ArticlePDF AvailableLiterature Review

Genetic and Epigenetic Studies in Nonsyndromic Oral Clefts

Authors:

Abstract

The etiology of non‐syndromic oral clefts (NSOFC) is complex with genetics, genomics, epigenetics and stochastics factors playing a role. Several approaches have been applied to understand the etiology of non‐syndromic oral clefts. These include linkage, candidate gene association studies, genome‐wide association studies, whole genome sequencing, copy number variations and epigenetics. In this review we shared these approaches, genes and loci reported in some studies.
Oral Diseases. 2022;00:1–12. wileyonlinelibrary.com/journal/odi
|
1© 2022 Wiley Periodicals LLC
1 | INTRODUCTIO N
Non- syndromic oral clefts (NSOFC) account for 70% of all clef ts
and are some of the most common birth defects in humans. The
global prevalence is about 1 in 700 live bir ths. (Mossey & Modell,
2012). However, there are population differences in the prevalence
which ranges from 1/500 in Asians to 0.5/1000 in Africans (Mossey
et al., 2009).
The etiology of non- syndromic clefts is complex with genetics,
genomics, epigenetics, and stochastics factors playing a role. Several
studies alluded to the possible genetic causes based on the segrega-
tion of clefts phenot ypes in families with multiple affected individuals.
Trew reported a f amily with several NSOFC affected members in 1751.
Andersen concluded that there is a presence of a strong genetic com-
ponent which did not align with the classic Mendelian dominant/reces-
sive inheritance pattern (Andersen, 1942). Attempts at uncovering the
inheritance pattern led to the suggestion of a multifactorial threshold
model (Fraser, 1976). This was later rejected in favor of a major locus/
gene (Melnick et al., 1986) and a mixed model, that is, major gene and
multifactorial model (Chung et al., 1986; Marazita et al., 1984).
Several a pproaches have be en applied to under stand the eti ology
of non- syndromic clef ts. These include linkage, candidate gene as-
sociation studies, genome- wide association studies, whole- genome
sequencing, copy number variations, and epigenetics (Table 1).
2 |LINKAGE STUDIES
The major gene effect model motivated early linkage studies
to understand the genetic basis of NSOFC . Linkage is a power-
ful method used to identify region(s) of the genome harboring a
disease- causing gene/locus by typing DNA markers to see whether
they co- segregates with the disease phenotype in related individu-
als (Rahimov et al., 2012; Wyszynski, 2002). This design is based
on the concept of linkage and identity by descent (IBD). Two loci
are in linkage if the recombination probability between them is less
than 50% during meiosis. IBD refers to genomic regions with iden-
tical nucleotide sequences inherited from a common ancestor and
not due to recombination. Linkage analysis can either be parametric
or non- parametric. Parametric linkage analyses are used for major
gene disorders with a pre- specified mode of inheritance. In con-
trast, non- parametric methods are used for complex diseases (e.g.,
NSOFC) where several genes contribute to disease risk (Kruglyak
et al., 1996). The non- parametric analysis approach looks for excess
IDB shared allele at any region/locus between affected sibling/rela-
tives pairs to identify regions/loci likely predisposing to disease (Shih
& Whittemore, 2001).
Through linkage analysis, over 20 chromosomal regions have
been linked to NSOFC. Notable ones include chr 1p, 1q21, 1q32-
42.3, 6p, 2p, 4q, and 17q (Carinci et al., 2000; Martinelli et al., 1998;
Received: 21 September 2021 
|
Revised: 11 January 2022 
|
Accepted: 20 January 2022
DOI : 10.1111/o di.14146
INVITED REVIEW
Genetic and epigenetic studies in non- syndromic oral clefts
Azeez Alade1,2,3| Waheed Awotoye1,2| Azeez Butali1,2
1Depar tment of Oral Patholog y, Radiolog y
and Medi cine, Colle ge of Dentistry,
University of Iowa, I owa City, Iowa, U SA
2Iowa Institute for Or al Health Research,
University of Iowa, I owa City, Iowa, U SA
3Depar tment of Epid emiology, College of
Public Health, University of Iowa, Iowa
City, Iowa , USA
Correspondence
Azeez B utali, Department of Oral
Pathology, Radiology and Medicine,
College of D entistr y, Universit y of Iowa,
Iowa Cit y, IA, USA.
Email: Azeez-butali@uiowa.edu
Funding information
Nationa l Institute of Dental and
Craniofacial Research, Grant/Award
Number: DE028300
Abstract
The etiology of non- syndromic oral clefts (NSOFC) is complex with genetics, genom-
ics, epigenetics, and stochastics factors playing a role. Several approaches have been
applied to understand the etiology of non- syndromic oral cleft s. These include linkage,
candidate gene association studies, genome- wide association studies, whole- genome
sequencing, copy number variations, and epigenetics. In this review, we shared these
approaches, genes, and loci reported in some studies.
KEY WORDS
epigenetics, genetics, Non- syndromic clefts
2 
|
    A LADE Et AL .
Mitchell & Risch, 1992). Some of these have been widely investi-
gated across different populations with varying results. For example,
Carinci et al. (2000) showed evidence for chr 2, 4, and 6 with cleft
lip with or without cleft palate (CL/P) in the Italian population, while
Wong et al. (2000) found no evidence for these regions in Swedish
multiplex CL/P families, suggesting a population- specific effect for
these loci— a phenomenon that has become increasingly apparent
with current knowledge. Although linkage studies have successfully
mapped genes for rare monogenic disorders, limited success was re-
corded with NSOFC. This is because NSOFC are complex disorders
caused by genes/loci with small effect sizes (Rahimov et al., 2012).
In line with the multifactorial inheritance model, linkage studies
were later ex tended to cover the whole genome via the use of mark-
ers placed at regular inter vals covering the whole genome. Prescott
et al. (2000) were the first to report a genome- wide significant link-
age. They carried out a two- stage genome- wide scan of 92 sibling
pairs with CL/P and identified 11 loci on eight chromosomes. Albeit,
these were not statistically significant genome wide. Two of the loci
had been previously reported as susceptible loci for CL/P, and one of
them harbors the TGFA gene. Other genes associated with NSOFCs
via linkage studies include COL11A1 , IRF6, EGF, MSX1, PTCH, TGFB1,
ROR2, FOXE1, TGFB3, RARA, APOC2, BCL3, and PVRL2.(Ardinger
et al., 1989; Chenevix- Trench et al.,1992; Kurihara et al., 1994; Lidral
et al., 1998; Nottoli et al.,1998; van den Boogaard et al., 20 00). The
major limitation to this approach is the broadness of the linked ge-
nomic region, making it difficult to pinpoint the disease- causing
gene or locus within the region (Rahimov et al., 2012).
3 |CANDIDATE GENES ASSOCIATION
STUDIES
While linkage studies require multiplex families with multiple af-
fected relatives/siblings, candidate gene association studies can be
carried out on isolated cleft cases without affected family members.
In general, association studies look for statistically significant differ-
ence in the frequency of an allele or haplotype between individuals
with the phenotype of interest and those without the phenotype in
order to identify the likely causative variant/marker (Kwon & Goate,
2000). Early candidate gene approaches leveraged on the premise
that common variants with less deleterious effec ts in known syndro-
mic cleft genes predispose to NSOFC (Stanier & Moore, 2004). This
hypothesis was tested for I RF6 gene implicated in Van der woude
syndrome and FOXE1 gene implicated in Bamforth Lazarus syn-
drome (Clif ton- Bligh et al., 1998; Zucchero et al., 20 04). Both genes
implicated in syndromic clefts were shown to be associated with
NSOFCs via candidate gene association studies. Other genes that
have been identified following their role in other craniofacial defects
(e.g., craniocynostosis) syndromes include FGFR1 and FGFR2 (Riley &
Murray, 2007). These observations were in line with the hypothesis
that variants in the non- coding regions of these genes are associated
with NSOFC while variants in the coding regions are responsible for
syndromic OFC (Stanier & Moore, 2004).
Candidate genes for NSOFC are also selected based on func-
tional properties, expression pattern, and or informative mouse
model. Several other candidate genes have been identified using
animal models. For instance, a MSX1 gene mouse knock- out model
showed clef t palate phenotype (Satokata & Maas, 1994) and can-
didate gene studies found an association between variants in this
gene and human NSOFC (Butali et al., 2011; Jezewski et al., 2003; Li
et al., 2017). Cleft palate phenotypes were observed for BMP mice
and chick embryos as well (Foppiano et al., 2007; Liu et al., 2005).
Human variants have also been reported for BMP4 in multiple pop-
ulations and in subclinical forms of clefts (Suzuki et al., 2004, 2009).
Other genes have been reported using several approaches such as
genome- wide scans for CRISPLD2 (Chiquet et al., 2007), fluorescent
in situ hybridization (FISH) of a balanced reciprocal translocation
for SUMO1 (Alkuraya et al., 2006), and case– control analyses for
TFGβ (Ardinger et al.1989).
The association of some candidate genes is inconsistent across
populations. The association with some genes has not been repli-
cated, while others show opposite effects. For instance, increased
risk for NSOFC was reported for BMP4 rs17563 polymorphism in
Asians and Caucasians and reduced risk was observed in Brazilian
population. Similarly, inconsistent association has been reported for
MTHFR. Some studies reported increased risk for CP (Mills et al.,
2008; Zhu et al., 2006), some reported a reduced risk for CL(P)
(Jugessur et al., 2003; Little et al., 2008), while another did not find
any associated risk with maternal MTHFR CT or TT genotypes for
either CL(P) or CP (Boyles et al., 2008).
4 |GENOME- WIDE ASSOCIATION
STUDIES
The need to identify novel risk loci/genes contributing to the risk
of NSOFCs ne cessitated the shi ft toward genome- wide as sociation
studies (GWAS). Unlike candidate gene association studies which
are hypothesis driven, GWAS is an unbiased and hypothesis free
approach used to identify genotype– phenotype associations by
testing multiple genetic variants across the genome (Christensen
& Murray, 2007). Since inception, It has revealed multiple loci con-
tributing to several complex traits (Shaffer et al., 2012). The first
GWAS on NSOFC was published by Birnbaum et al. (2009). They
reported a genome- wide significant association between NSCL/P
and three markers in 8q24- 21; a gene deser t devoid of any known
protein- coding gene (Birnbaum et al., 2009). That same year, Gant
et al. found a genome- wide association with the same region in
an independent cohor t of NSCL/P patients (Grant et al., 2009).
Beaty et al. (2010), conducted the first case- parent trios GWAS
on NSOFC. This study replicated the findings from the previous
GWAS and identified two new loci near MAFB and ABCA4 genes.
Further, Mangold et al. (2010) reported two additional loci in
17q22 and 10q25.3 with NOG and VAX1 identified as the possi-
ble candidate genes. Sun et al. (2015) conducted the first Chinese
GWAS on NSCL/P. This study replicated four previously published
   
|
 3
ALAD E Et AL.
Candidate genes
and genomic loci Technology and Methodology
SHH Association, Mutation screen, Targeted sequencing
TP63 Mutation screen, Targeted sequencing, Whole- exome sequencing
GLI2 Targeted sequencing
MSX2 Mutation screen, Targeted sequencing, Linkage
SPRY2 Mutation screen, Targeted sequencing
SPRY1 GWAS
SU LT2A 1 GWAS
CTNNA2 GWA S
PDGFRA Mutation screen, Targeted sequencing, Epigenetics
TBX1 Mutation screen, Copy number variation, Epigenetics
CTNNB1 Association, Mutation screen
PAX9 Targeted sequencing
PVRL1 Targeted sequencing, Mutation screen
TBX22 Targeted sequencing, Mutation screen
CTNND1 Mutation screen, Exome sequencing, Targeted sequencing
RARA Association
FGF10 GWAS, Mutation screen
WNT9B GWAS, Mutation screen
KRT18 G WAS
TFAP 2A GWAS, Mutation screen, Whole- genome sequencing
IRF6 Linkage, Association, Targeted sequencing, GWAS, Exome sequencing,
Copy number variation, whole- genome sequencing
FOXE1 Linkage, Association, Targeted sequencing, GWAS
MSX1 Animal models, Targeted sequencing
BMP4 Animal models, Targeted sequencing
FGFR1 GWAS, Targeted sequencing
FGFR2 Targeted sequencing
CRISPLD2 Linkage and Association
SUMO1 FISH
TFGβAssociation
MAFB GWA S
PAX7 GWAS
VAX1 GWAS
ARHGAP29 GWAS, Mutation Screen, Whole- exome sequencing
Chr8q.24 GWAS
Chr16p13.3 GWAS
VAX1, GWAS
NOG GWAS
GRHL3 Linkage, Exome sequencing
CDH1 Exome sequencing, Targeted sequencing
MGAM Copy number variation
ADAM3A Copy number variation
ZFHX4 Whole- genome sequencing
Chr21q22 Whole- genome sequencing
ADAM5A Copy number variation
TABLE 1 Some candidate genes and
loci that play roles in the etiology of non-
syndromic clefts and the techniques used
in their discoveries
4 
|
    A LADE Et AL .
loci and identified a novel locus in Chr 16p13.3 between CREBBP
and ADCY9 genes. With a ver y large sample size consisting of
multiethnic groups, Yu et al. (2017) reported 26 GWAS signifi-
cant loci, 14 of which were known with several new potential cleft
candidate genes (RAD54B, T MEM19, KRT18, WNT9B, GSC/DICER1,
PTCH1, RPS26, OFCC1/TFAP 2A, TAF1B, FGF10, MSX1, LINC00640,
FGFR1, and S PRY1).
With an increasing number of independent GWAS studies, the
first GWAS meta- analysis was conducted by Ludwig et al. (2012).
The study replicated most of the previously identified loci and
identified six novel regions. Following the success recorded by
this meta- analysis, several other meta- analyses were conducted
on NSCL/P (Leslie et al., 2017a; Yang et al., 2020; Yu et al., 2017).
All these studies (discovery and meta- analyses) put together led
to the identification of over 40 risk loci associated with NSCL/P.
The first study to report genome- wide significant loci associated
with non- syndromic cleft palate only was by Leslie et al., (2016).
They identified three novel loci with GRHL3, Yap1, and PARK2 as
possible candidate genes. The signal in GRHL3 was replicated by
another GWAS for CPO by Mangold et al. (2016). In 2019, The
African only GWAS by Butali et al. (2019) and the Chinese GWAS
by Huang et al. (2019), respectively, identified additional NSCPO
risk loci near CTNNA2, SULT2A 1, DLK1, DOCK9, FOXC2/FOXL1,
IRF6, MAU2, PAX9, POMG, NT2, and WHSC1 genes (Butali et al.,
2019; Huang et al., 2019). Despite the tremendous successes re-
corded by GWAS in identifying NSOFCs susceptible genes, all the
genes/loci identified so far only account for 20%– 30% of the tot al
heritability of NSOFC (Leslie et al., 2017a).
The missing heritability could be explained by low frequency/
rare variants, gene– gene interactions, and epigenetics among
others. In a bid to determine the missing heritability, rare variant
studies of NSOFC are becoming increasingly popular. Initially,
through candidate genes resequencing in small samples to iden-
tify private variants and subsequently via genome- wide rare vari-
ants analysis. The first genome- scale study of rare variants was by
Bureau et al in 2014 (Bureau et al., 2014). They performed whole-
exome sequencing and looked for shared rare variants in 348 es-
tablished cleft candidate genes among NSOFC affected relatives
from 55 multiplex families. The study reported five novels shared
variants in the CDH1 gene and showed statistically significant ev-
idence of co- segregation in an Indian family. Leslie et al. (2017b)
examined the role of rare coding variants applying a genome- wide
rare variants aggregate association test and found a statistically
significant association with low frequency and rare variants in
N4BPH gene in the Latin American population. With a multieth-
nic sample of 11,727 participants, Shaffer et al., 2019 focused on
the rare non- coding variants in previously identified craniofacial
enhancer regions. They found a significant association between
mm60 on chr 3p13 (an enhancer element located bet ween MITF
and FOXP1 gene) and CPO (Shaffer et al., 2019). The candidate
gene approach is still ver y relevant following GWAS, especially
as replication for novel genes identified through next- generation
omics approaches.
5 |WHOLE- EXOME SEQUENCING
Whole- exome sequencing (WES) study has long been applied
in the investigation of the genetic risk of common birth defects
(Feliciano et al., 2019; Yu et al., 2013). This next- generation se-
quencing technique has helped to identify pathogenic genetic
mutations that were undetected by earlier screening approaches.
Earlier use of this technique in gene discover y focused on rare
Mendelian disorders (Antonarakis & Beckmann, 2006); however, it
has also been used for complex traits. This sequencing approach is
usually used to identify genetic variants within the coding regions
of the human genome (exome). A large number of mutations within
the exome af fect the amino acid sequence which have a direct ef-
fect on the protein structures and function (Kryukov et al., 2007).
This knowledge forms the foundation of the exome sequencing
studies designed to identify protein- altering mutations that led to
the disorders.
This next- generation sequencing technique has been applied
in the study of NSOFC across multiple populations(Aylward et al.,
2016; Basha et al., 2018; Cai et al., 2017; Liu et al., 2017; Machado
et al., 2021; Pengelly et al., 2016). One of such studies used WES
approach to screen for damaging mutations segregating with the
phenotype across multiple pedigrees in a family with cleft pal-
ate. This approach was used to identify a novel loss- of- function
mutation in ARHGAP29 as the etiologic factor in the family (Liu
et al., 2017). Families with multiple pedigrees (multiplex) with
affected relatives provide a huge opportunity for the identifica-
tion of the genetic risk driving birth defect or trait (Bureau et al.,
2019; D'Netto et al., 2009). This is because the unaffected rel-
atives ser ve as more accurate controls compared to the use of
population- based controls. Thus, this requires a deep phenotyp-
ing of all individuals within the pedigree and using the appropriate
inheritance model for the genetic screens. Other factors that must
be considered when studying the Mendelian disorders and com-
plex traits using WES are penetrance, identification of disease-
causing mutation, and heterogeneity of genetic variants to the risk
of the disorder or trait.
The use of WES in family- based study design helps to inves-
tigate the numerous inheritance models possible in the genetic
risk of cleft. This sequencing strategy can also be applied to case–
control study design while applying the different inheritance
model in the genetic screens. Although WES helps to investigate
high impact mutations that are potentially pathogenic, it is only
limited to discovery of protein- altering mutations within a re-
stricted 1%– 2% of the entire human genome also known as the
protein- coding region of the genome. Nonetheless, this miniscule
region harbors 85% of the disease- causing mutations (Majewski
et al., 2011). Regardless, there is a missing 15% disease- causing
mutation which are potentially harbored within the non- coding
region (~99% of the human genome). This uninvestigated region
(as well as the exome) can, however, be studied by using a whole-
genome sequencing approach with better precision and accuracy
(Belkadi et al., 2015).
   
|
 5
ALAD E Et AL.
6 |WHOLE- GENOME SEQUENCING
STUDIES
Whole- genome sequencing (WGS) is one of the next- generation
sequencing approaches that is now being used to study the eti-
opathogenesis of genetic disorders. Unlike whole- exome sequenc-
ing (WES), another next- generation sequencing that detects genetic
variants within the coding region (exome) of the genome, WGS can
be used to detect variant s within the non- coding and coding regions.
These non- coding regions account for 99% of the entire genome
and contain regulatory elements that control the gene expression.
Recent studies have investigated and improved our understanding
of the roles of these non- coding variants in the etiopathogenesis of
genetic disorders (French & Edwards, 2020; Li & Montgomery, 2013;
Wells et al., 2019; Zhang & Lupski, 2015).
A report that compared the performance of WGS and WES in
the detec tion of coding variants which are easy to interpret found
that WGS outperforms WES. The broader coverage of WGS gives
us the power to detect potential variants that would have been
missed by using WES (Gilissen et al., 2014; Saunders et al., 2012).
The study showed that WGS could detect about 3% coding vari-
ants that are missed by WES (Belkadi et al., 2015). Additionally,
WGS is more reliable in the detection of structural variants within
the coding region when compared to WES (Belkadi et al., 2015).
The comparable costs of WES and WGS, coupled with the other
factors, make WGS a more reliable choice in genetic variants
screening. As diagnostic tool, trio based WGS has a diagnostic
yield of about 42% which is comparable to that of WES 40% diag-
nostic yield (Wright et al., 2018).
In our quest to underst and the source and identify the missing
heritability of NSOFC, we have begun to explore next- generation
sequencing studies. A larger proportion of the missing heritability is
due to rare variants (Wainschtein et al., 2021). Prior investigation of
these rare pathogenic variants has been limited to candidate gene
sequencing approach which investigated the mutations in known
cleft genes and whole- exome sequencing which identifies only cod-
ing mutations. Whole- genome sequencing studies provide us with
the ability to investigate the entire genome for both coding and non-
coding, common and rare mutations that can explain the genetic risk
of NSOFC. In a single genomic data per individual, we can query the
entire 3 billion base pairs for variations that are pathogenic and can
affect the normal develop of the lip and palate. This genomic se-
quence data can be used to answer every research question as it
concerns the genetics of NSOFC.
One strategy that has been reported recently used the genomic
sequence to identify a new risk locus associated with NSOFC in a
multiethnic population (Mukhopadhyay et al., 2020). Unlike the
traditional GWAS, this strategy does not require imputation and
provides a deeper and more reliable sequence. Another strategy
is the investigation of de novo mutations (DNMs) that increase
the risk of this defect. DNMs are those germline mutations that
are found in the offspring but not carried by either parent. These
DNMs have been reported to increase the risk of congenital defects
(Alonso- Gonzalez et al., 2018; Homsy et al., 2015; Ji et al., 2020),
but limited studies are available for NSOFC. Majorit y of cleft cases
are sporadic thus explains the role of DNMs in the etiopathogen-
esis. One study that investigated the DNMs using whole- genome
sequence data in a NSOFC case- parent trios study design identified
excess loss- of- function DNMs in genes (IRF6, TFAP2A, and ZFHX4)
involved in craniofacial development and identified ZFHX4 as a novel
cleft candidate gene (Bishop et al., 2020).
Thus, with the WGS data and powerful study designs, we can
identify structural variants, common and rare variations that in-
crease the genetic risk of NSOFC. As we race toward improved
management, this genetic study approach will help reduce the
knowledge gap in disease etiology and take us a step closer to-
ward translation.
7 |COPY NUMBER VARIATIONS
In addition to the other methods for identifying risk variants for
orofacial clefts, copy number variations (CNVs) have been reported
in the etiology of complex traits like NSOFC. CNVs modif y gene
expression, disrupt gene, and alter gene dosage leading to syndro-
mic and NSOFC (Maarse et al., 2012). A genome- wide deletion-
association analysis identified a locus near ch7p14.1 associated with
NSOFC (Younkin et al., 2014). Using the same GWAS data set that
was used for the Beaty et al. (2010) study, genome- wide inherited
deletions identified a 67 kb deletion on Chr7q34 and a 200 kb de-
letion on chr8p11 (Younkin et al. 2015). In another CNV analyses
of 23 unrelated individuals with clefts and 200 controls, TCEB3 and
KIF7 were identified as new clef t candidate genes (Simioni et al.,
2015). A large cohort of 312 OFC patients analyzed 249 genomic
deletions and 226 duplications in two publicly accessible databases
(DECIPHER and ECARUCA) of chromosome imbalance. The follow-
ing known cleft candidate genes were identified in these deleted
and duplicated regions: SATB2, MEIS2, DGCR6, FGF2, FRZB, LETM1,
MAPK3, SPRY1, THB S1, TSHZ1, TTC28, TULP4, WHSC1, and WHSC2
(Conte et al., 2018).
Isthmin 1 was identified as a new OFC candidate gene when CNV
analyses was done for 97 NSOFC and 43 cases with CPO. Lansdon
et al. (2018) identified a heterozygous deletion of in one affected
case and deletion in a second case. The deletion removed the puta-
tive 3’ regulatory information from Isthmin 1 (Lansdon et al., 2018).
Another study replicated the findings in the 62kb deletion region
of 7p14 in 399 patients and 1318 controls and reported an asso-
ciation with NSOFC (p = 0.024). They also identified de novo de-
letion in three sporadic families, and both incomplete segregation
and incomplete penetrance in multiplex families. An investigation of
whole- genome high- resolution SNP in 33 patients with syndromic
and NSOFC with in normal karyotypes identified six novel CNVs
(3 in patients with NSOFC and 3 in patients with syndromic clefts).
The authors also identified two novel candidate genes for NSOFC;
K AT6 B and MACROD2 and confirmed the role of CNVs in the etiol-
ogy of NSOFC (Lei et al., 2016).
6 
|
    A LADE Et AL .
8 |EPIGENETICS
Epigenetics is the reversible change in the expression of genes with-
out changes in the DNA sequence. These include DNA methylation,
histone modifications, and non- coding RNA. The role of epigenetics
changes has been well documented in the etiology of several dis-
eases including birth defects (Keil & Vezina, 2015; Wahlqvist et al.,
2015).
8.1  | DNA methylation and orofacial clefts
There are several studies that have conducted epigenetic- wide
association studies (EWAS) for orofacial clefts. A study in the UK
conducted EWAS using blood and lip tissues to test the association
between methylation at each site and cleft subtype (cleft lip only
(CLO) n = 50; cleft palate only (CPO) n = 50; cleft lip and palate (CLP)
n = 50). They found four genomic regions in blood differentially
methylated in CLO compared to CLP, 17 in CPO compared to CLP
and 294 in CPO compared to CLO. Interestingly, they found regions
that mapped to known clefts candidate genes like TBX1, COL11A 2,
HOXA2, and PDGFRA. In addition, they reported 250 novel differen-
tially methylated loci. They also found similar methylated regions in
blood and lip tissues (Sharp et al., 2017).
Another study used a Brazilian cohor t (67 NSCLP, 59 controls)
for EWAS and found 578 methylation variable positions (MVPs) or
differentially methylated regions were significantly associated with
NSCLP. They observed that these MVPs were enriched in regula-
tory and active regions of the genome and in known pathways for
craniofacial development. Four of the 11 MVP were replicated in an
independent UK cohor t (171 NSCLP, 177 controls). Like the study
by Sharp et al. (2017), they demonstrated a significant positive
correlation between blood and lip tissue DNA methylation suppor t-
ing the suitability of blood tissue for NSCLP methylation studies
(Alvizi et al., 2017). In a study that compared whole- genome DNA
methylation in six MZ twin pairs discordant for NSCLP, they found
differential methylation in MAFB and ZEB2 in two independent MZ
twin pairs. They also found common dif ferential methylation in
genes belonging to the Hippo signaling pathway.
Long interspersed nucleotide element- 1 (LINE- 1) is a marker of
global DNA methylation. Studies have reported differential global
DNA methylation patterns between NSOFC and controls using the
methylation levels of LINE- 1 (Cáceres- Rojas et al., 2020; Khan et al.,
2018, 2019; Li et al., 2019). There is evidence to suggest that ge-
netic variations (c.C677T and c.A1298C) in the MTHFR gene lead to
low DNA methylation levels (Frosst et al., 1995; van der Put et al.,
1998). Further evidence for the role of MTHFR gene variant in DNA
methylation has been reported for NSOFC where higher levels of
LINE- 1 methylation were obser ved on the medial side of cleft lip in
individuals with c.C677T genotypes (Khan et al., 2019).
Differentially methylated regions maybe enriched in clefts co-
hort, and these could explain the missing heritability for clefts.
While there may be similar methylated regions across populations,
there possibility of population- specific regions is high considering
the various types of environmental exposures within each popula-
tion. Therefore, exploring the methylation profiles of cleft families in
populations across the world will lead to a clear unders tanding of the
role of epigenetics in the etiology of orofacial clefts.
8.2  | Histone modification and microRNAs
The evidence supporting the role of histone modification in the
etiolog y of orofacial clefts in humans is sparse. Few studies have
FIGURE 1 Critical pathways and the
IRF6 gene- regulatory network relevant to
craniofacial development and reported for
non- syndromic orofacial clefts (NSOFCs).
Some clef t candidate genes in black fonts
within the pathways and net work. The
gene list, pathways, and network are
not exhaustive but represent current
knowledge and a starting point for further
investigations
   
|
 7
ALAD E Et AL.
explored the role in the development of cleft palate in the presence
of drugs and other exposures in mice (Cuiping et al., 2014; Yuan
et al., 2016). In contrast, the role of long non- coding RNAs (lncR-
NAs) and microRNAs in the etiology of orofacial clefts in humans
is becoming clearer and the evidence is growing. For instance, a
study identified 36 lncRNAs, 1,341 mRNAs, and 60 miRNAs to be
differentially expressed in the CL/P group compared to the control
group, and 57 lncRNAs, 1,255 mRNAs, and 162 miRNAs to be dif fer-
entially expressed in the CPO group compared to the control group.
These were identified when they conducted next- generation RNA
sequencing (RNA- seq) to identify mRNAs, lncRNAs, and miRNAs in
patients with CL/P and CPO (Gao et al., 2019). Another study com-
bined the DEMs during mouse embryonic palatal development with
DEMs in NSCL/P patients and identified a let- 7c- 5p- PIGA and miR-
193 a- 3p - TGFB2 net work that may play important roles in the devel-
opment of NSCL/P (Fu et al., 2021).
As we explore the missing heritabilit y in the etiology of orofa-
cial clefts, a comprehensive understanding of the role of histone
modifications, lncRNAs, and microRNAs will be extremely useful
and potentially translational toward the development of prevention
strategies.
9 |PATHWAYS AND GENE- REGULATORY
NETWORKS
Several pathways have been reported to play a role in palate devel-
opment, and genes in these pathways are potential clefting genes.
The WNT pathway is critical for craniofacial development, and WNT
pathway genes such as AXIN1 and WNT9B (Figure 1) have been
associated with NSOFC (Mani et al., 2010; Menezes et al., 2010).
Fgf10/Fgfr2/Shh signaling pathway is also essential for palate de-
velopment (Rice et al., 2004). Genes in this pathway such as FGFR1
and FGF2 have been associated with NSOFC (Riley & Murray, 2007).
Other pathways that have been reported to be important for pal-
ate development include the MSX1 pathway, Folate pathway, and
TGFB pathway (Figure 1). The IRF6 gene- regulatory network is also
critical in the development of the palate (Figure 1), and studies have
shown genes in this network to be associated with NSOFC (Kousa &
Schutte, 2016; Liu et al., 2016).
10  | POPULATION
Accurate phenotyping is the foundation for identif ying genetics
and genomics risks for NSOFC. Furthermore, population and geo-
graphical location of affected individuals with NSOFC are also very
important factors for considerations. Evidence from some genetic
studies shows that some loci are population- specific while other
loci have associations with multiple populations (Beat y et al., 2010).
This is mainly due to differences in allele frequencies and population
specificity between ancestral populations which was demonstrated
by the identification of risk loci at the chr8q24 locus in Caucasians
in Europe and North America and MAFB in Asians. Furthermore,
population specificity has been demonstrated for 15q22, which
reached genome- wide significance in Europeans and Mexicans only
but not in Asians,16p13.3, significant only in the Chinese population
and 2p13.1 where a significant association was identified in Native
Americans and not in Europeans or Asians (Ludwig et al., 2012,
2014; Masotti et al., 2018; Sun et al., 2015).
Finally, the inheritance mode needs to be clearly understood
for any investigation to be meaningful. For instance, a multiplex
family appears to be Mendelian, and approach will prioritize seg-
regation of risk variants. For sporadic cases, priority should be
placed on identifying de novo variants. However, investigators
and the community need to be aware of incomplete penetrance
of the phenotypes and variants as well as the polygenic and multi-
factorial inheritance for NSOFC. Bringing all these into the design
and strategies will improve our understanding and opportunities
for discover y.
11  | CHALLENGES AND FUTURE
DIRECTIONS
Discovery is the foundation for a successful clinical translational
process. For NSOFC, just like other polygenic disorders, the main
challenges will be the identification of the critical pathways for devel-
opment where variants / polymorphisms in candidate genes within
these pathways will lead to the disruptions at critical time points dur-
ing the development of the lip and palate. The challenges in discov-
ery may include access to populations around the world and accurate
phenotyping in resource limited populations. Future directions for
NSOFC should be focused on taking the knowledge from discovery
to translation. This will involve the development of diagnostic tools
for pregnancy screening to enable counseling and pregnancy plan-
ning. It should also involve in utero interventions to rescue the clefts.
While this is theoretically possible and showing promise in animal
models, the success in humans will be a game changer.
ACKNOWLEDGMENTS
This work was supported by grants from the National Institutes of
Health (NIH DE028300).
CONFLICT OF INTEREST
None.
AUTHOR CONTRIBUTIONS
Azeez Alade: Writing – original draft; Writing – review & editing.
Waheed Awotoye: Writing – original draft; Writing – review & edit-
ing. Azeez Butali: Conceptualization; Methodology; Project adminis-
tration; Writing – original draft; Writing – review & editing.
DATA AVAIL ABILI TY STATEMENT
Data sharing not applicable to this article as no datasets were gener-
ated or analyzed during the current study.
8 
|
    A LADE Et AL .
ORCID
Azeez Butali https://orcid.org/0000-0002-1229-5964
REFERENCES
Alkuraya, F. S., Saadi, I., Lund, J. J., Turbe- Doan, A., Morton, C. C., &
Maas, R. L. (2006). SUMO1 haploinsufficiency leads to cleft lip and
palate. Science, 313, 1751.
Alonso- Gonzalez, A., Rodriguez- Fontenla, C., & Carracedo, A. (2018). De
novo Mutations (DNMs) in autism spectrum disorder (asd): path-
way and net work analysis. Frontiers in Genetics, 9, 406. https://doi.
org/10.3389/fgene.2018.00406
Alvizi, L., Ke, X., Br ito, L. A., Seselgy te, R., Moore, G. E., Stanier, P., &
Passos- Bueno, M. R. (2017). Differential methylation is asso-
ciated with non- syndromic cleft lip and palate and contributes
to penetrance effects. Scientific Reports, 7(1), 2441. https://doi.
org /10.1038/s 4159 8- 017- 02721 - 0
Antonarakis, S. E., & Beckmann, J. S. (2006). Mendelian disorders de-
serve more attention. Nature Reviews Genetics, 7(4), 277– 282.
htt ps://doi.o rg/10.10 38/nrg1826
Ardinger, H. H., Buetow, K. H., Bell, G. I., Bardach, J., VanDemark, D.
R., & Murray, J. C. (1989). Association of genetic variation of the
transforming grow th factor- alpha gene with cleft lip and palate.
American Journal of Human Genetics, 45(3), 348– 353.
Aylward, A., Cai, Y., Lee, A., Blue, E., Rabinowitz, D., & Haddad, J. Jr
(2016). Using whole exome sequencing to identify candidate genes
with rare variants in nonsyndromic cleft lip and palate. Genetic
Epidemiology, 40(5), 432441. https://doi.org/10.1002/gepi.21972
Basha, M., Demeer, B., Revencu, N., Helaers, R., Theys, S., Bou Saba, S.,
Boute, O., Devauchelle, B., Francois , G., Bayet, B., & Vikkula , M.
(2018). Whole exome sequencing identifies mutations in 10% of
patient s with familial non- syndromic cleft lip and/or palate in genes
mutated in well- known syndromes. Journal of Medical Genetics,
55(7), 449– 458. https://doi.org/10.1136/jmedg enet- 2017- 105110
Beaty, T. H., Murray, J. C., Marazita, M. L., Munger, R. G., Ruczinski,
I., Hetmanski, J. B., Liang, K. Y., Wu, T., Murray, T., Fallin, M. D.,
Redett, R. A., Raymond, G., Schwender, H., Jin, S.- C., Cooper,
M. E., Dunnwald, M., Mansilla, M. A ., Leslie, E ., Bullard, S., …
Scott, A. F., (2010). A genome- wide association study of cleft
lip with and without cleft palate identifies risk variants near
MAFB and ABCA4. Nature Genetics, 42(6), 525– 529. https://doi.
org /10.1038/ng.58 0
Belkadi, A., Bolze, A., Itan, Y., Cobat, A., Vincent, Q. B., Antipenko, A .,
Shang, L., Boisson, B., Casanova, J.- L., & Abel, L. (2015). Whole-
genome sequencing is more powerful than whole- exome se-
quencing for detecting exome variants. Proceedings of the National
Academy of Sciences, 112(17), 5473– 5 478. https ://doi.org/10.1073/
pna s.14186 31112
Birnbaum, S., Ludwig, K. U., Reut ter, H., Herms, S., Steffens , M., Rubini,
M., Baluardo, C., Fer rian, M., Almeida de Assis, N., Alblas, M. A.,
Barth, S., Freudenberg, J., Lauster, C., Schmidt, G., Scheer, M.,
Braumann, B., Bergé, S. J., Reich, R. H., Schiefke, F., … Mangold, E.
(2009). Key susceptibility locus for nonsyndromic cleft lip with or
without cleft palate on chromosome 8q24. Nature Genetics, 41(4),
473– 47 7. ht tps://doi.org/10.1038/ng.33 3
Bishop, M. R., Diaz Perez, K. K., Sun, M., Ho, S., Chopra, P.,
Mukhopadhyay, N., Hetmanski, J. B., Taub, M. A ., Moreno- Uribe,
L. M., Valencia- Ramirez, L. C., Restrepo Muñeton, C. P., Wehby, G.,
Hecht, J. T., Deleyiannis, F., Weinberg, S. M., Wu- Chou, Y. H., Chen,
P. K., Brand, H., Epstein, M. P., … Leslie, E. J. (2020). Genome- wide
Enrichment of De Novo Coding Mutations in Orofacial Cleft Trios.
The American Journal of Human Genetics, 107 (1), 124– 136. https://
doi.org/10.1016/j.ajhg.2020.05.018
Boyles, A. L., Wilcox, A. J., Taylor, J. A., Meyer, K., Fredriksen, Å., Ueland,
P. M., Drevon, C. A ., Vollset, S. E., & Lie, R. T. (2008). Folate and
one- carbon metabolism gene polymorphisms and their associa-
tions with oral facial clefts. American Journal of Medical Genetics
Part A, 146A (4), 440– 449. https://doi.org/10.1002/ajmg.a.32162
Bureau, A., Begum, F., Taub, M. A., Hetmanski, J. B., Parker, M. M.,
Albacha- Hejazi, H., Scott, A. F., Murray, J. C., Marazita, M. L.,
Bailey- Wilson, J. E., Beaty, T. H., & Ruczinski, I. (2019). Inferring
disease risk genes from sequencing data in multiplex pedigrees
through sharing of rare variants. Genetic Epidemiology, 43(1), 37– 49.
https://doi.org/10.1002/gepi.22155
Bureau, A., Parker, M. M., Ruczinski, I., Taub, M. A., Marazita, M .
L., Murray, J. C., Mangold, E., Noethen, M. M., Ludwig, K. U.,
Hetmanski, J. B., Bailey- Wilson, J. E., Cropp, C. D., Li, Q., Szymczak,
S., Albacha- Hejazi, H., Alqosayer, K., Field, L . L., Wu- Chou, Y.- H.,
Doheny, K. F., … Beat y, T. H. (2014). Whole exome sequencing of
distant relatives in multiplex families implicates rare variants in can-
didate genes for oral clefts. Genetics, 197 (3), 1039– 104 4. https://
doi.org/10.1534/genet ics.114.165225
Butali, A., Mossey, P. A., Adeyemo, W. L., Eshete, M. A., Gowans, L. J. J.,
Busch, T. D., Jain, D., Yu, W., Huan, L., Laurie, C. A., Laurie, C. C.,
Nelson, S., Li, M., Sanchez- L ara, P. A., Magee, W. P., Magee, K . S.,
Auslander, A., Brindopke, F., Kay, D. M., … Adeyemo, A . A. (2019).
Genomic analyses in African populations identify novel risk loci for
cleft palate. Human Molecular Genetics, 28(6), 1038– 1051. https://
doi.org/10.1093/hmg/ddy402
Butali, A ., Mossey, P. A., Adeyemo, W. L., Jezewski, P. A., Onwuamah,
C. K., Ogunlewe, M. O., Ugboko, V. I., Adejuyigbe, O., Adigun, A. I.,
Abdur- Rahman, L. O., Onah, I. I., Audu, R. A., Idigbe, E. O., Mansilla,
M. A., D ragan, E. A ., Petrin, A. L., Bullard, S. A., Uduezue, A. O.,
Akpata, O., … NigeriaCRAN Collaboration (2011). Genetic studies
in the Nigerian population implicate an MSX1 mutation in complex
oral facial clefting disorders. The Cleft Palate- Craniofacial Journal,
48(6), 646– 653.
Cáceres- Rojas, G., Salamanca, C., Krause, B. J., Recabarren, A. S.,
Recabarren, P. A., Pantoja, R., & Suazo, J. (2020). Nonsyndromic
orofacial clefts in Chile: LINE- 1 methylation and MTHFR variants.
Epigenomics, 12(20), 1783– 1791.
Cai, Y., Patterson, K. E., Reinier, F., Keesecker, S. E., Blue, E., Bamshad,
M., & Haddad, J. Jr (2017). Copy number changes identified using
whole exome sequencing in nonsyndromic cleft lip and palate in a
Honduran population. Birth Defec ts Research, 109(16), 1257– 1267.
htt ps://doi.o rg/10.10 02/bdr2.1063
Carinci, F., Pezzetti, F., Scapoli, L., Martinelli, M., Carinci, P., & Tognon, M.
(2000). Genetics of nonsyndromic cleft lip and palate: a review of
international studies and data regarding the Italian population. The
Cleft Palate- Craniofacial Journal, 37(1), 33– 40. https://doi.org/10.15
97/1545- 1569_2000_037_0033_goncla_2.3.co_2
Chenevix- Trench, G., Jones, K., Green, A., Duffy, D., & Martin, N. (1992).
Cleft lip with or without cleft palate: associations with transform-
ing growth factor alpha and retinoic acid receptor loci. American
Journal of Human Genetics, 51(6) , 13 77.
Chiquet, B. T., Lidral, A. C., Stal, S., Mulliken, J. B., Moreno, L. M., Arcos-
Burgos, M., & Hecht, J. T. (2007). CRISPLD2: a novel NSCLP candi-
date gene. Human Molecular Genetics, 16, 2241– 2248.
Christensen, K., & Murray, J. C. (2007). What genome- wide associa-
tion studies can do for medicine. New England Journal of Medicine,
356(11), 1094– 1097. https://doi.org/10.1056/NEJMp 068126
Chung, C . S., Bixler, D., Watanabe, T., Koguchi, H., & Fogh- Andersen, P.
(1986). Segregation analysis of cleft lip with or without cleft pal-
ate: a comparison of Danish and Japanese data. American Journal of
Human Genetics, 39(5), 6 03– 611.
Clifton- Bligh, R. J., Wentworth, J. M., Heinz, P., Crisp, M. S., John, R.,
Lazarus, J. H., Ludgate, M., & Chatterjee, V. K. (1998). Mutation of
the gene encoding human TTF- 2 associated with thyroid agenesis,
cleft palate and choanal atresia. Nature Genetics, 19(4), 399– 401.
htt ps://doi .org /10.1038/1294
   
|
 9
ALAD E Et AL.
Conte, F., Oti, M., Dixon, J., Carels, C. E., Rubini, M., & Zhou, H. (2016).
Systematic analysis of copy number variants of a large cohort of
orofacial cleft patients identifies candidate genes for orofacial
cleft s. Human Genetics, 135(1), 41– 59. ht tps://doi.org/10.1007/
s0043 9- 015- 1606- x
Cuiping , L., Xingan g, Y., Yuexi an, F., Li n, Q., Xiaofei, T., Yan, L., & G uanghui,
W. (2014). The role of histone H3 acetylation on cleft palate in mice
induced by 2, 3, 7, 8- tetrachlorodibenzopdioxin. Zhonghua Zheng
Xing Wai Ke Za Zhi, 30 (5), 369– 372.
D'Netto, M. J., Ward, H., Morrison, K. M., Ramagopalan, S. V., Dyment,
D. A., DeLuca, G. C ., Handunnetthi, L., Sadovnick, A. D., & Ebers,
G. C. (2009). Risk alleles for multiple sclerosis in multiplex fami-
lies. Neurology, 72(23), 1984– 1988. https://doi.org/10.1212/
WNL.0b013 e3181 a92c25
Felician o, P., Zho u, X., Ast rovskaya, I. , Turn er, T. N., Wang , T., Brueggema n,
L., Bar nard, R., Hsieh, A., Snyder, L. A. G., Muzny, D. M., Sabo, A .,
Gibbs, R. A., Eichler, E. E., O’Roak, B. J., Michaelson, J. J., Volfovsky,
N., Shen, Y., & Chung, W. K. (2019). Exome sequencing of 457 au-
tism families recruited online provides evidence for autism risk
genes. NPJ Genomic Medicine, 4(1), 19. http s://doi.org/10.1038/
s4152 5- 019- 0093- 8
Fogh- A ndersen, P. (1942). Inheritance harelip and cleft palate. Opara
Exdome Biologiae Hareditariae Universities Hafniensis, 4, 1.
Foppiano, S., Hu, D., & Marcucio, R. S. (2007). Signaling by bone morpho-
genetic proteins direc ts formation of an ectodermal signaling cen-
ter that regulates craniofacial development. Developmental Biology,
312, 103– 114. https://doi.org/10.1016/j.ydbio.2007.09.016
Fraser, F. C. (1976). The multifactorial/threshold concept – uses and
misuses. Teratology, 14(3), 267– 280. https://doi.or g/10.1002/
ter a.14201 40302
French, J. D., & Edwards, S. L. (2020). The role of noncoding variants in
heritable disease. Trends in Genetics, 36(11), 880– 891. https://doi.
org/10.1016/j.tig.2020.07.004
Frosst, P., Blom, H. J., Milos, R., Goyette, P., Sheppard, C. A., Matthews,
R. G., Boers, G. J. H., den Heijer, M., Kluijtmans, L. A. J., van den
Heuve, L . P., & Rozen, R. A. (1995). Candidate genetic risk factor
for vascular disease: A common mutation in methylenetetrahy-
drofolate reductase. Nature Genetics, 10(1) , 111– 113. h ttp s://doi.
org /10.10 38/ng 059 5- 111
Fu, C., Lou, S., Zhu, G., Fan, L., Yu, X., Zhu, W., & Pan, Y. (2021).
Identif ication of new miRNA- mRNA net works in the development
of non- syndromic cleft lip with or without cleft palate. Frontiers in
Cell and Developmental Biology, 9, 631057. htt ps://doi.org/10.3389/
fcell.2021.631057
Gao, Y., Zang, Q., Song, H., Fu, S., Sun, W., Zhang, W., & Jiao, X. (2019).
Comprehensive analysis of differentially expressed prof iles of non-
coding RNAs in peripheral blood and ceRNA regulatory networks
in non- syndromic orofacial clefts . Molecular Medicine Reports, 20(1),
513– 528. https://doi. org/10 .3892/mmr.2019.10261
Gilissen, C., Hehir- Kwa, J. Y., Thung, D. T., van de Vorst, M., van Bon, B.
W., Willemsen, M. H., & Veltman, J. A. (2014). Genome sequenc-
ing identifies major c auses of severe intellectual disability. Nature,
511(7509), 344– 347.
Grant, S. F., Wang, K., Zhang, H., Glaberson, W., Annaiah, K., Kim, C.
E., & Garris, M. (2009). A genome- wide association study identi-
fies a locus for nonsyndromic clef t lip with or without cleft pal-
ate on 8q24. The Journal of Pediatrics, 155(6), 909– 913. https://doi.
org/10.1016/j.jpeds.2009.06.020
Homsy, J., Zaidi, S., Shen, Y., Ware, J. S., Samocha, K. E., Karczewski, K.
J., & Chung, W. K. (2015). De novo mutations in congenital heart
disease with neurodevelopmental and other congenital anomalies.
Science, 350(6265), 1262– 1266.
Huang, L., Jia, Z., Shi, Y. I., Du, Q., Shi, J., Wang, Z., Mou, Y., Wang, Q.,
Zhang, B., Wang, Q., Ma, S., Lin, H. E., Duan, S., Yin, B., Lin, Y.,
Wang, Y., Jiang, D., Hao, F., Zhang, L., … Yang, Z. (2019). Genetic
factors define CPO and CLO subtypes of nonsyndromicorofacial
cleft. PLoS Genetics, 15(10), e100 8357. https://doi.org/10.1371/
journ al.pgen.1008357
Jezewski, P., Vieira, A., Nishimura, C., Ludwig, B., Johnson, M., O'Brien, S.,
Daack- Hirsch, S., Schultz, R., Weber, A., Nepomucena, B., Romitti,
P., Christensen, K., Orioli, I., Castilla, E., Machida, J., Natsume, N.,
& Murray, J. (2003). Complete sequencing shows a role for MSX1
in non- syndromic cleft lip and palate. Journal of Medical Genetics,
40(6), 399– 4 07. https://doi.org/10.1136/jmg.40.6.399
Ji, W., Ferdman, D., Copel, J., Scheinost, D., Shabanova, V., Brueckner,
M., Khokha, M. K., & Ment, L. R. (2020). De novo damaging variants
associated with congenital hear t diseases contribute to the con-
nectome. Scientific Reports, 10(1), 7046. ht tps: //do i.or g/10.10 38/
s4159 8- 020- 63928 - 2
Jugessur, A., Wilcox, A. J., Lie, R. T., Murray, J. C., Taylor, J. A., Ulvik, A.,
Drevon, C . A., Vindenes, H. A ., & Abyholm, F. E. (2003). Exploring
the effects of methylenetetrahydrofolate reductase gene variants
C677T and A1298C on the risk of orofacial clefts in 261 Norwegian
case- parent triads. American Journal of Epidemiology, 157(12),
1083 1091 .
Keil, K. P., & Vezina, C . M. (2015). DNA methylation as a dynamic reg-
ulator of development and disease processes: Spotlight on the
prostate. Epigenomics, 7(3), 413– 425. https://doi.org/10.2217/
epi.15.8
Khan, M. F. J., Little, J., Aleotti, V., Mossey, P. A., Steegers- Theunissen, R.
P. M., Autelitano, L., & Rubini, M. (2019). LINE- 1 methylation in cleft
lip tissues: influence of infant MTHFR c.677C >T genot ype. Oral
Diseases, 25(6), 166 8– 1671.
Khan, M. F. J., Little, J., Mossey, P. A., Steegers- Theunissen, R. P. M.,
Autelitano, L., Lombardo, I., Andreasi, R . B., & Rubini, M . (2018).
Evaluating LINE- 1 methylation in cleft lip tissues and its associa-
tion with early pregnancy exposures. Epigenomics, 10 (1), 105– 113.
htt ps://doi .org /10.2217/epi- 2017- 0081
Kousa, Y. A., & Schutte, B. C. (2016). Toward an orofacial gene regulatory
network. Developmental Dynamics, 245(3), 220– 232.
Kruglyak, L., Daly, M. J., Reeve- Daly, M. P., & Lander, E. S. (1996).
Parametric and nonparametric linkage analysis: A unified multipoint
approach. American Journal of Human Genetics, 58(6), 1347– 1363.
Kryukov, G. V., Pennacchio, L . A., & Sunyaev, S. R. (2007). Most rare
missense alleles are deleterious in humans: Implications for com-
plex disease and association studies. American Journal of Human
Genetics, 80(4), 727739. https://doi.org/10.1086/513473
Kurihara, Y., Kurihara, H., Suzuki, H., Kodama, T., Maemura, K., Nagai, R.,
Oda, H., Kuwaki, T., Cao, W. H., Kamada, N., & Jishage, K. (1994).
Elevated blood pressure and craniofacial abnormalities in mice de-
ficient in endothelin- 1. Nature, 368( 6 473) , 7 03– 710 .
Kwon, J. M., Goate, A. M. (2000). The candidate gene approach. Alcohol
Research & Health, 24(3), 164– 168.
Lansdon, L. A., Darbro, B. W., Petrin, A. L., Hulstrand, A. M., Standley,
J. M., Brouillette, R. B., Long, A., Mansilla, M. A., Cornell, R. A.,
Murray, J. C., Houston, D. W., & Manak, J. R. (2018). Identification
of isthmin 1 as a novel clefting and craniofacial pat terning gene in
humans. Genetics, 208(1), 283– 296. ht tps://doi.org/10.1534/genet
ic s . 117. 3 0 0 5 35
Lei, T. Y., Wang, H. T., Li, F., Cui, Y. Q., Fu, F., Li, R., & Liao, C. (2016).
Applic ation of high resolution SNP arrays in patients with congen-
ital oral clefts in south China. Journal of Genetics, 95(4), 801– 809.
https://doi.org/10.1007/s1204 1- 016- 0696- 0
Leslie, E. J., Carlson, J. C., Shaffer, J. R., Butali, A., Buxó, C. J., Castilla, E.
E., Christensen, K., Deleyiannis, F. W. B., Leigh Field, L ., Hecht, J.
T., Moreno, L., Orioli, I. M., Padilla, C., Vieira, A. R., Wehby, G. L.,
Feingold, E., Weinberg, S. M., Murray, J. C., Beaty, T. H., & Marazita,
M. L. (2017a). Genome- wide meta- analyses of nonsyndromic oro-
facial clefts identify novel associations between FOXE1 and all
orofacial clefts, and TP63 and cleft lip with or without clef t palate.
Human Genetics, 136(3), 275– 286. https://doi.org/10.1007/s0043
9- 016- 175 4- 7
10 
|
    A LADE Et AL .
Leslie, E. J., Carlson, J. C., Shaffer, J. R., Buxó, C. J., Castilla, E. E.,
Christensen, K., Deleyiannis, F. W. B., Field, L. L., Hecht, J. T.,
Moreno, L ., Orioli, I. M., Padilla, C., Vieira, A. R., Wehby, G. L.,
Feingold, E., Weinberg, S. M., Murray, J. C., & Marazita, M. L.
(2017b). Association studies of low- frequency coding variants
in nonsyndromic clef t lip with or without clef t palate. American
Journal of Medical Genetics. Part A , 173 (6), 15311538. https://doi.
org /10.1002 /ajmg.a.3 8210
Leslie, E. J., Carlson, J. C., Shaffer, J. R., Feingold, E., Wehby, G., Laurie,
C. A., Jain, D., Laurie, C. C., Doheny, K. F., McHenry, T., Resick, J.,
Sanchez, C., Jacobs, J., Emanuele, B., Vieira, A. R., Neiswanger,
K., Lidral, A. C., Valencia- Ramirez, L. C., Lopez- Palacio, A. M., …
Marazit a, M. L. (2016). A multi- ethnic genome- wide association
study identifies novel loci for non- syndromic clef t lip with or with-
out cleft palate on 2p24.2, 17q23 and 19q13. Human Molecular
Genetics, 25(13), 2862– 2872. https://doi.org/10.1093/hmg/ddw104
Li, X., & Montgomer y, S. (2013). Detection and impac t of rare regulatory
variants in human disease. Frontiers in Genetic s, 4, 67. https://doi.
org/10.3389/fgene.2013.00067
Li, Y., Deng, Y., Deng, C., Xie, L ., Yu, L. I., Liu, L., Yuan, Y., Liu, H., & Dai,
L. I. (2019). Association of long interspersed nucleotide element- 1
and inter feron regulatory fac tor 6 methylation changes with non-
syndromic cleft lip with or without cleft palate. Oral Diseases, 25(1),
215– 222. https://doi.org/10.1111/odi.12965
Li, Y. H., Yang, J., Zhang, J. L., Liu, J. Q., Zhneg, Z., & Hu, D. H. (2017).
BMP4 rs17563 polymorphism and nonsyndromic cleft lip with
or without cleft palate: A meta- analysis. Meta- Analysis Medicine
(Baltimore), 96(31), e7676. https://doi.org/10.1097/MD.0 000 0
00000 007676
Lidral, A. C., Romitti, P. A., Basart, A. M ., Doetschman, T., Leysens,
N. J., Daack- Hirsch, S., Semina, E. V., Johnson, L. R., Machida, J.,
Burds, A., Parnell, T. J., Rubenstein, J. L. R., & Murray, J. C. (1998).
Association of MSX1 and TGFB3 with nonsyndromic clefting in
humans. The American Journal of Human Genetics, 63(2), 557– 568.
htt ps://doi.o rg/10.10 86/301956
Little, J., Gilmour, M., Mossey, P. A., Fitzpatrick, D., Cardy, A., Clayton-
Smith, J., Hill, A., Duthie, S. J., Fryer, A. E., Molloy, A. M., Scott, J. M.,
& ITS MAGIC Collaboration (2008). Folate and clef ts of the lip and
palate- a U. K.- based case- control study: Part II: Biochemical and
genetic analysis. The Cleft palate- Craniofacial Journal, 45, 428– 438.
htt ps://doi.org/10.1597/06- 151.1
Liu, H., Busch, T., Eliason, S., Anand, D., Bullard, S., Gowans, L. J. J., &
Butali, A. (2017). Exome sequencing provides additional evidence
for the involvement of ARHGAP29 in Mendelian orofacial clefting
and extends the phenotypic spectrum to isolated cleft palate. Birth
Defects Research, 109 (1), 27– 37.
Liu, H., Leslie, E. J., Jia, Z., Smith, T., Eshete, M., Butali, A., & Cornell, R. A.
(2016). Irf6 dir ectly regul ates Klf17 in zebrafi sh periderm an d Klf4 in
murine oral epithelium, and dominant- negative KLF4 variants
are present in patient s with cleft lip and palate. Human Molecular
Genetics, 25(4): 766– 776. https://doi.org/10.1093/hmg/ddv614
Liu, W., Sun, X ., Braut, A., Mishina, Y., Behringer, R. R ., Mina, M., &
Martin, J. F. (2005). Distinct functions for Bmp signaling in lip and
palate fusion in mice. Development, 132, 1453– 1461. htt ps://do i.
org/10.1242/dev.01676
Ludwig, K. U., Böhmer, A. C., Rubini, M., Mossey, P. A., Herms, S., Nowak,
S., Reut ter, H., Alblas, M. A., Lippke, B., Barth, S., Paredes- Zenteno,
M., Muñoz- Jimenez, S. G., Ortiz- Lopez, R., Kreusch, T., Hemprich,
A., Martini, M., Braumann, B., Jäger, A., Pötzsch, B., … Mangold, E. J.
(2014). Strong association of variants around FOXE1 and orofacial
clefting. Journal of Dental Research, 93(4) , 376– 381 .
Ludwig, K. U., Mangold, E., Herms, S., Nowak, S., Reutter, H., Paul, A.,
Becker, J., Herberz, R., AlChawa, T., Nasser, E., Böhmer, A. C.,
Mattheisen, M., Alblas, M. A., Barth, S., Kluck, N., Lauster, C.,
Braumann, B., Reich, R. H., Hemprich, A., … Nöthen, M. M . (2012).
Genome- wide meta- analyses of nons yndromic cleft lip with or
without cleft palate identify six new risk loci. Nature Genetics, 44(9),
968– 971. https://doi.org/10.1038/ng.2360
Maarse, W., Rozendaal, A. M., Pajkrt, E., Vermeij- Keer s, C., Mink van
der Molen, A. B., & van den Boogaard, M. J. (2012). A system-
atic review of associated structur al and chromosomal defects in
oral clefts: when is prenatal genetic analysis indicated? Journal of
Medical Genetics, 49(8), 490– 498. https://doi.org/10.1136/jmedg
enet- 2012- 101013
Machado, R. A., Martelli- Junior, H., Reis, S. R. D. A., Küchler, E. C., Scariot,
R., das Neves, L. T., & Colet ta, R. D. (2021). Identific ation of novel
variant s in cleft palate- associated genes in B razilian patients with
non- syndromic clef t palate only. Frontiers in Cell and Developmental
Biology, 9, 1334. https://doi.org/10.3389/fcell.2021.638522
Majewski, J., Schwartzentruber, J., Lalonde, E., Montpetit, A ., & Jabado,
N. (2011). What can exome sequencing do for you? Journal of
Medical Genetics, 48(9), 580– 589. https://doi.org/10.1136/jmedg
enet- 2011- 100223
Mangold, E., hmer, A. C., Ishorst, N., Hoebel, A.- K., Gültepe, P.,
Schuenke, H., Klamt, J., Hofmann, A., Gölz, L., Raff, R., Tessmann,
P., Nowak, S., Reutter, H., Hemprich, A., Kreusch, T., Kramer, F.-
J., Braumann, B., Reich, R., Schmidt, G., Ludwig, K. U. (2016).
Sequencing the GRHL3 coding region reveals rare truncating mu-
tations and a common susceptibility variant for nonsyndromic cleft
palate. American Journal of Human Genetics, 98(4), 755– 762. https://
doi.org/10.1016/j.ajhg.2016.02.013
Mangold, E., Ludwig, K. U., Birnbaum, S., Baluardo, C., Ferrian, M.,
Herms, S ., Reutter, H., de Assis, N. A ., Chawa, T. A., Mattheisen,
M., Steffens, M., Barth, S., Kluck, N., Paul, A., Becker, J., Lauster,
C., Schmidt, G., Braumann, B., Scheer, M., … Nöt hen, M. M. (2010).
Genome- wide association study identifies two susceptibility loci
for nonsyndromic cleft lip with or without clef t palate. Nature
Genetics, 42(1), 24– 26. https://doi.org/10.1038/ng.506
Mani, P., Jarrell, A., Myers, J., & Atit, R. (2010). Visualizing canonical Wnt
signaling during mouse craniofacial development. Developmental
Dynamics, 239(1), 354– 363.
Marazit a, M. L., Spence, M. A ., & Melnick, M. (1984). Genetic analysis of
cleft lip with or without cleft palate in Danish kindreds. American
Journal of Medical Genetics, 19(1), 9– 18. https://doi.o rg/10 .1002 /
ajmg.13201 90104
Martinelli, M., Scapoli, L., Pezzetti, F., Carinci, F., Carinci, P., Baciliero,
U., & Tognon, M. (1998). Suggestive linkage between markers on
chromosome 19q13. 2 and nonsyndromic orofacial cleft mal-
formation. Genomics, 51(2), 177– 181. ht tps: //do i.or g/10.10 06/
geno.1998.5384
Masotti, C., Brito, L. A., Nica, A. C., Ludwig, K. U., Nunes, K., Savastano,
C. P., Malcher, C., Ferreira, S. G., Kobayashi, G. S., Bueno, D. F.,
Alonso, N., Franco, D., Rojas- Martinez, A., Dos Santos, S. E.,
Galante, P. A., Meyer, D., Hünemeier, T., Mangold, E., Dermitzakis,
E. T., & Passos- Bueno, M. R. (2018). MRPL53, a new candidate gene
for orofacial clefting, identif ied using an eQTL approach. Journal
of Dental Research, 97(1), 33– 4 0. https ://doi.o rg/10.1177/002 20
34 517 7358 05
Melnick , M., Marazita, M. L., & Hu, D. N. (1986). Genetic analysis of
cleft lip with or without cleft palate in Chinese kindreds. American
Journal of Medical Genetics, 25, 183190. https://doi.org/10.1002/
ajmg.13202 50622
Menezes, R., Letra, A., Kim, A. H., Küchler, E. C., Day, A., Tannure, P.
N., Gomes da Motta, L., Paiva, K. B., Granjeiro, J. M., & Vieira, A.
R. (2010). Studies with Wnt genes and non- syndromic cleft lip
and palate. Birth Defects Research . Part A, Clinical and Molecular
Teratology, 88(11), 995– 1000.
Mills, J. L ., Molloy, A. M., Parle- McDermott, A ., Troendle, J. F., Brody,
L. C., Conley, M. R., & Kirke, P. N. (2008). Folate- related gene
polymorphisms as risk factors for cleft lip and cleft palate. Birth
Defects Research. Par t A, Clinical and Molecular Teratolog y, 2008(82),
636– 643.
   
|
11
ALAD E Et AL.
Mitchell, L. E., & Risch, N. (1992). Mode of inheritance of nonsyndromic
cleft lip with or without cleft palate: A reanalysis. American Journal
of Human Genetics, 51(2), 323.
Mossey, P. A., Little, J., Munger, R. G., Dixon, M. J., & Shaw, W. C.
(2009). Cleft lip and palate. Lancet, 374(9703), 1773– 1785. https://
pubmed.ncbi.nlm.nih.gov/19747 722/#:~:text=PMID%3A%20
197 47722 - ,DOI%3A%2010.1016/S0140 %2D673 6(09)60695
%2D4,- Abstract
Mossey, P. A., & Modell, B. (2012). Epidemiology of oral cleft s 2012: An
international perspective. Frontiers of Oral Biology, 16, 1– 1 8 .
Mukhopadhyay, N., Bishop, M., Mortillo, M., Chopra, P., Hetmanski, J. B.,
Taub, M. A., Moreno, L. M., Valencia- Ramirez, L. C., Restrepo, C.,
Wehby, G. L., Hecht, J. T., Deleyiannis, F., Butali, A., Weinberg, S.
M., Beaty, T. H., Murray, J. C., Leslie, E. J., Feingold, E., & Marazita,
M. L. (2020). Whole genome sequencing of orofacial cleft trios
from the Gabriella Miller Kids Fir st Pediatric Research Consortium
identif ies a new locus on chromosome 21. Human Genetics, 139(2),
215– 226. htt ps://doi.org /10.1007/s0043 9- 019- 02 099 - 1
Nottoli, T., Hagopian- Donaldson, S., Zhang, J., Perkins, A., & Williams,
T. (1998). AP- 2- null cells disrupt morphogenesis of the eye, face,
and limbs in chimeric mice. Proceedings of the National Academy
of Sciences, 95(23), 13714– 13719. https://doi.org/10.1073/
pnas .95.23 .13714
Pengelly, R. J., Arias, L., Martínez, J., Upstill- Goddard, R., Seaby, E. G.,
Gibson, J., Ennis, S., Collins, A., & Briceño, I. (2016). Deleterious
coding variants in multi- case families with non- syndromic cleft lip
and/or palate phenotypes. Scientific Repor ts, 6(1), 30457. https://
doi.org/10.1038/srep3 0457
Prescott, N. J., Lees, M. M., Winter, R. M., & Malcolm, S. (20 00).
Identif ication of susceptibility loci for nonsyndromic cleft lip with
or without cleft palate in a two stage genome scan of affected sib-
pairs. Human Genetics, 10 6(3), 345– 350. https://doi.org/10.1007/
s0043 90000239
Rahimov, F., Jugessur, A., & Murray, J. C. (2012). Genetics of nonsyn-
dromic orofacial clef ts. The Cleft Palate- Craniofacial Journal, 49(1),
73– 91. https ://doi.o rg/10.1597/10- 178
Rice, R., Spencer- D ene, B., Connor, E. C., Gritli- Linde, A., McMahon, A.
P., Dickson, C., & Rice, D. P. (2004). Disruption of Fgf10/Fgfr2b-
coordinated epithelial- mesenchymal interactions causes cleft pal-
ate. Journal of Clinical Investigation, 113, 1692.
Riley, B. M., & Murray, J. C. (2007). Sequence evaluation of FGF and
FGFR gene conserved non- coding elements in non- syndromic cleft
lip and palate cases. American Journal of Medical Genetics. Part A,
143A(24), 3228– 3234.
Satokat a, I., & Maas, R . (1994). Msx1 deficient mice exhibit cleft palate
and abnormalities of craniofacial and tooth development. Nature
Genetics, 6(4), 348– 356. ht tps://doi.org/10.1038/ng049 4- 34 8
Saunders, C. J., Miller, N. A., Soden, S. E., Dinwiddie, D. L., Noll, A.,
Alnadi, N. A., Andraws, N., Patterson, M. L. A., K rivohlavek, L.
A., Fellis, J., Humphray, S., Saffrey, P., Kingsbur y, Z., Weir, J. C.,
Betley, J., Grocock, R. J., Margulies, E. H., Farrow, E. G., Artman,
M., … King smore, S. F. (2012). Rapid whole- genome sequencing for
genetic disease diagnosis in neonatal intensive care units. Science
Translational Medicine, 4(154), 154ra135. https://doi.org/10.1126/
scitr anslm ed.3004041
Shaffer, J. R., Feingold, E., & Marazita, M. L . (2012). Genome- wide as-
sociation studies: Prospects and challenges for oral health. Journal
of Dental Research, 91(7), 637– 641. https://doi.org/10.1177/00220
34512 44 6968
Shaffer, J. R., LeClair, J., Carlson, J. C., Feingold, E., Buxó, C. J., Christensen,
K., Deleyiannis, F. W. B., Field, L. L., Hecht, J. T., Moreno, L., Orioli,
I. M., Padilla, C., Vieira, A. R., Wehby, G. L., Murray, J. C., Weinberg,
S. M., Marazita, M. L ., & Leslie, E. J. (2019). Association of low-
frequency genetic variants in regulatory regions with nonsyn-
dromic orofacial clef ts. American Journal of Medical Genetics Part A,
179(3), 467– 474. https://doi.org/10.1002/ajmg.a.61002
Sharp, G . C., Ho, K., D avies, A., Ste rgiakouli, E., H umphries, K ., McArdl e, W.,
Sandy, J., Davey Smith, G., Lewis, S. J., & Relton, C. L. (2017). Dis tinct
DNA methylation profiles in subtypes of orofacial cleft. Clinical
Epigenetics, 9, 63– 63. https://doi.org/10.1186/s1314 8- 017- 0362- 2
Shih, M. C., & Whittemore, A. S. (2001). Allele- sharing among af-
fected relatives: Non- parametric methods for identifying genes.
Statistical Methods in Medical Research, 10(1), 27– 55. ht tps://doi.
org /10.1177/09622 8 0201 010 0010 3
Simioni, M., Araujo, T. K., Monlleo, I. L ., Maurer- Morelli, C. V., & Gil- da-
Silva- Lopes, V. L. (2015). Investig ation of genetic factors underly-
ing typical orofacial clefts: mutational screening and copy number
variation. Journal of Human Genetics, 60(1), 1725. https://doi.
org /10.1038/ jhg. 2014.96
Stanier, P., & Moore, G. E. (2004). Genetics of cleft lip and palate:
Syndromic genes contribute to the incidence of non- syndromic
cleft s. Human Molecular Genetics, 13 (90001), 73R– 81. ht tps://doi.
org /10.1093/hmg/ddh052
Sun, Y., Huang, Y., Yin, A., Pan, Y., Wang, Y., Wang, C., Du, Y., Wang, M.,
Lan, F., Hu, Z., Wang, G., Jiang, M., Ma, J., Zhang, X., Ma, H., Ma, J.,
Zhang, W., Huang, Q., Zhou, Z., … Yang, Y. (2015). Genome- wide
association study identifies a new susceptibility locus for cleft lip
with or without a cleft palate. Nature Communications, 6(1), 6414.
https://doi.org/10.1038/ncomm s7414
Suzuki, S., Marazita, M. L., Cooper, M. E., Miwa, N., Hing, A., Jugessur,
A., Nat sume, N., Shimozato, K., Ohbayashi, N ., Suzuki, Y., Niimi,
T., Minami, K., Yamamoto, M., Altannamar, T. J., Erkhembaatar, T.,
Furukawa, H., Daack- Hirsch, S., L'Heureux, J., Brandon, C. A.,
Murray, J. C. (2009). Mutations in BMP4 are associated with sub-
epithelial, microform, and overt cleft lip. Am erican Journal of Human
Genetics, 84(3), 406– 411.
Suzuki, Y., Jezewski, P. A., Machida, J., Watanabe, Y., Shi, M., Cooper,
M. E., Viet le, T., Nguyen, T. D., Hai, H., Natsume, N., Shimozato,
K., Marazita, M. L., & Murray, J. C. (2004). In a Vietnamese popu-
lation, MSX1 variants contribute to cleft lip and palate. Genetics in
Medicine, 6(3), 117– 125.
van den Boogaard, M.- J., Dorland, M., Beemer, F. A., & van Amstel, H. K.
P. (2000). MSX1 mutation is associated with orofacial clefting and
tooth agenesis in humans . Nature Genetics, 24(4), 342– 343. https://
doi.org/10 .1038/74155
van der Put, N. M. J., Gabreëls, F., Stevens, E. M. B., Smeitink, J. A. M.,
Trijbels, F. J. M., Eskes, T. K. A. B., van den Heuvel, L. P., & Blom, H.
J. (1998). A second common mutation in the methylenetetrahydro-
folate reductase gene: An additional risk fac tor for neural- tube de-
fec ts? The American Journal of Human Genetics, 62(5), 10 44– 1051.
htt ps://doi.o rg/10.10 86/301825
Wahlqvist, M. L., Krawetz, S. A ., Rizzo, N. S., Dominguez- Bello, M. G.,
Szymanski, L. M., Barkin, S., Yatkine, A., Waterland, R. A., Mennella,
J. A ., Desai, M., Ross, M. G., Krebs, N. F., Young, B. E., Wardle, J.,
Wrann, C. D., & Kral, J. G. (2015). Early- life influences on obesity:
From preconception to adolescence. Annals of the Ne w York Academy
of Sciences, 1347(1) , 1. https://doi.org/10.1111/ny as.12778
Wainschtein, P., Jain, D., Yengo, L., Zheng, Z. TOPMed Anthropometry
Working Group, Trans- Omics for Precision Medicine Consortium,
Visscher, P. M. (2021). Recovery of trait heritability from
whole genome sequence data, bioRxiv, 588020. https://doi.
org /10.1101/588020
Wells, A., Heckerman, D., Torkamani, A., Yin, L., Sebat, J., Ren, B., & di Iulio,
J. (2019). Ranking of non- coding pathogenic variants and putative
essential regions of the human genome. Nature Communications,
10(1), 5241. ht tps://doi.org/10.1038/s4146 7- 019- 13212 - 3
Wong, F. K., Hagberg, C., Karsten, A ., Larson, O., Gustavsson, M.,
Huggare, J., Larsson, C., Teh, B. T., & Linder- Aronson, S. (2000).
Linkage analysis of candidate regions in Swedish nonsyndromic
cleft lip with or without cleft palate families. The Cleft Palate-
Craniofacial Journal, 37(4), 357– 362. https://doi.org/10.1597/154
5- 1569_2000_037_0357_laocri_2.3.co_2
12 
|
    A LADE Et AL .
Wright, C . F., FitzPatrick, D. R., & Firth, H . V. (2018). Paediatric genomics:
diagnosing rare disease in children. Nature Reviews Genetics, 19(5),
253– 268. https://doi.org/10.1038/nrg.2017.116
Wyszynski, D. F. (Ed.). (2002). Locating genes for oral clefts in humans. In
Cleft lip and palate: From origin to treatment (pp. 255– 264). Oxford
University Press.
Yang, Y., Suzuki, A., Iwata, J., & Jun, G. (2020). Secondary genome- wide
association study using novel analytical strategies disentangle ge-
netic components of cleft lip and/or cleft palate in 1q32.2. Genes,
11(11), 1280 . https://do i.or g/10.339 0/gene s 1111128 0
Younkin, S. G ., Scharpf, R. B., Schwender, H., Parker, M. M., Scott, A. F.,
Marazit a, M. L., Beaty, T. H., & Ruczinski, I. (2014). A genome- wide
study of de novo deletions identifies a candidate locus for non-
syndromic isolated cleft lip/palate risk. BMC Genetics, 15, 24– 24.
htt ps://doi .org /10.1186/1471- 2156- 15- 24
Younkin, S. G ., Scharpf, R. B., Schwender, H., Parker, M. M., Scott, A.
F., Marazita, M. L., Beat y, T. H., & Ruczinski, I . (2015). A genome-
wide study of inherited deletions identified two regions associated
with nonsyndromic isolated oral clefts. Birth Defect s Research. Part
A, Clinical and Molecular Teratology, 103(4), 276– 283. https://doi.
org/10.1002/bdra.23362
Yu, T., Chahrour, M., Coulter, M., Jiralerspong, S., Okamura- Ikeda,
K., Ataman, B., Schmitz- Abe, K., Harmin, D., Adli, M., Malik, A.,
D’Gama, A., Lim, E., Sanders, S., Mochida, G., Partlow, J., Sunu, C.,
Felie, J., Rodriguez, J., Nasir, R., … Walsh, C. (2013). Using whole-
exome sequencing to identify inherited causes of autism. Neuron,
77(2), 259273. https://doi.org/10.1016/j.neuron.2012.11.002
Yu, Y., Zuo, X., He, M., Gao, J., Fu, Y., Qin, C., Meng, L., Wang, W., Song,
Y., Cheng, Y., Zhou, F., Chen, G., Zheng , X., Wang, X., Liang, B. O.,
Zhu, Z., Fu, X., Sheng, Y., Hao, J., … Bian, Z. (2017). Genome- wide
analyses of non- syndromic cleft lip with palate identify 14 novel
loci and genetic heterogeneity. Nature Communications, 8(1), 1– 11.
https://doi.org/10.1038/ncomm s14364
Yuan, X., Qiu, L., Pu, Y., Liu, C., Zhang, X ., Wang, C., Pu, W., & Fu, Y.
(2016). Histone acetylation is involved in TCDD- induced cleft pal-
ate format ion in fetal mice. Molecular Medicine Repor ts, 14(2), 1139–
1145. https://doi.org/10.3892/mmr.2016.5348
Zhang, F., & Lupski, J. R. (2015). Non- coding genetic variants in human
disease. Human Molecular Genetics, 24(R1), R 102– 110. https://doi.
org /10.1093/hmg/ddv259
Zhu, J., Ren, A., Hao, L., Pei, L., Liu, J., Zhu, H., Li, S., Finnell, R. H., & Li, Z.
(2006). Variable contribution of the MTHFR C677T polymorphism
to non- s yndromic cleft lip and palate risk in China. American Journal
of Medical Genetics. Part A, 2006(140), 551– 557.
Zucchero, T. M., Cooper, M. E., Maher, B. S., Daack- Hirsch, S.,
Nepomuceno, B., Ribeiro, L., & Murray, J. C. (2004). Interferon
regulatory factor 6 (IRF6) gene variants and the risk of iso-
lated clef t lip or palate. New England Journal of Medicine, 351(8),
769– 780 .
How to cite this article: Alade, A., Awotoye, W., & Butali, A.
(2022). Genetic and epigenetic studies in non- syndromic oral
clefts. Oral Diseases, 00, 1– 12. ht tps://doi.org /10.1111/
odi.14146
... While many syndromes involving cleft lips, affecting or not the cleft palate, are recognized in humans, the majority of oral clefts fall into the category of 'non-syndromic oral clefts' and their etiology remains poorly understood. However, the general agreement is that oral clefts are of multifactorial origin, with both genetic and environmental factors playing a role (Schutte & Murray, 1999;Alade et al., 2022). ...
... Syndromic cases of oral clefts in humans can be subdivided into chromosomal anomalies, Mendelian syndromes, teratogens, and uncategorized syndromes (Schutte & Murray, 1999;Alade et al., 2022). Genetic factors are related to gene and chromosome alterations, with specific depletion or duplication, where the predisposition of the fissure depends on the integration of polygenic factors associated with environmental conditions. ...
... The karyotype analysis indicated that in this case, this anomaly did not represent a chromosomal syndrome. However, other genetic disorders related to oral cleft were not evaluated and cannot be discarded (Kohli & Kohli, 2012;Alade et al., 2022). Similarly, viral infections or nutritional disorders during the mother's pregnancy in this case are unknown and difficult to identify during the calf evaluation. ...
Article
Since 1976, Amazonian manatees have been examined either alive or through necropsies as part of a long-term program to rescue and rehabilitate orphan manatee calves whose mothers were either victims of illegal hunting or accidental catch in fishing nets in the Brazilian Amazon region. Rescued manatees have been found to have several types of diseases, but to our knowledge, there is no record of a congenital anomaly of the face, known as an oral cleft, in Amazonian manatees. In this paper, we report the occurrence of an oral cleft in an Amazonian manatee calf, describing the clinical evaluation and discussing the potential causes.
... 11 This complexity is attributed to the interplay between genomics, genetics, epigenetics and environmental factors. 12 The specific roles of genetics and genomics in the etiology of nsOFC have been investigated using several approaches. These approaches include linkage studies, candidate gene association studies, genome wide association studies (GWAS), whole genome sequencing, whole exome sequencing (WES) amongst others. ...
... These approaches include linkage studies, candidate gene association studies, genome wide association studies (GWAS), whole genome sequencing, whole exome sequencing (WES) amongst others. 12 Some cleft genes and risk loci have been identified and associated with nsOFC using these approaches. 13 However, the magnitude of progress on the search of the genetic causes of nsOFC has been modest. ...
Article
Objectives The interaction between genomics, genetic and environmental factors have been implicated in non-syndromic orofacial cleft development. In the current study, we investigated the contributions of rare and novel genetic variants in known cleft genes using whole exome sequencing (WES) data of Indonesians with non-syndromic orofacial clefts. Design WES was conducted on 6 individuals. Variants in their exons were called and annotated. These variants were filtered for novelty and rarity using MAF of 0 and 1%. Setting Hospital in Indonesia. Patients/Participants Indonesians with non-syndromic orofacial clefts. Interventions Deleterious variants were prioritized. Pathogenic amino acid changes effect on protein structure and function were identified using HOPE. Main Outcome Measure(s) Rare and novel variants in known cleft genes were filtered and their deleteriousness were predicted using polyphen, SIFT and CADD. Results We identified rare (MAF <1%) deleterious variants in 4 craniofacial genes namely MMACHC (rs371937044, MAF = 0.00011). SOS1 (rs190222208, MAF = 0.00045), TULP4 (rs199583035, MAF = 0.067), and MTHFD1L (rs143492706, MAF = 0.0044). MMACHC has a mouse knockout model with facial cleft and failure of palatal fusion. The individual with variant in MMACHC presented with nsCPO. Conclusions Our study provides additional evidence for the role of TULP4, SOS1, MTHFD1L and MMACHC genes in nsOFC development. This is the first time MMACHC is implicated in nsOFC development in humans.
... 5 SOC represents a genetic defect (monogenic or chromosomal aberration), and the majority of their causal genes have been successfully identified. 6 On the other hand, 70% of all clefts are NSOC, 7 which have a multifactorial etiology resulting from both genetic and environmental factors identified. 6 Socioeconomic status, family, neighborhood, and social network context are some environmental factors that can influence epigenetic regulation through DNA methylation, histone modifications, and non-coding RNAs, thereby altering gene expression and resulting in clefts. ...
... 6 On the other hand, 70% of all clefts are NSOC, 7 which have a multifactorial etiology resulting from both genetic and environmental factors identified. 6 Socioeconomic status, family, neighborhood, and social network context are some environmental factors that can influence epigenetic regulation through DNA methylation, histone modifications, and non-coding RNAs, thereby altering gene expression and resulting in clefts. 8,9 Our hypothesis is that differences in lifestyle and the environment that exist between geographic living areas (coast vs. inland, rural vs. urban) are associated with differences in the frequency of OC. [10][11][12] These differences are generally related to geographic characteristics, such as air and water quality, health services and food availability, green space, and working conditions. ...
Article
Full-text available
Objetive: To analyze if differences in lifestyle and environment between coastal and inland areas are associated with differences in frequency of orofacial cleft types. Design: Populational cross-sectional study. Setting: All live borns with orofacial cleft registered at Brazilian Live Birth Information System between 1999 and 2020. Participants: 33,699 live borns with orofacial cleft. Intervention: Data from borns with orofacial cleft were collected at Brazilian Live Birth Information System. Main outcome measure: Differences in frequencies between the cleft types and covariates were determined using chi-square. Bivariate analysis was done to obtain the prevalence ratio of types of clefts by geographic origin. Multiple logistic regression analysis was used to determine adjusted odds ratios, controlling for covariates, establishing a significance level of p value <0.05. Results: The frequency of cleft types was statistically significant different according to geographic origin (inland x coast). For syndromic clefts, the prevalence ratio for cleft lip with/without palate was 3.6 times higher inland (p value = 0.000). Regarding non-syndromics, the prevalence ratio for cleft lip with/without palate was two times higher inland (p value = 0.000). Logistic regression suggested cleft lip with/without palate was 6.33 more likely to occur in inland regions (p value = 0.000). Conclusion: Geographic origin was associated with the type of cleft in Brazil, with a higher prevalence of cleft lip with/without palate in inland areas, compared to cleft palate, which was higher in the coast.
... Several studies have identified genetic loci and candidate gene polymorphisms associated with OFCs. 57 Epigenetics, involving reversible changes in gene expression without altering the DNA sequence, can be impacted by metabolic status, potentially contributing to congenital anomalies, including OFCs. 58 Additional investigation is required to specifically examine the link between maternal environment risk factors, including metabolic status, and susceptibility genes contributing to the development of OFCs. ...
... As with many complex traits, the etiology of NSOFCs is multifactorial, with genetic factors playing a considerable role 6 . According to a recent review, over 60 (> 40 associated with NSCL/P) risk loci have been implicated mainly through common variant association studies 7 . However, all identi ed loci/genes are estimated to explain only a small fraction (~ 25% for NSCL/P and even less for NSCPO) of the estimated heritability 8 . ...
Preprint
Full-text available
Non-syndromic orofacial clefts (NSOFCs) are common birth defects with a complex etiology. While over 60 common risk loci have been identified, they explain only a small proportion of the heritability for NSOFC. Rare variants have been implicated in the missing heritability. Thus, our study aimed to identify genes enriched with nonsynonymous rare coding variants associated with NSOFCs. Our sample included 814 non-syndromic cleft lip with or without palate (NSCL/P), 205 non-syndromic cleft palate only (NSCPO), and 2150 unrelated control children from Nigeria, Ghana, and Ethiopia. We conducted a gene-based analysis separately for each phenotype using three rare-variants collapsing models: (1) protein-altering (PA), (2) missense variants only (MO); and (3) loss of function variants only (LOFO). Subsequently, we utilized relevant transcriptomics data to evaluate associated gene expression and examined their mutation constraint using the gnomeAD database. In total, 13 genes showed suggestive associations (p = E-04). Among them, eight genes (ABCB1, ALKBH8, CENPF, CSAD, EXPH5, PDZD8, SLC16A9, and TTC28) were consistently expressed in relevant mouse and human craniofacial tissues during the formation of the face, and three genes (ABCB1, TTC28, and PDZD8) showed statistically significant mutation constraint. These findings underscore the role of rare variants in identifying candidate genes for NSOFCs. Main documents (excluding the methods section) word count: 2145
... The etiology of the nonsyndromic cleft palate is multifactorial, involving both genetic and environmental factors (Gritli-Linde, 2008), and recent genome-wide association studies and gene knockout animal experiments have identified many genetic variants (Alade et al., 2022). Genetic variants in STAT3 have been found to be positively associated [nonspecific positive correlation (rs1905339, P=0.01; rs744166, P=0.02)] with nonsyndromic cleft lip and/or palate (Vieira et al., 2008). ...
Article
Full-text available
Cleft palate has a multifactorial etiology. In palatal fusion, the contacting medial edge epithelium (MEE) forms the epithelial seam, which is subsequently removed with the reduction of p63. Failure in this process results in a cleft palate. We herein report the involvement of janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling in palatal fusion and that folic acid rescues the fusing defect by reactivating JAK2/STAT3. In closure of bilateral palatal shelves, STAT3 phosphorylation was activated at the fusing MEE and mesenchyme underlying the MEE. JAK2 inhibition by AG490 inhibited STAT3 phosphorylation and resulted in palatal fusion failure without removal of the epithelial seam, in which p63 and keratin 17 (K17) periderm markers were retained. Folic acid application restored STAT3 phosphorylation in AG490-treated palatal explants and rescued the fusion defect, in which the p63- and K17-positive epithelial seam were removed. The AG490-induced palatal defect was also rescued in p63 haploinsufficient explants. These findings suggest that JAK2/STAT3 signaling is involved in palatal fusion by suppressing p63 expression in MEE and that folate restores the fusion defect by reactivating JAK2/STAT3.
... 2 In 70% of cases, Cleft Lip and/or Palate present as Non-Syndromic (NSCL/P), meaning they occur without structural malformations in other organs or behavioral and cognitive alterations. 3,4 Based on epidemiological features and embryologic timing, NSCL/P are traditionally classified into Cleft Lip (NSCL), Cleft Lip and Palate (NSCLP), and Cleft Palate (NSCP). 5 The incidence of NSCL/P is approximately 1 in 500-2,000 live births, varying according to the geographical location, ethnicity, and socioeconomic status of the population studied. 1 In Brazil, studies on the incidence of NSCL/P are scarce and show considerable variation. ...
Article
Full-text available
Objective: The aim of this case-control study was to investigate environmental factors, such as caffeine, folic acid, nutritional iron supplementation, multivitamin complexes, alcohol, and tobacco (second-hand smoking), which have been described as risk factors for the development of oral clefts. Methods: This case-control study employed convenience sampling and included 409 mothers: 132 with children with oral clefts (cases) and 277 with children without oral clefts (controls). The age range of the children in both groups was 0 to 2 years. A questionnaire was administered to each mother to inquire about their habits and food consumption during the first trimester of pregnancy. Results: Folic acid supplementation was observed in 116 (87.8%) of the case group (p < 0.001) and 271 (97.8%) of the control group. Regarding the use of ferrous sulfate, 114 (86.3%) of the case group and 271 (97.8%) of the control group reported using it. In the case group, 84 (63.6%) mothers reported being exposed to second-hand smoke, and 5 (3.7%) reported alcohol consumption (p = 0.797). In terms of caffeine consumption, 127 mothers (95.4%) in the case group consumed it (p = 0.13), while 247 (88.8%) reported consumption in the control group. Conclusions: The results suggest a direct relationship between secondhand smoke, alcohol consumption, and the lack of maternal supplementation with oral clefts.
... However, a hereditary component in the development of cleft formation is undeniable even though the exact genetic mechanism is not yet fully understood, and more than 50 genes have been described to be associated with non-syndromic patients with a CL/P [35]. For instance, T-box and MSX genes are believed to play a major role in palatogenesis [36][37][38]. Interestingly, these gene families are also involved in heart development [39][40][41][42]. ...
Article
Full-text available
Objective The aim was to provide epidemiological and clinical data on patients with orofacial clefts in Lower Saxony in Germany. Materials and methods The records of 404 patients with orofacial clefts treated surgically at the University Medical Center Goettingen from 2001 to 2019 were analyzed in this retrospective study. Prevalence of orofacial clefts in general, orofacial clefts as manifestation of a syndrome, sex distribution, and prevalence of different cleft types was evaluated and associated with the need for corrective surgery, family history, pregnancy complications, and comorbidities. Results The prevalence of orofacial clefts for Goettingen in Lower Saxony was 1:890. 231 patients were male and 173 were female. CLP was most common (39.1%) followed by CP (34.7%), CL (14.4%), CLA (9.9%), and facial clefts (2%). The left side was more frequently affected and unilateral cleft forms occurred more often than bilateral ones. Almost 10% of the population displayed syndromic CL/P. 10.9% of all patients had a positive family history regarding CL/P, predominantly from the maternal side. Pregnancy abnormalities were found in 11.4%, most often in the form of preterm birth. Comorbidities, especially of the cardiovascular system, were found in 30.2% of the sample. 2.2% of patients treated according to the University Medical Center Goettingen protocol corrective surgery was performed in form of a velopharyngoplasty or residual hole closure. Conclusions The epidemiological and clinical profile of the study population resembled the expected distributions in Western populations. The large number of syndromic CL/P and associated comorbidities supports the need for specialized cleft centers and interdisciplinary cleft care.
Article
Full-text available
Non-syndromic orofacial clefts (NSOFCs) are common birth defects with a complex etiology. While over 60 common risk loci have been identified, they explain only a small proportion of the heritability for NSOFCs. Rare variants have been implicated in the missing heritability. Thus, our study aimed to identify genes enriched with nonsynonymous rare coding variants associated with NSOFCs. Our sample included 814 non-syndromic cleft lip with or without palate (NSCL/P), 205 non-syndromic cleft palate only (NSCPO), and 2150 unrelated control children from Nigeria, Ghana, and Ethiopia. We conducted a gene-based analysis separately for each phenotype using three rare-variants collapsing models: (1) protein-altering (PA), (2) missense variants only (MO); and (3) loss of function variants only (LOFO). Subsequently, we utilized relevant transcriptomics data to evaluate associated gene expression and examined their mutation constraint using the gnomeAD database. In total, 13 genes showed suggestive associations (p = E−04). Among them, eight genes (ABCB1, ALKBH8, CENPF, CSAD, EXPH5, PDZD8, SLC16A9, and TTC28) were consistently expressed in relevant mouse and human craniofacial tissues during the formation of the face, and three genes (ABCB1, TTC28, and PDZD8) showed statistically significant mutation constraint. These findings underscore the role of rare variants in identifying candidate genes for NSOFCs.
Preprint
Full-text available
Background Children born with non-syndromic orofacial clefts are an at-risk population for neuropsychiatric disorders. In a previous study, we have observed a significantly higher proportion of previous psychiatric care, special education needs, and cognitive difficulties in children with cleft compared to their non-cleft controls. Our goal was to strengthen these observations by screening these children with a clinically well-established neuropsychiatric tool. Methods A follow-up study of our previous cohort was carried out in the Department of Pediatrics of the University of Pécs. The cohort included children with non.syndromic orofacial clefts and healthy controls. The M.I.N.I Kid semi-structured interview was used to screen for neuropsychiatric disorders. A parental questionnaire was used to collect demographicand clinical data. Chi2-test or Fischer's Exact test were used to analyze categorical data. Two-sided independent samples Student’s t-test were used for quantitative variables. Results The data of 32 non-syndromic cleft and 41 control participants were analyzed. Children with non-syndromic orofacial clefts represented a higher proportion of psychiatric diagnoses compared to their controls, significantly major depressive episode (12,5%), panic disorder (15,6%), and conduct disorder (9,4%). Subgroup analysis revealed higher proportions of diagnoses in children with complex (combined and/or bilateral) and left-sided clefts. Conclusions A higher proportion of neuropsychiatric diagnoses were observed in children with non-syndromic orofacial clefts compared to controls. Screening this population at an early age is an important part of their overall clinical care.
Article
Full-text available
The identification of genetic risk factors for non-syndromic oral clefts is of great importance for better understanding the biological processes related to this heterogeneous and complex group of diseases. Herein we applied whole-exome sequencing to identify potential variants related to non-syndromic cleft palate only (NSCPO) in the multiethnic Brazilian population. Thirty NSCPO samples and 30 sex- and genetic ancestry-matched healthy controls were pooled (3 pools with 10 samples for each group) and subjected to whole-exome sequencing. After filtering, the functional affects, individually and through interactions, of the selected variants and genes were assessed by bioinformatic analyses. As a group, 399 variants in 216 genes related to palatogenesis/cleft palate, corresponding to 6.43%, were exclusively identified in the NSCPO pools. Among those genes are 99 associated with syndromes displaying cleft palate in their clinical spectrum and 92 previously related to cleft lip palate. The most significantly biological processes and pathways overrepresented in the NSCPO-identified genes were associated with the folic acid metabolism, highlighting the interaction between LDL receptor-related protein 6 (LRP6) and 5-methyltetrahydrofolate-homocysteine methyltransferase (MTR) that interconnect two large networks. This study yields novel data on characterization of specific variants and complex processes and pathways related to NSCPO, including many variants in genes of the folate/homocysteine pathway, and confirms that variants in genes related to syndromic cleft palate and cleft lip-palate may cause NSCPO.
Article
Full-text available
Objective: To identify new microRNA (miRNA)-mRNA networks in non-syndromic cleft lip with or without cleft palate (NSCL/P). Materials and Methods: Overlapping differentially expressed miRNAs (DEMs) were selected from cleft palate patients (GSE47939) and murine embryonic orofacial tissues (GSE20880). Next, the target genes of DEMs were predicted by Targetscan, miRDB, and FUNRICH, and further filtered through differentially expressed genes (DEGs) from NSCL/P patients and controls (GSE42589), MGI, MalaCards, and DECIPHER databases. The results were then confirmed by in vitro experiments. NSCL/P lip tissues were obtained to explore the expression of miRNAs and their target genes. Results: Let-7c-5p and miR-193a-3p were identified as DEMs, and their overexpression inhibited cell proliferation and promoted cell apoptosis. PIGA and TGFB2 were confirmed as targets of let-7c-5p and miR-193a-3p, respectively, and were involved in craniofacial development in mice. Negative correlation between miRNA and mRNA expression was detected in the NSCL/P lip tissues. They were also associated with the occurrence of NSCL/P based on the MGI, MalaCards, and DECIPHER databases. Conclusions: Let-7c-5p-PIGA and miR-193a-3p-TGFB2 networks may be involved in the development of NSCL/P.
Article
Full-text available
Orofacial cleft (OFC) is one of the most prevalent birth defects, leading to substantial and long-term burdens in a newborn’s quality of life. Although studies revealed several genetic variants associated with the birth defect, novel approaches may provide additional clues about its etiology. Using the Center for Craniofacial and Dental Genetics project data (n = 10,542), we performed linear mixed-model analyses to study the genetic compositions of OFC and investigated the dependence among identified loci using conditional analyses. To identify genes associated with OFC, we conducted a transcriptome-wide association study (TWAS) based on predicted expression levels. In addition to confirming the previous findings at four loci, 1q32.2, 8q24, 2p24.2 and 17p13.1, we untwined two independent loci at 1q32.2, TRAF3IP3 and IRF6. The sentinel SNP in TRAF3IP3 (rs2235370, p-value = 5.15 × 10−9) was independent of the sentinel SNP at IRF6 (rs2235373, r2 < 0.3). We found that the IRF6 effect became nonsignificant once the 8q24 effect was conditioned, while the TRAF3IP3 effect remained significant. Furthermore, we identified nine genes associated with OFC in TWAS, implicating a glutathione synthesis and drug detoxification pathway. We identified some meaningful additions to the OFC etiology using novel statistical methods in the existing data.
Preprint
Full-text available
Heritability, the proportion of phenotypic variance explained by genetic factors, can be estimated from pedigree data ¹ , but such estimates are uninformative with respect to the underlying genetic architecture. Analyses of data from genome-wide association studies (GWAS) on unrelated individuals have shown that for human traits and disease, approximately one-third to two-thirds of heritability is captured by common SNPs 2–5 . It is not known whether the remaining heritability is due to the imperfect tagging of causal variants by common SNPs, in particular if the causal variants are rare, or other reasons such as over-estimation of heritability from pedigree data. Here we show that pedigree heritability for height and body mass index (BMI) appears to be fully recovered from whole-genome sequence (WGS) data on 21,620 unrelated individuals of European ancestry. We assigned 47.1 million genetic variants to groups based upon their minor allele frequencies (MAF) and linkage disequilibrium (LD) with variants nearby, and estimated and partitioned variation accordingly. The estimated heritability was 0.79 (SE 0.09) for height and 0.40 (SE 0.09) for BMI, consistent with pedigree estimates. Low-MAF variants in low LD with neighbouring variants were enriched for heritability, to a greater extent for protein altering variants, consistent with negative selection thereon. Cumulatively variants in the MAF range of 0.0001 to 0.1 explained 0.54 (SE 0.05) and 0.51 (SE 0.11) of heritability for height and BMI, respectively. Our results imply that the still missing heritability of complex traits and disease is accounted for by rare variants, in particular those in regions of low LD.
Chapter
Major advances in the diagnosis and treatment of oral clefts have been made in the past 50 years, and recent genetics and epidemiological studies have led to new theories about the causes of cleft lip and palate. Addressing issues that are relevant to clinicians, researchers and family members, this book is a comprehensive, well-illustrated, and up-to-date account of the many facets of this common disorder. The authors describe the embryological and molecular mechanisms of cleft causation, present and illustrate the genetic and epidemiological methods used to identify risk factors for oral clefts, and describe treatments by the various professionals of the cleft team. A section is also devoted to the integration of research findings into public health practice, including ethical and financial considerations. The book draws together such diverse disciplines as craniofacial development, gene mapping, epidemiology, medicine, ethics, health economics, and health policy and management, and it will be an invaluable reference work.
Article
Aim: To evaluate the risk of nonsyndromic orofacial clefts (NSOFCs) associated with LINE-1 methylation, as a marker of global DNA methylation, and the effect of MTHFR functional variants on this variable. Patients & methods: LINE-1 methylation was evaluated by bisulfite modification coupled to DNA pyrosequencing in 95 NSOFC cases and 95 controls. In these subjects, MTHFR genotypes for variants c.C677T (rs1801133) and c.A1298C (rs1801131) were obtained. Results: Middle levels (second tertile) of LINE-1 methylation increase the risk of NSOFCs. In addition, LINE-1 methylation depends on c.A1298C genotypes in controls but not in cases. Conclusion: A nonlinear association between global DNA methylation and NSOFCs was detected in this Chilean population, which appears to be influenced by MTHFR functional variants.
Article
The aims of this review are (1) to illustrate current knowledge of the mode of inheritance and the loci involved in the cleft lip and palate and (2) to summarize the results of our investigations, which were carried out in Italy. It is well established that nonsyndromic cleft of the lip with or without the palate (CL±P) and cleft palate only (CPO) are separate entities. Genetic heterogeneity has been observed in CL±P, which involves different chromosome regions, mainly 6p23 (OFC1), 2q13 (OFC2), and 19q13.2 (OFC3), as well as other loci, such as 4q25-4q31.3 and 17q21. Furthermore, an interaction between different genes has been suggested in the oligogenic model. In one case at least, an OFC1 and OFC2 interaction has been demonstrated. The mode of inheritance of CPO is compatible with a recessive single major gene model, while an association with a candidate gene, mapping on the chromosome region 2q13/TGF α, remains to be confirmed.
Article
Nonsyndromic cleft lip with or without cleft palate (CL/P) is a complex disorder of multigenic origin involving between two and ten loci. Linkage and association studies of CL/P have implicated a number of candidate genes and regions but have often proved difficult to replicate. Here, we report the findings from a two-stage genome-wide scan of 92 affected sib-pairs to identify susceptibility loci to CL/P. An initial set of 400 microsatellite markers was used, with an average spacing of 10 cM throughout the genome. Eleven regions on eight chromosomes were found to have a P-value smaller than 0.05. These eight chromosomes were then further mapped with a second set of markers to increase the average map density to 5 cM. In seven out of eleven areas densely mapped, significance was markedly increased by decreasing the marker interval. Excessive allele sharing was found at 1p (NPL=2.35, P=0.009, MLS=1.51), 2p (NPL=1.77, P=0.04, MLS=0.66), 6p (NPL=2.35, P=0.009, MLS=1.34), 8q (NPL=2.15, P=0.015, MLS=1.51) 11cen (NPL=2.70, P=0.003, MLS=2.10), 12q (NPL=2.08, P=0.02, MLS=1.5), 16p (NPL=2.1, P=0.018, MLS=0.97) and Xcen-q (NPL=2.40, P=0.008, MLS=2.68). Although none reached the level required for significant susceptibility loci, two of these areas have previously been implicated in CL/P, viz. 2p13, an area harbouring the TGFA gene, and 6p23–24. We also demonstrate highly suggestive linkage to a susceptibility locus for nonsyndromic clefting on the X chromosome. Further studies are currently underway to replicate these findings in a larger cohort of affected sib-pairs.
Article
The genetic basis of disease has largely focused on coding regions. However, it has become clear that a large proportion of the noncoding genome is functional and harbors genetic variants that contribute to disease etiology. Here, we review recent examples of inherited noncoding alterations that are responsible for Mendelian disorders or act to influence complex traits. We explore both rare and common genetic variants and discuss the wide range of mechanisms by which they affect gene regulation to promote disease. We also debate the challenges and progress associated with identifying and interpreting the functional and clinical significance of genetic variation in the context of the noncoding regulatory landscape.
Article
Although de novo mutations (DNMs) are known to increase an individual’s risk of congenital defects, DNMs have not been fully explored regarding orofacial clefts (OFCs), one of the most common human birth defects. Therefore, whole-genome sequencing of 756 child-parent trios of European, Colombian, and Taiwanese ancestry was performed to determine the contributions of coding DNMs to an individual’s OFC risk. Overall, we identified a significant excess of loss-of-function DNMs in genes highly expressed in craniofacial tissues, as well as genes associated with known autosomal dominant OFC syndromes. This analysis also revealed roles for zinc-finger homeobox domain and SOX2-interacting genes in OFC etiology.