The terminal differentiation of hypertrophic chondrocytes is a tightly regulated process that plays a pivotal role in endochondral ossification. As a negative regulator, Sox9 is essentially downregulated in terminally differentiated hypertrophic chondrocytes. However, the underlying mechanism of Sox9 silencing is undefined. Here we show that the zinc finger protein Zbtb20 regulates the terminal differentiation of hypertrophic chondrocytes by repressing Sox9. In the developing skeleton of the mouse, Zbtb20 protein is highly expressed by hypertrophic chondrocytes from late embryonic stages. To determine its physiological role in endochondral ossification, we have generated chondrocyte-specific Zbtb20 knockout mice and demonstrate that disruption of Zbtb20 in chondrocytes results in delayed endochondral ossification and postnatal growth retardation. Zbtb20 deficiency caused a delay in cartilage vascularization and an expansion of the hypertrophic zone owing to reduced expression of Vegfa in the hypertrophic zone. Interestingly, Sox9, a direct suppressor of Vegfa expression, was ectopically upregulated at both mRNA and protein levels in the late Zbtb20-deficient hypertrophic zone. Furthermore, knockdown of Sox9 greatly increased Vegfa expression in Zbtb20-deficient hypertrophic chondrocytes. Our findings point to Zbtb20 as a crucial regulator governing the terminal differentiation of hypertrophic chondrocytes at least partially through repression of Sox9.

The terminal differentiation of hypertrophic chondrocytes is essential for endochondral ossification. During endochondral bone development, chondrocytes proliferate and then differentiate into hypertrophic chondrocytes. Hypertrophic chondrocytes express specific extracellular matrix molecules, such as collagen type X (Col10a1) (Elima et al., 1993; Linsenmayer et al., 1991), and normally undergo a further maturation process. Terminal hypertrophic chondrocytes at the chondro-osseous junction actively express additional molecules, such as matrix metalloproteinase 13 (Mmp13) (Stickens et al., 2004), osteopontin (Opn; also known as Spp1) (Chen et al., 1993) and vascular endothelial growth factor (Vegfa) (Gerber et al., 1999), which are crucial for the invasion of blood vessels, osteoclasts and osteoblast precursors from the perichondrium. Defects in the terminal differentiation of hypertrophic chondrocytes result in severely delayed formation of the primary ossification center (POC) and secondary ossification center (SOC) (Hattori et al., 2010).

Hypertrophic conversion of proliferating chondrocytes during development of the POC is regulated by Indian hedgehog (Ihh) and parathyroid hormone related peptide (PTHrP; also known as Pthlh) (Vortkamp et al., 1996) and several transcription factors including Runx2 and Runx3 (Takeda et al., 2001; Yoshida et al., 2004; Zheng et al., 2003) and Mef2c (Arnold et al., 2007). However, the molecular mechanisms regulating the late and terminal differentiation of hypertrophic chondrocytes are still poorly understood.

Sox9 is a multifunctional factor that regulates multiple processes in endochondral ossification. It plays an essential role in early chondrogenesis (Akiyama et al., 2002), promotes the hypertrophy of prehypertrophic chondrocytes (Dy et al., 2012) and inhibits the subsequent terminal differentiation and Vegfa expression of hypertrophic chondrocytes (Hattori et al., 2010; Ikegami et al., 2011; Kim et al., 2011). Sox9 mRNA is highly expressed in chondrocytes of the proliferating and prehypertrophic zones but declines abruptly in the hypertrophic zone. Downregulation of Sox9 in the hypertrophic zone of the normal growth plate is essential to allow vascular invasion, bone marrow formation and endochondral ossification. However, to our knowledge, the mechanism underlying its downregulation in hypertrophic chondrocytes remains largely unknown.

Zinc finger and BTB domain-containing protein 20 (Zbtb20, also known as DPZF, Hof and Zfp288) is a member of a subfamily of zinc finger proteins containing C2H2 Krüppel-type zinc fingers and BTB/POZ domains (Mitchelmore et al., 2002; Zhang et al., 2001). We and others have reported that Zbtb20 functions primarily as a transcriptional repressor and plays an essential role in the specification of pyramidal neurons in the developing hippocampus (Nielsen et al., 2007; Xie et al., 2010,, 2008; Zhang et al., 2012). Zbtb20 null mice exhibit severe postnatal growth retardation, metabolic dysfunction and lethality, suggesting that Zbtb20 plays nonredundant roles in multiple organ systems (Sutherland et al., 2009). Furthermore, conditional gene targeting demonstrates that Zbtb20 regulates insulin secretion in pancreatic β cells (Zhang et al., 2012), promotes Toll-like receptor-mediated innate immune response in macrophages (Liu et al., 2013), and modulates pain sensation in nociceptive sensory neurons (Ren et al., 2014).

The growth retardation of Zbtb20 null mice prompted us to investigate its potential role in skeletal development. Here, we demonstrate previously undescribed expression of Zbtb20 in developing cartilage. Conditional deletion of Zbtb20 in developing cartilage resulted in delayed endochondral ossification. Molecular analysis showed that Sox9 mRNA and protein are ectopically upregulated in the Zbtb20-deleted late hypertrophic zone, which impeded the terminal differentiation of hypertrophic chondrocytes, subsequent growth plate vascularization and endochondral ossification. Thus, our findings suggest that Zbtb20 regulates the terminal differentiation of hypertrophic chondrocytes at least partially through repressing Sox9.

Expression of Zbtb20 in the developing mouse skeleton

To investigate the potential role of Zbtb20 in skeletal development, we first characterized its expression pattern in the developing cartilage by immunostaining. Before embryonic day (E) 13.5, Zbtb20 protein was undetectable in the cartilage anlage of the long bones (supplementary material Fig. S1A). It was initially detected as early as E14.5, when the chondrocytes in the center of the cartilage anlage start to undergo hypertrophic differentiation (Fig. 1A). Zbtb20 protein was specifically expressed in the nuclei of hypertrophic chondrocytes (characterized by an increase in cell size and by expression of Col10a1), but was undetectable in the reserve and proliferating columnar chondrocytes as well as in Co1la1-positive osteoblasts in the perichondrium at embryonic stages. After birth, the reserve zone began to show expression of Zbtb20 protein (supplementary material Fig. S1B). The staining intensity was very weak at birth [postnatal day (P) 0.5] but rapidly increased thereafter. At P4, Zbtb20 protein was expressed at high levels in both the reserve and hypertrophic zone, and to a much lesser extent in the proliferating zone (Fig. 1B). This expression pattern of Zbtb20 in the growth plate was maintained at 3 weeks and 3 months of age (Fig. 1C; supplementary material Fig. S1C). These data suggest that Zbtb20 might play an important role in bone formation.

Fig. 1.

Generation of chondrocyte-specific Zbtb20 knockout mice. (A) Zbtb20 protein expression in hypertrophic chondrocytes in the normal femur at E14.5. Arrows indicate Zbtb20+ cells (red). Col10a1 and Col1a1 mRNA expression, as shown by in situ hybridization, indicates the differentiation of hypertrophic chondrocytes and osteoblasts at E14.5, respectively. (B,C) Zbtb20 protein was differentially expressed by growth plate chondrocytes in normal tibia at P4 (B) and P21 (C). Arrows indicate Zbtb20+ cells. Zbtb20 was highly expressed in the reserve and hypertrophic zones, and to a much lesser extent in the proliferating zone. (D,E) Specific disruption of Zbtb20 in chondrocytes from Col-ZB20KO mice at E15.5 (D) and P6 (E). Absence of Zbtb20 staining is indicated by arrowheads in the hypertrophic zone (D,E) and reserve zone (E) in the mutant tibia. HZ, hypertrophic zone; PZ, proliferating zone; RZ, reserve zone. Scale bars: 100 µm.

Fig. 1.

Generation of chondrocyte-specific Zbtb20 knockout mice. (A) Zbtb20 protein expression in hypertrophic chondrocytes in the normal femur at E14.5. Arrows indicate Zbtb20+ cells (red). Col10a1 and Col1a1 mRNA expression, as shown by in situ hybridization, indicates the differentiation of hypertrophic chondrocytes and osteoblasts at E14.5, respectively. (B,C) Zbtb20 protein was differentially expressed by growth plate chondrocytes in normal tibia at P4 (B) and P21 (C). Arrows indicate Zbtb20+ cells. Zbtb20 was highly expressed in the reserve and hypertrophic zones, and to a much lesser extent in the proliferating zone. (D,E) Specific disruption of Zbtb20 in chondrocytes from Col-ZB20KO mice at E15.5 (D) and P6 (E). Absence of Zbtb20 staining is indicated by arrowheads in the hypertrophic zone (D,E) and reserve zone (E) in the mutant tibia. HZ, hypertrophic zone; PZ, proliferating zone; RZ, reserve zone. Scale bars: 100 µm.

Chondrocyte-specific deletion of Zbtb20 results in delayed endochondral ossification and in postnatal growth retardation

To explore the role of Zbtb20 in skeletal development in vivo, we deleted the gene specifically in the cartilaginous template of endochondral bones by mating mice with a floxed Zbtb20 allele (Xie et al., 2008) with Col2-Cre transgenic mice, a line that was reported to mediate loxP recombination specifically in cartilaginous structures at E13.5 (Yang et al., 2008; Zhang et al., 2005), when Zbtb20 protein expression is not detectable in chondrocytes. Immunohistochemical analysis showed that Zbtb20 protein is undetectable in chondrocytes of Zbtb20 chondrocyte-specific knockout embryos and postnatal mice (Zbtb20flox/flox;Col2-Cre, hereafter referred to as Col-ZB20KO) (Fig. 1D,E), confirming the efficient disruption of the Zbtb20 gene in chondrocytes.

Born at the expected Mendelian ratios, Col-ZB20KO mice did not differ in size or weight from their littermate controls at birth (supplementary material Fig. S2A,B), with normal patterning of hindlimbs and forelimbs by visual inspection. From P14 onwards, Col-ZB20KO mice displayed significantly shortened tails, although they still had similar body weights to wild-type littermates (Fig. 2A,B; supplementary material Fig. S2B). From P21 onwards, the mutants showed a slight but significant decrease in the length of long bones (Fig. 2C,D). There was no significant difference between female and male mice in phenotype. Heterozygous mice were morphologically indistinguishable from wild-type littermates in the appearance and length of long bones (supplementary material Fig. S2C).

Fig. 2.

Delayed endochondral ossification and growth retardation caused by conditional deletion of Zbtb20 in chondrocytes. (A) Representative skeletal preparations from control and mutant mice at P21. Mutant mice had shortened tails (arrowhead). (B) Reduced tail lengths of mutant mice at the indicated ages (n=6 pairs). (C) Representative images of femur and tibia from control and mutant mice at P21. Both femur and tibia were shorter in mutants. (D) Reduced femur lengths in mutant mice at the indicated ages (n=6 pairs). (E) Ossification defects in mutant mice demonstrated by whole-mount Alcian Blue (cartilage) and Alizarin Red (bone) staining. At P0.5, the ossification areas were absent or reduced in the mutant cervical vertebra, tail and hindlimbs, as indicated by arrowheads. C1 and C2 indicate cervical vertebrae. (F) At P14 and P21, the formation of the tibial secondary ossification center (SOC) was delayed in the mutant (arrowheads). **P<0.01; *P<0.05. Scale bars: 10 mm in A; 5 mm in C; 1 mm in E,F.

Fig. 2.

Delayed endochondral ossification and growth retardation caused by conditional deletion of Zbtb20 in chondrocytes. (A) Representative skeletal preparations from control and mutant mice at P21. Mutant mice had shortened tails (arrowhead). (B) Reduced tail lengths of mutant mice at the indicated ages (n=6 pairs). (C) Representative images of femur and tibia from control and mutant mice at P21. Both femur and tibia were shorter in mutants. (D) Reduced femur lengths in mutant mice at the indicated ages (n=6 pairs). (E) Ossification defects in mutant mice demonstrated by whole-mount Alcian Blue (cartilage) and Alizarin Red (bone) staining. At P0.5, the ossification areas were absent or reduced in the mutant cervical vertebra, tail and hindlimbs, as indicated by arrowheads. C1 and C2 indicate cervical vertebrae. (F) At P14 and P21, the formation of the tibial secondary ossification center (SOC) was delayed in the mutant (arrowheads). **P<0.01; *P<0.05. Scale bars: 10 mm in A; 5 mm in C; 1 mm in E,F.

We then performed whole-mount Alcian Blue/Alizarin Red staining to examine skeletal development in detail. The mutant newborns revealed a remarkable defect in the ossification of several cartilage-based structures. In the axial skeleton, the defects in endochondral ossification were evident in the mutant vertebral column and sternum (Fig. 2E; supplementary material Fig. S2D). In the appendicular skeleton at the level of the hindlimbs, Col-ZB20KO newborns showed an impairment or absence of endochondral ossification centers in the calcaneus, the talus and middle phalanges (Fig. 2E). The ossification of calvaria, clavicle and mandible, however, was indistinguishable between mutant and wild-type control mice, suggesting that intramembranous ossification is unaffected in Col-ZB20KO mice (supplementary material Fig. S2E-G). At P14 and P21, the impairment of endochondral ossification was still apparent in the mutants and additionally manifested by a severe delay in the formation of the SOC (Fig. 2F; supplementary material Fig. S2H).

Delayed vascular invasion and expanded hypertrophic zone in Col-ZB20KO embryos and postnatal mice

To investigate the mechanism underlying the delayed endochondral ossification in Col-ZB20KO mice, we first performed a histological examination of the growth plates at different stages. At E15.5, the central portion of the femur is composed of hypertrophic chondrocytes, with proliferative chondrocytes in each epiphyseal region. H&E staining showed that the hypertrophic domains in the wild-type and Col-ZB20KO femurs were similar, which was confirmed by the similar expression domains of Col10a1 and Opn (a marker for mature hypertrophic chondrocytes and osteoblasts) (McKee et al., 1992) (supplementary material Fig. S3A,B). This observation indicates normal timing in the initiation of chondrocyte hypertrophy during the formation of the POC. At E16, however, the mutants exhibited delayed vascular invasion in tibia [demonstrated by immunostaining for the endothelial marker CD31 (Pecam1)] (Cao et al., 2009), resulting in a delay in the formation of the POC when compared with wild-type littermates (Fig. 3A). By E17.5, the POCs in both control and mutant tibia had developed but the Col-ZB20KO tibia contained an expanded hypertrophic zone with a reduced domain of vascularization (Fig. 3B). At P2, the mutant hypertrophic zone was 74% longer than that in control mice, whereas the reserve and proliferating zones were indistinguishable between the two groups (Fig. 3C,D). This abnormality persisted, but to a lesser extent, during the growth of the bones (supplementary material Fig. S3C).

Fig. 3.

Zbtb20 deletion leads to delayed vascular invasion and an expanded hypertrophic zone. (A) Immunostaining of the endothelial marker CD31 (red) showing impaired vascular invasion in the mutant tibia at E16. Nuclei are counterstained with DAPI (blue). Star indicates the lack of CD31 staining in the center of mutant tibia. (B) Mutant tibias contained an expanded hypertrophic zone (arrowheads) with a reduced domain of vascularization at E17.5. (C) Representative images of H&E-stained tibia sections from control and mutant mice at P2. Mutant tibias had a lengthened hypertrophic zone (arrowhead). (D) The increased hypertrophic zone length in P2 mutant mice. n=5 pairs; **P<0.01. Scale bars: 100 µm in A; 500 µm in B; 200 µm in C.

Fig. 3.

Zbtb20 deletion leads to delayed vascular invasion and an expanded hypertrophic zone. (A) Immunostaining of the endothelial marker CD31 (red) showing impaired vascular invasion in the mutant tibia at E16. Nuclei are counterstained with DAPI (blue). Star indicates the lack of CD31 staining in the center of mutant tibia. (B) Mutant tibias contained an expanded hypertrophic zone (arrowheads) with a reduced domain of vascularization at E17.5. (C) Representative images of H&E-stained tibia sections from control and mutant mice at P2. Mutant tibias had a lengthened hypertrophic zone (arrowhead). (D) The increased hypertrophic zone length in P2 mutant mice. n=5 pairs; **P<0.01. Scale bars: 100 µm in A; 500 µm in B; 200 µm in C.

These findings suggest that the hypertrophic conversion of columnar chondrocytes remains largely normal in Zbtb20 mutants during the development of the POC, but subsequent vascular invasion and ossification are significantly delayed.

Decreased columnar chondrocyte proliferation in Col-ZB20KO mice

To determine whether the expansion of the hypertrophic zone was associated with increased chondrocyte proliferation in the Col-ZB20KO growth plate, we evaluated the proliferation rate of chondrocytes in vivo by a 2-h BrdU labeling approach. At P2, BrdU incorporation was comparable between Col-ZB20KO and control mice in both the reserve and proliferating zones (Fig. 4A,B). At P22, however, the mutant mice showed a marked decrease in BrdU incorporation in chondrocytes in the proliferating zone compared with control mice (Fig. 4A,B). The decreased proliferation rate of the mutant columnar chondrocytes was consistent with the observation of decreased long bone length in the mutant mice at P21.

Fig. 4.

Decreased proliferation rates in Col-ZB20KO columnar chondrocytes at P22. (A) Representative images of BrdU-stained tibia at the indicated ages. BrdU+ cells in the boxed areas were counted. For P22, boxed areas are also magnified in insets. Scale bars: 100 µm. (B) Percentage of BrdU+ cells within boxed areas in A. Mutant mice showed a marked decrease in BrdU+ chondrocytes in the proliferating zone at P22 but not at P2. **P<0.01; n=5 pairs.

Fig. 4.

Decreased proliferation rates in Col-ZB20KO columnar chondrocytes at P22. (A) Representative images of BrdU-stained tibia at the indicated ages. BrdU+ cells in the boxed areas were counted. For P22, boxed areas are also magnified in insets. Scale bars: 100 µm. (B) Percentage of BrdU+ cells within boxed areas in A. Mutant mice showed a marked decrease in BrdU+ chondrocytes in the proliferating zone at P22 but not at P2. **P<0.01; n=5 pairs.

To exclude the possibility that the expanded hypertrophic zone resulted from decreased cell death, we examined apoptosis in hypertrophic chondrocytes in the growth plate using the TUNEL assay. Very few apoptotic cells were observed at P22, and no difference was apparent between Col-ZB20KO and control mice (supplementary material Fig. S4). Together, these data suggest that the expanded hypertrophic zone in the mutant growth plate is not due to increased proliferation or decreased apoptosis of columnar chondrocytes. Therefore, we reason that the hypertrophic zone phenotype may be caused by the differentiation and/or ossification defect.

Impaired terminal differentiation of hypertrophic chondrocytes in Col-ZB20KO mice

To determine whether the impaired vascularization in the mutant hypertrophic zone is associated with the alteration of chondrocyte biology, we analyzed the differentiation state of hypertrophic chondrocytes. As mentioned above, hypertrophic chondrocytes normally undergo a further maturation process. The late differentiated hypertrophic chondrocytes in the lower hypertrophic zone are capable of producing a mineralized matrix (Kirsch et al., 1997). von Kossa staining revealed an expanded mineralized hypertrophic zone in the mutant (Fig. 5A).

Fig. 5.

Impaired terminal differentiation of chondrocytes in Col-ZB20KO mice. (A) von Kossa staining showing the expanded mineralization of the hypertrophic zone (mHZ) in the growth plate of mutant tibia at P2. (B-E) In situ hybridization showing mRNA expression of Col10a1 (B), Ihh (B), Mmp13 (C) and Vegfa (D,E) in tibia sections from control and mutant animals at the indicated ages. Note that in the mutant the expression domain of Col10a1 and Ihh was expanded and Vegfa expression was decreased in the HZ (arrowheads), whereas Mmp13 expression was comparable between the two groups. Scale bars: 200 µm. (F) Real-time RT-PCR analysis of Vegfa mRNA expression in P5 hypertrophic chondrocytes. The control Vegfa expression level was set at 1. **P<0.01; n=3 pairs. PreH, prehypertrophic zone.

Fig. 5.

Impaired terminal differentiation of chondrocytes in Col-ZB20KO mice. (A) von Kossa staining showing the expanded mineralization of the hypertrophic zone (mHZ) in the growth plate of mutant tibia at P2. (B-E) In situ hybridization showing mRNA expression of Col10a1 (B), Ihh (B), Mmp13 (C) and Vegfa (D,E) in tibia sections from control and mutant animals at the indicated ages. Note that in the mutant the expression domain of Col10a1 and Ihh was expanded and Vegfa expression was decreased in the HZ (arrowheads), whereas Mmp13 expression was comparable between the two groups. Scale bars: 200 µm. (F) Real-time RT-PCR analysis of Vegfa mRNA expression in P5 hypertrophic chondrocytes. The control Vegfa expression level was set at 1. **P<0.01; n=3 pairs. PreH, prehypertrophic zone.

We next examined the expression of a set of differentiation markers in the Col-ZB20KO mice. At P4, the expression domain of Col10a1 mRNA was expanded in accordance with a lengthened hypertrophic zone in the tibia (Fig. 5B). The prehypertrophic zone, as characterized by high expression of Ihh mRNA (Vortkamp et al., 1996), appeared normal in the mutant (Fig. 5B). The expression domains and intensities of Mmp13 and Opn, which are markers for terminally differentiated hypertrophic chondrocytes at the chondro-osseous border (the lowest region of the growth plate) (Chen et al., 1993; Mark et al., 1988; Stickens et al., 2004), were comparable between mutant and control (Fig. 5C; supplementary material Fig. S5A). We then analyzed angiogenesis-related factors. Interestingly, in situ hybridization analysis revealed a marked decrease in Vegfa mRNA expression in the mutant hypertrophic zone at E16 and P4, which was confirmed by quantitative RT-PCR analysis in mutant primary hypertrophic chondrocytes at P5 (Fig. 5D-F). However, expression levels of Lect1, which encodes an angiogenesis-inhibiting factor (Hiraki et al., 1997), were comparable between control and mutant mice (supplementary material Fig. S5A). The downregulation of Vegfa was compatible with delayed cartilage vascularization in the mutant long bone at E16. Thus, Zbtb20 deletion impairs the terminal differentiation of hypertrophic chondrocytes.

An expanded hypertrophic zone could also result from a defect in osteoblast and/or osteoclast function. Therefore, we examined the expression of osteoblast markers, including Opn for immature osteoblasts and osteocalcin (also known as Bglap) for terminally differentiated osteoblasts, in the primary spongiosum (Mark et al., 1988). The mutant showed normal expression domains and intensities for these genes, as indicative of normal osteoblast development (supplementary material Fig. S5A). Similarly, the expression domain of Mmp9, a marker for chondroclast/osteoclast (Vu et al., 1998), and the number of TRAP+ multinuclear osteoclasts at the chondro-osseous junction (10.7±4.2 versus 9.4±1.5 TRAP+ cells/section; P=0.6, n=5 pairs) were comparable between control and mutant (supplementary material Fig. S5A-C).

Together, these data demonstrated that the absence of Zbtb20 causes an impairment in the final steps of chondrocyte terminal maturation but has little or no impact on the development of osteoblasts or osteoclasts.

Ectopic dysregulated expression of Sox9 in the late hypertrophic zone in Col-ZB20KO mice

To elucidate the mechanism by which Zbtb20 regulates the terminal differentiation and Vegfa expression of hypertrophic chondrocytes, we examined the expression of some key regulators of chondrocyte maturation. The expression domains of Runx2 and Mef2c mRNA, which are positive regulators of chondrocyte maturation that are normally expressed in the prehypertrophic and hypertrophic zones (Arnold et al., 2007; Zheng et al., 2003), were broadened in the mutant, similar to that of Col10a1, whereas the staining intensities of these genes were indistinguishable between the mutant and control growth plates (supplementary material Fig. S6).

Sox9 is required for hypertrophy but negatively regulates terminal differentiation (Dy et al., 2012; Hattori et al., 2010). Consistent with a previous report (Hattori et al., 2010), in the control growth plate Sox9 mRNA was highly expressed in chondrocytes of the proliferating and prehypertrophic zones but downregulated abruptly in the hypertrophic zone. In the mutant, however, the expression domain extended to the whole hypertrophic zone at P4 (Fig. 6A). This observation was confirmed by immunohistochemistry showing that the expression of Sox9 protein expanded to the chondro-osseous border in the mutant, whereas it was excluded from the late hypertrophic zone in control mice (Fig. 6B). Ectopic Sox9 expression in the late hypertrophic zone was also observed in the mutant embryos at E15.5 (Fig. 6C). The expression domain of Sox5 protein, which is reported to coexpress and cooperate with Sox9 in chondrogenesis (Han and Lefebvre, 2008), remained unchanged in the mutant (Fig. 6D). Thus, Zbtb20 is required specifically for Sox9 downregulation in the hypertrophic chondrocytes. As Sox9 is a direct suppressor of Vegfa expression (Hattori et al., 2010), the abnormal downregulation of Vegfa and upregulation of Sox9 in Zbtb20-deleted hypertrophic zones supports the hypothesis that Zbtb20 inhibits Sox9 expression, thereby positively regulating Vefga expression and chondrocyte terminal maturation.

Fig. 6.

Ectopic upregulation of Sox9 expression in the late hypertrophic zone in Col-ZB20KO mice. (A) In situ hybridization showing the expression domain of Sox9 mRNA in control and mutant tibia at P4. In contrast to the control, Sox9 mRNA expression was expanded to the late hypertrophic zone in the mutant (arrowhead). (B-D) Immunostaining of Sox9 and Sox5 (red) in the hypertrophic zone of the tibia at P4 and E15.5. Dotted lines demarcate the prehypertrophic and hypertrophic zones. In the mutant, Sox9 protein expression was upregulated in the late hypertrophic zone (B,C), whereas Sox5 protein was unaffected (D). Nuclei were counterstained with DAPI (blue). Scale bars: 200 µm.

Fig. 6.

Ectopic upregulation of Sox9 expression in the late hypertrophic zone in Col-ZB20KO mice. (A) In situ hybridization showing the expression domain of Sox9 mRNA in control and mutant tibia at P4. In contrast to the control, Sox9 mRNA expression was expanded to the late hypertrophic zone in the mutant (arrowhead). (B-D) Immunostaining of Sox9 and Sox5 (red) in the hypertrophic zone of the tibia at P4 and E15.5. Dotted lines demarcate the prehypertrophic and hypertrophic zones. In the mutant, Sox9 protein expression was upregulated in the late hypertrophic zone (B,C), whereas Sox5 protein was unaffected (D). Nuclei were counterstained with DAPI (blue). Scale bars: 200 µm.

To further examine the functional involvement of Sox9 in the regulation of chondrocyte terminal maturation by Zbtb20, we next investigated the effects of siRNA-mediated knockdown of Sox9 in wild-type and Zbtb20-deleted hypertrophic chondrocytes. Primary hypertrophic chondrocytes isolated from both wild-type and Col-ZB20KO mice expressed high levels of Col10a1 and alkaline phosphatase (Alpl) mRNA (Miao and Scutt, 2002), two widely used markers for hypertrophic chondrocytes, and their hypertrophic characteristics were maintained at 48 h post-seeding (supplementary material Fig. S7). The wild-type hypertrophic chondrocytes transfected with Sox9 siRNA failed to show a significant increase in Vegfa mRNA levels, probably owing to their relatively low basal level of Sox9 expression (Fig. 7). By contrast, the mutant hypertrophic chondrocytes treated with Sox9 siRNA exhibited a dramatic increase in Vegfa mRNA levels (Fig. 7). Taken together, these findings indicate that Zbtb20 positively regulates chondrocyte terminal maturation at least partly via downregulation of Sox9 expression.

Fig. 7.

Knockdown of Sox9 restores Vegfa expression in Col-ZB20KO hypertrophic chondrocytes. Primary wild-type (WT) and mutant hypertrophic chondrocytes were transfected with control siRNA or Sox9 siRNA and Sox9 and Vegfa expression levels were analyzed after 48 h. (A) Western blot analysis showed that Sox9 protein levels are markedly reduced by Sox9 siRNA in wild-type or mutant hypertrophic chondrocytes. The blot is representative of three independent experiments, with semi-quantification shown to the right. (B) mRNA expression levels of Sox9 and Vegfa were determined by quantitative RT-PCR. Data are presented as fold increase relative to control siRNA-treated cells (set at 1) for each group. The mean value of three independent experiments is shown. **P<0.01, *P<0.05.

Fig. 7.

Knockdown of Sox9 restores Vegfa expression in Col-ZB20KO hypertrophic chondrocytes. Primary wild-type (WT) and mutant hypertrophic chondrocytes were transfected with control siRNA or Sox9 siRNA and Sox9 and Vegfa expression levels were analyzed after 48 h. (A) Western blot analysis showed that Sox9 protein levels are markedly reduced by Sox9 siRNA in wild-type or mutant hypertrophic chondrocytes. The blot is representative of three independent experiments, with semi-quantification shown to the right. (B) mRNA expression levels of Sox9 and Vegfa were determined by quantitative RT-PCR. Data are presented as fold increase relative to control siRNA-treated cells (set at 1) for each group. The mean value of three independent experiments is shown. **P<0.01, *P<0.05.

Indirect regulation of Sox9 expression by Zbtb20

To explore the molecular mechanisms underlying the regulation of Sox9 expression by Zbtb20, we first performed chromatin immunoprecipitation (ChIP) analysis to determine whether Zbtb20 directly binds the Sox9 promoter in vivo. Using ChIP grade anti-Zbtb20 antibodies (Xie et al., 2008), we did not detect any significant occupancy by Zbtb20 in the 1 kb region upstream of the Sox9 transcription start site in isolated mouse primary chondrocytes (supplementary material Fig. S8A-C), whereas the positive control with anti-acetyl-histone H3 antibody showed robust enrichment of acetyl-H3 in the Sox9 promoter. A reporter assay using a luciferase reporter construct (Sox9-Luc) harboring the mouse Sox9 promoter region (−2196 to +27) showed that overexpression of Zbtb20 in HEK293T cells had no significant effects on the transcriptional activity of the Sox9 promoter (supplementary material Fig. S8D). Taken together, these data suggest that Zbtb20 regulates Sox9 expression via an indirect, as yet unidentified, mechanism.

In this study, we examined the role of Zbtb20 in chondrocytes. Our findings provide compelling evidence that Zbtb20 is a novel essential regulator of cartilage development. First, Zbtb20 is expressed in developing chondrocytes, with expression most abundant in hypertrophic chondrocytes. Second, and most importantly, conditional deletion of Zbtb20 in developing cartilage results in delayed endochondral ossification and postnatal growth retardation, with a delay in cartilage vascularization and an expansion of the hypertrophic zone associated with reduced expression of Vegfa in the hypertrophic zone. Lastly, Sox9, a direct suppressor of Vegfa expression, is ectopically upregulated in the late hypertrophic zone by the loss of Zbtb20, which at least partially impairs the terminal differentiation of hypertrophic chondrocytes and subsequent growth plate vascularization.

The initiation of chondrocyte hypertrophy during the development of the POC is regulated by genes including Ihh, Runx2/3 and Mef2c. However, the most terminally differentiated hypertrophic chondrocytes actively express genes that allow the invasion of blood vessels, osteoclasts and osteoblast precursors from the perichondrium, suggesting additional regulatory mechanisms at this late stage. Sox9 has previously been reported to promote hypertrophy but to inhibit terminal differentiation, indicating that its downregulation is a necessary event to allow this process to occur (Dy et al., 2012; Hattori et al., 2010). Interestingly, loss of Zbtb20 resulted in the ectopic expression of Sox9 in the late hypertrophic zone, suggesting that Zbtb20 positively regulates terminal maturation, at least partially, by inhibiting Sox9 expression. Although this fits the notion that Zbtb20 functions primarily as a transcriptional repressor (Liu et al., 2013; Xie et al., 2008; Zhang et al., 2012), ChIP and luciferase reporter assays did not support direct binding of Zbtb20 to the Sox9 promoter to regulate transcriptional activity. One possibility is that Zbtb20 regulates Sox9 transcription via a cis-acting element that is located outside of the sequence that we analyzed, such as a distal enhancer. Alternatively, Zbtb20 might regulate Sox9 expression via an indirect mechanism. These possibilities are currently under further investigation.

However, Col-ZB20KO mice showed less severe retardation in vascular invasion, when compared with transgenic mice misexpressing Sox9 in hypertrophic chondrocytes under the control of a BAC Col10a1 promoter (Hattori et al., 2010). Indeed, the expression of Vegfa was completely inhibited in the Sox9-overexpressing hypertrophic zones but only downregulated in corresponding Zbtb20-deleted areas. Similarly, expression domains of Mmp13 and Opn were decreased in the Sox9 transgenic hypertrophic zones but appeared unchanged in corresponding Col-ZB20KO zones. These discrepancies could result from differences in Sox9 expression levels and domain between the two animal models. In Zbtb20-deleted hypertrophic zones, Sox9 protein levels were increased but were similar to those in the proliferating zones, as shown by comparable immunostaining intensity in these two regions in the same section (Fig. 6B). In the Sox9 overexpression model, however, the Sox9 immunostaining intensity was stronger than that in the proliferating zone in the same section, suggesting a much higher dosage of Sox9 in the hypertrophic zones. Furthermore, in Col-ZB20KO mice upregulation of Sox9 was restricted to the late hypertrophic zones, whereas in the transgenic mice Sox9 was highly overexpressed by all chondrocytes expressing Col10a1, which spans from the prehypertrophic to the terminal hypertrophic zones; indeed, the prehypertrophic zone, as characterized by strong expression of Ihh and Runx2, was extended and hypertrophic chondrocytes were retained in a prehypertrophic/early hypertrophic state in the Sox9 transgenic cartilage (Hattori et al., 2010). In addition, the other reported hypertrophic chondrocyte-specific Sox9 overexpression mouse model, which was constructed by mating CAG-mRFP1floxed-Sox9-EGFP transgenic mice and Col10a1-Cre mice, exhibited delayed chondrocyte terminal differentiation but did not show Mmp13 downregulation (Kim et al., 2011). These discrepancies between the different animal models indicate that some of the functions of Sox9 might be dosage dependent.

A microdeletion in human chromosome 3q13.31, which encompasses five RefSeq genes including ZBTB20, has recently been reported (Molin et al., 2012). Patients with this microdeletion share several major characteristics, including significant developmental delay. We thus speculate that ZBTB20 might play crucial roles in human skeletal development, similar to that in mice.

Generation of Zbtb20 conditional knockout mice

Mouse strains carrying floxed Zbtb20 were described previously (Xie et al., 2008). To achieve chondrocyte-specific deletion of floxed Zbtb20, mice were crossed with transgenic mice expressing Cre recombinase under the control of regulatory regions of the mouse Col2a1 gene (Zhang et al., 2005). All animal experimental procedures were preformed according to institutional guidelines of the Second Military Medical University, Shanghai, China. Sex-matched littermates carrying floxed Zbtb20 were used as controls.

Immunohistochemistry

Tissue was fixed with 4% paraformaldehyde (pH 7.4) and decalcified in 0.5 M EDTA from P5 onwards, then embedded in paraffin or OCT (Thermo Scientific, 22-110-617). Immunostaining for paraffin sections or cryosections (CD31) was performed using the protocol described previously (Xie et al., 2010). The following antibodies were used: Zbtb20 monoclonal antibody 9A10 (made in our lab, 1:1000) (Xie et al., 2008), CD31 (Abcam, ab28364, 1:2000), Sox5 and Sox9 (Santa Cruz, sc-20091, sc-20095; 1:2000), All secondary antibodies were from Vector Laboratories (PI-1000, PI-2000; 1:500).

In situ hybridization

Riboprobes labeled with digoxygenin-UTP were transcribed from cDNA clones (supplementary material Table S1). Tissue samples were fixed overnight in 4% paraformaldehyde (pH 9.0) and decalcified in 0.5 M EDTA from P5 onwards. In situ hybridization of cryosections was performed as previously described (Xie et al., 2010).

Skeletal analyses

Fetal and postnatal skeletons were freed from adherent tissue, fixed in 95% ethanol and stained for cartilage with Alcian Blue and counterstained for bone with Alizarin Red as described previously (Ovchinnikov, 2009).

Histological techniques

Tissue was fixed and decalcified as for immunohistochemistry. Paraffin sections (4 µm) were stained with Hematoxylin and Eosin (H&E), Alcian Blue or Safranin O as described (Schmitz et al., 2010). To measure growth plate length, five semi-serial sections (20 µm between each level) through the center of proximal tibia from each mouse were cut, stained with H&E, and photographed for measurement using Image-Pro Plus software (Media Cybernetics). For von Kossa staining, nondecalcified tissue was used. Sections were deparaffinized and immersed in 1% silver nitrate solution under ultraviolet light for 45 min. Slides were rinsed in distilled water and immersed in 3% sodium thiosulfate for 5 min. Slides were rinsed again in distilled water and counterstained with Nuclear Fast Red for 5 min. Osteoclast activity was detected by staining for tartrate-resistant acid phosphatase (TRAP) using a leukocyte acid phosphatase kit (Sigma-Aldrich). At least five semi-serial sections through the center of proximal tibia from each mouse were used to reveal mean TRAP+ cell counts at the chondro-osseous border.

Proliferation and apoptosis assays

Cell proliferation was analyzed using BrdU (5-bromo-2ʹ-deoxyuridine) incorporation. BrdU was administered by intraperitoneal injection (100 mg/kg) 2 h prior to sacrifice. BrdU immunoperoxidase staining of paraffin knee joint sections was performed using a similar protocol to that described previously (Xie et al., 2010), except that antigen retrieval used 95% (v/v) formamide. BrdU+ cells were counted in designated areas on five semi-serial sections through the center of proximal tibia or distal femur from each animal. Apoptotic chondrocytes were detected using the In Situ Cell Death Detection Kit (Roche Diagnostics).

Real-time RT-PCR

Hypertrophic cartilage was microdissected from P5 long bones and ribs, and digested with collagenase as described (Belluoccio et al., 2010). Total RNA was extracted using TRIzol (Life Technologies) and digested with DNase I to eliminate any contaminating genomic DNA. cDNA was synthesized by reverse transcription and amplified in triplicate using the SYBR Green PCR assay (QuantiFast SYBR Green PCR Kit, Qiagen, 204057). RNA expression levels were normalized to that of internal control 36B4 (Rplp0). The primers used are listed in supplementary material Table S2.

RNA interference analysis

Pooled primary resting and hypertrophic chondrocytes from P2 wild-type or P7 Col-ZB20KO mice were isolated as described above and subjected to further digestion with 1.0 mg/ml of pronase (Roche) for 3 h at 37°C with slow agitation. The resting chondrocytes were harvested, while the hypertrophic chondrocytes were washed twice in PBS and electroporated with 500 nM Sox9 small interfering RNA (siRNA; sc-36534, Santa Cruz) or control siRNA (sc-36869, Santa Cruz) diluted in BTXpress High Performance Electroporation Solution (Harvard Apparatus). Electroporation was performed in a 2 mm cuvette with settings of one pulse of 170 Volts for 15 ms using a BTX ECM830 electroporator. Chondrocytes were then plated and cultured in DMEM supplemented with 10% FBS (Hyclone), 25 μg/ml L-ascorbic acid 2-phosphate, 10 mM β-glycerophosphate and 1× penicillin/streptomycin/amphotericin B solution (Sigma). The cells were collected 48 h after electroporation and mRNA or total proteins were extracted. Sox9, Vegfa, Col10a1 and Alpl mRNA levels were analyzed by real-time RT-PCR, standardized to 36B4. Sox9 protein levels were assessed by western blotting. The intensity of the signals was evaluated by densitometry and semi-quantified using the ratio between the protein of interest and β-actin for each experiment.

Reporter gene assay

The Sox9 luciferase reporter was constructed by cloning the mouse Sox9 promoter region (−2196 to +27) into the XhoI and BglII sites of the pGL4.10 vector (Promega). HEK293T cells were co-transfected with 1 µg reporter construct, 100 ng β-galactosidase expression vector as internal control and 1 µg either Zbtb20 expression vector or a mock vector. Luciferase and β-galactosidase were measured 48 h post-transfection as described previously (Xie et al., 2008).

ChIP assay

Fragmented chromatin from mouse P5 primary chondrocytes was incubated overnight with specific antibodies: anti-acetylated histone H3 (Millipore, 06-942), anti-Zbtb20 (Xie et al., 2010), or normal mouse IgG (Upstate, 12-371), followed by incubation with Dynabeads Protein G (Invitrogen). Purified chromatin DNA was subjected to conventional and quantitative PCR with primers for core promoter regions of Sox9 (supplementary material Table S2). Two independent ChIP reactions were performed.

Statistical analysis

Results are expressed as mean±s.d. Statistical comparison between genotypes was performed with a two-tailed Student's t-test. Proliferative index was analyzed by one-way analysis of variance (ANOVA). Data from the RNA interference experiment were analyzed using a two-way ANOVA. P<0.05 was considered significant.

We thank Y. Zhang, Q Hao and L Gao for technical assistance.

Author contributions

Z.X. and W.J.Z. conceived and designed the experiments. Z.X., G.Z., X.J., H.Z., Y.L., X.M., A.L., G.Y., R.Y. and H.S. performed the experiments. J.Z., Y.H. and X.Y. analyzed the data. Z.X. and W.J.Z. wrote the paper.

Funding

This work was supported by grants from the China National Natural Science Foundation [31171395, 31025013, 81130084, 31470759] and the National Key Basic Science Research and Development Program [2012CB524904, 2013CB530603].

Akiyama
,
H.
,
Chaboissier
,
M.-C.
,
Martin
,
J. F.
,
Schedl
,
A.
and
de Crombrugghe
,
B.
(
2002
).
The transcription factor Sox9 has essential roles in successive steps of the chondrocyte differentiation pathway and is required for expression of Sox5 and Sox6
.
Genes Dev.
16
,
2813
-
2828
.
Arnold
,
M. A.
,
Kim
,
Y.
,
Czubryt
,
M. P.
,
Phan
,
D.
,
McAnally
,
J.
,
Qi
,
X.
,
Shelton
,
J. M.
,
Richardson
,
J. A.
,
Bassel-Duby
,
R.
and
Olson
,
E. N.
(
2007
).
MEF2C transcription factor controls chondrocyte hypertrophy and bone development
.
Dev. Cell
12
,
377
-
389
.
Belluoccio
,
D.
,
Etich
,
J.
,
Rosenbaum
,
S.
,
Frie
,
C.
,
Grskovic
,
I.
,
Stermann
,
J.
,
Ehlen
,
H.
,
Vogel
,
S.
,
Zaucke
,
F.
,
von der Mark
,
K.
, et al. 
(
2010
).
Sorting of growth plate chondrocytes allows the isolation and characterization of cells of a defined differentiation status
.
J. Bone Miner. Res.
25
,
1267
-
1281
.
Cao
,
G.
,
Fehrenbach
,
M. L.
,
Williams
,
J. T.
,
Finklestein
,
J. M.
,
Zhu
,
J.-X.
and
DeLisser
,
H. M.
(
2009
).
Angiogenesis in platelet endothelial cell adhesion molecule-1-null mice
.
Am. J. Pathol.
175
,
903
-
915
.
Chen
,
J.
,
Singh
,
K.
,
Mukherjee
,
B. B.
and
Sodek
,
J.
(
1993
).
Developmental expression of osteopontin (OPN) mRNA in rat tissues: evidence for a role for OPN in bone formation and resorption
.
Matrix
13
,
113
-
123
.
Dy
,
P.
,
Wang
,
W.
,
Bhattaram
,
P.
,
Wang
,
Q.
,
Wang
,
L.
,
Ballock
,
R. T.
and
Lefebvre
,
V.
(
2012
).
Sox9 directs hypertrophic maturation and blocks osteoblast differentiation of growth plate chondrocytes
.
Dev. Cell
22
,
597
-
609
.
Elima
,
K.
,
Eerola
,
I.
,
Rosati
,
R.
,
Metsaranta
,
M.
,
Garofalo
,
S.
,
Perala
,
M.
,
De Crombrugghe
,
B.
and
Vuorio
,
E.
(
1993
).
The mouse collagen X gene: complete nucleotide sequence, exon structure and expression pattern
.
Biochem. J.
289
,
247
-
253
.
Gerber
,
H.-P.
,
Vu
,
T. H.
,
Ryan
,
A. M.
,
Kowalski
,
J.
,
Werb
,
Z.
and
Ferrara
,
N.
(
1999
).
VEGF couples hypertrophic cartilage remodeling, ossification and angiogenesis during endochondral bone formation
.
Nat. Med.
5
,
623
-
628
.
Han
,
Y.
and
Lefebvre
,
V.
(
2008
).
L-Sox5 and Sox6 drive expression of the aggrecan gene in cartilage by securing binding of Sox9 to a far-upstream enhancer
.
Mol. Cell. Biol.
28
,
4999
-
5013
.
Hattori
,
T.
,
Muller
,
C.
,
Gebhard
,
S.
,
Bauer
,
E.
,
Pausch
,
F.
,
Schlund
,
B.
,
Bosl
,
M. R.
,
Hess
,
A.
,
Surmann-Schmitt
,
C.
,
von der Mark
,
H.
, et al. 
(
2010
).
SOX9 is a major negative regulator of cartilage vascularization, bone marrow formation and endochondral ossification
.
Development
137
,
901
-
911
.
Hiraki
,
Y.
,
Inoue
,
H.
,
Iyama
,
K.-i.
,
Kamizono
,
A.
,
Ochiai
,
M.
,
Shukunami
,
C.
,
Iijima
,
S.
,
Suzuki
,
F.
and
Kondo
,
J.
(
1997
).
Identification of chondromodulin I as a novel endothelial cell growth inhibitor: purification and its localization in the avascular zone of epiphyseal cartilage
.
J. Biol. Chem.
272
,
32419
-
32426
.
Ikegami
,
D.
,
Akiyama
,
H.
,
Suzuki
,
A.
,
Nakamura
,
T.
,
Nakano
,
T.
,
Yoshikawa
,
H.
and
Tsumaki
,
N.
(
2011
).
Sox9 sustains chondrocyte survival and hypertrophy in part through Pik3ca-Akt pathways
.
Development
138
,
1507
-
1519
.
Kim
,
Y.
,
Murao
,
H.
,
Yamamoto
,
K.
,
Deng
,
J. M.
,
Behringer
,
R. R.
,
Nakamura
,
T.
and
Akiyama
,
H.
(
2011
).
Generation of transgenic mice for conditional overexpression of Sox9
.
J. Bone Miner. Metab.
29
,
123
-
129
.
Kirsch
,
T.
,
Nah
,
H.-D.
,
Shapiro
,
I. M.
and
Pacifici
,
M.
(
1997
).
Regulated production of mineralization-competent matrix vesicles in hypertrophic chondrocytes
.
J. Cell Biol.
137
,
1149
-
1160
.
Linsenmayer
,
T. F.
,
Chen
,
Q. A.
,
Gibney
,
E.
,
Gordon
,
M. K.
,
Marchant
,
J. K.
,
Mayne
,
R.
and
Schmid
,
T. M.
(
1991
).
Collagen types IX and X in the developing chick tibiotarsus: analyses of mRNAs and proteins
.
Development
111
,
191
-
196
.
Liu
,
X.
,
Zhang
,
P.
,
Bao
,
Y.
,
Han
,
Y.
,
Wang
,
Y.
,
Zhang
,
Q.
,
Zhan
,
Z.
,
Meng
,
J.
,
Li
,
Y.
,
Li
,
N.
, et al. 
(
2013
).
Zinc finger protein ZBTB20 promotes Toll-like receptor-triggered innate immune responses by repressing IkappaBalpha gene transcription
.
Proc. Natl. Acad. Sci. USA
110
,
11097
-
11102
.
Mark
,
M. P.
,
Butler
,
W. T.
,
Prince
,
C. W.
,
Finkelman
,
R. D.
and
Ruch
,
J.-V.
(
1988
).
Developmental expression of 44-kDa bone phosphoprotein (osteopontin) and bone gamma-carboxyglutamic acid (Gla)-containing protein (osteocalcin) in calcifying tissues of rat
.
Differentiation
37
,
123
-
136
.
McKee
,
M. D.
,
Glimcher
,
M. J.
and
Nanci
,
A.
(
1992
).
High-resolution immunolocalization of osteopontin and osteocalcin in bone and cartilage during endochondral ossification in the chicken tibia
.
Anat. Rec.
234
,
479
-
492
.
Miao
,
D.
and
Scutt
,
A.
(
2002
).
Histochemical localization of alkaline phosphatase activity in decalcified bone and cartilage
.
J. Histochem. Cytochem.
50
,
333
-
340
.
Mitchelmore
,
C.
,
Kjaerulff
,
K. M.
,
Pedersen
,
H. C.
,
Nielsen
,
J. V.
,
Rasmussen
,
T. E.
,
Fisker
,
M. F.
,
Finsen
,
B.
,
Pedersen
,
K. M.
and
Jensen
,
N. A.
(
2002
).
Characterization of two novel nuclear BTB/POZ domain zinc finger isoforms: association with differentiation of hippocampal neurons, cerebellar granule cells, and macroglia
.
J. Biol. Chem.
277
,
7598
-
7609
.
Molin
,
A.-M.
,
Andrieux
,
J.
,
Koolen
,
D. A.
,
Malan
,
V.
,
Carella
,
M.
,
Colleaux
,
L.
,
Cormier-Daire
,
V.
,
David
,
A.
,
de Leeuw
,
N.
,
Delobel
,
B.
, et al. 
(
2012
).
A novel microdeletion syndrome at 3q13.31 characterised by developmental delay, postnatal overgrowth, hypoplastic male genitals, and characteristic facial features
.
J. Med. Genet.
49
,
104
-
109
.
Nielsen
,
J. V.
,
Nielsen
,
F. H.
,
Ismail
,
R.
,
Noraberg
,
J.
and
Jensen
,
N. A.
(
2007
).
Hippocampus-like corticoneurogenesis induced by two isoforms of the BTB-zinc finger gene Zbtb20 in mice
.
Development
134
,
1133
-
1140
.
Ovchinnikov
,
D.
(
2009
).
Alcian blue/alizarin red staining of cartilage and bone in mouse
.
Cold Spring Harb. Protoc.
2009
, ppdb.prot5170.
Ren
,
A.-J.
,
Wang
,
K.
,
Zhang
,
H.
,
Liu
,
A.
,
Ma
,
X.
,
Liang
,
Q.
,
Cao
,
D.
,
Wood
,
J. N.
,
He
,
D. Z.
,
Ding
,
Y.-Q.
, et al. 
(
2014
).
ZBTB20 regulates nociception and pain sensation by modulating TRP channel expression in nociceptive sensory neurons
.
Nat. Commun.
5
,
4984
.
Schmitz
,
N.
,
Laverty
,
S.
,
Kraus
,
V. B.
and
Aigner
,
T.
(
2010
).
Basic methods in histopathology of joint tissues
.
Osteoarthritis Cartilage
18
Suppl. 3
,
S113
-
S116
.
Stickens
,
D.
,
Behonick
,
D. J.
,
Ortega
,
N.
,
Heyer
,
B.
,
Hartenstein
,
B.
,
Yu
,
Y.
,
Fosang
,
A. J.
,
Schorpp-Kistner
,
M.
,
Angel
,
P.
and
Werb
,
Z.
(
2004
).
Altered endochondral bone development in matrix metalloproteinase 13-deficient mice
.
Development
131
,
5883
-
5895
.
Sutherland
,
A. P. R.
,
Zhang
,
H.
,
Zhang
,
Y.
,
Michaud
,
M.
,
Xie
,
Z.
,
Patti
,
M.-E.
,
Grusby
,
M. J.
and
Zhang
,
W. J.
(
2009
).
Zinc finger protein Zbtb20 is essential for postnatal survival and glucose homeostasis
.
Mol. Cell. Biol.
29
,
2804
-
2815
.
Takeda
,
S.
,
Bonnamy
,
J.-P.
,
Owen
,
M. J.
,
Ducy
,
P.
and
Karsenty
,
G.
(
2001
).
Continuous expression of Cbfa1 in nonhypertrophic chondrocytes uncovers its ability to induce hypertrophic chondrocyte differentiation and partially rescues Cbfa1-deficient mice
.
Genes Dev.
15
,
467
-
481
.
Vortkamp
,
A.
,
Lee
,
K.
,
Lanske
,
B.
,
Segre
,
G. V.
,
Kronenberg
,
H. M.
and
Tabin
,
C. J.
(
1996
).
Regulation of rate of cartilage differentiation by Indian hedgehog and PTH-related protein
.
Science
273
,
613
-
622
.
Vu
,
T. H.
,
Shipley
,
J. M.
,
Bergers
,
G.
,
Berger
,
J. E.
,
Helms
,
J. A.
,
Hanahan
,
D.
,
Shapiro
,
S. D.
,
Senior
,
R. M.
and
Werb
,
Z.
(
1998
).
MMP-9/gelatinase B is a key regulator of growth plate angiogenesis and apoptosis of hypertrophic chondrocytes
.
Cell
93
,
411
-
422
.
Xie
,
Z.
,
Zhang
,
H.
,
Tsai
,
W.
,
Zhang
,
Y.
,
Du
,
Y.
,
Zhong
,
J.
,
Szpirer
,
C.
,
Zhu
,
M.
,
Cao
,
X.
,
Barton
,
M. C.
, et al. 
(
2008
).
Zinc finger protein ZBTB20 is a key repressor of alpha-fetoprotein gene transcription in liver
.
Proc. Natl. Acad. Sci. USA
105
,
10859
-
10864
.
Xie
,
Z.
,
Ma
,
X.
,
Ji
,
W.
,
Zhou
,
G.
,
Lu
,
Y.
,
Xiang
,
Z.
,
Wang
,
Y. X.
,
Zhang
,
L.
,
Hu
,
Y.
,
Ding
,
Y.-Q.
, et al. 
(
2010
).
Zbtb20 is essential for the specification of CA1 field identity in the developing hippocampus
.
Proc. Natl. Acad. Sci. USA
107
,
6510
-
6515
.
Yang
,
G.
,
Sun
,
Q.
,
Teng
,
Y.
,
Li
,
F.
,
Weng
,
T.
and
Yang
,
X.
(
2008
).
PTEN deficiency causes dyschondroplasia in mice by enhanced hypoxia-inducible factor 1alpha signaling and endoplasmic reticulum stress
.
Development
135
,
3587
-
3597
.
Yoshida
,
C. A.
,
Yamamoto
,
H.
,
Fujita
,
T.
,
Furuichi
,
T.
,
Ito
,
K.
,
Inoue
,
K.-i.
,
Yamana
,
K.
,
Zanma
,
A.
,
Takada
,
K.
,
Ito
,
Y.
, et al. 
(
2004
).
Runx2 and Runx3 are essential for chondrocyte maturation, and Runx2 regulates limb growth through induction of Indian hedgehog
.
Genes Dev.
18
,
952
-
963
.
Zhang
,
W.
,
Mi
,
J.
,
Li
,
N.
,
Sui
,
L.
,
Wan
,
T.
,
Zhang
,
J.
,
Chen
,
T.
and
Cao
,
X.
(
2001
).
Identification and characterization of DPZF, a novel human BTB/POZ zinc finger protein sharing homology to BCL-6
.
Biochem. Biophys. Res. Commun.
282
,
1067
-
1073
.
Zhang
,
J.
,
Tan
,
X.
,
Li
,
W.
,
Wang
,
Y.
,
Wang
,
J.
,
Cheng
,
X.
and
Yang
,
X.
(
2005
).
Smad4 is required for the normal organization of the cartilage growth plate
.
Dev. Biol.
284
,
311
-
322
.
Zhang
,
Y.
,
Xie
,
Z.
,
Zhou
,
L.
,
Li
,
L.
,
Zhang
,
H.
,
Zhou
,
G.
,
Ma
,
X.
,
Herrera
,
P. L.
,
Liu
,
Z.
,
Grusby
,
M. J.
, et al. 
(
2012
).
The zinc finger protein ZBTB20 regulates transcription of fructose-1,6-bisphosphatase 1 and beta cell function in mice
.
Gastroenterology
142
, p1571-1580.e6.
Zheng
,
Q.
,
Zhou
,
G.
,
Morello
,
R.
,
Chen
,
Y.
,
Garcia-Rojas
,
X.
and
Lee
,
B.
(
2003
).
Type X collagen gene regulation by Runx2 contributes directly to its hypertrophic chondrocyte-specific expression in vivo
.
J. Cell Biol.
162
,
833
-
842
.

Competing interests

The authors declare no competing financial interests.

Supplementary information