Skip to main content
Advertisement
  • Loading metrics

Anteroposterior Limb Skeletal Patterning Requires the Bifunctional Action of SWI/SNF Chromatin Remodeling Complex in Hedgehog Pathway

  • Shin Jeon,

    Affiliation School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea

  • Rho Hyun Seong

    rhseong@snu.ac.kr

    Affiliation School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea

Abstract

Graded Sonic hedgehog (Shh) signaling governs vertebrate limb skeletal patterning along the anteroposterior (AP) axis by regulating the activity of bifunctional Gli transcriptional regulators. The genetic networks involved in this patterning are well defined, however, the epigenetic control of the process by chromatin remodelers remains unknown. Here, we report that the SWI/SNF chromatin remodeling complex is essential for Shh-driven limb AP patterning. Specific inactivation of Srg3/mBaf155, a core subunit of the remodeling complex, in developing limb buds hampered the transcriptional upregulation of Shh/Gli target genes, including the Shh receptor Ptch1 and its downstream effector Gli1 in the posterior limb bud. In addition, Srg3 deficiency induced ectopic activation of the Hedgehog (Hh) pathway in the anterior mesenchyme, resulting in loss of progressive asymmetry. These defects in the Hh pathway accompanied aberrant BMP activity and disruption of chondrogenic differentiation in zeugopod and autopod primordia. Notably, our data revealed that dual control of the Hh pathway by the SWI/SNF complex is essential for spatiotemporal transcriptional regulation of the BMP antagonist Gremlin1, which affects the onset of chondrogenesis. This study uncovers the bifunctional role of the SWI/SNF complex in the Hh pathway to determine the fate of AP skeletal progenitors.

Author Summary

Anteroposterior (AP) limb skeletal patterning is directed by morphogen Sonic hedgehog (Shh) signaling. Modulation of Shh responsiveness and repression of Shh pathway activity in distinct limb bud regions are essential for proper limb skeletal formation. Although the genetic networks involved in these processes have been identified, epigenetic control by chromatin remodeler remains unknown. We have unraveled the function of the SWI/SNF chromatin remodeling complex in Shh signaling during limb patterning. The complex activates the responses of the posterior limb progenitors to Shh, however, it represses the signaling in the anterior limb progenitors. Here we provide genetic evidence for the dual requirement of the SWI/SNF complex in Shh signaling to pattern AP limb skeletal elements.

Introduction

Vertebrate limb anteroposterior (AP) patterning is controlled by a diffusible morphogen, Sonic hedgehog (Shh), that is produced from the posteriorly located zone of polarizing activity (ZPA) [1]. Cell fate marking studies on mouse limb buds have revealed that Shh signaling regulates identities of limb skeletal elements, such as the ulna and digits 2 to 5, depending on the signal concentration and time of exposure to that signal [24]. During limb bud outgrowth, Shh promotes FGF signaling in the apical ectodermal ridge (AER) by mediating the BMP antagonist Gremlin1 (Grem1) that maintains low BMP activity [5].

In vertebrates, binding of Shh to its receptor Patched1 (Ptch1) enables the signal transduction through derepression of signal transducer Smoothened, allowing Gli transcription factors (Gli1−3) to function as activators (GliA) [6]. The transcriptional upregulation of Ptch1 serves as a sensitive readout of Shh activity and is required for sequestering diffusible ligands to restrain their spread within the target range [7, 8]. Notably, the spatiotemporal regulation of Ptch1 expression is important to prevent aberrant activation of Hedgehog (Hh) signaling, indicating that Ptch1 functions as a negative regulator of Hh signaling [9, 10]. Meanwhile, the full-length activators Gli2A and Gli3A contribute to the activation of Shh target genes such as Gli1, which might act as an indicator of the Shh signaling range in limb development [1113]. The absence of Shh signaling allows proteolytic processing of bifunctional Gli2 and Gli3 to form the truncated repressors Gli2R and Gli3R (GliR) [14, 15].

Gli3 functions as a major regulator of AP digit patterning, whereas Gli2 has compensatory roles of Gli3 activity [4, 1618]. During early limb bud development, Gli3 is required to establish AP polarity through mutual antagonism with Hand2 and is involved in the formation of two signaling centers, the ZPA and AER, by restraining GliA activity [10, 1921]. In addition, constitutive Gli3 expression during anterior digit patterning is mediated by repressing cell-cycle genes implicated in the proliferative expansion of Shh-dependent mesenchymal progenitors and by terminating Grem1 expression to initiate chondrogenic differentiation [22, 23].

Despite recent progress in identifying networks of trans-acting regulators interacting with multiple cis-regulatory modules (CRM) that orchestrate limb development, epigenetic control of the developmental process, especially the role of chromatin remodelers, remains poorly understood. The mammalian SWI/SNF chromatin remodeling complex is an ATP-dependent chromatin remodeler that uses the energy of ATP hydrolysis to alter nucleosomal structure [24]. The SWI/SNF complex is a multisubunit complex including core factors such as ATPase Brg1, tumor suppressor Snf5, and scaffolding subunit Srg3/mBaf155 (hereafter referred to as Srg3) [25]. In differentiation pathways, SWI/SNF complexes cooperate with histone-modifying factors and transcriptional regulators to mediate both transcriptional activation and repression in response to extracellular stimuli [26].

Here, we show that the SWI/SNF complex is essential for limb AP skeletal patterning. Specific inactivation of limb mesenchymal Srg3, resulting in defects in SWI/SNF complex activity [27], fails to upregulate posterior Shh/Gli target gene expression and induces the ectopic activation of target genes in the anterior limb bud after intact establishment of the ZPA. The SWI/SNF complex-mediated modulation of Shh responsiveness and repression of the ectopic Hh pathway determine the AP identities of limb progenitors and regulate the spatiotemporal expression of Grem1. Thus, bifunctional action of the SWI/SNF complex in the Hh pathway is essential to pattern AP limb skeletal elements.

Results

Srg3 is essential for anteroposterior limb skeletal patterning

To study the specific function of the SWI/SNF complex in developing limb buds, we used a conditional loss-of-function allele of Srg3 (Srg3f/f) [28] and a Prx1Cre transgene encoding a Cre recombinase that is activated in the early limb bud mesenchyme [29]. Prx1Cre-mediated inactivation of Srg3 in the limb bud mesenchyme was confirmed by measuring the expression of the transcript and protein in control and Srg3f/f;Prx1Cre (hereafter shortened as Srg3 CKO) limb buds. Whole-mount RNA in situ hybridization showed the specific clearance of Srg3 transcripts throughout the mesenchyme and western blot analysis confirmed the downregulation of Srg3 proteins with a time lapse between the fore- and hindlimb buds (S1A and S1B Fig). In addition, the downregulation of Brg1 observed in Srg3 CKO limb buds revealed the structural function of Srg3 that stabilizes the SWI/SNF complex (S1B Fig) [27].

Skeletal analysis of Srg3 CKO limbs at birth (P0) revealed the requirement of Srg3 for limb development (Fig 1). In Srg3 CKO forelimbs, the scapula developed poorly with bifurcated or enlarged foramen, aplastic clavicle, stylopod (humerus) lacking deltoid tuberosity, and radial agenesis were observed (Fig 1A and 1B). In Srg3 CKO hindlimbs, the proximal skeletons (pelvic girdle and femur) were retained normally, whereas zeugopod elements (tibia and fibula) were shortened to a similar extent (Fig 1C and 1D and S1C Fig). Both Srg3 CKO fore- and hindlimbs had rudimentary digits that were connected by ossified tissues in the anterior digital tips (syndactyly) and exhibited more severe ossification defects in anterior digits than those in posterior digits (Fig 1B and 1D and S1D Fig). Unlike predominant preaxial polydactyly in Srg3 CKO hindlimbs, digit number was variable in Srg3 CKO forelimbs (4 or less, 28%; 5, 34%; 6 or more, 38%, n = 84) (Fig 1E). The discrepancy in severity between fore- and hindlimbs lacking Srg3 is a likely consequence of Srg3 deficiency mediated by the onset timing of Prx1Cre activity, which is first activated in the prospective forelimb bud prior to hindlimb budding [29]. Taken together, the malformation of zeugopod elements and variable digit numbers observed in Srg3-deficienct limbs suggest that mesenchymal Srg3 is involved in AP limb skeletal patterning.

thumbnail
Fig 1. Srg3 is essential for anteroposterior limb skeletal patterning.

(A−D) Skeletal preparations of control and Srg3 CKO limbs at P0. The inset in (B) shows another scapula phenotype. Red arrowheads denote hypoplastic scapulae and black arrowheads indicate the loss of clavicle, deltoid tuberosity, and radius in the Srg3 CKO forelimb. The insets in (C) and (D) show a dorsal view of a hindlimb autopod marked with digit numbers. Red arrows point to the fused digits with soft tissues. cv, clavicle; dt, deltoid tuberosity; fe, femur; fi, fibula; hu, humerus; pg, pelvic girdle; r, radius; sc, scapula; ti, tibia; u, ulna; 1−5, digit identity. Scale bars: 1mm. (E) Percentages of digit number in Srg3 CKO forelimbs and hindlimbs. Upper panels show various types of cartilage structures in Srg3 CKO forelimb digits compared with control digits. Asterisks indicate hypoplastic digits.

https://doi.org/10.1371/journal.pgen.1005915.g001

Srg3 CKO forelimb buds establish distinct Hh pathways in the anterior and posterior mesenchyme

Given that limb bud development requires formation of the ZPA and AER [5], we first analyzed the formation of ZPA and AER signaling centers at early stages. In E10 Srg3 CKO forelimb buds, ZPA-Shh expression levels was similar with control expression levels (n = 8 limb buds analyzed), whereas AER-Fgf8 expression was slightly reduced in Srg3 CKO forelimb buds relative to controls (n = 6) (S2A Fig). Although Srg3 inactivation did not significantly alter the formation of signaling centers, subtle changes in the AER suggest that the SWI/SNF complex functions in initial limb development. To understand the mechanism underlying Srg3-mediated limb AP patterning controlled by the counteraction of Shh and Gli3 [16, 17], we examined the expression of Shh/Gli target genes. In Srg3 CKO forelimb buds, the expression domains of Gli1 and Ptch1 were normal up to at least E10 (Gli1, n = 12; Ptch1, n = 8), but were ectopically activated at E10.25 and at E10.75, respectively, in the anterior mesenchyme (Gli1 and Ptch1, n = 6) (Fig 2A and 2B). In addition, Gli1 and Ptch1 expression was activated in a graded manner along the AP axis in control forelimb buds, whereas their expression domains including ectopic regions were confined to the distal region in Srg3 CKO forelimb buds over time (Fig 2A and 2B; Gli1, n = 5; Ptch1, n = 6). Importantly, Ptch1 transcripts were not detected in the core mesenchyme of Srg3 CKO forelimb buds (Fig 2B). Gli1 was ectopically activated from around E11 in Srg3 CKO hindlimb buds, but its expression was comparable to control hindlimb buds in the posterior region (S2B Fig). These data suggest that Srg3 both activates and represses Shh/Gli target gene expression in distinct regions.

thumbnail
Fig 2. Srg3 CKO forelimb buds establish distinct Hh pathways in the anterior and posterior mesenchyme.

(A, B) Spatiotemporal distribution of Gli1 and Ptch1 transcripts in control and Srg3 CKO forelimb buds at indicated stages. Arrowheads denote the ectopic activation of Shh target genes in the anterior mesenchyme. Scale bars: 100 μm. (C−F) Spatial distribution of Shh, Grem1, Bmp4, and Fgf4 in E10.5 control and Srg3 CKO forelimb buds. Brackets in (C) mark the spatial extent of the Shh expression domain. The black arrow in (D) indicates the anterior expansion of Grem1 expression. Black arrowheads in (F) indicate the anterior and posterior end of the AER. Scale bars: 100 μm. (G−K) Spatial distribution of Hand2, Gli3, Hoxd13, and Alx4 in control and Srg3 CKO forelimb buds at indicated stages. Arrows in (I) and (J) indicate the anterior expansion of Hand2 and Hoxd13. Scale bars: 50 μm in (G−H); 100 μm in (I−K).

https://doi.org/10.1371/journal.pgen.1005915.g002

To define whether bifunctional action of Srg3 in the Hh pathway affects the interlinked signaling between the ZPA and the AER [30], we examined the expression pattern of epithelial-mesenchymal signaling genes during limb bud outgrowth. In Srg3 CKO forelimb buds, the size of the Shh expression domain was subtly reduced (Fig 2C; n = 6). Grem1 expression expanded anteriorly (n = 7), whereas Bmp4 expression was reduced in the anterior and posterior mesenchyme of Srg3 CKO forelimb buds (n = 4) (Fig 2D and 2E). AER-Fgf4 expression shifted anteriorly in Srg3 CKO forelimb buds (Fig 2F; n = 6). Taken together, these data suggest that distinct Hh pathways established by Srg3 deficiency differentially impacted epithelial-mesenchymal signaling in the anterior and posterior mesenchyme.

The polarization of nascent limb mesenchyme and establishment of the ZPA are controlled by antagonistic interactions between Hand2 and Gli3 in the posterior and anterior regions, respectively [1921]. To assess whether Srg3 deficiency in the limb bud mesenchyme affects AP polarity at the prepatterning stage, we examined the expression domains of Hand2 and Gli3 at E9.5. Consistent with the formation of an intact ZPA up to at least E10 (S2A Fig), the expression domains of these positional markers remained comparable to controls in Srg3 CKO forelimb buds (Fig 2G and 2H; Hand2, n = 8; Gli3, n = 9). During limb bud outgrowth, the distribution of posterior markers Hand2 and Hoxd13 was more posteriorly restricted or reduced in Srg3 CKO forelimb buds than in control limb buds, whereas their expression was activated in the anterior region at E10.75 (Fig 2I and 2J; Hand2, n = 7; Hoxd13, n = 5). By contrast, the expression domains of anterior markers Alx4 and Pax9 exhibited progressive decreases in Srg3 CKO forelimb buds (Fig 2K and S3A Figs; Alx4, n = 6; Pax9, n = 6) [31]. Consistently, the expression of anterior markers was mildly downregulated in Srg3 CKO hindlimb buds (S3B Fig), suggesting that the loss of anterior identity in Srg3-deficient limb buds correlates with the timing of Srg3 inactivation. Taken together, these data indicate that Srg3 deficiency progressively decreased the AP identities of limb progenitors, leading to a disruption of asymmetry after early specification of the AP axis.

Mesenchymal Srg3 deficiency induces ectopic Shh expression and distalizes epithelial-mesenchymal signaling at late stages

Inactivation of Srg3 in the limb bud mesenchyme caused progressive alterations in Shh/Gli target gene expression and in AP identity (Fig 2). To gain further insights into the regulation of Shh/Gli target genes by the SWI/SNF complex, we reexamined the distribution of epithelial-mesenchymal signaling genes at subsequent stages. In Srg3 CKO limb buds, Shh expression was ectopically induced in the anterior margin and subsequently expanded along the distal margin (Fig 3A and S4A Fig; n = 6 per stage). Ectopic Shh signaling reduced Gli3R protein levels by inhibiting Gli3 processing in the anterior mesenchyme of Srg3 CKO forelimb buds (Fig 3B). To test whether the SWI/SNF complex is directly implicated in repressing Shh through the regulation of limb-specific Shh enhancer ZRS (ZPA regulatory sequence), which is responsible for localized expression of Shh [19, 32], we performed a chromatin immunoprecipitation (ChIP) assay. We did not observe the enrichment of Srg3 at any regions on the ZRS (S4B Fig). This suggests that ectopic Shh expression is indirectly induced in Srg3 CKO limb buds.

thumbnail
Fig 3. Mesenchymal Srg3 deficiency induces ectopic Shh expression and distalizes epithelial-mesenchymal signaling at late stages.

(A) Expression pattern of Shh in control and Srg3 CKO forelimb buds at indicated stages. Arrowheads and arrow denote the anterior ectopic expression of Shh and its anterior expansion, respectively. (B) Western blot analysis of Gli3FL and Gli3R in lysates from the anterior (Ant) and posterior (Post) halves of E11 control and Srg3 CKO forelimb buds. Values represent the relative levels of Gli3R protein. α-tubulin served as a loading control. (C−E) Spatial distribution of Grem1, Fgf8, and Hoxd13 in control and Srg3 CKO forelimb buds. Red arrowheads denote the anterior ectopic expression. In (C), the black bracket indicates the dissociation of Grem1 expression domains and red brackets mark the distance between the distal expression of Grem1 and the AER. Black arrowheads in (D) point to the posterior end of Fgf8 expression. Asterisks in (E) indicate the distalized expression domain of Hoxd13. Scale bars in (A) and (C−E): 200 μm.

https://doi.org/10.1371/journal.pgen.1005915.g003

After ectopic Shh expression was activated, the anteriorly expanded domain of Grem1 at E10.5 was divided into two parts: the anterior domain and the posterior domain (Figs 2D and 3C; n = 7). In E11.5 Srg3 CKO forelimb buds, the derepressed expression of Grem1 in the anterior was remarkably reduced in the distal mesenchyme, whereas its posterior domain was distally shifted (Fig 3C). As the posterior domain of Grem1 closer to the AER reflects loss of FGF signaling repressing Grem1 [33], we assessed AER-Fgf8 expression and found the thinning and posterior loss of AER together with ectopic upregulation in the anterior end (Fig 3D; n = 6). Hoxd13 expression was also anteriorly expanded and confined to the distal mesenchyme in Srg3 CKO forelimb buds (Fig 3E; n = 6). Likewise, the expression of Grem1, Fgf8 and Hoxd13 was ectopically upregulated in the anterior margin of Srg3 CKO hindlimb buds (S4C–S4E Fig). Particularly, distalization of Grem1 and Hoxd13 expression domains was also observed in Srg3 CKO hindlimb buds (S4C and S4E Fig). These data reveal that low Shh response and anterior Hh pathway activity by Srg3 deficiency distalized epithelial-mesenchymal signaling and expanded the anterior digit progenitors.

Srg3-containing SWI/SNF complexes are required for the transcriptional activation and repression of Gli1 and Ptch1 in developing limb buds

To verify whether Srg3 directly regulates the expression of Shh/Gli target genes in developing limbs, we examined the effects of Srg3 deficiency by transducing a Cre-expressing retroviral vector into Srg3f/f mouse embryonic fibroblasts (MEFs). We focused our analyses on the transcriptional regulation of Shh/Gli target genes Gli1 and Ptch1. Quantitative real-time PCR (qPCR) showed that Srg3-deficient MEFs expressed higher levels of Gli1 and Ptch1, suggesting that the SWI/SNF complex represses Shh/Gli target genes (Fig 4A). To exclude the possibility that the SWI/SNF complex indirectly represses Shh/Gli target genes by other factors in the MEFs, we treated Srg3-deficient MEFs with the Hh pathway inhibitor cyclopamine [34]. Although cyclopamine reduced Gli1 and Ptch1 expression in control and Srg3-deficient MEFs, Srg3-deficient MEFs expressed higher levels of Gli1 and Ptch1 than control MEFs (Fig 4A). This indicates that the Srg3-containing SWI/SNF complex represses Shh/Gli target genes in, at least, a Hh-free condition. Thus, this finding could corroborate the derepression of Shh/Gli target genes in the anterior mesenchyme of Srg3 CKO limb buds. Next, we examined whether Srg3 is involved in the activation of Gli1 and Ptch1 expression upon Shh stimulation. In the presence of Shh-conditioned medium, Srg3-deficient MEFs displayed severely reduced activation levels of Gli1 (101- vs. 16.1-fold) and Ptch1 (7.79- vs. 2.96-fold), relative to controls (Fig 4B). These data suggest that Srg3 is required for responses to Shh, supporting findings that the distribution of Shh/Gli target genes was confined to the distal-posterior mesenchyme in Srg3 CKO forelimb buds.

thumbnail
Fig 4. Srg3 is required for the transcriptional activation and repression of Gli1 and Ptch1 in developing limbs.

(A, B) Quantitative PCR analysis of Gli1 and Ptch1 mRNA in Srg3f/f MEF infected with empty vector (Empty) or Cre-expressing viral vector (Cre). Each of infected MEFs was treated with an ethanol vehicle (EtOH) or cyclopamine (Cyc) (A), or incubated in Shh-conditioned media (+) or control media (−) (B). Error bars represent SD from six independent experiments. (*)P < 0.05; (**)P < 0.01. (C) Reciprocal immunoprecipitation of Srg3 with Gli2 and Gli3 proteins in E11.5 control limb buds. (D) Schematic representation of the relative positions of primer sets in the Gli1 locus (g1, g2) and Ptch1 locus (p1−p5) around the putative Gli-binding sites (red boxes) is shown on the top. Direction of transcription (arrows) and exons (numbers above boxes) are indicated. ChIP−qPCR analyses of DNA fragments precipitated with anti-Brg1 (b), anti-Srg3 (s), and IgG (i) in E11.5 control and Srg3 CKO limb buds. (E, F) ChIP−qPCR analyses of DNA fragments precipitated with anti-Gli2 (left) or anti-Gli3 (right) in MEFs (E) and in E11.5 limb buds (F). (G, H) ChIP−qPCR analyses of DNA fragments precipitated with anti-H3K27me3 from MEFs cultured in a basal condition (G) or in Shh-conditioned media (H). All data in (D−H) are represented as the percentage of input DNA, normalized to the value of nonspecific binding to the Gapdh promoter. Error bars represent SD from three independent experiments.

https://doi.org/10.1371/journal.pgen.1005915.g004

During limb development, Gli1 expression requires both transcription factors Gli2 and Gli3 [11], and Gli proteins regulate the expression of Ptch1 [23, 35]. We asked whether the bifunctional action of Srg3 requires an interaction with Gli2 and Gli3 to regulate Gli1 and Ptch1 expression in developing limbs. Reciprocal coimmunoprecipitation of Srg3 with Gli2 and Gli3 from E11.5 limb bud lysates revealed that Srg3 formed a complex with endogenous Gli2, Gli3FL, and Gli3R (Fig 4C). Using previously reported Gli-binding sites [23], we assessed Brg1 and Srg3 occupancy at the regulatory regions of Gli1 and Ptch1 by performing chromatin immunoprecipitation followed by qPCR (ChIP−qPCR) in E11.5 limb bud extracts. ChIP−qPCR analysis showed that both Brg1 and Srg3 proteins were enriched at the promoter regions of Gli1 and Ptch1 around Gli-binding regions in control limb buds, whereas their occupancies were considerably diminished in Srg3 CKO limb buds (Fig 4D). Furthermore, Brg1 and Srg3 were also enriched near the limb specific enhancer of Ptch1, which might be required for sensing graded Shh activity (Fig 4D, region p5) [35].

We next investigated whether loss of Srg3 affects the recruitment of Gli2 and Gli3 proteins to the regulatory region of Shh target genes. The occupancy of Gli2 and Gli3 proteins was not significantly changed at the regulatory regions of Gli1 and Ptch1 in Srg3-deficient MEFs (Fig 4E). However, we found that Gli2 occupancy of Gli-binding sites was increased and the occupancy of Gli3 was reduced in E11.5 Srg3 CKO limb buds at the promoter regions of Gli1 and Ptch1 relative to controls (Fig 4F, regions g1−g2 and p1−p4). These data indicate that Gli proteins bound to the Gli-binding sites were not affected by Srg3 deficiency and suggest that their enrichment was differentially influenced by ectopic Shh activity in E11.5 Srg3 CKO limb buds. By contrast, we also found the decreased occupancy of Gli2 and Gli3 proteins near the limb specific enhancer of Ptch1 in E11.5 Srg3 CKO limb buds (Fig 4F, region p5), suggesting that GliA contributed by ectopic Shh signals might have no significant effect on this region. We hypothesized that Srg3 deficiency affects histone modification at the promoter regions of Gli1 and Ptch1 because the expression domains of Gli1 and Ptch1 were not expanded throughout Srg3 CKO forelimb buds at E11.5, despite the high GliA and the low GliR condition. Indeed, SWI/SNF complexes functionally interact with histone modifying proteins [3638]. Furthermore, Shh signaling induces a loss of a repressive mark, trimethylation of histone 3 at lysine 27 (H3K27me3), by switching histone modifiers from methyltransferase Ezh2 to demethylase Jmjd3 [39]. To test our hypothesis, we compared H3K27me3 enrichment at the promoter regions of Gli1 and Ptch1 upon Srg3 deficiency against a previously reported distribution of H3K27me3 in MEFs [40]. Although Ezh2 and Suz12, components of the Polycomb repressive complex 2 (PRC2), were immunoprecipitated with both Gli2 and Gli3 in developing limbs (S5A Fig), there was no global change in H3K27me3 levels in Srg3-deficient MEFs or in the anterior and posterior mesenchyme of Srg3 CKO limb buds (S5B and S5C Fig). At the enriched regions of H3K27me3 on Gli1 and Ptch1 promoters, however, Srg3 deficiency resulted in decreased H3K27me3 level in a basal condition (Fig 4G, regions g1−g2 and p1−p2). Upon Shh stimulation, on the contrary, H3K27me3 levels at these regions in Srg3-deficient MEFs were significantly higher than in controls (Fig 4H). Taken together, these data suggest that Srg3-containing SWI/SNF complexes contribute to the activation and repression of Shh target genes through changes in the chromatin status of Gli binding regions.

Loss of mesenchymal Srg3 disrupted BMP signaling and caused defective chondrogenesis in forelimb buds

Posterior Shh signaling establishes limb skeletal structures including posterior zeugopod elements (ulna/fibula) and digits 2 to 5 [2, 4]. By contrast, loss of Gli3R or ectopic Shh signaling is detrimental to the formation of anterior skeletal structures [17, 31, 41]. To determine the role of bifunctional Srg3 in skeletal patterning, we assessed BMP activity, which promotes chondrogenesis at late stages [42]. In the anterior mesenchyme of Srg3 CKO forelimb buds, Msx2 expression, which marks BMP activity [30], was reduced at E10.75 and greatly abolished in the proximal region excluding the distal mesenchyme after ectopic Shh was induced (Fig 5A; n = 8 per stage). By contrast, posterior BMP activity remained low in Srg3 CKO forelimb buds (Fig 5A). Among Bmp ligands, the expression of both Bmp2 and Bmp4, but not Bmp7, was diminished in the posterior mesenchyme of Srg3 CKO forelimb buds at E11.75 (Fig 5B and S6 Fig; n = 6 per gene). Concurrent downregulation of Bmp2 and Bmp4, which are required to form the ulna and posterior digits 4 and 5 [43], could be causally implicated in the hypoplastic posterior skeletal elements of Srg3 CKO forelimbs (Fig 1). These results indicate that Srg3 deficiency disrupted BMP activities in the anterior and posterior mesenchyme.

thumbnail
Fig 5. Loss of mesenchymal Srg3 disrupted BMP signaling and caused defective chondrogenesis in forelimb buds.

(A, B) Spatiotemporal distribution of Msx2 and Bmp4 expression domains in control and Srg3 CKO forelimb buds at indicated stages. Red and black arrowheads indicate the anterior and posterior domains, respectively. (C) Spatiotemporal distribution of Sox9 in control and Srg3 CKO forelimb buds at indicated stages. Red and black arrowheads indicate the primordia corresponding to scapula and zeugopod, respectively. a, autopod; h, humerus; r, radius; sc, scapula; u, ulna; asterisks, digit rays. (D) Expression of Hoxa13 in E11.75 control and Srg3 CKO forelimb buds. Red and black arrowheads point to anterior expansion and posterior decrease, respectively. Scale bars in (A−D): 200 μm.

https://doi.org/10.1371/journal.pgen.1005915.g005

We next examined the distribution of Sox9, which marks the condensation of chondrogenic progenitors [44]. Consistent with partially developed proximal skeletal elements, Sox9 expression was diminished in the stylopod primordia of Srg3 CKO forelimb buds prior to E10.75 (Fig 5C, left panel; n = 6). The expression of Hoxa9, Hoxd9, and Hoxd10 of paralogous hox groups Hox9 and Hox10, which control the formation of proximal skeletal elements [45, 46], was reduced in the proximal region of Srg3 CKO forelimb buds (S7A Fig; n = 4 per gene). The expression of Irx3 and Irx5, which are essential for patterning proximal and anterior skeletal structures [31], was also downregulated in the proximal anterior region of Srg3 CKO forelimb buds (S7B Fig; n = 4 per gene). These data suggest that the Srg3-containing complexes might be required to pattern proximal skeletons. As limb bud outgrowth distally proceeds, Sox9-expressing progenitors were also decreased in the zeugopod and autopod primordia of Srg3 CKO forelimb buds (Fig 5C, middle and right panel; n = 8). Particularly, Sox9-expressing autopod progenitors in Srg3 CKO forelimb buds did not initiate mesenchymal condensation. Furthermore, Hoxa13, which delineates the presumptive autopod territories [47], was distalized and relatively enhanced in the anterior region (Fig 5D; n = 5). Taken together, deficiency of mesenchymal Srg3 progressively resulted in the loss of Sox9-positive progenitors in zeugopod and autopod primordia and this loss was paralleled by alterations in BMP activity.

Genetic interaction between Srg3 and Twist1 reveals synergism in the anterior zeugopod development

We tested whether ectopic Shh activity impacts anterior zeugopod development, as demonstrated by the absence of the radius and hypoplastic tibia in Srg3 CKO limbs. We introduced a single conditional allele of Twist1 (Twist1f/+), which represses Shh expression in the anterior mesenchyme [41], into the Srg3 CKO background. Twist1f/+;Prx1Cre forelimbs were phenotypically similar to Srg3 CKO forelimbs, except for more severe defects in the scapula (n = 13/13) (compare S8A Fig with Fig 1B). However, Twist1f/+;Srg3f/f;Prx1Cre hindlimbs displayed ossification defects and syndactyly in the anterior autopods similar to those of Srg3 CKO hindlimbs (Fig 1D and S8B Fig, arrow), but Twist1f/+;Srg3f/f;Prx1Cre hindlimbs exhibited a complete absence of tibia (S8B Fig, arrowhead). Consistent with this skeletal phenotype, Sox9-positive progenitors of the tibia primordia were reduced in Twist1f/+;Srg3f/f;Prx1Cre hindlimbs relative to Twist1f/+;Prx1Cre hindlimbs (S8C Fig; n = 6). In addition, ectopic expression of Gli1 in Twist1f/+;Srg3f/f;Prx1Cre hindlimb buds was activated earlier than in Twist1f/+;Prx1Cre and Srg3 CKO hindlimb buds (compare S8D Fig with S2B Fig; n = 7). However, early activation of ectopic Shh expression was not detected in Twist1f/+;Srg3f/f;Prx1Cre hindlimb buds (S8E Fig; n = 6). These data suggest that anterior zeugopod development might be affected by SWI/SNF complex-mediated epigenetic changes including the ectopic Hh pathway and not simply because of the Shh ligand-dependent pathway. This finding supports the idea that the fate of anterior skeletal progenitors is progressively determined.

Bifunctional action of SWI/SNF complex in the Hh pathway regulates the spatiotemporal expression of Grem1

The low to high transition of BMP activity by the timely termination of Grem1 expression is required to initiate condensation and chondrogenic differentiation of proliferative digit progenitors [30, 43]. To determine the effect of bifunctional action of the SWI/SNF complex on chondrogenic differentiation, Grem1 expression and BMP activity were analyzed at later stages. By E11.75, Grem1 expression began to be downregulated throughout the entire mesenchyme of control forelimb buds, but its decline was not observed and its separated domains became closer than those at E11.5 in Srg3 CKO forelimb buds (compare Fig 6A left panel with Fig 3C, black brackets; n = 8). At E12.5, Grem1 expression was cleared from the presumptive digit territories and confined to the interdigital mesenchyme in control forelimbs, but these spatial pattern changes were not observed in the autopods of Srg3 CKO forelimbs (Fig 6A, right panel; n = 6). Although Msx2 expression in the anterior margin of Srg3 CKO forelimb autopods was comparable to that in controls, it was undetectable in the interdigital mesenchyme (Fig 6B; n = 8). By contrast, the increased expression of Grem1 and low BMP activity were observed in the anterior region of Srg3 CKO hindlimb autopods (S9A Fig). We next examined whether delayed temporal kinetics of Grem1 in Srg3 CKO autopods is correlated with chondrogenesis of digit primordia and with digit separation. In Srg3 CKO autopods, the distributions of Sox9 and its target gene Col2a1 revealed delayed mesenchymal condensations, and anterior digit progenitors were relatively less condensed than posterior ones (Fig 6C and S9B Fig; n = 8 per gene). In addition, the comparison of Col2a1 distributions in Srg3 CKO fore- and hindlimb autopods revealed that both the extent of Grem1 propagation and its anterior upregulation caused the sequential onset of chondrogenesis in the posterior and anterior autopods. At this stage, Lysotracker Red staining in Srg3 CKO forelimb autopods showed increases of apoptotic cells in the distal mesenchyme underlying the AER, likely as a consequence of growth defects (Fig 6D, left panel; n = 6 per stage). Furthermore, the reduction of cell death in the interdigital mesenchyme, resulting in soft tissue syndactyly, was observed in the anterior autopods of Srg3 CKO forelimbs at E13.5 and hindlimbs at E12.5 (Fig 6D, right panel and S9C Fig). Taken together, these data indicate that the spatiotemporal regulation of Grem1 by the SWI/SNF complex is involved in digit determinative processes as well as in cell survival of expanding autopod progenitors.

thumbnail
Fig 6. Bifunctional action of the SWI/SNF complex in the Hh pathway regulates the spatiotemporal expression of Grem1.

(A, B) Distribution of Grem1 and Msx2 transcripts in control and Srg3 CKO forelimb autopods at indicated stages. Black bracket points to the distance between two domains of Grem1. Spatial restriction to the interdigital mesenchyme (arrowheads), combined region of separate domains (arrow), and enhanced region (red bracket) of Grem1 are indicated. (C) Expression pattern of Sox9 and Col2a1 in E12.5 control and Srg3 CKO forelimb autopods. Brackets indicate the delayed region of chondrogenic differentiation. Numbers 1 to 5 indicate the primordia of digit rays 1 to 5. Asterisks indicate the preceding chondrogenesis of shortened digit primordia in the posterior. (D) Distribution of apoptotic cells by Lysotracker Red staining in control and Srg3 CKO forelimb autopods at E12.5 and E13.5. Arrow points to increased cell death. Arrowheads indicate the reduction of interdigital cell death. Scale bars in (A−D): 200 μm.

https://doi.org/10.1371/journal.pgen.1005915.g006

Discussion

Our genetic analysis has shown that the SWI/SNF complex is required to modulate Shh responsiveness and repress the ectopic Hh pathway. Although specification of the AP limb bud axis is not affected by conditional inactivation of Srg3 in the limb bud mesenchyme, Srg3 CKO posterior progenitors fail to respond to graded Shh activity, leading to the redistribution of epithelial-mesenchymal signaling to the distal region. In parallel, loss of Srg3 causes the activation of ligand-independent and subsequent ligand-dependent Hh pathway in the anterior mesenchyme, resulting in the loss of anterior identity over time. Our analysis also reveals the dual requirement of the SWI/SNF complex in the Hh pathway for spatiotemporal regulation of Grem1.

Posterior limb skeletal elements are patterned depending on Shh signaling [2, 4]. By contrast, recent reports have shown that formation of proximal and anterior limb skeletons is inhibited by early Hh activity prior to establishment of the ZPA and by activation of the anterior Hh pathway during limb patterning [10, 31]. Skeletal phenotypes in Srg3 CKO forelimbs suggest that the Srg3-containing SWI/SNF complex is required for these distinct responses to Hh signaling. It has been known that SWI/SNF complexes and Polycomb group (PcG) proteins have antagonistic functions in repressing differentiation-related genes of embryonic stem cells [38]. In anterior limb buds, however, the SWI/SNF complexes appear to function synergistically with PcG proteins to repress the basal expression of Shh target genes. Consistent with our findings, deletion of H3K27 methyltransferase Ezh2, a catalytic subunit of PRC2, leads to ectopic expression of Shh target genes in anterior limb buds as well as derepression of Shh target genes in MEFs [39, 48]. Given that the PRC2 interacts with Gli proteins in developing limbs, PRC2 complexes are also likely to be involved in Gli-mediated repression of Shh target genes in anterior limb buds. In addition to the repressive function in the anterior limb bud, it is assumed that the SWI/SNF complexes also act cooperatively with H3K27 demethylases in activating Shh-induced target genes. It has been demonstrated that the SWI/SNF complexes functionally interact with H3K27 demethylases such as Jmjd3 and Utx in various tissues such as developing lungs and hearts [36, 37]. Particularly, a recent report showed changes in the epigenetic environment by switching Ezh2-PRC2 to Jmjd3 for Shh-induced target gene activation [39]. This implies that cooperative action between the SWI/SNF complex and Jmjd3 might be required for Shh target gene activation during limb development. Previous studies regarding SWI/SNF components have demonstrated that Snf5 deficiency leads to ectopic expression of Gli1 in developing limbs [49], and ATPase Brg1 is involved in the regulation of Shh target genes in an ATPase activity-independent manner during neural development [50]. However, we have presented genetic evidence showing bifunctional action of the SWI/SNF complex in distinct territories of limb bud mesenchyme. We do not exclude the possibility that the SWI/SNF complex acts cooperatively with other chromatin regulators such as histone deacetylase (HDAC) that is associated with Shh/Gli pathway in developing limbs [50, 51]. In addition, the phenotypes observed in Srg3 CKO limbs raise the possibility that the SWI/SNF complex likely controls the expression of other transcriptional regulators not specific to the Shh signaling pathway, such as Bmp and Hox genes. Further studies, including genome wide mapping of a H3K27Ac enhancer mark from the anterior and posterior limb buds of Srg3 CKO embryos, will help to elucidate the distinct regulatory functions of the SWI/SNF complex in chondrogenic differentiation and proximal patterning.

In Srg3 CKO forelimbs, one notable phenotype is the formation of variable digits, unlike polydactyly in hindlimbs. Concomitant deletion of Gli2 and Gli3 completely eliminates Gli1 expression but does not lead to digit loss in developing limbs [4, 11, 18]. Prx1Cre-mediated early deletion of Ptch1, however, causes oligodactyly and is accompanied by activation of the Hh pathway, whereas late Ptch1 depletion causes polydactyly [9, 10]. Importantly, we have uncovered the requirement of the SWI/SNF complex for robust expression of Ptch1. Thus, the core mesenchymal deficiency of Ptch1 expression, resulting from its posterior restriction, may lead to reduced Shh activity sensing and restrain posterior digit formation in Srg3 CKO forelimbs. In Srg3 CKO forelimb buds, the reduced sensing of Shh causes distalization of epithelial-mesenchymal signaling and Hoxa13/Hoxd13-positive presumptive autopod regions, markedly similar to limb buds conditionally lacking Ptch1 [9, 35]. Recent studies on the mammal species with two to four digits might support variable digit patterning by altered Ptch1 expression observed in Srg3 CKO forelimb buds [35, 52]. We assume that the extent of digit loss might be dependent on the integrity of the SWI/SNF complex controlled by Srg3. Meanwhile, ectopic Shh expression was induced in Srg3 CKO limb buds, although there is no enrichment for Srg3 on the ZRS. It has been shown that ectopic expression of Hoxd13 and Hand2 leads to misexpression of Shh in anterior limb buds [5355]. These molecular changes observed in Srg3 CKO limb buds may result in ectopic expression of Shh, causing preaxial polydactyly. Taken together, variable digit patterning in Srg3 CKO forelimbs appears to occur through combinatorial actions of altered Ptch1 expression and ectopic anterior Hh activity.

Both the proximal and distal BMP activities in the anterior mesenchyme of Srg3 CKO forelimb buds are distinct from those of Gli3-deficient limb buds [22]. The comparison of anterior zeugopod development and digit numbers between Srg3 CKO fore- and hindlimbs showed that the dose and exposed duration of ectopic Hh activity negatively impact the differentiation of anterior prechondrogenic progenitors. Our data and previous reports have demonstrated that the expansion of Hh signaling has an inhibitory effect on the formation of proximal and anterior skeletal elements [10, 31, 41]. In this regard, the proliferative expansion of anterior progenitors negatively controlled by Gli3 might require time to ensure a sufficient population such as both Irx3- and Irx5-positive early progenitors [22, 31]. Particularly, the genetic interaction between Srg3 and Twist1 showed synergism in limb skeletal formation such as in anterior zeugopod development. Twist1 not only functions as a Shh repressor but also controls the onset of osteoblast differentiation [41, 56]. It is possible that the repressive roles of Twist1 in developmental processes might contribute to recruit chromatin regulators such as the PcG, for example, in promoting the epithelial-mesenchymal transition and in suppressing mesenchymal stem cell senescence [57, 58]. The functional interaction of the SWI/SNF complex with transcriptional regulators acting either as activators or as repressors, which can recruit enzymes that modify active or repressive histone marks, may reveal synergistic and antagonistic actions of gene regulation at the chromatin level.

Derepression is one of the regulatory mechanisms underlying limb bud patterning. Our data highlight the sustained requirement of the SWI/SNF complex for transcriptional regulation of Grem1, a major Gli target gene controlled by derepression [23]. The expression of Grem1 in the limb bud is severely reduced in Shh−/− mutants and symmetrically expanded in both Gli3−/− and Shh−/−;Gli3−/− mutants [16, 17, 59]. Compared with previous observations, Grem1 expression in Srg3 CKO forelimb buds is dynamically redistributed, possibly a consequence of the reconstitution of the GliA/GliR gradient by low Shh responsiveness and ectopic Shh activity. Consistently, it has recently been suggested that limb-specific enhancers integrated by multiple posterior GliA- and anterior GliR-dependent CRMs regulate the transcriptional activity of Grem1 [60]. Furthermore, the combined region of Grem1 expression domains in Srg3 CKO forelimb buds indicates that the definitive digit identity in this region could be progressively determined by altered Hh activity (Fig 6). Thus, our analysis suggests that bifunctional action of the SWI/SNF complex in the Hh pathway is essential for spatiotemporal regulation of Grem1 that mediates AP skeletal patterning elicited by GliA and GliR functions [18, 22].

We have demonstrated that the SWI/SNF complex plays decisive roles in conferring graded Shh signaling upon developing limb progenitor cells. The SWI/SNF complex influences the progression of interlinked morphogen signaling pathways by modulating Shh responsiveness in the posterior limb bud and by repressing the Hh pathway in Shh-free regions. Our study showing the effects of epigenetic regulation by the SWI/SNF chromatin remodeling complex on limb patterning provides insights into deciphering developmental processes directed by morphogen gradients.

Materials and Methods

Ethics statement

All experiments with animals were performed according to the guidelines established by the Seoul National University Institutional Animal Care and Use Committees (SNUIACUC). SNUIACUC approved this study (approval number: SNU-130503-2). CO2 gas was used for animal euthanasia.

Mice and embryos

Generation of mice carrying a conditional allele of Srg3 (Srg3f/f) was previously described [28]. Srg3f/f, Prx1Cre [29], and Twist1f/f mice [41] were bred and maintained on a C57BL/6J genetic background. For all experiments, Srg3+/+;Prx1Cre and Srg3f/+;Prx1Cre mice and embryos harboring a Prx1Cre transgene were used as wild-type controls.

Whole-mount in situ hybridization

The transcript distributions were assessed by whole-mount in situ hybridization according to the standard procedures as described [61] with the following minor modifications: embryos were permeabilized in proteinase K (10 μg/ml) in PBST at room temperature for 11 min (E9.5−E10.5), 14 min (E10.5−E11.5) or 17 min (E11.5−E12.5) for analysis of limb mesenchyme and briefly for 3 min regardless of age for analysis of AER. All probes were linearized with the appropriate restriction enzyme and labeled using digoxigenin RNA labeling mix (Roche) with the appropriate polymerase (T7, T3 or SP6). Shh, Gli1, Bmp2, Bmp4 and Bmp7 probes were kindly provided by Y. Kong (Seoul National University). Fgf4 (Addgene plasmid #22085) [62] and Fgf8 (Addgene plasmid #22088) [63] probes were gifts from G. Martin. Hoxa9, Hoxd9 and Hoxd10 probes were generously provided by D. Wellik and Irx3 and Irx5 probes were provided by C. Hui. Other probes were amplified by PCR from cDNA fragments encompassing at least two exons (about 400−600 bp) of target genes and cloned into pGEM-T vectors (Promega). All representative expression patterns were obtained by analyzing at least three independent embryos per probe.

Skeletal staining and detection of apoptotic cells

Skeletal preparations and detection of apoptotic cells were performed as previously described [19, 30]. For analysis of skeletal structures, samples were collected at E14.5 and P0 and cartilages and bones were stained with Alcian Blue and Alizarin Red, respectively. Distribution of apoptotic cells in whole limb buds was analyzed using Lysotracker Red (Molecular Probes L-7528, Invitrogen).

Cell culture

Primary Mouse Embryonic Fibroblasts (MEFs) prepared from E13.5 Srg3f/f embryos, HEK293T, and Phoenix-eco cells were grown in DMEM medium (WelGENE) supplemented with 10% fetal bovine serum (FBS). For generation of Srg3-deficient MEFs, Phoenix-eco packaging cells were transfected with retroviral vectors expressing GFP alone (Empty) as a control or Cre-recombinase (Cre) by calcium phosphate method and their retroviral supernatants were harvested 2 d after transfection. MEFs were infected with the retroviral supernatant by spin infection for 90 min at 2500 rpm in the presence of 8 μg/ml polybrene. For inhibition of Hh signaling, MEFs were treated with 5 μM cyclopamine dissolved in ethanol vehicle for 24 h. For Shh stimulation, HEK293T cells were transiently transfected with ShhN expressing vector (kindly provided by M. Kang, Korea University Guro Hospital). Shh conditioned medium produced from transfected HEK293T cells was replaced with DMEM containing 2% FBS 24 h before harvesting and filtering of medium, and then added to MEFs for 24 h. Shh stimulated or cyclopamine treated MEFs were harvested for qPCR.

Immunoprecipitation (IP) and western blotting

IP and western blotting were performed as previously described [19, 28]. Limb bud lysates were immunoprecipitated or detected with following antibodies: Gli2 (R&D systems), Gli3 (R&D systems), α-tubulin (Sigma), Ezh2 (BD transduction), Suz12 (Cell signaling), H3K27me3 (Millipore), Histone H3 (Abcam), and rabbit polyclonal IgG (Millipore). Antisera for Brg1 and Srg3 were raised from rabbits in our laboratory. The band density of Gli3R level was quantified using ImageJ software (NIH) and normalized to α-tubulin as a loading control.

Chromatin immunoprecipitation (ChIP)

E11.5 control and Srg3f/f;Prx1Cre limb buds were dissected in cold PBS and minced with a douncer and MEFs were trypsinized. Dissociated tissues and MEFs were crosslinked in 1% formaldehyde (Sigma) for 10 min on a rotator at RT and were lysed for 10 min on ice with SDS lysis buffer (1% SDS, 50mM Tris-Cl (pH 8.1), 10mM EDTA). Lysates were sonicated to an average length of 200–500 bp using a Bioruptor sonicator and diluted 10-folds in dilution buffer (20mM Tris-Cl (pH 8.1), 150mM NaCl, 1% Triton X-100, 2mM EDTA). To reduce nonspecific background, samples were precleared for minimally 1 h with salmon-sperm DNA/Protein-A or G agarose (50% slurry, Millipore). Precleared lysates were incubated overnight on a rotator at 4°C with anti-Brg1, anti-Srg3, anti-H3K27me3 (Millipore), anti-Gli2 (abcam), anti-Gli3 (R&D systems) or with isotype-control anti-rabbit IgG (Millipore) as a negative control. Washing, elution and reverse-crosslinking of DNA-immunocomplexes and DNA purification were enriched as previously described [28]. Purified DNA was analyzed by qPCR with the following primers:

g1 forward: 5’-CCGGCACCCCCTCTCTAG-3’,

g1 reverse: 5’- GGCTCTTCCCGCTCACTTC-3’,

g2 forward: 5’-TTGCTCCCCGCTCTGAATC-3’,

g2 reverse: 5’-CTTGATGCTGTTCCCAAAGCT-3’,

p1 forward: 5’-AGGACACAATGCACCTGAGG-3’

p1 reverse: 5’-AGGTCTTGTGGGTGCCTCTA-3’

p2 forward: 5’-TAGTGGCGAGAATGACAGCG-3’

p2 reverse: 5’-TTTCTCCCTACCAACCGCAG-3’,

p3 forward: 5’-ACACACTGGCGCACTATCCA-3’,

p3 reverse: 5’-CCTCAAGCTGCAGCAAATACTG-3’,

p4 forward: 5’-GAATGGGAGAGGGAGGAAAGAT-3’,

p4 reverse: 5’-GCGGGAGCTCAGTTAGGAAA-3’,

p5 forward: 5’-TCTTCCAGCATGCTTACCTCTTT-3’,

p5 reverse: 5’-GCTTGGCCGCTGTAATCAAA-3’.

Supporting Information

S1 Fig. Prx1Cre-mediated inactivation of Srg3 in the limb bud mesenchyme and skeletal phenotypes of Srg3 CKO limbs.

(A) Whole-mount in situ hybridization reveals the distribution of Srg3 transcripts in E9.5 forelimb buds and E10.5 hindlimb buds of control and Srg3 CKO embryos. Scale bars: 100 μm. (B) Immunoblot analysis of Brg1 and Srg3 proteins in E10.5 control and Srg3 CKO forelimb buds and hindlimb buds. α-tubulin was used as loading control. (C) Skeletal structures of zeugopod elements in hindlimbs of control and Srg3 CKO pups (P0) and embryos (E14.5). Tibia (ti) and fibula (fi) were shortened in Srg3 CKO hindlimbs compared with control. (D) Bright-field images of control and Srg3 CKO autopods at E16. Arrows indicate syndactyly in the anterior region of Srg3-deficient autopods.

https://doi.org/10.1371/journal.pgen.1005915.s001

(TIFF)

S2 Fig. Formation of limb signaling centers and anterior ectopic activation of Hh pathway in Srg3 CKO limb buds.

(A) The expression of Shh and Fgf8 sensing the ZPA and the AER, respectively, in control and Srg3 CKO forelimb buds at E10. Arrowhead indicates the reduced activity of AER. (B) The distribution of Gli1 and Ptch1 transcript in control and Srg3 CKO hindlimb buds at indicated stages. Anterior ectopic expression of Gli1 in Srg3 CKO hindlimb buds was observed later than that in mutant forelimb buds. Arrowheads indicate anterior ectopic expression. Scale bars in (A−B): 100 μm.

https://doi.org/10.1371/journal.pgen.1005915.s002

(TIFF)

S3 Fig. The extent of loss of anterior identity in Srg3 CKO limb buds correlates with the timing of Srg3 inactivation.

(A, B) The expression of anterior marker genes Alx4 and Pax9 in E11.5 control and Srg3 CKO forelimb buds (A) and hindlimb buds (B) is indicated. Scale bars: 100 μm.

https://doi.org/10.1371/journal.pgen.1005915.s003

(TIFF)

S4 Fig. Expression pattern of epithelial-mesenchymal signaling genes in control and Srg3 CKO hindlimb buds at late stages and the enrichment of Srg3 on the ZRS.

(A, C−E) Spatial distribution of Shh, Grem1, Fgf8 and Hoxd13 in control and Srg3 CKO hindlimb buds. Red arrowheads denote anterior ectopic expression. The anterior expansion of Shh (A, arrow) and the distalized expression of Grem1 (C, red brackets) and Hoxd13 (E, asterisk) were observed in Srg3 CKO hindlimb buds, similarly to forelimb buds. Scale bars: 100 μm. (B) ChIP−qPCR analyses of DNA fragments precipitated with anti-Srg3 and IgG in E11.5 control limb buds. Bottom panel is a schematic representation of the relative positions of primer sets (blue lines: z1-z10) using the sequence of ZRS region within intron 5 of Lmbr1 gene. Gli binding region (g1) was used as a positive DNA control and anti-IgG as a negative antibody control.

https://doi.org/10.1371/journal.pgen.1005915.s004

(TIFF)

S5 Fig. Loss of Srg3 does not lead to global change in the level of H3K27me3 in MEFs and in the anterior and posterior limb buds.

(A) Gli2 and Gli3 proteins interact with PRC2 components Ezh2 and Suz12 in developing limbs. PC indicates the preclear beads as a negative control. (B, C) Immunoblot analysis of H3K27me3 in Srg3-deficient MEFs (B) and in the anterior and posterior regions of Srg3 CKO forelimb buds (C). Histone H3 was used as a loading control.

https://doi.org/10.1371/journal.pgen.1005915.s005

(TIFF)

S6 Fig. Expression pattern of Bmp2 and Bmp7 in E11.75 control and Srg3 CKO forelimb buds.

Arrowheads indicate the posterior domain of Bmp2 transcript. Scale bars: 200 μm.

https://doi.org/10.1371/journal.pgen.1005915.s006

(TIFF)

S7 Fig. Expression pattern of proximal skeletal patterning-related genes in control and Srg3 CKO forelimb buds.

(A) The expression of Hoxa9, Hoxd9, Hoxd10 in control and Srg3 CKO hindlimb buds at indicated stages. (B) The expression of Irx3 and Irx5 in control and Srg3 CKO hindlimb buds at indicated stages. Scale bars in (A−B): 200 μm.

https://doi.org/10.1371/journal.pgen.1005915.s007

(TIFF)

S8 Fig. Genetic interaction between Srg3 and Twist1 reveals the synergistic effect in the anterior zeugopod development.

(A) Skeletal preparations from Twist1f/+;Prx1Cre and Twist1f/+;Srg3f/f;Prx1Cre forelimbs at P0. Red arrowhead indicates the defect in scapula development. (B) Skeletal preparations from Twist1f/+;Prx1Cre and Twist1f/+;Srg3f/f;Prx1Cre hindlimbs at P0. Red arrowhead indicates loss of tibia. Red arrow indicates the ossification defects and syndactyly in anterior autopod. fe, femur; fi, fibula; pg, pelvic girdle; ti, tibia. Scale bars in (A−B): 1mm. (C−E) Expression pattern of Sox9, Gli1 and Shh in Twist1f/+;Prx1Cre and Twist1f/+;Srg3f/f;Prx1Cre hindlimb buds at indicated stages. Red arrowhead indicates the reduction of Sox9 expression in the tibia primordia. Scale bars: 100 μm

https://doi.org/10.1371/journal.pgen.1005915.s008

(TIFF)

S9 Fig. The increase of Grem1 expression in the anterior mesenchyme induces the sequential onset of chondrogenesis in the posterior and anterior autopods of Srg3 CKO hindlimbs.

(A, B) Distribution of Grem1, Msx2, Sox9 and Col2a1 transcripts in control and Srg3 CKO hindlimbs at indicated stages. Red brackets in (A) indicate the upregulated region of Grem1 expression and the downregulated region of Msx2 expression in Srg3 CKO hindlimb autopods. Arrowheads in (B) mark the initiation of mesenchymal condensation giving rise to digit ray primordia. Red brackets denote the region of delayed chondrogenesis. Numbers 1 to 5 indicate the primordia of digits 1 to 5. Asterisk indicates reduced digit primordia. (C) Lysotracker Red staining reveals the decrease of apoptotic cells in the anterior interdigital mesenchyme of Srg3 CKO hindlimb autopod (arrowheads). Scale bars in (A−C): 100 μm

https://doi.org/10.1371/journal.pgen.1005915.s009

(TIFF)

Acknowledgments

We thank G. Martin for Fgf4 and Fgf8 in situ probes. We are grateful to D. Wellik for generously providing Hoxa9, Hoxd9 and Hoxd10 in situ probes and to C. Hui for Irx3 and Irx5 in situ probes. We are also grateful to S.K. Im for in situ hybridization technical advice.

Author Contributions

Conceived and designed the experiments: SJ RHS. Performed the experiments: SJ. Analyzed the data: SJ RHS. Contributed reagents/materials/analysis tools: SJ RHS. Wrote the paper: SJ RHS.

References

  1. 1. Riddle RD, Johnson RL, Laufer E, Tabin C. Sonic hedgehog mediates the polarizing activity of the ZPA. Cell. 1993;75(7):1401–1416. pmid:8269518
  2. 2. Harfe BD, Scherz PJ, Nissim S, Tian H, McMahon AP, Tabin CJ. Evidence for an expansion-based temporal Shh gradient in specifying vertebrate digit identities. Cell. 2004;118(4):517–528. pmid:15315763
  3. 3. Chiang C, Litingtung Y, Harris MP, Simandl BK, Li Y, Beachy PA, et al. Manifestation of the limb prepattern: limb development in the absence of sonic hedgehog function. Developmental biology. 2001;236(2):421–435. pmid:11476582
  4. 4. Ahn S, Joyner AL. Dynamic changes in the response of cells to positive hedgehog signaling during mouse limb patterning. Cell. 2004;118(4):505–516. Epub 2004/08/19. pmid:15315762
  5. 5. Zeller R, Lopez-Rios J, Zuniga A. Vertebrate limb bud development: moving towards integrative analysis of organogenesis. Nature reviews Genetics. 2009;10(12):845–858. pmid:19920852
  6. 6. Hui CC, Angers S. Gli proteins in development and disease. Annual review of cell and developmental biology. 2011;27:513–537. pmid:21801010
  7. 7. Chen Y, Struhl G. Dual roles for patched in sequestering and transducing Hedgehog. Cell. 1996;87(3):553–563. Epub 1996/11/01. pmid:8898207
  8. 8. Briscoe J, Chen Y, Jessell TM, Struhl G. A hedgehog-insensitive form of patched provides evidence for direct long-range morphogen activity of sonic hedgehog in the neural tube. Molecular cell. 2001;7(6):1279–1291. pmid:11430830
  9. 9. Butterfield NC, Metzis V, McGlinn E, Bruce SJ, Wainwright BJ, Wicking C. Patched 1 is a crucial determinant of asymmetry and digit number in the vertebrate limb. Development. 2009;136(20):3515–3524. pmid:19783740
  10. 10. Zhulyn O, Li D, Deimling S, Vakili NA, Mo R, Puviindran V, et al. A switch from low to high Shh activity regulates establishment of limb progenitors and signaling centers. Developmental cell. 2014;29(2):241–249. pmid:24726283
  11. 11. Bai CB, Stephen D, Joyner AL. All mouse ventral spinal cord patterning by hedgehog is Gli dependent and involves an activator function of Gli3. Developmental cell. 2004;6(1):103–115. pmid:14723851
  12. 12. Park HL, Bai C, Platt KA, Matise MP, Beeghly A, Hui CC, et al. Mouse Gli1 mutants are viable but have defects in SHH signaling in combination with a Gli2 mutation. Development. 2000;127(8):1593–1605. pmid:10725236
  13. 13. Dai P, Akimaru H, Tanaka Y, Maekawa T, Nakafuku M, Ishii S. Sonic Hedgehog-induced activation of the Gli1 promoter is mediated by GLI3. The Journal of biological chemistry. 1999;274(12):8143–8152. Epub 1999/03/13. pmid:10075717
  14. 14. Wang B, Fallon JF, Beachy PA. Hedgehog-regulated processing of Gli3 produces an anterior/posterior repressor gradient in the developing vertebrate limb. Cell. 2000;100(4):423–434. pmid:10693759
  15. 15. Pan Y, Bai CB, Joyner AL, Wang B. Sonic hedgehog signaling regulates Gli2 transcriptional activity by suppressing its processing and degradation. Molecular and cellular biology. 2006;26(9):3365–3377. Epub 2006/04/14. pmid:16611981
  16. 16. te Welscher P, Zuniga A, Kuijper S, Drenth T, Goedemans HJ, Meijlink F, et al. Progression of vertebrate limb development through SHH-mediated counteraction of GLI3. Science. 2002;298(5594):827–830. pmid:12215652
  17. 17. Litingtung Y, Dahn RD, Li Y, Fallon JF, Chiang C. Shh and Gli3 are dispensable for limb skeleton formation but regulate digit number and identity. Nature. 2002;418(6901):979–983. pmid:12198547
  18. 18. Bowers M, Eng L, Lao Z, Turnbull RK, Bao X, Riedel E, et al. Limb anterior-posterior polarity integrates activator and repressor functions of GLI2 as well as GLI3. Developmental biology. 2012;370(1):110–124. pmid:22841643
  19. 19. Galli A, Robay D, Osterwalder M, Bao X, Benazet JD, Tariq M, et al. Distinct roles of Hand2 in initiating polarity and posterior Shh expression during the onset of mouse limb bud development. PLoS genetics. 2010;6(4):e1000901. pmid:20386744
  20. 20. Osterwalder M, Speziale D, Shoukry M, Mohan R, Ivanek R, Kohler M, et al. HAND2 targets define a network of transcriptional regulators that compartmentalize the early limb bud mesenchyme. Developmental cell. 2014;31(3):345–357. pmid:25453830
  21. 21. te Welscher P, Fernandez-Teran M, Ros MA, Zeller R. Mutual genetic antagonism involving GLI3 and dHAND prepatterns the vertebrate limb bud mesenchyme prior to SHH signaling. Genes & development. 2002;16(4):421–426.
  22. 22. Lopez-Rios J, Speziale D, Robay D, Scotti M, Osterwalder M, Nusspaumer G, et al. GLI3 constrains digit number by controlling both progenitor proliferation and BMP-dependent exit to chondrogenesis. Developmental cell. 2012;22(4):837–848. pmid:22465667
  23. 23. Vokes SA, Ji H, Wong WH, McMahon AP. A genome-scale analysis of the cis-regulatory circuitry underlying sonic hedgehog-mediated patterning of the mammalian limb. Genes & development. 2008;22(19):2651–2663.
  24. 24. Wang W, Cote J, Xue Y, Zhou S, Khavari PA, Biggar SR, et al. Purification and biochemical heterogeneity of the mammalian SWI-SNF complex. The EMBO journal. 1996;15(19):5370–5382. pmid:8895581
  25. 25. Phelan ML, Sif S, Narlikar GJ, Kingston RE. Reconstitution of a core chromatin remodeling complex from SWI/SNF subunits. Molecular cell. 1999;3(2):247–253. pmid:10078207
  26. 26. de la Serna IL, Ohkawa Y, Imbalzano AN. Chromatin remodelling in mammalian differentiation: lessons from ATP-dependent remodellers. Nature reviews Genetics. 2006;7(6):461–473. pmid:16708073
  27. 27. Sohn DH, Lee KY, Lee C, Oh J, Chung H, Jeon SH, et al. SRG3 interacts directly with the major components of the SWI/SNF chromatin remodeling complex and protects them from proteasomal degradation. The Journal of biological chemistry. 2007;282(14):10614–10624. pmid:17255092.
  28. 28. Choi J, Ko M, Jeon S, Jeon Y, Park K, Lee C, et al. The SWI/SNF-like BAF complex is essential for early B cell development. Journal of immunology. 2012;188(8):3791–3803.
  29. 29. Logan M, Martin JF, Nagy A, Lobe C, Olson EN, Tabin CJ. Expression of Cre Recombinase in the developing mouse limb bud driven by a Prxl enhancer. Genesis. 2002;33(2):77–80. pmid:12112875
  30. 30. Benazet JD, Bischofberger M, Tiecke E, Goncalves A, Martin JF, Zuniga A, et al. A self-regulatory system of interlinked signaling feedback loops controls mouse limb patterning. Science. 2009;323(5917):1050–1053. pmid:19229034
  31. 31. Li D, Sakuma R, Vakili NA, Mo R, Puviindran V, Deimling S, et al. Formation of proximal and anterior limb skeleton requires early function of Irx3 and Irx5 and is negatively regulated by Shh signaling. Developmental cell. 2014;29(2):233–240. pmid:24726282
  32. 32. Lettice LA, Heaney SJ, Purdie LA, Li L, de Beer P, Oostra BA, et al. A long-range Shh enhancer regulates expression in the developing limb and fin and is associated with preaxial polydactyly. Human molecular genetics. 2003;12(14):1725–1735. pmid:12837695
  33. 33. Verheyden JM, Sun X. An Fgf/Gremlin inhibitory feedback loop triggers termination of limb bud outgrowth. Nature. 2008;454(7204):638–641. pmid:18594511
  34. 34. Chen JK, Taipale J, Cooper MK, Beachy PA. Inhibition of Hedgehog signaling by direct binding of cyclopamine to Smoothened. Genes & development. 2002;16(21):2743–2748. Epub 2002/11/05.
  35. 35. Lopez-Rios J, Duchesne A, Speziale D, Andrey G, Peterson KA, Germann P, et al. Attenuated sensing of SHH by Ptch1 underlies evolution of bovine limbs. Nature. 2014;511(7507):46–51. pmid:24990743
  36. 36. Li Q, Wang HY, Chepelev I, Zhu Q, Wei G, Zhao K, et al. Stage-dependent and locus-specific role of histone demethylase Jumonji D3 (JMJD3) in the embryonic stages of lung development. PLoS genetics. 2014;10(7):e1004524. pmid:25079229
  37. 37. Lee S, Lee JW, Lee SK. UTX, a histone H3-lysine 27 demethylase, acts as a critical switch to activate the cardiac developmental program. Developmental cell. 2012;22(1):25–37. pmid:22192413
  38. 38. Ho L, Miller EL, Ronan JL, Ho WQ, Jothi R, Crabtree GR. esBAF facilitates pluripotency by conditioning the genome for LIF/STAT3 signalling and by regulating polycomb function. Nature cell biology. 2011;13(8):903–913. pmid:21785422
  39. 39. Shi X, Zhang Z, Zhan X, Cao M, Satoh T, Akira S, et al. An epigenetic switch induced by Shh signalling regulates gene activation during development and medulloblastoma growth. Nat Commun. 2014;5:5425. pmid:25370275
  40. 40. Bilodeau S, Kagey MH, Frampton GM, Rahl PB, Young RA. SetDB1 contributes to repression of genes encoding developmental regulators and maintenance of ES cell state. Genes & development. 2009;23(21):2484–2489.
  41. 41. Zhang Z, Sui P, Dong A, Hassell J, Cserjesi P, Chen YT, et al. Preaxial polydactyly: interactions among ETV, TWIST1 and HAND2 control anterior-posterior patterning of the limb. Development. 2010;137(20):3417–3426. pmid:20826535
  42. 42. Karamboulas K, Dranse HJ, Underhill TM. Regulation of BMP-dependent chondrogenesis in early limb mesenchyme by TGFbeta signals. Journal of cell science. 2010;123(Pt 12):2068–2076. pmid:20501701.
  43. 43. Bandyopadhyay A, Tsuji K, Cox K, Harfe BD, Rosen V, Tabin CJ. Genetic analysis of the roles of BMP2, BMP4, and BMP7 in limb patterning and skeletogenesis. PLoS genetics. 2006;2(12):e216. pmid:17194222
  44. 44. Akiyama H, Kim JE, Nakashima K, Balmes G, Iwai N, Deng JM, et al. Osteo-chondroprogenitor cells are derived from Sox9 expressing precursors. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(41):14665–14670. pmid:16203988
  45. 45. Wellik DM, Capecchi MR. Hox10 and Hox11 genes are required to globally pattern the mammalian skeleton. Science. 2003;301(5631):363–367. pmid:12869760
  46. 46. Fromental-Ramain C, Warot X, Lakkaraju S, Favier B, Haack H, Birling C, et al. Specific and redundant functions of the paralogous Hoxa-9 and Hoxd-9 genes in forelimb and axial skeleton patterning. Development. 1996;122(2):461–472. pmid:8625797
  47. 47. Lu P, Yu Y, Perdue Y, Werb Z. The apical ectodermal ridge is a timer for generating distal limb progenitors. Development. 2008;135(8):1395–1405. pmid:18359901
  48. 48. Wyngaarden LA, Delgado-Olguin P, Su IH, Bruneau BG, Hopyan S. Ezh2 regulates anteroposterior axis specification and proximodistal axis elongation in the developing limb. Development. 2011;138(17):3759–3767. pmid:21795281
  49. 49. Jagani Z, Mora-Blanco EL, Sansam CG, McKenna ES, Wilson B, Chen D, et al. Loss of the tumor suppressor Snf5 leads to aberrant activation of the Hedgehog-Gli pathway. Nature medicine. 2010;16(12):1429–1433. pmid:21076395
  50. 50. Zhan X, Shi X, Zhang Z, Chen Y, Wu JI. Dual role of Brg chromatin remodeling factor in Sonic hedgehog signaling during neural development. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(31):12758–12763. pmid:21768360
  51. 51. Canettieri G, Di Marcotullio L, Greco A, Coni S, Antonucci L, Infante P, et al. Histone deacetylase and Cullin3-REN(KCTD11) ubiquitin ligase interplay regulates Hedgehog signalling through Gli acetylation. Nature cell biology. 2010;12(2):132–142. pmid:20081843
  52. 52. Cooper KL, Sears KE, Uygur A, Maier J, Baczkowski KS, Brosnahan M, et al. Patterning and post-patterning modes of evolutionary digit loss in mammals. Nature. 2014;511(7507):41–45. pmid:24990742
  53. 53. Zakany J, Kmita M, Duboule D. A dual role for Hox genes in limb anterior-posterior asymmetry. Science. 2004;304(5677):1669–1672. Epub 2004/06/12. pmid:15192229
  54. 54. Buscher D, Bosse B, Heymer J, Ruther U. Evidence for genetic control of Sonic hedgehog by Gli3 in mouse limb development. Mechanisms of development. 1997;62(2):175–182. pmid:9152009
  55. 55. Charite J, McFadden DG, Olson EN. The bHLH transcription factor dHAND controls Sonic hedgehog expression and establishment of the zone of polarizing activity during limb development. Development. 2000;127(11):2461–2470. pmid:10804186
  56. 56. Bialek P, Kern B, Yang X, Schrock M, Sosic D, Hong N, et al. A twist code determines the onset of osteoblast differentiation. Developmental cell. 2004;6(3):423–435. PubMed pmid:15030764
  57. 57. Cakouros D, Isenmann S, Cooper L, Zannettino A, Anderson P, Glackin C, et al. Twist-1 induces Ezh2 recruitment regulating histone methylation along the Ink4A/Arf locus in mesenchymal stem cells. Molecular and cellular biology. 2012;32(8):1433–1441. pmid:22290439
  58. 58. Yang MH, Hsu DS, Wang HW, Wang HJ, Lan HY, Yang WH, et al. Bmi1 is essential in Twist1-induced epithelial-mesenchymal transition. Nature cell biology. 2010;12(10):982–992. pmid:20818389
  59. 59. Panman L, Galli A, Lagarde N, Michos O, Soete G, Zuniga A, et al. Differential regulation of gene expression in the digit forming area of the mouse limb bud by SHH and gremlin 1/FGF-mediated epithelial-mesenchymal signalling. Development. 2006;133(17):3419–3428. pmid:16908629
  60. 60. Li Q, Lewandowski JP, Powell MB, Norrie JL, Cho SH, Vokes SA. A Gli silencer is required for robust repression of gremlin in the vertebrate limb bud. Development. 2014;141(9):1906–1914. pmid:24700818
  61. 61. Piette D, Hendrickx M, Willems E, Kemp CR, Leyns L. An optimized procedure for whole-mount in situ hybridization on mouse embryos and embryoid bodies. Nature protocols. 2008;3(7):1194–1201. pmid:18600225
  62. 62. Hebert JM, Basilico C, Goldfarb M, Haub O, Martin GR. Isolation of cDNAs encoding four mouse FGF family members and characterization of their expression patterns during embryogenesis. Developmental biology. 1990;138(2):454–463. pmid:2318343
  63. 63. Crossley PH, Martin GR. The mouse Fgf8 gene encodes a family of polypeptides and is expressed in regions that direct outgrowth and patterning in the developing embryo. Development. 1995;121(2):439–451. pmid:7768185