Elsevier

Differentiation

Volume 111, January–February 2020, Pages 60-69
Differentiation

SMAD2 overexpression rescues the TGF-β3 null mutant mice cleft palate by increased apoptosis

https://doi.org/10.1016/j.diff.2019.10.001Get rights and content

Abstract

During palatal development, medial edge epithelium (MEE) disappearance is one of the crucial steps in the process of fusion. The fate of these cells is still debated, and controversies remain. During secondary palate fusion, TGF-β3 signaling mediated in the cell through the SMAD2 protein plays an important role and leads to the disappearance of the midline epithelial seam (MES) and the confluence of the palatal mesenchyme. In mice, TGF-β3 knock-out is lethal and mice are born with a cleft in the secondary palate. This phenotype has been rescued by targeted overexpression of SMAD2 in the medial edge epithelium (MEE). The goal of this research was to understand the mechanism of palatal fusion in the rescue mice.

Methods

The heads of embryos with four different genotypes (wild-type, K14-SMAD2/TGF-β3(−/−), K14-SMAD2/TGF-β3(±), and TGF-β3 null) were collected at embryonic day E14.5, genotyped, fixed and embedded in paraffin. Serial sections were studied for detection of apoptosis and epithelial mesenchymal transition using immunofluorescence.

Results

TGF-β3 null mice developed a cleft in the secondary palate while both mice with K14-SMAD2 overexpression had fusion of the secondary palate. The MEE of both the rescue mice and K14-SMAD2 overexpression had a much higher ratio of apoptotic cells than wild-type mice. The increase in apoptosis was correlated with increased phospho-SMAD2 in the MEE.

Conclusion

SMAD2 overexpression rescued the cleft in the secondary palate by increasing apoptosis in the medial edge epithelium.

Introduction

The fetal development of the secondary palate is a tightly regulated process in mammals (Li et al., 2017; Ferguson, 1988). This process requires precise interaction between several cells and tissues (Nakajima, F. Shuler, Gulka & Hanai, 2018; Shuler et al., 1991). Temporal and spatial specialization of extracellular matrix (Nakajima, F. Shuler, Gulka & Hanai, 2018; Brinkley and Morris-Wiman, 1984), mesenchymal cells (Ferguson, 1988), and epithelial cells (Ferguson, 1988) are necessary in order to achieve palatal fusion prior to birth. Failure of this process results in cleft palate.

In a mouse model, palatal development begins with palatal shelves growing from the inner aspect of each maxillary process at embryonic day E11.5. Initially at day E13.5, the shelves protrude vertically along either side of the tongue. As the tongue begins to grow and form, the shelves reorient horizontally above the tongue at day E14.0. The shelves make contact in the midline at E14.5, initiating fusion and forming the temporary midline epithelial seam (MES) (Bush and Jiang, 2012). Histologically, the palatal shelves are composed of mesenchyme which is derived from the neural crest (Li et al., 2017; Bush and Jiang, 2012; Ferguson, 1988; Shuler, 1995). The mesenchyme is surrounded by a layer of oral epithelial cells, derived from the ectoderm. The MES must eventually be eliminated in order to permit mesenchymal confluence of the palatal shelves to complete fusion.

Recent studies have identified possible molecular and cellular mechanisms involved in the critical process of MES disappearance, some aspects remain controversial (Nakajima, F. Shuler, Gulka & Hanai, 2018; Gritli-Linde, 2007). Three different theories of processes regulating the fate of MES have been proposed; 1) apoptosis (DeAngelis and Nalbandian, 1968; Farbman, 1968; Hayward, 1969; Shapiro and Sweney, 1969; Cuervo and Covarrubias, 2004; Vaziri Sani et al., 2005; Dudas et al., 2006; Dudas et al., 2007; Xu et al., 2006); 2) epithelial mesenchymal transdifferentiation (EMT) (Fitchett and Hay, 1989; Griffith and Hay, 1992; Shuler et al., 1991, 1992; Kaartinen et al., 1997; Nawshad and Hay, 2003; Takigawa and Shiota, 2004; Kang and Svoboda, 2005; Jin and Ding, 2006); 3) cell migration (Carette and Ferguson (1992). Recent studies have shown that both apoptosis and EMT may be involved in the disappearance of the MES (Nawshad, 2008; Martinez-Alvarez et al., 2000; Ahmed et al., 2007).

Transforming growth factor beta 3 (TGF-β3) is a cytokine involved in many cellular functions including cell cycle control, cell development, differentiation, hematopoiesis and apoptosis (Zhang et al., 2016; Schuster and Krieglstein, 2002). TGF-β3 is involved in the repression of growth of epithelial cells and is expressed in the MES (Schuster and Krieglstein, 2002; Fitzpatrick et al., 1990). TGF-β3 null mutant mice have a phenotype that includes a cleft secondary palate due to the MES remaining intact during development (Kaartinen et al., 1995; Proetzel et al., 1995). TGF-β3 utilizes SMAD proteins (SMAD2/SMAD3) to transmit regulatory signals inside target cells (Schuster and Krieglstein, 2002). SMAD2 and SMAD3 are extremely homologous sharing nearly 95% of their amino acid sequence but they differ in function (Liu et al., 2016; Shiomi et al., 2006). The concept that SMAD3 null mutant mice develop normally while SMAD2 null mutant mice embryos are non-viable indicate the important role that SMAD2 plays during embryonic development (Brown et al., 2007). SMAD2 and SMAD3 are both expressed in the medial edge epithelium (MEE) but during the process of palatal fusion but only SMAD2 is phosphorylated (Cui et al., 2003). The inhibition and inactivation of SMAD signaling and specifically SMAD2 during palatal fusion by all-trans retinoic acid (atRA) resulted in maintenance of MEE and failure of palatal fusion (Wang et al., 2011). The inhibition of SMAD2 in MEE cells by SMAD2 siRNA maintained the proliferation of MEE and prevented the fusion of the palatal shelves (Shiomi et al., 2006). Furthermore, the addition of exogenous TGF-β3 to the siRNA treated MEE cells failed to rescue the palatal fusion indicating the critical role of SMAD2 signaling pathway during palatal fusion (Shiomi et al., 2006). Interestingly, SMAD2 overexpression in TGF-β3 null mutant mice has been found to rescue palatal fusion (Cui et al., 2005). SMAD2 overexpression has also been found to increase the apoptosis rate in junctional epithelium and downregulates Bcl-2 (Fujita et al., 2012), in prostate epithelial cells (Yang et al., 2009), human gingival epithelial cells (Yoshimoto et al., 2015), and human ocular lens epithelial cells (Lee et al., 2002).

We hypothesized that: SMAD2 overexpression rescues the TGF-β3 null mutant mice by increased apoptosis of MEE. The aim of this study was to investigate the TGF-β3 and SMAD2 regulated mechanism of MES disappearance in a rescue mouse model [K14-SMAD2/TGF-β3(−/−)] compared to wild type mice, null mutant [TGF-β3(−/−)] and SMAD2 overexpression [K14-SMAD2/TGF-β3(+/-)].

Section snippets

Increased MEE cell apoptotic rate

Cleaved caspase3 (Ccaspase3), an apoptosis marker, was used to detect apoptotic cells associated with SMAD2 overexpression in the palatal MEE cells. Ccaspase-3 protein was localized in the palatal MEE cells, in the anterior, middle and posterior regions of the secondary palate at day E14.5 (Fig. 1). Our results showed that Ccaspase-3 positive cells were detected in the nasal and oral MES triangles. The K14-SMAD2/TGF-β3(+/-) mice also had a much higher ratio of Ccaspase3 positive MEE (16.14%

Discussion

TGF-β3 has been known to play a role in the fate of MEE cells during palatal fusion (Kaartinen et al., 1995). TGF-β3 null mutant mice are born with a clefting of the secondary palate due to failure in MEE adhesion (Taya, O'Kane & Ferguson, 1999; Kaartinen et al., 1995). SMAD2 over expression in TGF-β3 null mutant mice rescued the cleft palatal fusion and mesenchymal confluence was established (Cui et al., 2005). Little is known about the precise MEE fate induced by SMAD2 overexpression in this

Conclusion

The present study has focused on the effect of SMAD2 over expression on medial edge epithelial cells during palatal fusion. The mechanism of MEE disappearance during palatal fusion was examined. This study is the first to have studied specifically the fate of MEE during palatal fusion in the K14-SMAD2/TGF-β3(−/−) mouse model. The available data indicate that the SMAD2 over expression rescues the palatal fusion in the TGF-β3(−/−) mice by increased MEE apoptosis. The same effect was seen in the

Animal breeding and genotyping

Methods within this study followed the guidelines of the Animal Care Committee of the University of British Columbia. To identify the K14-SMAD2 transgene a polymerase chain reaction (PCR) primer set was used to detect the cytokeratin 14 promoter region. To detect the TGF-β3 knockout gene another PCR primer set was used to detect the mutated allele (discussed in detail in section 3.2). Mice heterozygous with respect to the TGF-β3(+/−) gene were mated to produce TGF-β3(−/−) null embryos, as well

Acknowledgements

The research was supported by funding from the UBC Faculty of Dentistry research support mechanism. The authors wish to declare no potential conflicts of interests with respect to the authorship and publication of this article.

References (50)

  • C. Martinez-Alvarez et al.

    Medial edge epithelial cell fate during palatal fusion

    Dev. Biol.

    (2000)
  • C. Shuler et al.

    Medial edge epithelium fate traced by cell lineage analysis during epithelial-mesenchymal transformation in vivo

    Dev. Biol.

    (1992)
  • F. Vaziri Sani et al.

    Fate-mapping of the epithelial seam during palatal fusion rules out epithelial-mesenchymal transformation

    Dev. Biol.

    (2005)
  • X. Xu et al.

    Cell autonomous requirement for Tgfbr2 in the disappearance of medial edge epithelium during palatal fusion

    Dev. Biol.

    (2006)
  • T. Yoshimoto et al.

    Involvement of smad2 and Erk/Akt cascade in TGF-β1-induced apoptosis in human gingival epithelial cells

    Cytokine

    (2015)
  • K. Brown et al.

    A tale of two proteins: Differential roles and regulation of Smad2 and Smad3 in TGF-β signaling

    J. Cell. Biochem.

    (2007)
  • J. Bush et al.

    Palatogenesis: morphogenetic and molecular mechanisms of secondary palate development

    Development

    (2012)
  • M. Carette et al.

    The fate of medial edge epithelial cells during palatal fusion in vitro: an analysis by DiI labelling and confocal microscopy

    Development

    (1992)
  • R. Cuervo et al.

    Death is the major fate of medial edge epithelial cells and the cause of basal lamina degradation during palatogenesis

    Development

    (2004)
  • X. Cui et al.

    TGF-beta3-dependent SMAD2 phosphorylation and inhibition of MEE proliferation during palatal fusion

    Dev. Dyn.

    (2003)
  • V. DeAngelis et al.

    Ultrastructure of mouse and rat palatal processes prior to and during secondary palate formation

    Arch. Oral Biol.

    (1968)
  • M. Ferguson

    Palate development

    Development

    (1988)
  • D. Fitzpatrick et al.

    Differential expression of TGF beta isoforms in murine palatogenesis

    Development

    (1990)
  • C. Griffith et al.

    Epithelial-mesenchymal transformation during palatal fusion: carboxyfluorescein traces cells at light and electron microscopic levels

    Development

    (1992)
  • A. Hayward

    Ultrastructural changes in the epithelium during fusion of the palatal processes in rats

    Arch. Oral Biol.

    (1969)
  • Cited by (13)

    • Comprehensive analysis of plasma miRNA and related ceRNA network in non-syndromic cleft lip and/or palate

      2022, International Journal of Pediatric Otorhinolaryngology
      Citation Excerpt :

      Studies have shown that TGF-βfamily can extensively regulate a variety of cell biological behaviors involved in palate development [29], including cell proliferation, apoptosis, differentiation, migration, and EMT, etc. At present, many studies have confirmed that SMAD2 protein located in TGF-β/Smad signal pathway can participate in the development of palate, and the expression is down-regulated in the formation of cleft palate [30], which is consistent with the expression trend of this study. Studies have shown that overexpression of miR-200 b in mouse palatal fusion can induce apoptosis and proliferation in palatal fusion area by inhibiting SMAD2, ZEB1 and ZEB2 [31].

    • Crosstalk between sex-related genes and apoptosis signaling reveals molecular insights into sex change in a protogynous hermaphroditic teleost fish, ricefield eel Monopterus albus

      2022, Aquaculture
      Citation Excerpt :

      In further analyzing the mechanisms underlying sex change in the ricefield eel M. albus, we found that apoptosis-related signaling pathways may play crucial roles in sex change (Table 1, Fig. 6). Studies have shown that p53 signaling is a core pathway that integrates upstream elements of apoptosis signaling, such as cblb (Fujiwara et al., 2006; Jeong et al., 2008; Leszczynska et al., 2015; Li et al., 2011; Qu et al., 2010), erbb4 (Das et al., 2010; Yan et al., 2017), arap1 (Daniele et al., 2008; Yoon et al., 2008), TGF-β signaling components (AlMegbel and Shuler, 2020; Chen et al., 2019; Elston and Inman, 2012; Liu et al., 2017; Ying et al., 2017), ccnd3 (Fimognari et al., 2002), ptpn11 (Chitranshi et al., 2017; Ren et al., 2017) and numb (Liu et al., 2016). In fish, p53 has been demonstrated to promote oocyte apoptosis and spermatogenesis for sex change from female to male in the zebrafish D. rerio (Rodriguez-Mari et al., 2010; Rodriguez-Mari and Postlethwait, 2011).

    • TGF beta −1, −2 and −3 in the modulation of fibrosis in the cornea and other organs

      2021, Experimental Eye Research
      Citation Excerpt :

      Defects in epithelial–mesenchymal interactions also are prominent in TGF beta-3−/− null mice (Kaartinen et al., 1995). Interestingly, SMAD2 overexpression rescues the cleft in the secondary palate, and is associated with increased apoptosis in the medial edge epithelial cells (AlMegbel and Shuler, 2020). Some further support for differential effects of the TGF beta isoforms in wound healing and fibrosis have been published.

    • The cleft palate candidate gene BAG6 supports FoxO1 acetylation to promote FasL-mediated apoptosis during palate fusion

      2020, Experimental Cell Research
      Citation Excerpt :

      Apoptosis is an important mechanism ensuring proper palate fusion during palate development [2]. During palate fusion, the pro-apoptotic protease caspase-3 is activated in medial edge epithelial (MEE) cells located at the medial epithelial seam (MES) [3]. In both humans and rodents, the transforming growth factor-3 (TGF-β3) signaling pathway is required for palate fusion by promoting MEE apoptosis [3].

    View all citing articles on Scopus
    View full text