ArticlePDF Available

Expression of DLX6 Gene in Mandibular Deficiency (Retrognathic Mandible): A Randomized Clinical and Genetic Study

Authors:

Abstract and Figures

Introduction There are various genes that affect craniofacial development and among the important genes that affect jaw development is distal-less homeobox (DLX) 6 genes. The present study was carried out to determine the role of DLX6 gene variations in mandibular deficiency. Methods Thirty subjects having retrognathic mandible were evaluated by clinical examination and assessed using lateral cephalometric radiographs based on cephalometrics for orthognathic surgery (COGS) analysis of hard tissue with N-Pog parameters being less than -13 mm. For the same subjects, saliva samples were taken and sent to biotechnology labs for genetic evaluation. DNA was isolated from salivary samples using a DNA extraction kit and was subjected to polymerase chain reaction (PCR) amplification and sequencing. Single nucleotide polymorphisms (SNP) analysis was done to assess the role of DLX6 gene in these study subjects. Results All 30 subjects showed N-POG parameters of COGS analysis for hard tissue to be less than -13mm, confirming retrognathic mandible. SNP analysis of subjects showed no SNPs in any EXON of the DLX6 gene for all 30 study samples. Conclusion No variations in DLX6 gene were found in the present study. Further studies are required to investigate other genes that could be involved in the cause of retrognathic mandible with a larger sample size and to include subjects in the sample having features other than mandibular retrognathia like hearing loss, abnormal pinnae, ectrodactyly, cleft palate, developmental delay and abnormal teeth to determine the contribution of DLX6 gene variations in mandibular deficiency.
Content may be subject to copyright.
Review began 02/10/2021
Review ended 02/19/2021
Published 02/26/2021
© Copyright 2021
S J et al. This is an open access article
distributed under the terms of the
Creative Commons Attribution License
CC-BY 4.0., which permits unrestricted
use, distribution, and reproduction in any
medium, provided the original author and
source are credited.
Expression of DLX6 Gene in Mandibular
Deficiency (Retrognathic Mandible): A
Randomized Clinical and Genetic Study
Rajalakshmi S J , Nausheer Ahmed , Shashikala Kumari , Venkanna gudda Sreenivas Prasad , Lohit N.
Naik , Vinod Kumar
1. Orthodontics and Dentofaical Orthopedics, Government Dental College and Research Institute, Bangalore, IND 2.
Orthodontics and Dentofacial Orthopedics, Government Dental College and Research Institute, Bangalore, IND 3. Oral
and Maxillofacial Surgery, Yadgir Government District Hospital, Yadgir, IND 4. Pedodontics, Navodaya Dental College
and Hospital, Raichur, IND
Corresponding author: Vinod Kumar, dr.vinod990@gmail.com
Abstract
Introduction
There are various genes that affect craniofacial development and among the important genes that affect jaw
development is distal-less homeobox (DLX) 6 genes. The present study was carried out to determine the role
of DLX6 gene variations in mandibular deficiency.
Methods
Thirty subjects having retrognathic mandible were evaluated by clinical examination and assessed using
lateral cephalometric radiographs based on cephalometrics for orthognathic surgery (COGS) analysis of hard
tissue with N-Pog parameters being less than -13 mm. For the same subjects, saliva samples were taken and
sent to biotechnology labs for genetic evaluation. DNA was isolated from salivary samples using a DNA
extraction kit and was subjected to polymerase chain reaction (PCR) amplification and sequencing. Single
nucleotide polymorphisms (SNP) analysis was done to assess the role of DLX6 gene in these study subjects.
Results
All 30 subjects showed N-POG parameters of COGS analysis for hard tissue to be less than -13mm,
confirming retrognathic mandible. SNP analysis of subjects showed no SNPs in any EXON of the DLX6 gene
for all 30 study samples.
Conclusion
No variations in DLX6 gene were found in the present study. Further studies are required to investigate
other genes that could be involved in the cause of retrognathic mandible with a larger sample size and to
include subjects in the sample having features other than mandibular retrognathia like hearing loss,
abnormal pinnae, ectrodactyly, cleft palate, developmental delay and abnormal teeth to determine the
contribution of DLX6 gene variations in mandibular deficiency.
Categories: Genetics, Orthopedics, Anatomy
Keywords: dlx6 gene, genetic evaluation, retrognathic mandible
Introduction
There are various genes that affect craniofacial development and among the important genes that affect jaw
development is distal-less homeobox (DLX) 6 genes. DLX genes comprise a highly conserved family of
homeobox genes homologous to that distal-less (DLL) gene of drosophila. They are thought to act as
transcription factors. All DLX genes are expressed in spatially and temporally restricted patterns in
craniofacial primordia, basal telencephalon and diencephalon and in distal regions of extending appendages,
including the limb and the genital bud. DLX 5 and DLX 6 are expressed in differentiating osteoblasts [1].
Studies have shown that DLX 5 and DLX6 were expressed in all skeletal elements from initial cartilage
formation until ossification in mice [2]. In gnathostome jaws, DLX5 and DLX6 expression in neural crest
cells of pharyngeal arch 1 invariably determines lower jaw identity and maintains the myogenic program for
the formation of muscularized jaws [3]. In humans, three generations of the family showed five affected
members all of whom had hearing loss, micrognathia, abnormal pinnae and paracentric inversion of the
long arm of chromosome 7. Deregulation of DLX5 and DLX 6 genes were implicated in the pathogenesis of
the family’s phenotype [4].
The literature on the role of DLX6 gene as a single entity resulting in mandibular deficiency is
insufficient. This study will be helpful for the orthodontists to formulate treatment plans for mandibular
1 2 2 3
2 4
Open Access Original
Article DOI: 10.7759/cureus.13572
How to cite this article
S J R, Ahmed N, Kumari S, et al. (February 26, 2021) Expression of DLX6 Gene in Mandibular Deficiency (Retrognathic Mandible): A Randomized
Clinical and Genetic Study. Cureus 13(2): e13572. DOI 10.7759/cureus.13572
deficiency-associated malocclusion and it is also useful for genetic scientists and orthopedicians as a source
of complementary information. Hence this study centralized on evaluating DLX6 gene variations
contributing to mandibular deficiency.
Materials And Methods
It’s a pilot study. Thirty study subjects with retrognathic mandible (confirmed by cephalometric radiographs)
aged between 18 and 60 years were selected by a simple random sampling method. The syndrome patients,
systemic diseases patients, and uncooperative patients were excluded. Rights of human subjects were
protected; informed consent obtained from the patients and this study was approved by the institutional
ethical committee of Government Dental College and Research Institute, Bangalore, India. This study was
conducted from December 2010 to November 2012. Detailed case histories were elicited from the study
subjects. Subjects with retrognathic mandible were evaluated by clinical examination and confirmed using
lateral cephalometric radiographs based on cephalometrics for orthognathic surgery (COGS) analysis of hard
tissue with N-Pog parameters being less than -13 mm. Figure 1 shows the lateral cephalogram of a female
patient. Figure 2 shows cephalometric landmarks used in this study.
FIGURE 1: Lateral cephalogram.
2021 S J et al. Cureus 13(2): e13572. DOI 10.7759/cureus.13572 2 of 6
FIGURE 2: Cephalometric landmarks used in this study.
Landmarks used as shown in Figure 2: Sella (S) - The center of Pituitary Fossa, Nasion (N) - The most
anterior point of the nasofrontal suture in the mid-sagittal plane, supramentale (B) - The deepest point in
mid-sagittal plane on the concavity between infradentale and pogonion, Pogonion (Pg) - Most anterior mid-
sagittal point on the contour of the chin, Pterygomaxillary fissure (Ptm) - The most posterior point on the
anterior contour of the maxillary tuberosity, Articulare (Ar) - The intersection of basisphenoid and the
posterior border of the condyle, Gonion (Go) - Constructed by bisecting the posterior ramal plane and
mandibular plane.
For the same 30 subjects, unstimulated saliva samples were collected and stored in a refrigerator at -20
degree centigrade. 5-ml normal saline was given to the patient to hold it in mouth for one minute and then
spit it back to sterile sample collection falcon tube. Samples were sent to the biotechnology laboratory
within seven days (Chromous Biotech Pvt. Ltd., Bangalore). DNA was isolated from salivary samples using a
DNA extraction kit and was subjected to PCR (polymerase chain reaction) amplification. Once the gene was
amplified, it was subjected to DNA sequencing to check for single nucleotide polymorphisms (SNP). It was
done to see the role of DLX6 gene in these cases. DLX6 gene is a homeobox gene located on chromosome
7q22. It has 5,062 base pairs which include exons and introns. Exons are the protein-coding regions of the
gene. Introns are the non-coding regions of the gene. DLX6 gene has 3 exons. 1st exon has 82 base pairs, 2nd
exon has 200 base pairs and 3rd exon has 255 base pairs.
Procedure followed by the biotechnology lab for the gene evaluation:
Genomic DNA (gDNA) was isolated from the samples provided by using a DNA extraction kit. 2 µl of the
Genomic DNA isolated was loaded on 1 % Agarose gel.
PCR amplification of the gene was performed. Primers were designed by using the reference sequence of the
DLX6 gene of the human for all the 3 exons of DLX6 gene.
The PCR Mix of total 50µl was prepared which had 1µl template DNA, Forward Primer 2µl (200ng), Reverse
Primer 2µl (200ng), dNTPs (2.5mM each) 2 µl, 10X Taq DNA polymerase Assay Buffer 5 µl, Taq DNA
Polymerase Enzyme (3U/µl)-0.5µl, and Water 37.5µl.
The Exons 1, 2 and 3 of DLX6 gene was amplified by 35 cycles consisting of denaturation at 94°C for 30 sec,
with an initial denaturation for 5 min, annealing at 55°C for 30 sec, and primer extension at 72°C for 1 min,
with final extension for 5 minutes.
Reaction was cycled in a thermal cycler.
PCR products were then loaded on 1% Agarose gel and post-PCR products have been shown in Figure 3.
Sequencing of the PCR product was done by using ABI Sequencing machine ABI 3500 XL Genetic Analyzer.
2021 S J et al. Cureus 13(2): e13572. DOI 10.7759/cureus.13572 3 of 6
FIGURE 3: Post-PCR products on 1% agarose gel.
Grey bands represent the wells the prod uct (gDNA isolated from the saliva sample of the subject) was loaded
into, and the white bands represent DNA fragments produced by PCR. In this 100bp DNA ladders (Lanes 1, 2
and 3), the target fragments amplified by the primers were 300bp in size.
Lane 1 represents DNA ladder marker; Lanes 2 and 3 represent PCR amplified products of DLX6 gene of the
subject.
The demographic details of 30 study participants like age, gender and COGS were subjected to descriptive
statistics (percentage, mean, range, and standard deviation).
Results
The following were the results of this study: Out of 30 subjects, 15 were males and 15 were females.
For females, the age ranged from 18 years to 42 years with the mean age being 24 years and standard
deviation was 10.20 years. For males, the age ranged from 18years to 59 years with the mean age being 29.2
years and standard deviation was 14.62 years.
For females, the N-Pog parameter of COGS analysis ranged from -16 mm to -23 mm with the mean being -
20.40 mm and standard deviation was 3.21 mm. For males, the N-POG parameter of COGS analysis ranged
from -17 mm to -22 mm with the mean being -19.36 mm and standard deviation was 1.75 mm as shown in
Table 1.
Gender N-pog parameter in range (mm) Mean ± SD (mm)
Female -16 to -23 -20.40 ± 3.21
Male -17 to -22 -19.36 ± 1.75
TABLE 1: Mean N-pog parameter of study sample.
Genetic evaluation results: SNP analysis was done by comparing the reference data from NCBI (NCBI
reference sequence number for DLX6 gene on chromosome 7: NC_000007.13) with the sequence data
obtained from the 30 samples for all the three exons (exon 1, 2, 3) of DLX6 gene by using the Align tool of
Blast in NCBI and analyzing the mismatches if any to detect SNP (Figure 4). It showed no SNPs in any of the
exons of the DLX6 gene of all 30 samples indicating non-involvement of DLX6 gene in mandibular
2021 S J et al. Cureus 13(2): e13572. DOI 10.7759/cureus.13572 4 of 6
deficiency.
FIGURE 4: NCBI reference data for exon 1 of DLX6 gene in humans.
Discussion
Studies have shown that DLX5 and DLX6 are predominantly expressed by mandibular cranial neural crest
cells, which give rise to most bones and tendons of jaws [5]. It is documented in chick embryos that levels of
DLX5 and DLX6 are required for the maintenance of cellular proliferation during development and over
expression of DLX5 and DLX6 leads to generalized antagonism of cellular proliferation [6]. Evaluation of
phenotype of DLX5 and DLX6 double mutant mice suggest they specify mandibular identity [5]. A study on
gnathostome jaws shows that neural crest cells pharyngeal arch 1 specific DLX5 and DLX6 inactivation
generates severely hypomorphic lower jaws that have typical maxillary traits [3]. The role of DLX5 and DLX6
was evaluated in mice. The inactivation of DLX5 and DLX6 in mice had shown a multitude of craniofacial
and ear defects including the failure of Meckel’s cartilage, mandible, calvaria formation [7]. A study on dogs
has shown that a LINE-1 insertion within DLX6 is responsible for cleft palate and mandibular abnormalities,
which prompted sequencing of DLX5 and DLX6 in humans with Pierre Robin Sequence, where a missense
mutation within the highly conserved DLX5 homeobox was identified [8]. A study on humans has shown
multiple individuals with deletion of DLX5 and DLX6 who had reported severe phenotypes including
ectrodactyly, hearing loss, abnormal pinnae, cleft palate, lower jaw retrognathia, developmental delay and
abnormal teeth [9]. So DLX6 gene is an important developmental gene responsible for proper mandibular jaw
development in humans and its variations may lead to failure of proper development of mandibles in them.
In the present study, since no SNPs were found in DLX6 gene of the study subjects, the causative factor is
other than the SNPs, as retrognathic or a small mandible has a multifactorial etiology. Human study done
shows DLX6 gene affected members to be having features other than mandibular retrognathia like hearing
loss, abnormal pinnae, ectrodactyly, cleft palate, developmental delay and abnormal teeth. The samples
with any one of the above features if subjected to genetic evaluation probably would show mutations in
DLX6 gene.
Variation of the phenotype is a central issue in biology because it is the basis for individuality, adaptation of
populations to environmental fluctuations, and the evolution of biodiversity. Phenotypic variation can be
produced by genetic differences, environmental influences and stochastic developmental events [10]. The
concept that phenotype represents the consequence of genotype-environment interactions (GEI) is
universal and relates to all living organisms. In response to severe environmental changes, a genome can
respond by selectively regulating (increasing or decreasing) the expression of specific genes [11]. So in the
present study even though DLX6 gene variation or SNPs is not present, other genes or environmental factors
could have influenced and resulted in retrognathic mandible in these subjects.
Conclusions
It was concluded in the present study that DLX6 gene was not the cause for retrognathic mandible in the
2021 S J et al. Cureus 13(2): e13572. DOI 10.7759/cureus.13572 5 of 6
subjects of the present study. Retrognathic mandible has a multifactorial etiology so further studies are
required to investigate other genes that could be involved in the cause of retrognathic mandible and to
include subjects with different ethnicity in the sample who are also having features other than mandibular
retrognathia like hearing loss, abnormal pinnae, ectrodactyly, cleft palate, developmental delay and
abnormal teeth to determine the contribution of DLX6 gene variations in mandibular deficiency.
Additional Information
Disclosures
Human subjects: Consent was obtained or waived by all participants in this study. Institutional ethical
committee of Government Dental College and Research Institute, Bangalore, India issued approval NOT
APPLICABLE. Study has been approved . Animal subjects: All authors have confirmed that this study did
not involve animal subjects or tissue. Conflicts of interest: In compliance with the ICMJE uniform
disclosure form, all authors declare the following: Payment/services info: All authors have declared that no
financial support was received from any organization for the submitted work. Financial relationships: All
authors have declared that they have no financial relationships at present or within the previous three years
with any organizations that might have an interest in the submitted work. Other relationships: All authors
have declared that there are no other relationships or activities that could appear to have influenced the
submitted work.
References
1. Merlo GR, Zerega B, Paleari L, et al.: Multiple functions of DLX genes . Int J Dev Biol. 2000, 44:619-626.
2. Simeone A, Acampora D, Pannese M, et al.: Cloning and characterization of two members of the vertebrate
Dlx gene family. Proc NatI Acad Sci USA. 1994, 91:2250-2254. 10.1073/pnas.91.6.2250
3. Shimizu M, Narboux-Neme N, Gitton Y, et al.: Probing the origin of matching functional jaws: roles of
Dlx5/6 in cranial neural crest cells. Sci Rep. 2018, 8:14975. 10.1038/s41598-018-33207-2
4. Brown KK, Reiss JA, Crow K, et.al: Deletion of an enhancer near DLX5 and DLX6 in a family with hearing
loss, craniofacial defects and an. Human Genetics. 2010, 127:19. 10.1007/s00439-009-0736-4
5. Gitton Y, Narboux-Neme N and Levi G: Transitory expression of Dlx5 and Dlx6 in Maxillary arch epithelial
precursors is essential for upper jaw morphogenesis. F1000Res. 2014, 2:261. 10.12688/f1000research.2-
261.v3
6. Mackenzie RK, Sankar PR, Bendall AJ: Dlx5 and Dlx6 can antagonize cell division at G1/S checkpoint . BMC
Mol Cell Biol. 2019, 20:8. 10.1186/s12860-019-0191-6
7. Robledo RF, Rajan L, Li X, et al.: The Dlx5 and Dlx6 homeobox genes are essential for craniofacial, axial, and
appendicular skeletal development. Genes Dev. 2002, 16:1089-1101. 10.1101/gad.988402
8. Wolf ZT, Leslie EJ, Arzi B, et al.: A LINE-1 insertion in DLX6 is responsible for cleft palate and mandibular
abnormalities in a canine model of Pierre Robin sequence. PLoS Genet. 2014, 10:e1004257.
10.1371/journal.pgen.1004257
9. Scherer SW, Poorkaj P, Massa H, et al.: Physical mapping of the split hand/ split foot locus on chromosome 7
and implication in syndromicectrodactyly. Hum Mol Genet. 1994, 3:1345-1354. 10.1093/hmg/3.8.1345
10. Vogt G, Huber M, Thiemann M, et al.: Production of different phenotypes from the same genotype in the
same environment by developmental variation. J Exp Biol. 2008, 211:510-523. 10.1242/jeb.008755
11. Bayel TM, Abebe T,Wilke RA: Genotype-environment interactions and their translational implications . Per
Med. 2011, 8:59-70. 10.2217/pme.10.75
2021 S J et al. Cureus 13(2): e13572. DOI 10.7759/cureus.13572 6 of 6
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Background Dlx5 and Dlx6 stimulate differentiation of diverse progenitors during embryonic development. Their actions as pro-differentiation transcription factors includes the up-regulation of differentiation markers but the extent to which differentiation may also be stimulated by regulation of the cell cycle has not been addressed. Results We document that expression of Dlx5 and Dlx6 antagonizes cell proliferation in a variety of cell types without inducing apoptosis or promoting cell cycle exit. Rather, a variety of evidence indicates that elevated Dlx5 and Dlx6 expression reduces the proportion of cells in S phase and affects the length of the cell cycle. Conclusions Antagonism of S-phase entry by Dlx5 and Dlx6 proteins likely represents a lineage-independent function to effect Dlx-mediated differentiation in multiple progenitor cell types.
Article
Full-text available
Gnathostome jaws derive from the first pharyngeal arch (PA1), a complex structure constituted by Neural Crest Cells (NCCs), mesodermal, ectodermal and endodermal cells. Here, to determine the regionalized morphogenetic impact of Dlx5/6 expression, we specifically target their inactivation or overexpression to NCCs. NCC-specific Dlx5/6 inactivation (NCC∆Dlx5/6) generates severely hypomorphic lower jaws that present typical maxillary traits. Therefore, differently from Dlx5/6 null-embryos, the upper and the lower jaws of NCC∆Dlx5/6 mice present a different size. Reciprocally, forced Dlx5 expression in maxillary NCCs provokes the appearance of distinct mandibular characters in the upper jaw. We conclude that: (1) Dlx5/6 activation in NCCs invariably determines lower jaw identity; (2) the morphogenetic processes that generate functional matching jaws depend on the harmonization of Dlx5/6 expression in NCCs and in distinct ectodermal territories. The co-evolution of synergistic opposing jaws requires the coordination of distinct regulatory pathways involving the same transcription factors in distant embryonic territories.
Article
Full-text available
Asymmetric, articulated jaws support active predation in vertebrates; they derive from the first pharyngeal arch (PA1) which generates both maxillary and mandibular components. PA1 is colonized by cranial neural crest cells (CNCCs) which give rise to most bones and tendons of the jaws. The elements formed by different CNCCs contingents are specified by the combinatorial expression of Dlx genes. Dlx5 and Dlx6 are predominantly expressed by mandibular CNCCs. Analysis of the phenotype of Dlx5 and Dlx6 double mutant mice has suggested that they are necessary and sufficient to specify mandibular identity. Here, using 3D reconstruction, we show that inactivation of Dlx5 and Dlx6 does not only affect the mandibular arch, but results in the simultaneous transformation of mandibular and maxillary skeletal elements which assume a similar morphology with gain of symmetry. As Dlx5- and Dlx6 -expressing cells are not found in the maxillary bud, we have examined the lineage of Dlx5 -expressing progenitors using an in vivo genetic approach. We find that a contingent of cells deriving from precursors transiently expressing Dlx5 participate in the formation of the maxillary arch. These cells are mostly located in the distal part of the maxillary arch and might derive from its lambdoidal junction with the olfactory pit. Our findings extend current models of jaw morphogenesis and provide an explanation for the maxillary defects of Dlx5 and Dlx6 mutants. Our results imply that Dlx5 and Dlx6 model the upper and the lower PA1 components through different morphogenetic mechanisms which are, however, coordinated as they give rise to functional, articulated jaws.
Article
Full-text available
Asymmetric, articulated jaws are characteristic of most vertebrate species; they derive from the first pharyngeal arch (PA1) which generates both maxillary and mandibular components. PA1 is colonized by cranial neural crest cells (CNCCs) which give rise to most bones and tendons of the jaws. The elements formed by different CNCCs contingents are specified by the combinatorial expression of Dlx genes. Dlx5 and Dlx6 are predominantly expressed by mandibular CNCCs. Analysis of the phenotype of Dlx5 and Dlx6 double mutant mice has suggested that they are necessary and sufficient to specify mandibular identity. Here, using 3D reconstruction, we show that inactivation of Dlx5 and Dlx6 does not only affect the mandibular arch, but results in the simultaneous transformation of mandibular and maxillary skeletal elements which assume a similar morphology with gain of symmetry. As Dlx5- and Dlx6-expressing cells are not found in the maxillary bud, we have examined the lineage of Dlx5-expressing progenitors using an in vivo genetic approach. We find that a contingent of cells deriving from epithelial precursors transiently expressing Dlx5 participate in the formation of the maxillary arch. These cells are mostly located in the distal part of the maxillary arch and might derive from its lambdoidal junction with the olfactory pit. Our observations provide the first genetic demonstration of the ‘Hinge and Caps’ model[1]. We support the notion that ‘cap’ signals could originate from epithelial derivatives of Dlx5-expressing progenitors which migrate and colonize the maxillary arch epithelium. Our results imply that Dlx5 and Dlx6 control upper and lower jaw morphogenesis through different coordinated mechanisms to generate functional, articulated jaws.
Article
Full-text available
Cleft palate (CP) is one of the most commonly occurring craniofacial birth defects in humans. In order to study cleft palate in a naturally occurring model system, we utilized the Nova Scotia Duck Tolling Retriever (NSDTR) dog breed. Micro-computed tomography analysis of CP NSDTR craniofacial structures revealed that these dogs exhibit defects similar to those observed in a recognizable subgroup of humans with CP: Pierre Robin Sequence (PRS). We refer to this phenotype in NSDTRs as CP1. Individuals with PRS have a triad of birth defects: shortened mandible, posteriorly placed tongue, and cleft palate. A genome-wide association study in 14 CP NSDTRs and 72 unaffected NSDTRs identified a significantly associated region on canine chromosome 14 (24.2 Mb-29.3 Mb; praw = 4.64×10-15). Sequencing of two regional candidate homeobox genes in NSDTRs, distal-less homeobox 5 (DLX5) and distal-less homeobox 6 (DLX6), identified a 2.1 kb LINE-1 insertion within DLX6 in CP1 NSDTRs. The LINE-1 insertion is predicted to insert a premature stop codon within the homeodomain of DLX6. This prompted the sequencing of DLX5 and DLX6 in a human cohort with CP, where a missense mutation within the highly conserved DLX5 homeobox of a patient with PRS was identified. This suggests the involvement of DLX5 in the development of PRS. These results demonstrate the power of the canine animal model as a genetically tractable approach to understanding naturally occurring craniofacial birth defects in humans.
Article
Full-text available
Organisms frequently encounter different environmental conditions. The physiological and behavioral responses to these conditions depend on the genetic make up of individuals. Genotype generally remains constant from one environment to another, although occasional spontaneous mutations may occur which cause it to change. However, when the same genotype is subjected to different environments, it can produce a wide range of phenotypes. These phenotypic variations are attributable to the effect of the environment on the expression and function of genes influencing the trait. Changes in the relative performance of genotypes across different environments are referred to as genotype-environment interactions (GEI). A general argument for research on the impact of GEI in common diseases is that it provides insights into disease processes at the population, individual and molecular levels. In humans, GEI is complicated by multiple factors including phenocopies, genocopies, epigenetics and imprinting. A better understanding of GEI is essential if patients are to make informed health choices guided by their genomic information. In this article, we clarify the role of the environment on phenotype, we describe how human population structure can obscure the resolution of GEI and we discuss how emerging biobanks across the globe can be coordinated to further our understanding of genotype-phenotype associations within the context of varying environment.
Article
Full-text available
Precisely regulated temporal and spatial patterns of gene expression are essential for proper human development. Cis-acting regulatory elements, some located at large distances from their corresponding genes, play a critical role in transcriptional control of key developmental genes and disruption of these regulatory elements can lead to disease. We report a three generation family with five affected members, all of whom have hearing loss, craniofacial defects, and a paracentric inversion of the long arm of chromosome 7, inv(7)(q21.3q35). High resolution mapping of the inversion showed that the 7q21.3 breakpoint is located 65 and 80 kb centromeric of DLX6 and DLX5, respectively. Further analysis revealed a 5,115 bp deletion at the 7q21.3 breakpoint. While the breakpoint does not disrupt either DLX5 or DLX6, the syndrome present in the family is similar to that observed in Dlx5 knockout mice and includes a subset of the features observed in individuals with DLX5 and DLX6 deletions, implicating dysregulation of DLX5 and DLX6 in the family's phenotype. Bioinformatic analysis indicates that the 5,115 bp deletion at the 7q21.3 breakpoint could contain regulatory elements necessary for DLX5 and DLX6 expression. Using a transgenic mouse reporter assay, we show that the deleted sequence can drive expression in the inner ear and developing bones of E12.5 embryos. Consequently, the observed familial syndrome is likely caused by dysregulation of DLX5 and/or DLX6 in specific tissues due to deletion of an enhancer and possibly separation from other regulatory elements by the chromosomal inversion.
Article
Full-text available
A number of vertebrate genes of the Dlx gene family have been cloned in mouse, frog, and zebrafish. These genes contain a homeobox related to that of Distalless, a gene expressed in the developing head and limbs of Drosophila embryos. We cloned and studied the expression of two members of this family, which we named Dlx5 and Dlx6, in human and mouse. The two human genes, DLX5 and DLX6, are closely linked in an inverted convergent configuration in a region of chromosome 7, at 7q22. Similarly, the two human genes DLX1 and DLX2 are closely linked in a convergent configuration at 2q32, near the HOXD (previously HOX4) locus. In situ hybridization experiments in mouse embryos revealed expression of Dlx5 and Dlx6 mRNA in restricted regions of ventral diencephalon and basal telencephalon, with a distribution very similar to that reported for Dlx1 and Dlx2 mRNA. A surprising feature of Dlx5 and Dlx6 is that they are also expressed in all skeletal structures of midgestation embryos after the first cartilage formation. The expression pattern of these genes, together with their chromosome localization, may provide useful cues for the study of congenital disorders in which there is a combination of craniofacial and limb defects.
Article
Full-text available
Dlx genes comprise a highly conserved family of homeobox genes homologous to the distal-less (Dll) gene of Drosophila. They are thought to act as transcription factors. All Dlx genes are expressed in spatially and temporally restricted patterns in craniofacial primordia, basal telencephalon and diencephalon, and in distal regions of extending appendages, including the limb and the genital bud. Most of them are expressed during morphogenesis of sensory organs and during migration of neural crest cells and interneurons. In addition, Dlx5 and Dlx6 are expressed in differentiating osteoblasts. Gene targeting of Dlx1, Dlx2, Dlx3 and Dlx5 in the mouse germ-line has revealed functions in craniofacial patterning, sensory organ morphogenesis, osteogenesis and placental formation. However, no effect on limb development has yet been revealed from gene inactivation studies. A role for these genes in limb development is however suggested by the linkage of the Split Foot/Hand Malformation human syndrome to a region containing DLX5 and DLX6. As for most transcription factors, these genes seem to have multiple functions at different stages of development or in different tissues and cell types.
Article
Split hand/split foot (ectrodactyly; SHSF) is a human developmental malformation characterized by missing digits and claw-like extremities. An autosomal dominant form of this disorder has been mapped to 7q21.3-q22.1; the locus has been designated SHFD1. We have constructed a physical map consisting of overlapping yeast artificial chromosome clones for the entire region. Somatic cell hybrid and fluorescent in situ hybridization analyses were used to define SHSF-associated chromosomal rearrangements in twelve patients. An SHFD1 critical interval of 1.5 Mb was established by analysis of five patients with deletions. Translocation or inversion breakpoints found in six patients were mapped within 700 kb of each other in the critical region. Of note is that eight of the patients analyzed (67%) are in fact classified as having syndromic ectrodactyly. Thus, these mapping data establish a relationship between simple split hand/split foot and this more complex group of human birth defects. Finally, we have mapped DLX5, a member of the Distal-less homeobox gene family, to the SHFD1 critical interval.