. S3E). We then adjusted the amounts of the plasmid in H2A.Z knockdown cells such that they expressed comparable levels of Ac and Da as in the Luc knockdown cells (Fig. S2E). Expression of phyl and sca by comparable levels of Ac-Da was markedly decreased in H2A.Z knockdown cells (Fig. 6A).

We next performed transcriptomic analysis to assess the regulation of Ac-Da-induced genes by Arp6 and H2A.Z. We profiled the expression levels for genes induced by Ac-Da heterodimers. In the group of 111 Ac-Da-upregulated genes, most of their expression levels were compromised in both Arp6 knockdown and H2A.Z knockdown cells (Table S1; Fig. 6B). For comparison, we selected a collection of 117 genes expression levels of which were insensitive to Ac-Da heterodimer (expression changes less than 20%) (see Materials and Methods). Arp6 and H2A.Z knockdown caused a significantly greater reduction in the Ac-Da-upregulated genes than the non-Ac-Da-induced genes (Fig. 6C).

The preferential requirement of Arp6 and H2A.Z for Ac-Da-induced gene expression prompted us to examine whether Arp6 and H2A.Z promote proneural protein binding to their specific DNA-binding sites. The Ac-Da heterodimers bind to E-box sequences CAG(C/G)TG with high affinity (Singson et al., 1994). Chromatin immunoprecipitation (ChIP) analysis was performed for the Ac binding to two CAGCTG in the phyl upstream region (phyl E1 and E2) (Pi et al., 2004). The assay confirmed that Ac-Da preferentially bound the two E-box sites compared with the DNA-binding domain mutant AcR29G-Da (Fig. 6D). By contrast, no preferential binding was detected on the low-affinity binding site CACGTG (phyl E4) (Pi et al., 2004; Singson et al., 1994) (Fig. 6D), indicating the specificity of our assay. The sca proximal region contains a CAGGTG element (sca E1) bound by proneural proteins in the gel mobility shift assay (Singson et al., 1994). It was associated weakly with Flag-Ac in the ChIP analysis (Fig. 6D). The binding affinity to these sites was not significantly compromised by Arp6 knockdown (Fig. 6E) and was not reduced by H2A.Z knockdown (Fig. 6F), suggesting that the Ac-Da heterodimer binding to the E-box sites is likely independent of H2A.Z enhancer incorporation.

H2A.Z is enriched in the nucleosomes near the TSS, particularly the nucleosome immediately downstream of the TSS. The H2A.Z enrichment at the +1 nucleosome positively correlates with H2A.Z-dependent activation and facilitates Pol II-mediated transcription in Drosophila (Ibarra-Morales et al., 2021; Weber et al., 2014). To assay the H2A.Z incorporation to the proneural protein target gene, ChIP analysis for Myc-H2A.Z was performed in Myc-H2A.Z-transfected S2 cells and H2A.Z incorporation at the +1 nucleosome of phyl was assayed. The H2A.Z ChIP signal was specifically detected in cells transfected with Myc-H2A.Z, but not in mock-treated cells by anti-Myc antibodies from two different sources (Fig. 6G; Fig. S2C), indicating that phyl is intrinsically permissive for H2A.Z replacement. Because proneural proteins associated with Arp6, we next examined whether Ac influences H2A.Z incorporation of phyl. As shown in Fig. 6H, Ac-Da heterodimers elevated H2A.Z incorporation at the +1 nucleosome compared with AcR29G-Da, suggesting that recruitment of Ac-Da heterodimers to the E-box promotes robust H2A.Z replacement. The enhanced H2A.Z incorporation by Ac-Da was significantly diminished in Arp6 knockdown cells (Fig. 6H). Knockdown of Arp6 also decreased the H2A.Z occupancy in cells expressing the inactive AcR29G-Da (Fig. 6H), indicating that Arp6 is required for both Ac-Da-induced and Ac-Da-independent, basal H2A.Z incorporation of phyl at the +1 nucleosome.

Our analysis of SOP development suggests that Arp6 and H2A.Z are required for phyl transcription, but dispensable for Ac expression in proneural stripes (Figs 2A and 5B). Consistent with the in vivo data, Arp6 and H2A.Z knockdown did not reduce ac expression in S2 cells (Fig. 6A). Two high-affinity E-boxes in the ac proximal promoter were preferentially bound by Ac-Da relative to AcR29G-Da in the ChIP assay and the binding affinity was not significantly compromised in the Arp6 knockdown and H2A.Z knockdown (Fig. 6D-G). Compared with phyl, however, H2A.Z occupancy around the +1 nucleosome location of ac was undetected or significantly lower (Fig. 6F; Fig. S4), suggesting that the +1 nucleosome of ac is intrinsically inaccessible to H2A.Z incorporation.

To investigate further whether intrinsically differential H2A.Z replacement influences proneural protein target gene expression, we analyzed the correlation between H2A.Z replacement at the +1 nucleosome and Arp6- and H2A.Z-dependent gene expression. H2A.Z incorporation is initiated around zygotic genome activation preceding Ac- and Sc-dependent neurogenesis (Ibarra-Morales et al., 2021). By searching the database of H2A.Z replacement during zygotic genome activation, 41 Ac-Da-induced genes exhibited H2A.Z replacement at the +1 nucleosome and 62 genes did not. We analyzed the extent of downregulation in H2A.Z-positive genes and H2A.Z-negative genes by Arp6 knockdown. As shown in Fig. 6I, there was no significant difference in the downregulation of expression between H2A.Z-positive and -negative genes in both Ac-Da-induced and non-Ac-Da-induced gene groups. Upon elimination of H2A.Z by H2A.Z knockdown, however, we found that the Ac-Da-induced gene expression was much more decreased in the H2A.Z-positive genes than in the H2A.Z-negative genes (log2 fold change: −0.86 versus −0.34; P<0.01) (Fig. 6J). We conclude that the marked reduction was specific to Ac-Da-induced expression because the effects on the non-Ac-Da-induced, H2A.Z-positive genes were small (log2 fold change=−0.15) and there was no significant difference between H2A.Z-positive and -negative genes in non-Ac-Da induced gene expression (Fig. 6J). Taken together with the genetic interaction between Arp6 and ac sc, our data support the model that H2A.Z acts through its replacement at the +1 nucleosome to facilitate efficient onset and maximal activation of transcription upon proneural protein binding to E-box sites, thus promoting rapid differentiation of neural precursors (Fig. 6K).

bHLH proneural proteins are robust inducers of neurogenesis in vivo and in vitro. They act as pioneer factors to recognize cognate E-box sequence and activate target gene expression for neural determination and differentiation. Our data demonstrate that the histone variant H2A.Z is a crucial chromatin factor for proneural protein-induced maximal gene expression: (1) loss of Arp6 and H2A.Z led to prolonged SOP development and smaller ES organs, the phenotypes also observed in ac sc hypomorphic mutants; (2) SOPs and ES organs were almost eliminated in Arp6 and ac sc hypomorphic double mutants; (3) transcription of proneural protein target genes phyl and sca in SOPs were markedly reduced in Arp6 mutants; and (4) transcriptomic analysis revealed that loss of Arp6 and H2A.Z preferentially diminished gene expression induced by Ac-Da compared with the constitutively activated genes. Unlike the histone acetyltransferase CBP and the chromatin remodeler Brg1 in Xenopus and mouse, which are required for the generation of neural progenitors and neurons by proneural proteins (Koyano-Nakagawa et al., 1999; Lee et al., 2009; Seo et al., 2005), we found that Arp6-mediated H2A.Z incorporation specifically influences precursor differentiation rate without affecting fate determination and formation of SOPs. Therefore, histone variant H2A.Z exchange plays a distinct role in the regulation of proneural protein-induced neurogenesis compared with histone modification and nucleosome remodeling.

Our analyses suggest that Arp6-mediated H2A.Z replacement at the +1 nucleosome facilitates proneural protein target gene expression by both proneural protein-dependent and -independent mechanisms. From the S2 cell studies, we found that proneural proteins enhanced H2A.Z incorporation on its target gene phyl and loss of Arp6 diminished the elevated H2A.Z replacement. These results, together with the physical interaction between Arp6 and proneural proteins, suggest that proneural proteins act upstream and can recruit Arp6 to enhance H2A.Z incorporation. We also observed that H2A.Z enrichment at the +1 nucleosome before neurogenesis was highly correlated with the activation of proneural protein target genes by H2A.Z and Arp6 is required for this basal H2A.Z incorporation, at least to phyl. In addition, Arp6 and H2A.Z do not significantly influence E-box binding. These results suggest a parallel action between Arp6 and proneural proteins in the activation of proneural protein target genes. Genetic analyses showed that Ac overexpression failed to rescue timely SOP differentiation in Arp6 mutant clones, indicating that the absence of Arp6 could not be replaced by enhanced Ac expression. Our genetic and biochemical data support the suggestion that Arp6 acts both downstream and in parallel with proneural proteins to facilitate proneural protein-induced gene activation.

Studies in the past two decades have demonstrated that nuclear actin and nuclear ARPs are crucial components of chromatin remodeling and modifying complexes (reviewed by Klages-Mundt et al., 2018). The nuclear actin-Arp4 dimer participates in chromatin complexes, such as SWR1, Ino80 and BAF. We have shown that nuclear actin is important for proneural protein-mediated gene activation in SOPs and S2 cells; knockdown of both Actin 5C and Actin 42A disrupted SOP formation and overexpression of nuclear β-actin increased Ac/Da-dependent transcription in S2 cells (Hsiao et al., 2014). Given that nuclear actin, Arp4 and Arp6 are the core components of SWR1, our results suggest that one of the major functions of nuclear actin family proteins in neurogenesis involves H2A.Z exchange. A recent study also reveals that the β-actin level is particularly important for enhancing chromatin accessibility at the promoter region of genes involved in the regulation of neuron differentiation in mouse cells (Mahmood et al., 2021). Thus, our analyses of nuclear actin and ARPs in SOPs suggest a highly conserved mechanism of the nuclear actin family protein in the modulation of gene expression in neural development.

Proneural proteins are master regulators of ES organ development. Their spatial and temporal expression patterns determine where and when SOP fate determination and differentiation occur. In developing nota, Ac and Sc are expressed in longitudinal proneural stripes at 6-9 h APF in response to patterning cues from Delta-Notch signaling (Couturier et al., 2019). Our results showed that proneural patterning and ac sc expression in cultural cells was largely unaffected by loss of Arp6-mediated H2A.Z incorporation. At later stages, proneural protein is crucial for precursor selection from proneural cells; single SOPs are selected and arranged in regular arrays by several proneural protein-mediated mechanisms: (1) the proneural target gene phyl is expressed at a high level in SOPs to facilitate lateral inhibition by Notch signaling (Pi et al., 2004); (2) proneural protein target gene sca is expressed in proneural clusters and SOPs to mediate long-range precursor spacing; and (3) Notch target genes E(spl)m7 and m8 were activated by proneural proteins in proneural stripes to mediate SOP selection and spacing (Couturier et al., 2019). Following SOP selection, proneural proteins in single SOPs are gradually downregulated via negative feedback regulation by phyl to promote G2-M progression (Chang et al., 2008). SOPs also require proneural protein target gene sens for timely SOP division (Jafar-Nejad et al., 2006). Therefore, insufficient target gene expressions, caused either by reduced H2A.Z replacement in Arp6 and H2A.Z mutants or by low ac sc expression in acsbm mutants, lead to retarded SOP formation and division, and, ultimately, smaller ES organs.

Our results showed that phyl and sens are activated by Arp6 in SOPs. Although phyl and sens are also required for binary cell fate determination of SOP daughter cells, cell fate transformation was almost not observed in Arp6 and H2A.Z mutants. There are two possible explanations. First, phyl and sens were expressed in lower but sufficient levels in SOP daughters to sustain normal cell fate determination in Arp6 and H2A.Z mutants. Second, Arp6 and H2A.Z do not modulate phyl and sens expression in SOP daughter cells. We observed that once phyl and Sens reached sufficient levels in mature SOPs in Arp6 mutants, their levels in SOP daughter cells (pIIa and pIIb cells) were not reduced in mutant clones. Combined with the transcriptomic result that H2A.Z is largely dispensable for the expression of non-Ac-Da-inducible genes, we hypothesized that Arp6-mediated H2A.Z incorporation has little to no effect on phyl and sens expression in SOP daughter cells.

The transcriptomic analysis revealed that H2A.Z is preferentially required for Ac-Da-induced gene activation. By contrast, recent studies also showed that H2A.Z is essential for the expression of constitutively activated genes but not for transcriptional activator Zelda-dependent transcription during the zygotic genome activation (Ibarra-Morales et al., 2021). In addition, H2A.Z plays diverse roles in regulating DNA binding by transcriptional factors. H2A.Z could have positive (Murphy et al., 2020), negative (Cole et al., 2021), or no effect (this study) on the DNA binding.

Analyses of H2A.Z incorporation among Ac-induced genes support the hypothesis that differential H2A.Z replacement at the +1 nucleosome contributes to proneural target gene activation by H2A.Z. Several mechanisms have been proposed to mediate differential incorporation by SWR1. The yeast SWR1 complex preferentially binds promoters containing a more extended nucleosome-free region located between the +1 and −1 nucleosomes (Ranjan et al., 2013). DNA methylation prevents H2A.Z incorporation in Arabidopsis (Coleman-Derr and Zilberman, 2012; Zilberman et al., 2008). Histone H3 acetylation contributes to preferential SWR1 recruitment from yeast to human (Hsu et al., 2018; Klein et al., 2018; Ranjan et al., 2013). Therefore, intrinsic chromatin architecture and modification might further determine the responsiveness of proneural target genes to H2A.Z-dependent transcriptional activation.

Although Arp6 is essential for maintenance of H2A.Z levels in developing cells, as shown by the elimination of H2A.Z in a strong loss-of-function Arp61 clone, H2A.Z incorporation was only partially compromised in S2 cells by the knockdown of Arp6 (this study) and YL-1, another core component of the SWR1 complex (Weber et al., 2014). We hypothesize that RNAi-mediated silencing of Arp6 and YL-1 is not efficient enough to abolish most SWR1 activity in S2 cells, leading to incomplete elimination of H2A.Z from nucleosomes in knockdown cells.

In mammals and yeast, the catalytic subunits of the SWR1 complex and NuA4 complex are encoded by distinct genes. In Drosophila, recent studies have revealed that the core ATPase of SWR1 complex and NuA4 complex are encoded by different isoforms of dom, thus complicating the previous molecular interpretation of dom mutant phenotypes. Analysis of the SWR1 complex component has revealed that Arp6 is the sole SWR1-specific component from yeast to human (Scacchetti and Becker, 2021). Thus, our Arp6 mutants are an excellent tool with which to verify and further investigate SWR1-specific function in Drosophila.

Fly stocks

The following fly stocks were obtained from stock centers: acsbm [Bloomington Drosophila Stock Center (BDSC) #36539], His2Av810 (BDSC #9264), UAS-His2Av RNAi (BDSC #28966), UAS-RFP RNAi (BDSC #35785), scaA2–6 (sca-lacZ, BDSC #5403), Act5C-Gal4 (BDSC #4414), tubP-Gal4 (BDSC #5138), sca-Gal4 (BDSC #6479), Ubi-mRFP-nls FRT19A (BDSC #31416) and Ubx-FLP tubP-Gal80 FRT19A (BDSC #42731). sc-GFP was a gift from Dr Xi (Chen et al., 2018). dom-B-GFP was a gift from Dr Becker (Börner and Becker, 2016). phyl4.1-nGFP has been described by Pi et al. (2004). UAS-3XHA-Arp6 was generated in this study (see below for plasmid construction). Transgenic lines were generated by standard P-element mediated transformation. All fly stocks were raised at 18°C or 25°C.

Generation of mutant clones and transgenic flies

Negatively marked mutant clones were generated using the FLP/FRT technique (Xu and Rubin, 1993). For Arp6 mutant clones generated by hs-FLP, larvae were heat-shocked twice at 37°C for 1 h at the first and the second instar and returned to 25°C until dissection at the indicated time. For acsbm clones and Arp6 mutant clones generated by ubiquitously expressed ubi-FLP, larvae were raised at 25°C until dissection.

Construction of the UAS-plasmid

The UAS plasmids for Arps (Arp4, 5, 6 and 8), proneural proteins (Sc and Ato) and H2A.Z were constructed by cloning the open reading frames (ORFs) into Drosophila Gateway vectors pTFW or pTMW. The AcR29G substitution (codon change from AGA to GGA) was generated using the QuikChange site-directed mutagenesis kit (Stratagene).

CRISPR/Cas9-based genome editing of Arp6

The Arp61 mutant allele was generated by gRNA-1-guided DNA break followed by homology-directed repair (HDR) (see Fig. 1C). HDR donor plasmids harbor a DNA cassette containing the visible marker 3XP3-RFP and SV40 terminator. Homology sequences of Arp6 (∼500 bp in length) flank each side of the marker DNA cassette. We obtained an insertion allele Arp61 in which the 3XP3 marker cassette was inserted at the 11th residue of Arp6. For the Arp6D allele, the whole ORF was deleted via gRNA-1 and gRNA-2 (see Fig. 1C). The HA knock-in allele Arp6HA was generated by gRNA-2 guided DNA break followed by HDR via the donor plasmid containing the in-framed 3XHA tag at the 3′end of the ORF (see Fig. S2A). The gRNAs and HDR donor plasmids used for microinjection were purified using the Plasmid Midiprep kit (Invitrogen). The Arp61, Arp6D and Arp6HA alleles were validated by genomic PCR followed by Sanger sequencing.

Rescue of Arp6 mutant lethality

The rescue experiment was performed by a cross between Arp61/FM7C; Act5C-Gal4 adult females and UAS-3HA-Arp6 adult males. The numbers of progeny in each genotype group were examined.

Immunostaining and image analysis

Pupal nota were dissected in PBS, fixed in 4% paraformaldehyde, and permeabilized in 0.1% Triton X-100 in PBS (PBST). Pupal nota were incubated with primary antibodies overnight at 4°C and secondary antibodies for 2 h at room temperature. The following primary antibodies were used: mouse anti-Achaete [1:5, Developmental Studies Hybridoma Bank (DSHB)], guinea pig anti-Sens (1:2000, gift from Dr Bellen, Baylor College of Medicine, Houston, TX, USA), mouse anti-Pros MR1A (1:10, DSHB), mouse anti-Elav 9F8A9 (1:10, DSHB), mouse anti-Hnt 1G9 (1:5, DSHB), mouse anti-Cut 2B10 (1:25, DSHB), rabbit anti-Drosophila H2A.Z (1:2500, a gift from Dr Becker, Ludwig-Maximilians-University, Munich, Germany), mouse anti-FLAG M2 (1:1000, Sigma-Aldrich, F3165) and rabbit anti-GFP (1:2000, Thermo Fisher Scientific, A11122). All immunofluorescence images were obtained with a Zeiss LSM 780 microscope.

Cell culture and plasmid construction

Drosophila S2 cells were maintained in Schneider's Drosophila medium (Invitrogen) supplemented with 10% heat-inactivated fetal bovine serum (Gibco BRL). S2 cells were from the Drosophila Genomics Resource Center. UAS-based expression plasmids and the driver, pWA-GAL4, were transfected using TransIT-Insect transfection reagent (Mirus Bio) for gene expression. The transfected cells were harvested 48 h after transfection for Arp6 knockdown assays and 72 h after transfection for H2A.Z knockdown assays.

RNAi

Primers for dsRNA templates of Arp6, H2A.Z and firefly luciferase were designed using SnapDragon (https://www.flyrnai.org/snapdragon). For the myc-H2A.Z ChIP assay, the dsRNA template for H2A.Z was designed at the H2A.Z 5′ and 3′ UTRs. Templates for in vitro transcription were generated by PCR amplification using KOD FX Polymerase (Toyobo). dsRNAs were synthesized in vitro using the T7 RiboMAX Express RNAi System (Promega) according to the manufacturer's instructions. For Arp6 knockdown, cells were treated with 30 µg (12-well plate) or 300 µg (10-cm dish) dsRNA for 4 days. For H2A.Z knockdown, cells were treated with 5 µg (12-well plate) or 50 µg (10-cm dish) dsRNA for 6 days.

RNA extraction and RT-qPCR

Total RNA from Drosophila S2 cells was harvested using TRIzol reagent (Invitrogen) and purified as per the manufacturer's instructions. cDNA was synthesized from ∼2-3 µg of total RNA using random hexamers and MMLV reverse transcriptase (Thermo Fisher Scientific). qPCR was performed with 2X SYBR Green Master Mix (Bio-Rad) in a Bio-Rad iQ5 gradient Real-Time PCR system. Gapdh2 was the reference gene for expression quantification in all RT-PCR experiments. Results are shown as mean±s.e.m. of at least three biologically independent experiments. Sequences of the primers are listed in Table S2.

RNA preparation, transcriptomic analysis, and selection of Ac-Da-non-induced genes

For next-generation sequencing analysis, total RNA was extracted from S2 cells and 100 ng purified mRNA was used for cDNA library construction by NEBNext® UltraTM RNA Library Prep Kit. Raw sequencing data were analyzed by STAR (v2.7.3a) two-pass mapping strategy to align the raw FASTQ reads against the Drosophila melanogaster reference genome (BDGP6.32) downloaded from the Ensembl database. Ac-Da-induced genes were defined as genes with induction fold change ≥1.5 by Ac-Da relative to Ac-DaR29G. Non-Ac-Da-induced genes were genes with fold change between 0.8 and 1.2 by Ac-Da vs AcR29G-Da and expressed at 15-100 reads per kilobase of transcript per million reads mapped (RPKM), the levels at which Ac-Da-induced genes are usually expressed.

Co-immunoprecipitation and western blotting

Cells were washed twice with ice-cold PBS and fractioned into cytosolic and nuclear extracts as described (Hsiao et al., 2014). Protein extracts were diluted in mRIPA buffer [50 mM Tris-HCl (pH 7.8), 150 mM NaCl, 5 mM EDTA (pH 8.0), 0.5% Triton X-100, 0.5% NP-40] supplemented with cOmplete EDTA-free Protease Inhibitor (Roche). For immunoprecipitation, 20 µl FLAG-M2 affinity gel (Sigma-Aldrich, A2220) was incubated with 2 mg of lysate in 1 ml mRIPA buffer for 2 h. Beads were collected by centrifugation (2000 g for 3 min at 4°C) and washed five times with mRIPA buffer. The associated protein complexes were analyzed by SDS-PAGE followed by western blotting. The primary antibodies for western blot were: mouse anti-Drosophila H2A.Z (1:1000, Active Motif, 61752), mouse anti-c-Myc 9E10 (1:5000, Santa Cruz Biotechnology, sc-40), mouse anti-FLAG M2 (1:10,000, Sigma-Aldrich, F3165) and mouse anti-α-Tubulin (1:25,000, Sigma-Aldrich, T9026).

ChIP

The ChIP assay was performed as described (Chen et al., 2013; Riddle et al., 2011; Tettey et al., 2019) with the following modifications. S2 cells were cross-linked by adding formaldehyde (final concentration, 1%) directly to the culture for 10 min on ice. Freshly prepared 2.5 M glycine was added to stop the fixation at a final concentration of 0.125 M and incubated for 10 min on ice. Cells were pelleted at 500 g for 10 min at 4°C and washed twice with ice-cold PBS, followed by the preparation of cytosolic and nuclear protein extracts. Nuclei were suspended in RIPA buffer [50 mM Tris-HCl (pH 8.0), 150 mM NaCl, 5 mM EDTA (pH 8.0), 0.5% sodium deoxycholate, 0.1% SDS, 1% NP-40, 1× cOmplete EDTA-free Protease Inhibitor] for 30 min. Chromatin samples were sheared for ten cycles of 30 s ON/OFF with the Bioruptor® Plus sonication device to obtain 200-250 bp chromatin fragments, and insoluble chromatin was removed by centrifugation at 13,500 rpm (12,000 g) for 30 min at 4°C. An equal protein quantity (1 mg/ChIP) of soluble chromatin was pre-cleared by incubation with 15 µl Dynabeads™ protein G (Thermo Fisher Scientific, 10004D) for 1 h at 4°C. We added mouse anti-c-Myc 9E10 (3.5 µg/ChIP, Santa Cruz Biotechnology, sc-40), mouse anti-c-Myc 9E10 (3.5 µg/ChIP, Abcam, ab32), mouse anti-FLAG M2 (2 µg/ChIP, Sigma-Aldrich, F3165) or control normal mouse IgG (Millipore, CS200621); 30 µl protein G-coupled magnetic beads were added to each immunoprecipitation followed by rotation for 3 h at 4°C. After extensive washes, the immuno-complexes were treated with proteinase K and heated for de-crosslinking. Bound DNA in the ChIP and input DNA was extracted, purified, and subjected to quantitative real-time PCR analysis. Primer sequences are shown in Table S2.

Quantification and statistical analyses

Shaft length of ES organs

The shaft length of the ES organ was analyzed by measuring the distance of the shaft from the tip to the socket by ImageJ. In Figs 1I and 3B, ten ES organs in each notum were measured. In Fig. S2B, all ES organs in the dorsal-central region of five to eight nota were examined.

The ratio of Elav-positive and Pros-positive cells in Arp61 clones compared with controls

The number of positive cells in the RFP-negative, Arp61 area was considered to be the cell number in the mutant clones. For comparison, the RFP-negative, Arp61 mutant area was first outlined, and the outlined area was superimposed on the neighboring heterozygous tissue. The cell numbers in the superimposed area were counted as the numbers in the controls. The ratio was the numbers in Arp61 clones normalized to those in the controls.

Sens intensity in SOPs

The Sens intensity in SOPs was calculated as the fluorescence intensity of anti-Sens staining in SOPs subtracted by the background fluorescence intensity in the neighboring area. The intensity of Sens in Arp61 SOPs was normalized to the mean intensity of control SOPs in the neighboring heterozygous tissue.

Gene expression of qRT-PCR and RNA-seq

mRNA levels of phyl, sca and endogenous ac were normalized to Gapdh in the qRT-PCR analysis (Figs 4A and 6A). In Fig. 6C, each dot is the average of the two independent RNA-seq analyses.

ChIP

The binding percentage (% input) is shown as the percentage of DNA bound by the specific antibody subtracted by that bound by IgG.

Statistical analyses

Statistical analyses were performed using one-way ANOVA with Tukey's multiple comparison test for Figs 1I and 6C-J, Figs S2E and S4, and unpaired two-sample t-tests in Fig. 3B and Fig. S1D. Data were using by GraphPad Prism and Microsoft Excel. Statistical significance was defined as P<0.05. Data are represented as mean and s.e.m., except for Figs 1I and 3B in which data are represented as mean and s.d.

We thank Dr Cheng-Ting Chien for his critical reading of the manuscript. We thank Dr Peter B Becker for sharing the anti-His2Av (H2A.Z) antibody. The authors also thank the Microscopy Center at Chang Gung University for technical assistance.

Author contributions

Conceptualization: H.P., Y.-L.H.; Methodology: Y.-L.H., H.-W.C., K.-H.C.; Software: Y.-L.H.; Validation: Y.-L.H., H.-W.C., K.-H.C., H.P.; Formal analysis: Y.-L.H., H.P.; Investigation: Y.-L.H., H.-W.C., K.-H.C.; Resources: H.P., B.C.-M.T., C.-H.C.; Data curation: H.P., Y.-L.H.; Writing - original draft: H.P., Y.-L.H.; Writing - review & editing: H.P., B.C.-M.T.; Visualization: Y.-L.H.; Supervision: H.P., B.C.-M.T.; Project administration: H.P.; Funding acquisition: H.P., C.-H.C.

Funding

This work is supported by grants from the Ministry of Science and Technology, Taiwan (MOST 109-2311-B-182-002-MY3) and Chang Gung Memorial Hospital (CMRPD1K0451-52) to H.P.

Data availability

Next Generation Sequencing data have been deposited in GEO under accession number GSE223416.

Adam
,
M.
,
Robert
,
F.
,
Larochelle
,
M.
and
Gaudreau
,
L.
(
2001
).
H2A.Z is required for global chromatin integrity and for recruitment of RNA polymerase II under specific conditions
.
Mol. Cell. Biol.
21
,
6270
-
6279
.
Barski
,
A.
,
Cuddapah
,
S.
,
Cui
,
K.
,
Roh
,
T.-Y.
,
Schones
,
D. E.
,
Wang
,
Z.
,
Wei
,
G.
,
Chepelev
,
I.
and
Zhao
,
K.
(
2007
).
High-resolution profiling of histone methylations in the human genome
.
Cell
129
,
823
-
837
.
Bertrand
,
N.
,
Castro
,
D. S.
and
Guillemot
,
F.
(
2002
).
Proneural genes and the specification of neural cell types
.
Nat. Rev. Neurosci.
3
,
517
-
530
.
Blochlinger
,
K.
,
Bodmer
,
R.
,
Jan
,
L. Y.
and
Jan
,
Y. N.
(
1990
).
Patterns of expression of cut, a protein required for external sensory organ development in wild-type and cut mutant Drosophila embryos
.
Genes Dev.
4
,
1322
-
1331
.
Bonnet
,
J.
,
Lindeboom
,
R. G. H.
,
Pokrovsky
,
D.
,
Stricker
,
G.
,
Celik
,
M. H.
,
Rupp
,
R. A. W.
,
Gagneur
,
J.
,
Vermeulen
,
M.
,
Imhof
,
A.
and
Muller
,
J.
(
2019
).
Quantification of proteins and histone marks in Drosophila embryos reveals stoichiometric relationships impacting chromatin regulation
.
Dev. Cell
51
,
632
-
644.e636
.
Börner
,
K.
and
Becker
,
P. B.
(
2016
).
Splice variants of the SWR1-type nucleosome remodeling factor Domino have distinct functions during Drosophila melanogaster oogenesis
.
Development
143
,
3154
-
3167
.
Campuzano
,
S.
,
Balcells
,
L.
,
Villares
,
R.
,
Carramolino
,
L.
,
García-Alonso
,
L.
and
Modolell
,
J.
(
1986
).
Excess function hairy-wing mutations caused by gypsy and copia insertions within structural genes of the achaete-scute locus of Drosophila
.
Cell
44
,
303
-
312
.
Cao
,
T.
,
Sun
,
L.
,
Jiang
,
Y.
,
Huang
,
S.
,
Wang
,
J.
and
Chen
,
Z.
(
2016
).
Crystal structure of a nuclear actin ternary complex
.
Proc. Natl. Acad. Sci. USA
113
,
8985
-
8990
.
Chang
,
P.-J.
,
Hsiao
,
Y.-L.
,
Tien
,
A.-C.
,
Li
,
Y.-C.
and
Pi
,
H.
(
2008
).
Negative-feedback regulation of proneural proteins controls the timing of neural precursor division
.
Development
135
,
3021
-
3030
.
Chen
,
H.-W.
,
Yang
,
C.-C.
,
Hsieh
,
C.-L.
,
Liu
,
H.
,
Lee
,
S.-C.
and
Tan
,
B. C.-M.
(
2013
).
A functional genomic approach reveals the transcriptional role of EDD in the expression and function of angiogenesis regulator ACVRL1
.
Biochim. Biophys. Acta
1829
,
1309
-
1319
.
Chen
,
J.
,
Xu
,
N.
,
Wang
,
C.
,
Huang
,
P.
,
Huang
,
H.
,
Jin
,
Z.
,
Yu
,
Z.
,
Cai
,
T.
,
Jiao
,
R.
and
Xi
,
R.
(
2018
).
Transient Scute activation via a self-stimulatory loop directs enteroendocrine cell pair specification from self-renewing intestinal stem cells
.
Nat. Cell Biol.
20
,
152
-
161
.
Chen
,
Z.
,
Gabizon
,
R.
,
Brown
,
A. I.
,
Lee
,
A.
,
Song
,
A.
,
Diaz-Celis
,
C.
,
Kaplan
,
C. D.
,
Koslover
,
E. F.
,
Yao
,
T.
and
Bustamante
,
C.
(
2019
).
High-resolution and high-accuracy topographic and transcriptional maps of the nucleosome barrier
.
eLife
8
,
e48281
.
Cole
,
L.
,
Kurscheid
,
S.
,
Nekrasov
,
M.
,
Domaschenz
,
R.
,
Vera
,
D. L.
,
Dennis
,
J. H.
and
Tremethick
,
D. J.
(
2021
).
Multiple roles of H2A.Z in regulating promoter chromatin architecture in human cells
.
Nat. Commun.
12
,
2524
.
Coleman-Derr
,
D.
and
Zilberman
,
D.
(
2012
).
Deposition of histone variant H2A.Z within gene bodies regulates responsive genes
.
PLoS Genet.
8
,
e1002988
.
Couturier
,
L.
,
Mazouni
,
K.
,
Corson
,
F.
and
Schweisguth
,
F.
(
2019
).
Regulation of Notch output dynamics via specific E(spl)-HLH factors during bristle patterning in Drosophila
.
Nat. Commun.
10
,
3486
.
Dambly-Chaudiére
,
C.
and
Ghysen
,
A.
(
1987
).
Independent subpatterns of sense organs require independent genes of the achaete-scute complex in Drosophila larvae
.
Genes Dev.
1
,
297
-
306
.
Giaimo
,
B. D.
,
Ferrante
,
F.
,
Herchenröther
,
A.
,
Hake
,
S. B.
and
Borggrefe
,
T.
(
2019
).
The histone variant H2A.Z in gene regulation
.
Epigenet. Chromatin
12
,
37
.
Guillemette
,
B.
,
Bataille
,
A. R.
,
Gévry
,
N.
,
Adam
,
M.
,
Blanchette
,
M.
,
Robert
,
F.
and
Gaudreau
,
L.
(
2005
).
Variant histone H2A.Z is globally localized to the promoters of inactive yeast genes and regulates nucleosome positioning
.
PLoS Biol.
3
,
e384
.
Hardy
,
S.
,
Jacques
,
P.-E.
,
Gévry
,
N.
,
Forest
,
A.
,
Fortin
,
M.-E.
,
Laflamme
,
L.
,
Gaudreau
,
L.
and
Robert
,
F.
(
2009
).
The euchromatic and heterochromatic landscapes are shaped by antagonizing effects of transcription on H2A.Z deposition
.
PLoS Genet.
5
,
e1000687
.
Hong
,
J.
,
Feng
,
H.
,
Wang
,
F.
,
Ranjan
,
A.
,
Chen
,
J.
,
Jiang
,
J.
,
Ghirlando
,
R.
,
Xiao
,
T. S.
,
Wu
,
C.
and
Bai
,
Y.
(
2014
).
The catalytic subunit of the SWR1 remodeler is a histone chaperone for the H2A.Z-H2B dimer
.
Mol. Cell
53
,
498
-
505
.
Hsiao
,
Y.-L.
,
Chen
,
Y.-J.
,
Chang
,
Y.-J.
,
Yeh
,
H.-F.
,
Huang
,
Y.-C.
and
Pi
,
H.
(
2014
).
Proneural proteins Achaete and Scute associate with nuclear actin to promote formation of external sensory organs
.
J. Cell Sci.
127
,
182
-
190
.
Hsu
,
C.-C.
,
Shi
,
J.
,
Yuan
,
C.
,
Zhao
,
D.
,
Jiang
,
S.
,
Lyu
,
J.
,
Wang
,
X.
,
Li
,
H.
,
Wen
,
H.
,
Li
,
W.
et al.
(
2018
).
Recognition of histone acetylation by the GAS41 YEATS domain promotes H2A.Z deposition in non-small cell lung cancer
.
Genes Dev.
32
,
58
-
69
.
Ibarra-Morales
,
D.
,
Rauer
,
M.
,
Quarato
,
P.
,
Rabbani
,
L.
,
Zenk
,
F.
,
Schulte-Sasse
,
M.
,
Cardamone
,
F.
,
Gomez-Auli
,
A.
,
Cecere
,
G.
and
Iovino
,
N.
(
2021
).
Histone variant H2A.Z regulates zygotic genome activation
.
Nat. Commun.
12
,
7002
.
Jafar-Nejad
,
H.
,
Acar
,
M.
,
Nolo
,
R.
,
Lacin
,
H.
,
Pan
,
H.
,
Parkhurst
,
S. M.
and
Bellen
,
H. J.
(
2003
).
Senseless acts as a binary switch during sensory organ precursor selection
.
Genes Dev.
17
,
2966
-
2978
.
Jafar-Nejad
,
H.
,
Tien
,
A.-C.
,
Acar
,
M.
and
Bellen
,
H. J.
(
2006
).
Senseless and Daughterless confer neuronal identity to epithelial cells in the Drosophila wing margin
.
Development
133
,
1683
-
1692
.
Klages-Mundt
,
N. L.
,
Kumar
,
A.
,
Zhang
,
Y.
,
Kapoor
,
P.
and
Shen
,
X.
(
2018
).
The nature of actin-family proteins in chromatin-modifying complexes
.
Front. Genet.
9
,
398
.
Klein
,
B. J.
,
Ahmad
,
S.
,
Vann
,
K. R.
,
Andrews
,
F. H.
,
Mayo
,
Z. A.
,
Bourriquen
,
G.
,
Bridgers
,
J. B.
,
Zhang
,
J.
,
Strahl
,
B. D.
,
Côté
,
J.
et al.
(
2018
).
Yaf9 subunit of the NuA4 and SWR1 complexes targets histone H3K27ac through its YEATS domain
.
Nucleic Acids Res.
46
,
421
-
430
.
Koyano-Nakagawa
,
N.
,
Wettstein
,
D.
and
Kintner
,
C.
(
1999
).
Activation of Xenopus genes required for lateral inhibition and neuronal differentiation during primary neurogenesis
.
Mol. Cell. Neurosci.
14
,
327
-
339
.
Ku
,
M.
,
Jaffe
,
J. D.
,
Koche
,
R. P.
,
Rheinbay
,
E.
,
Endoh
,
M.
,
Koseki
,
H.
,
Carr
,
S. A.
and
Bernstein
,
B. E.
(
2012
).
H2A.Z landscapes and dual modifications in pluripotent and multipotent stem cells underlie complex genome regulatory functions
.
Genome Biol.
13
,
R85
.
Lee
,
S.
,
Lee
,
B.
,
Lee
,
J. W.
and
Lee
,
S.-K.
(
2009
).
Retinoid signaling and neurogenin2 function are coupled for the specification of spinal motor neurons through a chromatin modifier CBP
.
Neuron
62
,
641
-
654
.
Lin
,
H.
,
Zhu
,
X.
,
Chen
,
G.
,
Song
,
L.
,
Gao
,
L.
,
Khand
,
A. A.
,
Chen
,
Y.
,
Lin
,
G.
and
Tao
,
Q.
(
2017
).
KDM3A-mediated demethylation of histone H3 lysine 9 facilitates the chromatin binding of Neurog2 during neurogenesis
.
Development
144
,
3674
-
3685
.
Ma
,
P. C. M.
,
Rould
,
M. A.
,
Weintraub
,
H.
and
Pabo
,
C. O.
(
1994
).
Crystal structure of MyoD bHLH domain-DNA complex: perspectives on DNA recognition and implications for transcriptional activation
.
Cell
77
,
451
-
459
.
Mahmood
,
S. R.
,
Xie
,
X.
,
Hosny El Said
,
N.
,
Venit
,
T.
,
Gunsalus
,
K. C.
and
Percipalle
,
P.
(
2021
).
β-actin dependent chromatin remodeling mediates compartment level changes in 3D genome architecture
.
Nat. Commun.
12
,
5240
.
Marcellini
,
S.
,
Gibert
,
J.-M.
and
Simpson
,
P.
(
2005
).
achaete, but not scute, is dispensable for the peripheral nervous system of Drosophila
.
Dev. Biol.
285
,
545
-
553
.
Martire
,
S.
and
Banaszynski
,
L. A.
(
2020
).
The roles of histone variants in fine-tuning chromatin organization and function
.
Nat. Rev. Mol. Cell Biol.
21
,
522
-
541
.
Mavrich
,
T. N.
,
Jiang
,
C.
,
Ioshikhes
,
I. P.
,
Li
,
X.
,
Venters
,
B. J.
,
Zanton
,
S. J.
,
Tomsho
,
L. P.
,
Qi
,
J.
,
Glaser
,
R. L.
,
Schuster
,
S. C.
et al.
(
2008
).
Nucleosome organization in the Drosophila genome
.
Nature
453
,
358
-
362
.
Mizuguchi
,
G.
,
Shen
,
X.
,
Landry
,
J.
,
Wu
,
W.-H.
,
Sen
,
S.
and
Wu
,
C.
(
2004
).
ATP-driven exchange of histone H2AZ variant catalyzed by SWR1 chromatin remodeling complex
.
Science
303
,
343
-
348
.
Mlodzik
,
M.
,
Baker
,
N. E.
and
Rubin
,
G. M.
(
1990
).
Isolation and expression of scabrous, a gene regulating neurogenesis in Drosophila
.
Genes Dev.
4
,
1848
-
1861
.
Muller
,
J.
,
Oma
,
Y.
,
Vallar
,
L.
,
Friederich
,
E.
,
Poch
,
O.
and
Winsor
,
B.
(
2005
).
Sequence and comparative genomic analysis of actin-related proteins
.
Mol. Biol. Cell
16
,
5736
-
5748
.
Murphy
,
K. E.
,
Meng
,
F. W.
,
Makowski
,
C. E.
and
Murphy
,
P. J.
(
2020
).
Genome-wide chromatin accessibility is restricted by ANP32E
.
Nat. Commun.
11
,
5063
.
Pi
,
H.
,
Huang
,
S.-K.
,
Tang
,
C.-Y.
,
Sun
,
Y. H.
and
Chien
,
C.-T.
(
2004
).
phyllopod is a target gene of proneural proteins in Drosophila external sensory organ development
.
Proc. Natl. Acad. Sci. USA
101
,
8378
-
8383
.
Pickup
,
A. T.
,
Lamka
,
M. L. R.
,
Sun
,
Q.
,
Yip
,
M. L.
and
Lipshitz
,
H. D.
(
2002
).
Control of photoreceptor cell morphology, planar polarity and epithelial integrity during Drosophila eye development
.
Development
129
,
2247
-
2258
.
Poch
,
O.
and
Winsor
,
B.
(
1997
).
Who's who among the Saccharomyces cerevisiae actin-related proteins? A classification and nomenclature proposal for a large family
.
Yeast
13
,
1053
-
1058
.
Ranjan
,
A.
,
Mizuguchi
,
G.
,
FitzGerald
,
P. C.
,
Wei
,
D.
,
Wang
,
F.
,
Huang
,
Y.
,
Luk
,
E.
,
Woodcock
,
C. L.
and
Wu
,
C.
(
2013
).
Nucleosome-free region dominates histone acetylation in targeting SWR1 to promoters for H2A.Z replacement
.
Cell
154
,
1232
-
1245
.
Redon
,
C.
,
Pilch
,
D.
,
Rogakou
,
E.
,
Sedelnikova
,
O.
,
Newrock
,
K.
and
Bonner
,
W.
(
2002
).
Histone H2A variants H2AX and H2AZ
.
Curr. Opin. Genet. Dev.
12
,
162
-
169
.
Riddle
,
N. C.
,
Minoda
,
A.
,
Kharchenko
,
P. V.
,
Alekseyenko
,
A. A.
,
Schwartz
,
Y. B.
,
Tolstorukov
,
M. Y.
,
Gorchakov
,
A. A.
,
Jaffe
,
J. D.
,
Kennedy
,
C.
,
Linder-Basso
,
D.
et al.
(
2011
).
Plasticity in patterns of histone modifications and chromosomal proteins in Drosophila heterochromatin
.
Genome Res.
21
,
147
-
163
.
Ruhl
,
D. D.
,
Jin
,
J.
,
Cai
,
Y.
,
Swanson
,
S.
,
Florens
,
L.
,
Washburn
,
M. P.
,
Conaway
,
R. C.
,
Conaway
,
J. W.
and
Chrivia
,
J. C.
(
2006
).
Purification of a human SRCAP complex that remodels chromatin by incorporating the histone variant H2A.Z into nucleosomes
.
Biochemistry
45
,
5671
-
5677
.
Scacchetti
,
A.
and
Becker
,
P. B.
(
2021
).
Variation on a theme: evolutionary strategies for H2A.Z exchange by SWR1-type remodelers
.
Curr. Opin. Cell Biol.
70
,
1
-
9
.
Scacchetti
,
A.
,
Schauer
,
T.
,
Reim
,
A.
,
Apostolou
,
Z.
,
Campos Sparr
,
A.
,
Krause
,
S.
,
Heun
,
P.
,
Wierer
,
M.
and
Becker
,
P. B.
(
2020
).
Drosophila SWR1 and NuA4 complexes are defined by DOMINO isoforms
.
eLife
9
,
e56325
.
Seo
,
S.
,
Richardson
,
G. A.
and
Kroll
,
K. L.
(
2005
).
The SWI/SNF chromatin remodeling protein Brg1 is required for vertebrate neurogenesis and mediates transactivation of Ngn and NeuroD
.
Development
132
,
105
-
115
.
Singson
,
A.
,
Leviten
,
M. W.
,
Bang
,
A. G.
,
Hua
,
X. H.
and
Posakony
,
J. W.
(
1994
).
Direct downstream targets of proneural activators in the imaginal disc include genes involved in lateral inhibitory signaling
.
Genes Dev.
8
,
2058
-
2071
.
Takahashi
,
D.
,
Orihara
,
Y.
,
Kitagawa
,
S.
,
Kusakabe
,
M.
,
Shintani
,
T.
,
Oma
,
Y.
and
Harata
,
M.
(
2017
).
Quantitative regulation of histone variant H2A.Z during cell cycle by ubiquitin proteasome system and SUMO-targeted ubiquitin ligases
.
Biosci. Biotechnol. Biochem.
81
,
1557
-
1560
.
Tettey
,
T. T.
,
Gao
,
X.
,
Shao
,
W.
,
Li
,
H.
,
Story
,
B. A.
,
Chitsazan
,
A. D.
,
Glaser
,
R. L.
,
Goode
,
Z. H.
,
Seidel
,
C. W.
,
Conaway
,
R. C.
et al.
(
2019
).
A role for FACT in RNA polymerase II promoter-proximal pausing
.
Cell Rep
27
,
3770
-
3779.e3777
.
Villares
,
R.
and
Cabrera
,
C. V.
(
1987
).
The achaete-scute gene complex of D. melanogaster: conserved domains in a subset of genes required for neurogenesis and their homology to myc
.
Cell
50
,
415
-
424
.
Wapinski
,
O. L.
,
Vierbuchen
,
T.
,
Qu
,
K.
,
Lee
,
Q. Y.
,
Chanda
,
S.
,
Fuentes
,
D. R.
,
Giresi
,
P. G.
,
Ng
,
Y. H.
,
Marro
,
S.
,
Neff
,
N. F.
et al.
(
2013
).
Hierarchical mechanisms for direct reprogramming of fibroblasts to neurons
.
Cell
155
,
621
-
635
.
Weber
,
C. M.
,
Ramachandran
,
S.
and
Henikoff
,
S.
(
2014
).
Nucleosomes are context-specific, H2A.Z-modulated barriers to RNA polymerase
.
Mol. Cell
53
,
819
-
830
.
Whittle
,
C. M.
,
McClinic
,
K. N.
,
Ercan
,
S.
,
Zhang
,
X.
,
Green
,
R. D.
,
Kelly
,
W. G.
and
Lieb
,
J. D.
(
2008
).
The genomic distribution and function of histone variant HTZ-1 during C. elegans embryogenesis
.
PLoS Genet.
4
,
e1000187
.
Willhoft
,
O.
,
Ghoneim
,
M.
,
Lin
,
C. L.
,
Chua
,
E. Y. D.
,
Wilkinson
,
M.
,
Chaban
,
Y.
,
Ayala
,
R.
,
McCormack
,
E. A.
,
Ocloo
,
L.
,
Rueda
,
D. S.
et al.
(
2018
).
Structure and dynamics of the yeast SWR1-nucleosome complex
.
Science
362
,
eaat7716
.
Wu
,
W.-H.
,
Alami
,
S.
,
Luk
,
E.
,
Wu
,
C.-H.
,
Sen
,
S.
,
Mizuguchi
,
G.
,
Wei
,
D.
and
Wu
,
C.
(
2005
).
Swc2 is a widely conserved H2AZ-binding module essential for ATP-dependent histone exchange
.
Nat. Struct. Mol. Biol.
12
,
1064
-
1071
.
Xu
,
T.
and
Rubin
,
G. M.
(
1993
).
Analysis of genetic mosaics in developing and adult Drosophila tissues
.
Development
117
,
1223
-
1237
.
Zilberman
,
D.
,
Coleman-Derr
,
D.
,
Ballinger
,
T.
and
Henikoff
,
S.
(
2008
).
Histone H2A.Z and DNA methylation are mutually antagonistic chromatin marks
.
Nature
456
,
125
-
129
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information