search
for
 About Bioline  All Journals  Testimonials  Membership  News


Indian Journal of Plastic Surgery
Medknow Publications on behalf of Indian Journal of Plastic Surgery
ISSN: 0970-0358 EISSN: 1998-376x
Vol. 42, Num. S1, 2009, pp. 35-50

Indian Journal of Plastic Surgery, Vol. 42, Supp. 1 , 2009, pp. 35-50

Review Article

Cleft lip and palate genetics and application in early embryological development

Department of Biomedical Sciences, Texas A&M Health Science Center, Baylor College of Dentistry, Dallas, TX 75246 USA

Correspondence Address: Dr. Kathy K. H. Svoboda, Biomedical Sciences Department, Baylor College of Dentistry, 3302 Gaston Ave., Dallas, TX 75246 USA, Ksvoboda@bcd.tamhsc.edu

Code Number: pl09077

PMID: 19884679

DOI: 10.4103/0970-0358.57185

Abstract

The development of the head involves the interaction of several cell populations and coordination of cell signalling pathways, which when disrupted can cause defects such as facial clefts. This review concentrates on genetic contributions to facial clefts with and without cleft palate (CP). An overview of early palatal development with emphasis on muscle and bone development is blended with the effects of environmental insults and known genetic mutations that impact human palatal development. An extensive table of known genes in syndromic and non-syndromic CP, with or without cleft lip (CL), is provided. We have also included some genes that have been identified in environmental risk factors for CP/L. We include primary and review references on this topic.

Keywords: Cleft lip/palate; Human/murine palate genetics; Palate development

Introduction

Disturbances at any stage during palate development, e.g., defective palatal shelf growth, failed or delayed elevation, and blocked fusion, can result in cleft palate (CP) [1],[2] with or without cleft lip (CL/P). As one of the most common congenital cranio-facial defects, CL/P occurs in approximately 1 per 750 live births in the United States [2],[3] Clefts occur more frequently among Asians (about 1:400) and certain American Indians than Europeans or European descendants. Clefts are relatively less common among Africans and African Americans (about 1:1500). [4] Cleft lip and palate may not be life-threatening but many functions such as feeding, digestion, speech, middle-ear ventilation, hearing, respiration and facial and dental development can be disturbed because of the structures involved. These problems can also cause emotional, psycho-social, and educational difficulties. In addition, CP is an economic burden.

The aetiology of cleft lip with or without palate (CL/P) is theorized to be a combination of factors associated with genes and environment. [5],[6] The advent of gene targeting technology and basic conventional techniques using animal models has led to the identification of genes associated with known and unknown etiologic factors. Characterization of the genomic sequences will greatly impact regulation of gene networks and pinpoint any variations in the different stages of craniofacial morphogenesis. In this article, emphasis is placed on different genes associated with the classifications of CL/P into syndromic [Table - 1] and nonsyndromic [Table - 2]. Each classification plays a significant role in understanding the molecular and genetic mechanisms affecting these types of craniofacial defects. [7],[8],[9] In addition to known genes there is strong evidence that several environmental factors (e.g., alcohol consumption, tobacco, and anti-convulsants) increase the risk of CL/P. [10],[11] In contrast, several studies have shown that folic acid may have a protective effect on CL/P and neural tube defects. [12],[13],[14],[15],[16] Recent data from the National Birth Defect Prevention Network have indicated a decrease in neural tube defects from 5/10,000 to less than 2/10,000 after the fortification of the food supply with folic acid, indicating that this vitamin and the proteins that facilitate the uptake and metabolism of folic acid may be candidate genes in cranio-facial development. [14],[17],[18],[19],[20]

This review will concentrate on genetic contributions to facial clefts with/without cleft palate. We will begin with an overview of early palatal development, concentrate on muscle and bone development, and incorporate the effects of environmental insults and known genetic mutations that impact human palatal development.

Embryonic Palate Development

The palatal structures are composed of the cranial neural crest (CNC)-derived mesenchyme and pharyngeal ectoderm. [21-24] Epithelia that cover the palatal shelves are regionally divided into oral, nasal and medial edge epithelia (MEE). The nasal and oral epithelia differentiate into pseudo-stratified and squamous epithelia, whereas MEE is removed from the fusion line [Figure - 1].

The secondary palate originates as an outgrowth of the maxillary prominences at approximately embryonic day 11.5 in the mouse (E11.5-m) [Figure - 1] and post coital six weeks in humans (p.c.6wk-h). The palate shelves initially grow vertically along the sides of the tongue (E13.5-m; p.c.7wk-h) and then rise above the tongue as the latter drops in the oral cavity due to the forward and downward growth of the mandible (E14.0-m; p.c.8wks-h). With continued growth, the shelves appose at the midline (E14.5-m; p.c.10wks-h) and eventually fuse (E15.5-m; p.c.13wk-h). [25] Numerous genes similar in mice [26] and humans [25],[27],[28] are expressed [Table - 1] during palatal development.

During fusion the epithelium covering the tip of the opposing palatal shelves, adheres, intercalates and thins into a single-layer midline epithelial seam (MES). [23] The disintegration of this seam results in the confluence of the palatal mesenchyme. Tremendous interest has arisen in cellular mechanisms underlying MES degradation. Epithelial-mesenchymal transition (EMT) is one of the proposed models that regulates medial edge epithelial (MEE) cell fate. [23],[29],[30],[31],[32],[33],[34],[35],[36] However, other mechanisms have been proposed, such as apoptosis, [37],[38],[39],[40] in which all MEE cells are theorized to die during fusion. Alternatively, it is hypothesized by some researchers that MES cells disappear by migrating from the midline towards the nasal and oral epithelia. [41],[42] Other investigators postulate that all events, including apoptosis, migration and EMT, may occur. [23],[39],[43] Interestingly, the fusion of the external surface of the bilateral maxillary processes with the naso-frontal prominence in the chick is similar to palatal fusion [Figure - 2]. [44] The outer periderm layer dies through apoptosis, and the lateral edge epithelium of the inter-maxillary segment of the naso-frontal process fuses with the medial edge epithelium of the external maxillary process to form a seam that transitions to a confluent mesenchyme [Figure - 2]. [44] Evidence supporting these theories, especially those involving EMT and apoptosis, will be presented and further discussed.

Molecular Signalling Events in Embryonic Palatal Development

As stated above, cleft palate with or without cleft lip is a complex trait caused by a combination of multiple genes and environmental factors. [5] Palatal shelf development defects will be divided into five categories for the purpose of this review:

Failure of palatal shelf formation

The failure of the palatal shelf formation is a rare severe defect. Recent studies have identified several molecular networks operating between the palatal shelf epithelium and mesenchyme during different steps of palatogenesis. These networks include signalling molecules and growth factors such as sonic hedgehog (Shh), members of the transforming growth factor β (TGfβ) super family, including bone morphogenetic proteins (Bmps) and Tgfβs, fibroblast growth factors (Fgfs) and their receptors (FgfR), effectors and targets. [25] Studies addressing the role of Fgf signalling during early palatal development by analyzing Fgf10 and FgfR2b mutants found altered cell proliferation within both mesenchyme and epithelium in the palatal shelves and increased apoptosis within the epithelium. It was reported that Fgf10 and FgfR2b mutations affected the initial development of palatal shelves, and the mouse pups had complete CP. [45] By signalling via its receptor, FgfR2b, in the palatal shelf epithelium, the mesenchymal derived Fgf10 supports epithelial proliferation and survival and also induces the expression of Shh within the epithelium. Shh, in turn, signals to the mesenchyme and stimulates cell proliferation.

In general, signalling activities are subject to tight spatio-temporal control, and, in many instances, too much or too little control is detrimental to the developing organ. This situation is well illustrated in anomalies caused by de-regulated hedgehog (hh) and Fgf signalling. [46],[47] While Fgf10/FgfR2b activity plays a crucial role during palatogenesis, it appears to be subject to the tight spatio-temporal regulation shown in mice lacking Shox2. Shox2 mutant mice develop a very rare type of CP that may also be found in humans [48] the soft palate is intact, whereas the hard palate has a cleft. Abnormal proliferation and apoptosis are theorized to be the cause of the cleft. Surprisingly, a number of protagonists implicated in palatogenesis, including Msx1, Bmp4, Pax9, Lhx8, Osr2, Tgfβ3 and Jag 2, were expressed normally. [48] In contrast, Fgf10 and Fgfr2b were expressed at ectopic sites within the mesenchyme of the Shox2 mutant mice. [49] These studies emphasize the importance of the precise timing and determination of sites of signalling activities necessary for normal development. Mutation of activin-bA causes a severe facial primordial development defect, which may be responsible for the retardation of palatal shelf development and complete cleft palate. In addition, other genes, including Msx1, Lhx8, Shox2 and Osr2, assume important roles in the palatal shelf growth. The targeted mutation of these genes in mice generates CP, indicating the intrinsic requirement of these factors during palatogenesis. [49]

Fusion of the palatal shelf with the tongue or mandible

Under normal conditions, palatal shelves do not fuse with other oral structures. However, in mice that do not express Fgf10, the palatal shelf epithelium fuses with the tongue and mandible. [45] The loss of function mutations of Fgf10 results in anterior palatal shelf fusion with the tongue, whereas the middle and posterior palatal shelf regions adhere to the mandible, thus preventing the elevation of the palatal shelf. [50] There is a severe reduction of the expression of Jagged 2 (Jag2), thereby encoding a ligand for the Notch family receptors and ectopic Tgfβ3 production in the nasal epithelia of these mice. The analysis of Jag2 mutant embryos indicates that Jag2-Notch signalling prevents inappropriate palatal shelf adhesion to other oral epithelia through the control of oral epithelial differentiation. Another gene has also been associated with inappropriate adhesions. Mutations in TBX22 have been reported in families with X-linked cleft palate and ankyloglossia. [51],[52],[53] Tbx22 is expressed in the developing palate and tongue in mice, suggesting an important role in regulating tongue and palate development.

Failure of palatal elevation

Palatal shelf elevation is a rapid movement triggered by both intrinsic forces within the palatal shelves proper and by influences from other craniofacial and oral structures, including the movement of the tongue, and growth of the cranium and mandible. [1],[54] The role of the extra-cellular matrix in palatal shelf elevation has been supported by some studies and is presently accepted as an important determinant of palatal shelf elevation. [55],[56] Those studies [1] suggested that a progressive differential accumulation of glycosaminoglycans, primarily hyaluran in the palatal shelves, plays a role in their elevation. [55],[56] Hyaluronan is a highly charged glycosaminoglycan that retains high amounts of water, forming hydrated gels leading to the expansion of the extracellular matrix. Other constituents of the palatal shelves including collagen fibers, vascularization, and the epithelial covering; the polarized alignment of the mesenchyme cells may also contribute to the intrinsic elevation force of the PS. Mutations of Pax9, Pitx1 or Osr2 can lead to failed palatal shelf elevation and cleft palate defect. [57],[58],[59],[60] The cellular defect is associated with the CNC-derived palatal mesenchyme, suggesting the important functions of these transcription factors in regulating the fate of the CNC cells during palatogenesis.

Early studies attributed a role to neuro-transmitters during palatal shelf elevation. [1] At present, it is widely accepted that neuro-transmitter γ-aminobutyric acid (GABA) regulates not only neuronal activities but also cell migration, survival, proliferation and differentiation of neuronal and non-neuronal cells. [61],[62],[63] Terratological studies in rodents showed that GABA or GABA agonists generate CP by inhibiting palatal shelf elevation, whereas GABA antagonists stimulate the process. [64] The implication of GABA in palate development was demonstrated by genetic studies of mice lacking the β3 subunit of the GABA receptor that developed CP without other craniofacial malformations. [65]

Failure of palatal shelves to meet after elevation

Fusion of the opposing palatal shelves is an important step taking place through a sequence of events that includes the removal of the flat peridermal cells, contact and adhesion of the opposing MEE, which creates the MES, and the degeneration of the MES. The mesenchymal confluence thus forms at the midline. [22],[23],[34] Failure of shelf fusion is the most common type of cleft palate defect documented in animal studies. Mutations in Msx1 and Lhx8 and conditional inactivation of Tgfbr2 in CNC cells or Shh in the epithelium all result in retarded palatal shelf development. [45]

In many transgenic animals, the palatal shelves fail to meet at the midline because of hindrance by the tongue. This is usually associated with cases when the lower jaw does not move forward and downward during development, keeping the tongue between the palatal shelves. These secondary defects were evident in the Hand2 mutant mice in which the enhancer driving the expression of the gene in the pharyngeal arches was inactivated by targeted mutagenesis. [66] In these mice, the mandible did not grow properly, blocking the descent of the tongue, thus hindering palate fusion. [66],[67]

Persistence of middle edge epithelium

Adhesion of the opposing MEE is an important event in both human and mouse embryos. [21],[27],[34],[44],[68] E-cadherin is expressed in the epithelia covering the fronto-nasal and medial nasal processes as well as during the different stages of palate development, including the epithelial islands, remnants of the MES. [69],[70],[71] Mutations of CDH1/E cadherin, which deletes the extracellular cadherin repeat domains required for cell-cell adhesion, have recently been associated with CL/P in families with hereditary diffuse cancer. [72] E-cadherins are known to form dimers, indicating that the mutant proteins may have trans-dominant negative effects over the normal proteins. [72]

Extensive efforts have been made to elucidate the role of Tgfβ3 during palatal fusion. [73],[74],[75],[76] Adhesion of the MEE upon palatal shelf contact is a necessary step for fusion. Tgfβ3 is expressed in the MEE before and during fusion, and mediates MEE adhesion of the opposing palatal shelves through filopodia. E-cadherin is required for fusion, whereas filopodia seem to be crucial for proper alignment and guidance of cell sheets that are fated to fuse, but not for fusion itself. [77] Tgfβ3 is implicated in controlling the re-modelling of the extracellular matrix through regulation of the expression of the matrix metaloproteinases (Mmps) Mmp13, Mmp2 and the tissue inhibitor of metaloproteinase-2 (Timp). [78] Tgfβ3 signalling functions in the MEE by mediating the epithelial-masenchymal interactions leading to tissue changes that regulate palatal fusion. For example, EMT of the MES has been proposed as the major mechanism underlying the disappearance of the MES to generate mesenchyme continuity, thus preventing palatal clefts. [34] The establishment of the concept of EMT as the prevailing mechanism of MES disappearance led to studies attributing roles to different molecules, including Tgfβ3, Lef1, Smad, RhoA, phosphatidylinositol 3-kinase (PI-3 kinase), Mmps Twist and Snail. [22],[33],[79] In Tgfβ3 or Egfr mutant mice, there is an alteration of the fate of MEE cells. [80],[81] In Tgfβ3 null mutant mice, MEE cells fail to undergo apoptosis and remain along the midline, preventing normal fusion.

Ossification of the Palate

Palatal fusion signals the start of the ossification process in the anterior two-thirds of the palate to form the hard palatal tissues. This process entails the successful fusion of the three embryonic structures - lateral edges of the primary palate with the two anterior edges of the secondary palate. This process requires the synchronization of shelf movements together with the growth and withdrawal of the tongue and growth of the mandible and head. [82] Any form of disruption during the formative stages results in a pathological cleft. The same is true when ossification occurs too early. Sox9 is a gene controlling cartilage development and blocking the expression of Runx2, a transcription factor essential for osteoblast differentiation and bone formation associated with cleidocranial dysplasia. In Sox9 mutant, Runx2 expression is not repressed and ossification begins prematurely. [83] Since the palatal shelves are prematurely ossified, they cannot grow toward the midline and fail to fuse.

A wide range of studies on cranio-facial skeletal maturation has shown that the fusion of the palatal shelves along their length to form the mid-palatal (MP) suture occurs during the ossification of the maxillae and palatine bones before the mandibular condyle develops. [48],[84],[85] Ossification is observed where mesenchymal cells condense, the surrounding tissue vascularizes and the cells differentiate into osteoblasts that will form bone by mineral deposition. In this process, several growth and differentiation factors such as Bmps, core binding proteins (Cbf), Fgfs, and hedgehog (hh) proteins that interact with various signalling pathways to regulate the patterning of the undifferentiated mesenchyme, are involved. The Bmp-6 and the transcription factor Gli1 are also expressed during intra-membranous bone formation. [86],[87] As in cranio-facial sutures, the MP and trans-palatal (TP) suture osteoblasts express Tgfβ1, 2 and 3, while the suture cells express primarily Tgfβ3. [88],[89]

It has been established that cranial sutures are the growth sites for the neuro-cranium and that the dura mater provides the signalling molecules to regulate suture patency. [90] The MP and TP sutures have different morphology, so they are not in contact with the dura mater. Opperman's group hypothesized that these facial sutures are growth centres [88],[89] and that the nasal capsular cartilage produces signalling molecules to regulate the fusion of MP and TP sutures [Figure - 3]. [89] They found that the nasal cartilage maintained the TP sutures as growth sites in experiments on rat palatal organ cultures (E20) with or without nasal cartilage. They theorized that the nasal cartilage may regulate mid-facial growth. [89] Animal models have been developed to understand the aetiology and pathogenesis of orofacial clefts and the mechanisms of normal palatal ossification. The application of cyclic forces is an effective mechanical stimulus for the regulation of osteogenesis and osteoclastogenesis in the sutural growth of neonatal rats. [91] The process of tissue response and regeneration in the palato-maxillary suture under tensile forces was examined histologically and with fluorescence. A cyst-like zone appeared in the conjuncture of the bony front and the sutural connective tissue at the early stage of sutural expansion with increased proliferating osteoblasts and fibroblasts. New bone was deposited along the nasal septum and the front of the cyst until the new bone front formed and the suture restored its original morphology. [92]

The approach of utilizing MP suture expansion in mice has provided new insights into mechanical stress modulation as an important factor for the skeletal remodelling of bones and cartilage. The expansive force across the MP suture promotes both bone resorption through the activation of osteoclasts and bone formation through the increased proliferation and differentiation of the periosteal cells. [93] Similarly, the use of orthodontic wire expansion in growing rats showed that secondary cartilage can undergo chondrogenic and osteogenic differentiation in the maxillary arch. Interestingly, these induced changes were attributed to the alteration of the differentiation pathway of progenitor cells from chondroblastic to osteoblastic, in which many sutures temporarily form secondary cartilage during early development. Histological observations at days 7, 10, and 14 indicated that intra-membranous bone formation, which is partially recognized as mature bone, [94] occurred at the boundary between the pre-cartilaginous and cartilaginous cell layers where the calcified matrix was positive for osteocalcin antibody. The cellular events taking place at the MP suture cartilage in rat models as a result of expansion force have been observed as endochondral bone formation at the boundary between the maxillary bone and cartilage, whereas intra-membranous osteogenesis has appeared at the internal side of the cartilaginous layer. [95] To stimulate new bone formation in defective tissues, rat organ cultures with distracted palatal sutures were treated with Bmp-7 and Nell-1 for 8 days in vitro. The presence of Nell-1 increased chondrocyte hypertrophy and endochondral bone formation while Bmp-7 enhanced both chondrocyte proliferation and differentiation in the distracted palates of four-week-old male rats. This study indicates that Nell-1 is involved in the rapid osteoblast differentiation in palate sutures. [96] In another study, the application of TGF-β1 during the early stages of rat MP expansion induced rapid bone formation at the suture site. [97]

Oral and Palatal Musculature and Related Deformities

Overt CL/P encompasses a broad spectrum of defects, ranging from so-called microform clefts to complete unilateral or bilateral clefts of the lip and palate. The orbicularis oris (OO) muscle consists of numerous differently oriented strata of muscular fibres that surround the orifice of the mouth. At approximately seven weeks post-conception (p.c.) in humans, the two maxillary prominences fuse with the medial nasal prominence; however, lip fusion is not complete until the epithelial seam disappears through EMT and/or apoptosis [82] [Figure - 2]A-C. By eight weeks p.c., a dense, continuous band of mesenchymal cells corresponding to the future OO muscle can be seen, with discernible OO muscle fibers present by 12 weeks. [98],[104] The complete OO muscle architecture forms by 16 weeks. Any delay in fusion may result in sub-epithelial OO defects, such as the altered migration of the mesenchymal cells. Sub-epithelial (non-visible) defects of the orbicularis oris muscle represent the mildest form of cleft lip, and such defects are part of the phenotypic spectrum of CL/P. This defect usually is visualized as a ridge of tissue resembling a scar on the upper lip along the philtrum. [98]

Histological studies have demonstrated that such defects extend to the muscle fibres of the superior OO muscle. A method using high-resolution ultrasonography (USG) was developed to visualize the OO muscle non-invasively. [99] Significant differences in the defects of the OO are found in the first-degree relatives of CL/P individuals and controls. The OO muscle defect detected by ultrasound is consistent with the histological examination of cadavers. [99] Interestingly, the Bmp4 knockout mouse model shows bilateral cleft lip at E14.5, although this condition occurs at a rate of 22% after birth, [100] suggesting the initial cleft lip is rescued or healed in utero, leaving only the subepithelial OO defect. Potential mutations in BMP4 were found in two individuals with OO defects and none in the controls. [101] The strong evidence that OO discontinuities are indeed part of the phenotypic spectrum of CL/P provides an important clue for the clinical recurrence risk estimation for families with members affected with CL/P.

The mildest form of CP is termed a "submucosal cleft palate," described as a bifid uvula, palatal muscle diastasis and a notch in the posterior surface of the hard palate. [102] Defects in the nasopharyngeal anatomy and/or physiology may lead to velopharyngeal incompetence (VPI). Although most VPI is caused by CP, the population prevalence of VPI due to other causes is estimated to be approximately 2.5%. [103] In such cases, VPI may be caused by submucosal muscular defects of the levator veli palatini or musculus uvulae. Most of the soft palate muscles are derived from myotome cells, which first invade pharyngeal arch 4 and then migrate to the palate, carrying their innervations from the vagus nerve. One muscle (tensor veli palatini) is derived from myotome cells that first invade arch 1 and are innervated by the trigeminal nerve. [104] In the mouse, the tensor veli palatine, levator veli palatini, medial pterygoid, and lateral pterygoid muscles are identified as myogenic fields as early as gestational day 15. The palatoglossus, palatopharyngeus, and musculus uvulae, however, are not clearly visible. [105] In principle, the presence of these anatomical features in unaffected individuals may signify an elevated risk for producing clefts in offspring. [106]

Sumo Modification of Signalling Pathways in Palatogenesis

The molecular understanding of NS CL/P is further complicated when one considers that large differences in penetrance often occur when the same mutations are placed on different mouse strains, indicating a potential role for both genetic and/or environmental modifiers in the pathogenesis of CL/P. Several lines of evidence point to the involvement of the small ubiquitin-like modifier (SUMO) posttranslational modification machinery. [107] A surprisingly specific role in oro-facial development has been revealed for protein modification by the SUMO, which might hint at a possible interaction with environmental factors. Small ubiquitin-related modifiers belong to the ubiquitin-related protein family, and SUMO proteins are ubiquitously expressed throughout the eukaryotic kingdom. [108] SUMO1 shows strong expression in the MEE of the secondary palate. [109] A translocation breakpoint interrupting SUMO1 was found in a patient with CLP. [109] The causative nature of the translocation defect has been confirmed in SUMO1-deficient mice having a distinct CP phenotype. [109] Furthermore, it was recently shown that mutations in TBX22 have a profound effect on its ability to be "sumoylated," which is at least partially responsible for its loss of function. [110] Other SUMO targets include Smad4, Msx1, p63, Pax9, Eya1 and FGF signalling. [107] It seems likely that some of these factors may manifest through the disturbance of the SUMO pathway. De-stabilizing the normal balance of expression and activity for genes such as TBX22, MSX1, SATB2, and P63 during early pregnancy is likely to provide a high-risk environment for the occurrence of CL/P. Elucidating the relationship among environmental factors, the SUMO pathway, and the networks of craniofacial genes influenced by this post-transcriptional modification may be crucial to our understanding of the idiopathic forms of oro-facial clefts.

A-P Gradient of Molecular Signalling in Palatal Development

Multiple genes are critical for the development of the anterior region of the palate. Msx1, Bmp4, Bmp2, Fgf10, and Shox2 have restricted expression patterns in the anterior region of the palate. [45] In addition to the differential gene expression patterns along the A-P axis of the developing palate, there is also mesenchymal heterogeneity between the medial and lateral regions of the palatal shelf. The odd, skipped related genes Osr1 and Osr2 are expressed in a medial-lateral gradient in the palatal shelf. The mutation of the Osr2 gene results in the compromised development of the medial aspect of the palatal shelf and retards palatal shelf elevation. [60],[111] The expression of Fgfr2 is focused on the medial aspect of the developing palatal shelf, suggesting a possible functional significance in regulating its development and elevation.

An important discovery has been the confirmation of genetic heterogeneity along the anterior-posterior and medial-lateral axes of the developing palate. [48] This heterogeneity may provide a differential regulatory mechanism for the fusion of the anterior vs. posterior region of the palate. MEE cells undergo apoptosis at different times during palatal fusion. It has been shown that the apoptosis of MEE cells is triggered by palatal shelf contact in the anterior region, whereas it is initiated before any contact between the opposing shelves in the posterior region. [38] This difference may be the result of dissimilar molecular signals in the palatal mesenchyme along the anteroposterior axis that instruct different fates to the palatal epithelium. [112] Recent studies have demonstrated that constant and reciprocal interactions between palatal epithelium and CNC-derived mesenchyme are responsible for setting up this genetic heterogeneity along the AP axis and are crucial for normal palatal development and fusion. [25],[45],[113] The specific gene expression patterns in the posterior region of the palatal mesenchyme are less understood. Fgfr2 is expressed in the epithelium, and the CNC-derived mesenchyme is found in the middle and posterior palate. FGF8 signalling selectively induces the expression of Pax9 in the posterior region of the palatal mesenchyme. The loss of Pax9 results in a palatal shelf development defect and a cleft palate [48],[58]

Conclusion

It is clear from this and other review of literature that CL/P is caused by many factors, including both genes and environment. Gene targeting technology and basic conventional techniques using animal models led to the identification of genes associated with known and unknown aetiologic factors. In some cases, the human gene deficiency was identified first and replicated in an animal model, but in other cases, animal models led the way to understand gene/environment interactions. It is also clear from this extensive list of possible contributing genes that the molecular and cellular interactions associated with CL/P are not all understood. Fortunately, some subclinical changes in facial features may lead to a greater understanding of the gene/environment interactions in cranio-facial development.[186]

Acknowledgments

We thank Dr. Lynne Opperman for discussions about palate ossification and for permitting the use of a figure from one of her publications on palatal suture development. We thank Jeanne Santa Cruz for editorial assistance. We also thank the publishers for permission to reprint figures from previous papers. The gene tables are an ongoing project.

References

1.Ferguson MW. Palate development. Develop 1988;103:41-60.  Back to cited text no. 1    
2.Christensen K, Juel K, Herskind AM, Murray JC. Long term follow up study of survival associated with cleft lip and palate at birth. BMJ 2004;328:1405.  Back to cited text no. 2    
3.Fogh-Andersen P. Epidemiology and etiology of clefts. In: Bergsma D, editor. Birth defects: Original article series. Baltimore: Williams and Wilkins Co.; 1971.  Back to cited text no. 3    
4.Slavkin HC. Incidence of cleft lips, palates rising. J Am Dent Assoc 1992;123:61-5.  Back to cited text no. 4    
5.Murray JC. Gene/environment causes of cleft lip and/or palate. [review] [122 refs]. Clinical Genetics 2002;61:248-56.  Back to cited text no. 5    
6.Gritli-Linde A. The etiopathogenesis of cleft lip and cleft palate usefulness and caveats of mouse models. Curr Top Dev Biol 2008;84:37-138.  Back to cited text no. 6    
7.Gritli-Linde A. Molecular control of secondary palate development. Developmental Biology 2007; 301:309-26.  Back to cited text no. 7    
8.Carinci F, Pezzetti F, Scapoli L, Martinelli M, Carinci P, Tognon M. Genetics of nonsyndromic cleft lip and palate: A review of international studies and data regarding the Italian population. Cleft Palate Craniofac J 2000; 37:33-40.  Back to cited text no. 8    
9.Chai Y, Maxson RE, Jr. Recent advances in craniofacial morphogenesis. Dev Dyn 2006;235:2353-75.  Back to cited text no. 9    
10.Wyszynski DF, Duffy DL, Beaty TH. Maternal cigarette smoking and oral clefts: A meta-analysis. Cleft Palate Craniofac J 1997;34:206-10.  Back to cited text no. 10    
11.Shaw GM, Lammer EJ, Zhu H, Baker MW, Neri E, Finnell RH. Maternal periconceptional vitamin use, genetic variation of infant reduced folate carrier (a80g), and risk of spina bifida. Am J Med Genet 2002;108:1-6.   Back to cited text no. 11    
12.Boot MJ, Steegers-Theunissen RP, Poelmann RE, Van Iperen L, Lindemans J, Gittenberger-de Groot AC. Folic acid and homocysteine affect neural crest and neuroepithelial cell outgrowth and differentiation in vitro. Dev Dyn 2003;227:301-8.  Back to cited text no. 12    
13.Briggs RM. Vitamin supplementation as a possible factor in the incidence of cleft lip/palate deformities in humans. Clin Plast Surg 1976;3:647-52.  Back to cited text no. 13    
14.Finnell RH, Shaw GM, Lammer EJ, Brandl KL, Carmichael SL, Rosenquist TH. Gene-nutrient interactions: Importance of folates and retinoids during early embryogenesis. Toxicol Appl Pharmacol 2004;198:75-85.  Back to cited text no. 14    
15.Itikala PR, Watkins ML, Mulinare J, Moore CA, Liu Y. Maternal multivitamin use and orofacial clefts in offspring. Teratology 2001; 63:79-86.  Back to cited text no. 15    
16.Lammer EJ, Shaw GM, Iovannisci DM, Finnell RH. Periconceptional multivitamin intake during early pregnancy, genetic variation of acetyl-n-transferase 1 (nat1), and risk for orofacial clefts. Birth Defects Res A Clin Mol Teratol 2004;70:846-52.  Back to cited text no. 16    
17.Zhu H, Curry S, Wen S, Wicker NJ, Shaw GM, Lammer EJ, et al. Are the betaine-homocysteine methyltransferase (bhmt and bhmt2) genes risk factors for spina bifida and orofacial clefts? Am J Med Genet A 2005;135:274-7.  Back to cited text no. 17    
18.Tang LS, Santillano DR, Wlodarczyk BJ, Miranda RC, Finnell RH. Role of folbp1 in the regional regulation of apoptosis and cell proliferation in the developing neural tube and craniofacies. Am J Med Genet C Semin Med Genet 2005;135:48-58.  Back to cited text no. 18    
19.Lammer EJ, Shaw GM, Iovannisci DM, Van Waes J, Finnell RH. Maternal smoking and the risk of orofacial clefts: Susceptibility with nat1 and nat2 polymorphisms. Epidemiology 2004;15:150-6.  Back to cited text no. 19    
20.Shaw GM, Zhu H, Lammer EJ, Yang W, Finnell RH. Genetic variation of infant reduced folate carrier (a80g) and risk of orofacial and conotruncal heart defects. Am J Epidemiol 2003;158:747-52.  Back to cited text no. 20    
21.Hay ED. An overview of epithelio-mesenchymal transformation. Acta Anat 1995;154:8-20.  Back to cited text no. 21    
22.Kang P, Svoboda KK. Epithelial-mesenchymal transformation during craniofacial development. J Dent Res 2005;84:678-90.  Back to cited text no. 22    
23.Nawshad A. Palatal seam disintegration: To die or not to die? That is no longer the question. Dev Dyn 2008;237:2643-56.  Back to cited text no. 23    
24.Shuler CF. Programmed cell death and cell transformation in craniofacial development. Crit Rev Oral Biol Med 1995;6:202-17.  Back to cited text no. 24    
25.Murray JC, Schutte BC. Cleft palate: Players, pathways, and pursuits. J Clin Invest 2004;113:1676-8.  Back to cited text no. 25    
26.Yu W, Ruest L, Svoboda K. Regulation of epithelial-mesenchymal transition in palatal fusion. Exp Biol Med (Maywood) 2009; 234:483-91.  Back to cited text no. 26    
27.Britto JA, Evans RD, Hayward RD, Jones BM. Toward pathogenesis of Apert cleft palate: Fgf, fgfr, and tgf beta genes are differentially expressed in sequential stages of human palatal shelf fusion. Cleft Palate Craniofac J 2002;39:332-40.  Back to cited text no. 27    
28.Shi M, Wehby GL, Murray JC. Review on genetic variants and maternal smoking in the etiology of oral clefts and other birth defects. Birth Defects Res C Embryo Today 2008;84:16-29.  Back to cited text no. 28    
29.Shuler CF, Halpern DE, Guo Y, Sank AC. Medial edge epithelium fate traced by cell lineage analysis during epithelial-mesenchymal transformation in vivo. Dev Biol 1992;154:318-30.  Back to cited text no. 29    
30.Hay ED. An overview of epithelio-mesenchymal transformation. Acta Anat (Basel) 1995;154:8-20.  Back to cited text no. 30    
31.Kaartinen V, Cui XM, Heisterkamp N, Groffen J, Shuler CF. Transforming growth factor-beta3 regulates transdifferentiation of medial edge epithelium during palatal fusion and associated degradation of the basement membrane. Dev Dyn 1997;209:255-60.  Back to cited text no. 31    
32.Kang Y, Massague J. Epithelial-mesenchymal transitions: Twist in development and metastasis. Cell 2004;118:277-9.  Back to cited text no. 32    
33.Nawshad A, Hay ED. Tgfbeta3 signaling activates transcription of the lef1 gene to induce epithelial mesenchymal transformation during mouse palate development. J Cell Biol 2003;163:1291-301.  Back to cited text no. 33    
34.Nawshad A, LaGamba D, Hay ED. Transforming growth factor beta (TGFbeta) signalling in palatal growth, apoptosis and epithelial mesenchymal transformation (EMT). Arch Oral Biol 2004;49:675-89.  Back to cited text no. 34    
35.Sun W, Vincent S, Settleman J, Johnson GL. Mek kinase 2 binds and activates protein kinase c-related kinase 2. Bifurcation of kinase regulatory pathways at the level of an mapk kinase kinase. J Biol Chem 2000;275:24421-8.  Back to cited text no. 35    
36.LaGamba D, Nawshad A, Hay ED. Microarray analysis of gene expression during epithelial-mesenchymal transformation. Dev Dyn 2005;234:132-42.  Back to cited text no. 36    
37.Cuervo R, Covarrubias L. Death is the major fate of medial edge epithelial cells and the cause of basal lamina degradation during palatogenesis. Develop 2004;131:15-24.  Back to cited text no. 37    
38.Cuervo R, Valencia C, Chandraratna RA, Covarrubias L. Programmed cell death is required for palate shelf fusion and is regulated by retinoic acid. Dev Biol 2002;245:145-56.  Back to cited text no. 38    
39.Mori C, Nakamura N, Okamoto Y, Osawa M, Shiota K. Cytochemical identification of programmed cell death in the fusing fetal mouse palate by specific labelling of DNA fragmentation. Anat Embryol (Berl) 1994;190:21-8.  Back to cited text no. 39    
40.Taniguchi K, Sato N, Uchiyama Y. Apoptosis and heterophagy of medial edge epithelial cells of the secondary palatine shelves during fusion. Arch Histol Cytol 1995;58:191-203.  Back to cited text no. 40    
41.Carette MJ, Ferguson MW. The fate of medial edge epithelial cells during palatal fusion in vitro: An analysis by dii labelling and confocal microscopy. Development 1992;114:379-88.  Back to cited text no. 41    
42.Jin J-Z, Ding J. Analysis of cell migration, transdifferentiation and apoptosis during mouse secondary palate fusion. Development 2006;133:3341-7.  Back to cited text no. 42    
43.Martinez-Alvarez C, Blanco MJ, Perez R, Rabadan MA, Aparicio M, Resel E, et al. Snail family members and cell survival in physiological and pathological cleft palates. Dev Biol 2004;265:207-18.  Back to cited text no. 43    
44.Sun D, Baur S, Hay ED. Epithelial-mesenchymal transformation is the mechanism for fusion of the craniofacial primordia involved in morphogenesis of the chicken lip. Dev Biol 2000;228:337-49.  Back to cited text no. 44    
45.Rice R, Spencer-Dene B, Connor EC, Gritli-Linde A, McMahon AP, Dickson C, et al. Disruption of fgf10/fgfr2b-coordinated epithelial-mesenchymal interactions causes cleft palate.[see comment]. J Clin Invest 2004;113:1692-700.  Back to cited text no. 45    
46.Rice R, Thesleff I, Rice DP. Regulation of Twist, Snail, and Id1 is conserved between the developing murine palate and tooth. Dev Dyn 2005;234:28-35.  Back to cited text no. 46    
47.Nie X, Luukko K, Kettunen P. FGF signalling in craniofacial development and developmental disorders. Oral Dis 2006;12:102-11.  Back to cited text no. 47    
48.Hilliard SA, Yu L, Gu S, Zhang Z, Chen YP. Regional regulation of palatal growth and patterning along the anterior-posterior axis in mice. J Anat 2005;207:655-67.  Back to cited text no. 48    
49.Yu L, Gu S, Alappat S, Song Y, Yan M, Zhang X, et al. Shox2-deficient mice exhibit a rare type of incomplete clefting of the secondary palate. Development 2005;132:4397-406.  Back to cited text no. 49    
50.Alappat SR, Zhang Z, Suzuki K, Zhang X, Liu H, Jiang R, et al. The cellular and molecular etiology of the cleft secondary palate in fgf10 mutant mice. Dev Biol 2005;277:102-13.  Back to cited text no. 50    
51.Braybrook C, Doudney K, Marcano AC, Arnason A, Bjornsson A, Patton MA, et al. The t-box transcription factor gene tbx22 is mutated in x-linked cleft palate and ankyloglossia. Nat Genet 2001;29:179-83.  Back to cited text no. 51    
52.Marcano AC, Doudney K, Braybrook C, Squires R, Patton MA, Lees MM, et al. Tbx22 mutations are a frequent cause of cleft palate. J Med Genet 2004;41: 68-74.  Back to cited text no. 52    
53.Wong FK, Hagg U. An update on the aetiology of orofacial clefts. Hong Kong Med J 2004;10:331-6.  Back to cited text no. 53    
54.Ferguson MW. Palatal shelf elevation in the wistar rat fetus. J Anat 1978;125:555-77.  Back to cited text no. 54    
55.Brinkley LL, Morris-Wiman J, Brinkley LL, Morris-Wiman J. The role of extracellular matrices in palatal shelf closure. Curr Top Dev Biol 1984;19:17-36.  Back to cited text no. 55    
56.Brinkley LL, Morris-Wiman J, Brinkley LL, Morris-Wiman J. Effects of chlorcyclizine-induced glycosaminoglycan alterations on patterns of hyaluronate distribution during morphogenesis of the mouse secondary palate. Development 1987;100:637-40.  Back to cited text no. 56    
57.Kist R, Greally E, Peters H. Derivation of a mouse model for conditional inactivation of pax9. Genesis 2007;45:460-4.  Back to cited text no. 57    
58.Peters H, Neubuser A, Kratochwil K, Balling R. Pax9-deficient mice lack pharyngeal pouch derivatives and teeth and exhibit craniofacial and limb abnormalities. Genes Dev 1998;12:2735-47.  Back to cited text no. 58    
59.Szeto DP, Rodriguez-Esteban C, Ryan AK, O'Connell SM, Liu F, Kioussi C, et al. Role of the bicoid-related homeodomain factor pitx1 in specifying hindlimb morphogenesis and pituitary development. Genes Dev 1999;13:484-94.  Back to cited text no. 59    
60.Gao Y, Lan Y, Ovitt CE ,Jiang R. Functional equivalence of the zinc finger transcription factors osr1 and osr2 in mouse development. Dev Biol 2009;328; 200-9.  Back to cited text no. 60    
61.Wee EL, Zimmerman EF. Involvement of GABA in palate morphogenesis and its relation to diazepam teratogenesis in two mouse strains. Teratology 1983;28:15-22.  Back to cited text no. 61    
62.Scapoli L, Martinelli M, Pezzetti F, Carinci F, Bodo M, Tognon M, et al. Linkage disequilibrium between gabrb3 gene and nonsyndromic familial cleft lip with or without cleft palate. Hum Genet 2002;110: 15-20.  Back to cited text no. 62    
63.Varju P, Katarova Z, Madarasz E, Szabo G. GABA signalling during development: New data and old questions. Cell Tissue Res 2001;305:239-46.  Back to cited text no. 63    
64.Ding R, Tsunekawa N, Obata K. Cleft palate by picrotoxin or 3-mp and palatal shelf elevation in GABA-deficient mice. Neurotoxicol Teratol 2004;26:587-92.  Back to cited text no. 64    
65.Condie BG, Bain G, Gottlieb DI, Capecchi MR. Cleft palate in mice with a targeted mutation in the gamma-aminobutyric acid-producing enzyme glutamic acid decarboxylase 67. Proc Natl Acad Sci U S A 1997;94:11451-5.  Back to cited text no. 65    
66.Yanagisawa H, Clouthier DE, Richardson JA, Charite J, Olson EN. Targeted deletion of a branchial arch-specific enhancer reveals a role of hand in craniofacial development. Development 2003;130:1069-78.  Back to cited text no. 66    
67.Barbosa AC, Funato N, Chapman S, McKee MD, Richardson JA, Olson EN, et al. Hand transcription factors cooperatively regulate development of the distal midline mesenchyme. Dev Biol 2007;310:154-68.  Back to cited text no. 67    
68.Fitchett JE, Hay ED. Medial edge epithelium transforms to mesenchyme after embryonic palatal shelves fuse. Dev Biol 1989;131:455-74.  Back to cited text no. 68    
69.Montenegro MA, Rojas M, Dominguez S, Vergara A. Cytokeratin, vimentin and e-cadherin immunodetection in the embryonic palate in two strains of mice with different susceptibility to glucocorticoid-induced clefting. J Craniofac Genet Dev Biol 2000;20:137-43.  Back to cited text no. 69    
70.Vaziri Sani F, Hallberg K, Harfe BD, McMahon AP, Linde A, Gritli-Linde A. Fate-mapping of the epithelial seam during palatal fusion rules out epithelial-mesenchymal transformation. Dev Biol 2005;285:490-5.  Back to cited text no. 70    
71.Yu W, Kamara H, Svoboda KH. The role of twist during palate development. Dev Dyn 2008;237:2716-25.  Back to cited text no. 71    
72.Frebourg T, Oliveira C, Hochain P, Karam R, Manouvrier S, Graziadio C, et al. Cleft lip/palate and cdh1/e-cadherin mutations in families with hereditary diffuse gastric cancer. J Med Genet 2006;43:138-42.  Back to cited text no. 72    
73.Cui XM, Shiomi N, Chen J, Saito T, Yamamoto T, Ito Y et al. Overexpression of smad2 in tgf-beta3-null mutant mice rescues cleft palate. Dev Biol 2005;278; 193-202.  Back to cited text no. 73    
74.Fitzpatrick DR, Denhez F, Kondaiah P, Akhurst RJ. Differential expression of tgf beta isoforms in murine palatogenesis. Development 1990;109:585-95.  Back to cited text no. 74    
75.Pelton RW, Dickinson ME, Moses HL, Hogan BL. In situ hybridization analysis of tgf beta 3 rna expression during mouse development: Comparative studies with tgf beta 1 and beta 2. Development 1990;110:609-20.  Back to cited text no. 75    
76.Pelton RW, Hogan BL, Miller DA, Moses HL. Differential expression of genes encoding tgfs beta 1, beta 2, and beta 3 during murine palate formation. Dev Biol 1990;141:456-60.  Back to cited text no. 76    
77.Schock F, Perrimon N. Molecular mechanisms of epithelial morphogenesis. Annu Rev Cell Dev Biol 2002;18:463-93.  Back to cited text no. 77    
78.Blavier L, Lazaryev A, Groffen J, Heisterkamp N, DeClerck YA, Kaartinen V. Tgf-beta3-induced palatogenesis requires matrix metalloproteinases. Mol Biol Cell 2001;12:1457-66.  Back to cited text no. 78    
79.Kaartinen V, Haataja L, Nagy A, Heisterkamp N, Groffen J. TGFbeta3-induced activation of rhoa/rho-kinase pathway is necessary but not sufficient for epithelio-mesenchymal transdifferentiation: Implications for palatogenesis. Int J Mol Med 2002;9:563-70.  Back to cited text no. 79    
80.Kaartinen V, Voncken JW, Shuler C, Warburton D, Bu D, Heisterkamp N, et al. Abnormal lung development and cleft palate in mice lacking tgf-beta 3 indicates defects of epithelial-mesenchymal interaction. Nat Genet 1995;11:415-21.  Back to cited text no. 80    
81.Miettinen PJ, Chin JR, Shum L, Slavkin HC, Shuler CF, Derynck R, et al. Epidermal growth factor receptor function is necessary for normal craniofacial development and palate closure. Nature Genetics 1999;22:69-73.  Back to cited text no. 81    
82.Sperber G, Craniofacial development. Hamilton and London: BC Decker Inc; 2001.  Back to cited text no. 82    
83.Mori-Akiyama Y, Akiyama H, Rowitch DH, de Crombrugghe B. Sox9 is required for determination of the chondrogenic cell lineage in the cranial neural crest. Proc Natl Acad Sci U S A 2003;100:9360-5.  Back to cited text no. 83    
84.Kjaer I. Human prenatal palatal shelf elevation related to craniofacial skeletal maturation. Eur J Orthod 1992;14:26-30.  Back to cited text no. 84    
85.Okano J, Suzuki S, Shiota K. Regional heterogeneity in the developing palate: Morphological and molecular evidence for normal and abnormal palatogenesis. Congenit Anom (Kyoto) 2006;46:49-54.  Back to cited text no. 85    
86.Kerrigan JJ, Mansell JP, Sengupta A, Brown N, Sandy JR. Palatogenesis and potential mechanisms for clefting. J R Coll Surg Edinb 2000;45:351-8.  Back to cited text no. 86    
87.Iwasaki M, Le AX, Helms JA. Expression of Indian hedgehog, bone morphogenetic protein 6 and gli during skeletal morphogenesis. 1997;69:197-202.  Back to cited text no. 87    
88.Adab K, Sayne JR, Carlson DS ,Opperman LA. Tgf-beta1, tgf-beta2, tgf-beta3 and msx2 expression is elevated during frontonasal suture morphogenesis and during active postnatal facial growth. Orthod Craniofac Res 2002;5; 227-37   Back to cited text no. 88    
89.Adab K, Sayne JR, Carlson DS, Opperman LA. Nasal capsular cartilage is required for rat transpalatal suture morphogenesis. Differentiation 2003;71:496-505.  Back to cited text no. 89    
90.Opperman LA. Cranial sutures as intramembranous bone growth sites. Dev Dyn 2000;219:472-85.  Back to cited text no. 90    
91.Vij K, Mao JJ. Geometry and cell density of rat craniofacial sutures during early postnatal development and upon in vivo cyclic loading. Bone 2006;38:722-30.  Back to cited text no. 91    
92.Liu C, Song R, Song Y. A serial histological study on suture expansion osteogenesis for cleft palate closure. Zhonghua Zheng Xing Wai Ke Za Zhi 2000;16:43-5.  Back to cited text no. 92    
93.Hou B, Fukai N, Olsen BR. Mechanical force-induced midpalatal suture remodeling in mice. Bone 2007;40:1483-93.  Back to cited text no. 93    
94.Takahashi I, Mizoguchi I, Nakamura M, Sasano Y, Saitoh S, Kagayama M, et al. Effects of expansive force on the differentiation of midpalatal suture cartilage in rats. Bone 1996;18:341-8.  Back to cited text no. 94    
95.Kobayashi ET, Hashimoto F, Kobayashi Y, Sakai E, Miyazaki Y, Kamiya T, et al. Force-induced rapid changes in cell fate at midpalatal suture cartilage of growing rats. J Dent Res 1999;78: 1495-504.  Back to cited text no. 95    
96.Cowan CM, Cheng S, Ting K, Soo C, Walder B, Wu B, et al. Nell-1 induced bone formation within the distracted intermaxillary suture. Bone 2006;38:48-58.  Back to cited text no. 96    
97.Sawada M, Shimizu N. Stimulation of bone formation in the expanding mid-palatal suture by transforming growth factor-beta 1 in the rat. Eur J Orthod 1996;18:169-79.  Back to cited text no. 97    
98.Akita S, Hirano A. Surgical modifications for microform cleft lip repairs. J Craniofac Surg 2005;16:1106-10.  Back to cited text no. 98    
99.Neiswanger K, Weinberg SM, Rogers CR, Brandon CA, Cooper ME, Bardi KM, et al. Orbicularis oris muscle defects as an expanded phenotypic feature in nonsyndromic cleft lip with or without cleft palate. Am J Med Genet A 2007;143:1143-9.  Back to cited text no. 99    
100.Jiang R, Bush JO, Lidral AC. Development of the upper lip: Morphogenetic and molecular mechanisms. Dev Dyn 2006;235:1152-66.  Back to cited text no. 100    
101.Marazita ML. Subclinical features in non-syndromic cleft lip with or without cleft palate (cl/p): Review of the evidence that subepithelial orbicularis oris muscle defects are part of an expanded phenotype for cl/p. Orthod Craniofac Res 2007;10:82-7.  Back to cited text no. 101    
102.Gosain AK, Conley SF, Marks S, Larson DL. Submucous cleft palate: Diagnostic methods and outcomes of surgical treatment. Plast Reconstr Surg 1996;97:1497-509.  Back to cited text no. 102    
103.Boorman JG, Varma S, Ogilvie CM. Velopharyngeal incompetence and chromosome 22q11 deletion. Lancet 2001;357:774.  Back to cited text no. 103    
104.Sweeney L. Basic concepts in embryology: A student's survival guide. McGraw-Hill Professional; 1998. p. 443.  Back to cited text no. 104    
105.Trotman CA, Hou D, Burdi AR, Cohen SR, Carlson DS. Histomorphologic analysis of the soft palate musculature in prenatal cleft and noncleft a/jax mice. Cleft Palate Craniofac J 1995;32:455-62.  Back to cited text no. 105    
106.Weinberg SM, Neiswanger K, Martin RA, Mooney MP, Kane AA, Wenger SL, et al. The Pittsburgh oral-facial cleft study: Expanding the cleft phenotype. Background and justification. Cleft Palate Craniofac J 2006;43:7-20.  Back to cited text no. 106    
107.Pauws E, Stanier P. Fgf signalling and sumo modification: New players in the aetiology of cleft lip and/or palate. Trends Genet 2007;23:631-40.  Back to cited text no. 107    
108.Zhang FP, Mikkonen L, Toppari J, Palvimo JJ, Thesleff I, Janne OA. Sumo-1 function is dispensable in normal mouse development. Mol Cell Biol 2008;28:5381-90.  Back to cited text no. 108    
109.Alkuraya FS, Saadi I, Lund JJ, Turbe-Doan A, Morton CC, Maas RL. Sumo1 haploinsufficiency leads to cleft lip and palate. Science 2006;313:1751.  Back to cited text no. 109    
110.Andreou AM, Pauws E, Jones MC, Singh MK, Bussen M, Doudney K, et al. Tbx22 missense mutations found in patients with x-linked cleft palate affect DNA binding, sumoylation, and transcriptional repression. Am J Hum Genet 2007;81:700-12.  Back to cited text no. 110    
111.Lan Y, Ovitt CE, Cho ES, Maltby KM, Wang Q, Jiang R. Odd-skipped related 2 (osr2) encodes a key intrinsic regulator of secondary palate growth and morphogenesis. Development 2004;131:3207-16.  Back to cited text no. 111    
112.Ferguson MW, Honig LS, Slavkin HC. Differentiation of cultured palatal shelves from alligator, chick, and mouse embryos. Anat Rec 1984;209:231-49.  Back to cited text no. 112    
113.Zhang Z, Song Y, Zhao X, Zhang X, Fermin C, Chen Y. Rescue of cleft palate in msx1-deficient mice by transgenic bmp4 reveals a network of bmp and shh signaling in the regulation of mammalian palatogenesis. Development 2002;129:4135-46.  Back to cited text no. 113    
114.Kreiborg S, Cohen MM Jr. The oral manifestations of Apert syndrome. J Craniofac Genet Dev Biol 1992;12:41-8.  Back to cited text no. 114    
115.Martelli H Jr, Paranaiba LM, de Miranda RT, Orsi J Jr, Coletta RD. Apert syndrome: Report of a case with emphasis on craniofacial and genetic features. Pediatr Dent 2008;30:464-8.  Back to cited text no. 115    
116.Park WJ, Theda C, Maestri NE, Meyers GA, Fryburg JS, Dufresne C, et al. Analysis of phenotypic features and fgfr2 mutations in apert syndrome. Am J Hum Genet 1995;57:321-8.  Back to cited text no. 116    
117.Wilkie AO, Slaney SF, Oldridge M, Poole MD, Ashworth GJ, Hockley AD, et al. Apert syndrome results from localized mutations of fgfr2 and is allelic with Crouzon syndrome. Nat Genet 1995;9:165-72.  Back to cited text no. 117    
118.Moloney DM, Slaney SF, Oldridge M, Wall SA, Sahlin P, Stenman G, et al. Exclusive paternal origin of new mutations in Apert syndrome. Nat Genet 1996;13:48-53.  Back to cited text no. 118    
119.Castanet M, Park SM, Smith A, Bost M, Leger J, Lyonnet S, et al. A novel loss-of-function mutation in ttf-2 is associated with congenital hypothyroidism, thyroid agenesis and cleft palate. Hum Mol Genet 2002;11:2051-9.  Back to cited text no. 119    
120.Clifton-Bligh RJ, Wentworth JM, Heinz P, Crisp MS, John R, Lazarus JH, et al. Mutation of the gene encoding human ttf-2 associated with thyroid agenesis, cleft palate and choanal atresia. Nat Genet 1998;19:399-401.  Back to cited text no. 120    
121.Milunsky JM, Maher TA, Zhao G, Roberts AE, Stalker HJ, Zori RT, et al. Tfap2a mutations result in branchio-oculo-facial syndrome. Am J Hum Genet 2008;82:1171-7.  Back to cited text no. 121    
122.Kallen B, Mastroiacovo P, Robert E. Major congenital malformations in Down syndrome. Am J Med Genet 1996;65:160-6.  Back to cited text no. 122    
123.Celli J, Duijf P, Hamel BC, Bamshad M, Kramer B, Smits AP, et al. Heterozygous germline mutations in the p53 homolog p63 are the cause of eec syndrome. Cell 1999;99:143-53.  Back to cited text no. 123    
124.McGrath JA, Duijf PH, Doetsch V, Irvine AD, de Waal R, Vanmolkot KR, et al. Hay-Wells syndrome is caused by heterozygous missense mutations in the sam domain of p63. Hum Mol Genet 2001;10: 221-9.  Back to cited text no. 124    
125.Abel EL. Fetal alcohol syndrome: A cautionary note. Curr Pharm Des 2006;12:1521-9.  Back to cited text no. 125    
126.Green ML, Singh AV, Zhang Y, Nemeth KA, Sulik KK, Knudsen TB. Reprogramming of genetic networks during initiation of the fetal alcohol syndrome. Dev Dyn 2007;236:613-31.  Back to cited text no. 126    
127.Seki M, Yoshida K, Kashimura M. [a case of fetal alcohol effects with orofacial cleft]. Nihon Arukoru Yakubutsu Igakkai Zasshi 2005;40:137-43.  Back to cited text no. 127    
128.Wattendorf DJ, Muenke M. Fetal alcohol spectrum disorders. Am Fam Physician 2005;72:279-82, 85.  Back to cited text no. 128    
129.Kokavec R. Goldenhar syndrome with various clinical manifestations. Cleft Palate Craniofac J 2006;43:628-34.  Back to cited text no. 129    
130.Vilkki SK, Hukki J, Nietosvaara Y, Hurmerinta K, Suominen E. Microvascular temporomandibular joint and mandibular ramus reconstruction in hemifacial microsomia. J Craniofac Surg 2002;13:809-15.  Back to cited text no. 130    
131.Fang J, Dagenais SL, Erickson RP, Arlt MF, Glynn MW, Gorski JL, et al. Mutations in foxc2 (mfh-1), a forkhead family transcription factor, are responsible for the hereditary lymphedema-distichiasis syndrome. Am J Hum Genet 2000;67:1382-8.  Back to cited text no. 131    
132.Dode C, Fouveaut C, Mortier G, Janssens S, Bertherat J, Mahoudeau J, et al. Novel fgfr1 sequence variants in Kallmann syndrome, and genetic evidence that the fgfr1c isoform is required in olfactory bulb and palate morphogenesis. Hum Mutat 2007;28:97-8.  Back to cited text no. 132    
133.Dode C, Levilliers J, Dupont JM, De Paepe A, Le Du N, Soussi-Yanicostas N, et al. Loss-of-function mutations in fgfr1 cause autosomal dominant Kallmann syndrome.[see comment]. Nature Genetics 2003;33:463-5.  Back to cited text no. 133    
134.Suzuki K, Hu D, Bustos T, Zlotogora J, Richieri-Costa A, Helms JA, et al. Mutations of pvrl1, encoding a cell-cell adhesion molecule/herpesvirus receptor, in cleft lip/palate-ectodermal dysplasia. Nat Genet 2000;25:427-30.  Back to cited text no. 134    
135.Prows CA, Bender PL. Beyond Pierre Robin sequence. Neonatal Netw 1999;18:13-9.  Back to cited text no. 135    
136.Cole A, Lynch P, Slator R. A new grading of Pierre Robin sequence. Cleft Palate Craniofac J 2008;45:603-6.  Back to cited text no. 136    
137.Muenke M. The pit, the cleft and the web. Nat Genet 2002;32:219-20.  Back to cited text no. 137    
138.Wassif CA, Maslen C, Kachilele-Linjewile S, Lin D, Linck LM, Connor WE, et al. Mutations in the human sterol delta7-reductase gene at 11q12-13 cause Smith-Lemli-Opitz syndrome. Am J Hum Genet 1998;63:55-62.  Back to cited text no. 138    
139.Snead MP, Yates JR. Clinical and molecular genetics of Stickler syndrome. J Med Genet 1999;36:353-9.  Back to cited text no. 139    
140.Wilkin DJ, Mortier GR, Johnson CL, Jones MC, de Paepe A, Shohat M, et al. Correlation of linkage data with phenotype in eight families with Stickler syndrome. Am J Med Genet 1998;80:121-7.  Back to cited text no. 140    
141.Dixon J, Jones NC, Sandell LL, Jayasinghe SM, Crane J, Rey JP, et al. Tcof1/treacle is required for neural crest cell formation and proliferation deficiencies that cause craniofacial abnormalities. Proc Natl Acad Sci U S A 2006;103:13403-8.  Back to cited text no. 141    
142.Valdez BC, Henning D, So RB, Dixon J, Dixon MJ. The Treacher Collins syndrome (tcof1) gene product is involved in ribosomal DNA gene transcription by interacting with upstream binding factor. Proc Natl Acad Sci U S A 2004;101:10709-14.  Back to cited text no. 142    
143.Kondo S, Schutte BC, Richardson RJ, Bjork BC, Knight AS, Watanabe Y, et al. Mutations in irf6 cause van der Woude and popliteal pterygium syndromes. Nat Genet 2002;32:285-9.  Back to cited text no. 143    
144.Cuneo BF. 22q11.2 deletion syndrome: Digeorge, velocardiofacial, and conotruncal anomaly face syndromes. Curr Opin Pediatr 2001;13:465-72.  Back to cited text no. 144    
145.Moreno F, Zuazo E, Gonzalez S, Bereciartu P. 22q11 deletion syndrome: An expanding phenotype.. Neurologia 2009;24:69-71.  Back to cited text no. 145    
146.Sanlaville D, Etchevers HC, Gonzales M, Martinovic J, Clement-Ziza M, Delezoide AL, et al. Phenotypic spectrum of CHARGE syndrome in fetuses with chd7 truncating mutations correlates with expression during human development. J Med Genet 2006;43: 211-7.  Back to cited text no. 146    
147.Vissers LE, van Ravenswaaij CM, Admiraal R, Hurst JA, de Vries BB, Janssen IM, et al. Mutations in a new member of the chromodomain gene family cause charge syndrome. Nat Genet 2004;36:955-7.  Back to cited text no. 147    
148.Schuffenhauer S, Leifheit HJ, Lichtner P, Peters H, Murken J, Emmerich P. De novo deletion (14)(q11.2q13) including pax9: Clinical and molecular findings. J Med Genet 1999;36:233-6.  Back to cited text no. 148    
149.Abidi FE, Miano MG, Murray JC, Schwartz CE. A novel mutation in the phf8 gene is associated with x-linked mental retardation with cleft lip/cleft palate. Clin Genet 2007;72:19-22.  Back to cited text no. 149    
150.Ming JE, Kaupas ME, Roessler E, Brunner HG, Golabi M, Tekin M, et al. Mutations in patched-1, the receptor for sonic hedgehog, are associated with holoprosencephaly. Hum Genet 2002;110:297-301.  Back to cited text no. 150    
151.Ribeiro LA, Murray JC, Richieri-Costa A. Ptch mutations in four Brazilian patients with holoprosencephaly and in one with holoprosencephaly-like features and normal mri. Am J Med Genet A 2006;140:2584-6.  Back to cited text no. 151    
152.Leoyklang P, Suphapeetiporn K, Siriwan P, Desudchit T, Chaowanapanja P, Gahl WA, et al. Heterozygous nonsense mutation satb2 associated with cleft palate, osteoporosis, and cognitive defects. Hum Mutat 2007;28:732-8.  Back to cited text no. 152    
153.Yagi H, Furutani Y, Hamada H, Sasaki T, Asakawa S, Minoshima S, et al. Role of tbx1 in human del22q11.2 syndrome. Lancet 2003;362:1366-73.  Back to cited text no. 153    
154.Loeys BL, Chen J, Neptune ER, Judge DP, Podowski M, Holm T, et al. A syndrome of altered cardiovascular, craniofacial, neurocognitive and skeletal development caused by mutations in tgfbr1 or tgfbr2. Nature Genetics 2005;37:275-81.  Back to cited text no. 154    
155.Hines RN, McCarver DG. The ontogeny of human drug-metabolizing enzymes: Phase I oxidative enzymes. J Pharmacol Exp Ther 2002;300:355-60.  Back to cited text no. 155    
156.Stoilov I, Jansson I, Sarfarazi M, Schenkman JB. Roles of cytochrome p450 in development. Drug Metabol Drug Interact 2001;18: 33-55.  Back to cited text no. 156    
157.van Rooij IA, Wegerif MJ, Roelofs HM, Peters WH, Kuijpers-Jagtman AM, Zielhuis GA, et al. Smoking, genetic polymorphisms in biotransformation enzymes, and nonsyndromic oral clefting: A gene-environment interaction. Epidemiology 2001;12:502-7.  Back to cited text no. 157    
158.An S, Dickens MA, Bleu T, Hallmark OG, Goetzl EJ. Molecular cloning of the human edg2 protein and its identification as a functional cellular receptor for lysophosphatidic acid. Biochem Biophys Res Commun 1997;231: 619-22.  Back to cited text no. 158    
159.Shi M, Christensen K, Weinberg CR, Romitti P, Bathum L, Lozada A, et al. Orofacial cleft risk is increased with maternal smoking and specific detoxification-gene variants. Am J Hum Genet 2007;80:76-90.  Back to cited text no. 159    
160.Hartsfield JK Jr, Hickman TA, Everett ET, Shaw GM, Lammer EJ, Finnell RA. Analysis of the ephx1 113 polymorphism and gstm1 homozygous null polymorphism and oral clefting associated with maternal smoking. Am J Med Genet 2001;102:21-4.  Back to cited text no. 160    
161.Hozyasz KK, Mostowska A, Surowiec Z, Jagodzinski PP. [genetic polymorphisms of gstm1 and gstt1 in mothers of children with isolated cleft lip with or without cleft palate]. Przegl Lek 2005;62:1019-22.  Back to cited text no. 161    
162.Landi S. Mammalian class theta gst and differential susceptibility to carcinogens: A review. Mutat Res 2000;463:247-83.  Back to cited text no. 162    
163.Raijmakers MT, Steegers EA, Peters WH. Glutathione s-transferases and thiol concentrations in embryonic and early fetal tissues. Hum Reprod 2001;16:2445-50.  Back to cited text no. 163    
164.Sim E, Payton M, Noble M, Minchin R. An update on genetic, structural and functional studies of arylamine n-acetyltransferases in eucaryotes and procaryotes. Hum Mol Genet 2000;9:2435-41.  Back to cited text no. 164    
165.Smelt VA, Upton A, Adjaye J, Payton MA, Boukouvala S, Johnson N, et al. Expression of arylamine n-acetyltransferases in pre-term placentas and in human pre-implantation embryos. Hum Mol Genet 2000;9:1101-7.  Back to cited text no. 165    
166.Martinelli M, Scapoli L, Pezzetti F, Carinci F, Carinci P, Stabellini G, et al. C677t variant form at the mthfr gene and cl/p: A risk factor for mothers? Am J Med Genet 2001;98:357-60.  Back to cited text no. 166    
167.Blanton SH, Kolle BS, Hecht JT, Mulliken JB, Martin ER. No evidence supporting mthfr as a risk factor in the development of familial nsclp. Am J Med Genet 2000;92:370-1.  Back to cited text no. 167    
168.Botto LD, Yang Q. 5,10-methylenetetrahydrofolate reductase gene variants and congenital anomalies: A huge review. Am J Epidemiol 2000;151:862-77.  Back to cited text no. 168    
169.Jugessur A, Wilcox AJ, Lie RT, Murray JC, Taylor JA, Ulvik A, et al. Exploring the effects of methylenetetrahydrofolate reductase gene variants c677t and a1298c on the risk of orofacial clefts in 261 Norwegian case-parent triads. Am J Epidemiol 2003;157:1083-91.  Back to cited text no. 169    
170.Shaw GM, Rozen R, Finnell RH, Todoroff K, Lammer EJ. Infant c677t mutation in mthfr, maternal periconceptional vitamin use, and cleft lip. Am J Med Genet 1998;80:196-8.  Back to cited text no. 170    
171.Beaty TH, Wang H, Hetmanski JB, Fan YT, Zeiger JS, Liang KY, et al. A case-control study of nonsyndromic oral clefts in Maryland. Ann Epidemiol 2001;11:434-42.  Back to cited text no. 171    
172.Pepe G, Camacho Vanegas O, Giusti B, Brunelli T, Marcucci R, Attanasio M, et al. Heterogeneity in world distribution of the thermolabile c677t mutation in 5,10-methylenetetrahydrofolate reductase. Am J Hum Genet 1998;63:917-20.  Back to cited text no. 172    
173.Collier AC, Tingle MD, Paxton JW, Mitchell MD, Keelan JA. Metabolizing enzyme localization and activities in the first trimester human placenta: The effect of maternal and gestational age, smoking and alcohol consumption. Hum Reprod 2002;17:2564-72.  Back to cited text no. 173    
174.Shaw GM, Iovannisci DM, Yang W, Finnell RH, Carmichael SL, Cheng S, et al. Risks of human conotruncal heart defects associated with 32 single nucleotide polymorphisms of selected cardiovascular disease-related genes. Am J Med Genet A 2005;138:21-6.  Back to cited text no. 174    
175.Hwang SJ, Beaty TH, Panny SR, Street NA, Joseph JM, Gordon S, et al. Association study of transforming growth factor alpha (tgf alpha) taqi polymorphism and oral clefts: Indication of gene-environment interaction in a population-based sample of infants with birth defects. Am J Epidemiol 1995;141:629-36.  Back to cited text no. 175    
176.Maestri NE, Beaty TH, Hetmanski J, Smith EA, McIntosh I, Wyszynski DF, et al. Application of transmission disequilibrium tests to nonsyndromic oral clefts: Including candidate genes and environmental exposures in the models. Am J Med Genet 1997;73:337-44.  Back to cited text no. 176    
177.Miettinen PJ, Perheentupa J, Otonkoski T, Lahteenmaki A, Panula P. Egf- and tgf-alpha-like peptides in human fetal gut. Pediatr Res 1989;26:25-30.  Back to cited text no. 177    
178.Hwang SJ. Association study of transforming growth factor alpha (tgf alpha) taqi polymorphism and oral clefts: Indication of gene-environment interaction in a population-based sample of infants with birth defects. Amer J Epidem 1992;135:1000-11.  Back to cited text no. 178    
179.Mitchell LE, Murray JC, O'Brien S, Christensen K. Evaluation of two putative susceptibility loci for oral clefts in the Danish population. American Journal of Epidemiology 2001;153:1007-15.  Back to cited text no. 179    
180.Romitti PA, Lidral AC, Munger RG, Daack-Hirsch S, Burns TL, Murray JC. Candidate genes for nonsyndromic cleft lip and palate and maternal cigarette smoking and alcohol consumption: Evaluation of genotype-environment interactions from a population-based case-control study of orofacial clefts. Teratology 1999;59:39-50.  Back to cited text no. 180    
181.Beiraghi S, Zhou M, Talmadge CB, Went-Sumegi N, Davis JR, Huang D, et al. Identification and characterization of a novel gene disrupted by a pericentric inversion inv(4)(p13.1q21.1) in a family with cleft lip. Gene 2003;309:11-21.  Back to cited text no. 181    
182.Felix TM, Hanshaw BC, Mueller R, Bitoun P, Murray JC. Chd7 gene and non-syndromic cleft lip and palate. Am J Med Genet A 2006;140:2110-4.  Back to cited text no. 182    
183.Osoegawa K, Vessere GM, Utami KH, Mansilla MA, Johnson MK, Riley BM, et al. Identification of novel candidate genes associated with cleft lip and palate using array comparative genomic hybridisation. J Med Genet 2008;45:81-6.  Back to cited text no. 183    
184.Riley BM, Mansilla MA, Ma J, Daack-Hirsch S, Maher BS, Raffensperger LM, et al. Impaired fgf signaling contributes to cleft lip and palate. Proc Natl Acad Sci U S A 2007;104:4512-7.  Back to cited text no. 184    
185.Vieira AR, Avila JR, Daack-Hirsch S, Dragan E, Felix TM, Rahimov F, et al. Medical sequencing of candidate genes for nonsyndromic cleft lip and palate. PLoS Genet 2005;1: e64.  Back to cited text no. 185    
186.Inoue H, Kayano S, Aoki Y, Kure S, Yamada A, Hata A, et al. Association of the gabrb3 gene with nonsyndromic oral clefts. Cleft Palate Craniofac J 2008;45:261-6.  Back to cited text no. 186    

Copyright 2009 - Indian Journal of Plastic Surgery


The following images related to this document are available:

Photo images

[pl09077f3.jpg] [pl09077t2.jpg] [pl09077f2.jpg] [pl09077t1.jpg] [pl09077f1.jpg]
Home Faq Resources Email Bioline
© Bioline International, 1989 - 2024, Site last up-dated on 01-Sep-2022.
Site created and maintained by the Reference Center on Environmental Information, CRIA, Brazil
System hosted by the Google Cloud Platform, GCP, Brazil