#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Two novel mutations in MSX1 causing oligodontia


Authors: Le Yang aff001;  Jia Liang aff001;  Haitang Yue aff001;  Zhuan Bian aff001
Authors place of work: The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei Province, China aff001
Published in the journal: PLoS ONE 15(1)
Category: Research Article
doi: https://doi.org/10.1371/journal.pone.0227287

Summary

Tooth agenesis is one of the most common developmental anomalies in humans and can affect dental occlusion and speech pronunciation. Research has identified an association between mutations in MSX1, PAX9, EDA, AXIN2, WNT10A, WNT10B and LRP6 and human tooth agenesis. Two unrelated individuals with non-syndromic tooth agenesis and their families were enrolled in this study. Using Sanger sequencing of the candidate genes, we identified two novel mutations: a missense mutation c.572 T>C and a frameshift mutation c.590_594 dup TGTCC, which were both detected in the homeodomain of MSX1. After identifying the mutations, structural modeling and bioinformatics analysis were used to predict the resulting conformational changes in the MSX1 homeodomain. Combined with 3D-structural analysis of other MSX1 mutations, we propose that there is a correlation between the observed phenotypes and alterations in hydrogen bond formation, thereby potentially affecting protein binding.

Keywords:

Teeth – Dentition – Hydrogen bonding – Mutation databases – Insertion mutation – Missense mutation – Homeobox – Frameshift mutation

Introduction

Tooth agenesis is a common developmental anomaly of the human dentition. In the general population, the incidence of tooth agenesis of permanent teeth ranges from 2.2% to 10.1% [1]. Several terms are used to describe tooth agenesis: hypodontia is referred as an absence of less than six teeth excluding the third molars; oligodontia is an absence of six or more teeth excluding the third molars; and anodontia is the complete absence of teeth [2]. Tooth agenesis is classified as syndromic when it is accompanied by other inherited abnormalities including nail dysplasia [3], sparse hair, lack of sweat glands or a cleft lip and palate [48], and classified as non-syndromic when the absence of teeth is an isolated characteristic. Tooth agenesis occurs either in a sporadic or in a hereditary manner, in autosomal dominant [9], autosomal recessive [10] or X-linked patterns [11]. The etiology of tooth agenesis is complex and not yet completely elucidated [2]. Environmental factors potentially influencing the dental development include trauma, chemotherapeutic drugs, radiotherapy or thalidomide use during pregnancy [12, 13]. The majority of cases are caused by genetic mutations, and to date, mutations in the MSX1, PAX9, AXIN2, WNT10A, EDA, EDAR, EDARADD, WNT10B and LRP6 genes have been associated with non-syndromic tooth agenesis (NSTA) cases [1422].

MSX1 and PAX9 are the first genes identified causing NSTA [17, 19]. Both genes are transcription factors that play crucial part during the bud to cap stages of odontogenic development. Msx1 and Pax9 can interact with each other and they also act synergistically to activate the Bmp4 which is critical for tooth development [23]. EDA, EDAR and EDARADD are candidate genes of both NSTA and STA. EDA/EDAR/EDARADD signaling has been shown to play an important role in NSTA [24]. Recently, more and more genes in Wnt signaling pathway have been found to be related to NSTA. The first gene of Wnt signaling pathway involved in NSTA was AXIN2 [20]. This gene was also found to be associated with a variety of cancers [25]. Axin2 plays a critical role in regulating the stability of β-catenin in the Wnt signaling pathway [26]. The second gene was WNT10A which encodes a secreted signaling protein. WNT10A mutations have been reported to be linked to a majority of NSTA cases [16]. Wnt10a is a key mediator of Wnt signaling which is required for proper tooth development [27]. In 2015, mutations in the WNT co-receptor LRP6 were identified in families with autosomal-dominant non-syndromic oligodontia [15]. In 2016, WNT10B mutations causing oligodontia had been reported in a Chinese population [14].

Among those genes, NSTA is mostly associated with mutations in PAX9, MSX1, WNT10A, AXIN2 and EDA [2830]. Therefore, in this study, we searched for mutations in the above five genes in two unrelated individuals with non-syndromic oligodontia. The study objectives were: 1) to identify the mutations responsible for the tooth agenesis in two Chinese patients; 2) to use 3D-structure modeling to explain the reasons why these mutations can lead to tooth agenesis.

Materials and methods

Subjects

Two unrelated Chinese oligodontia patients were identified in the orthodontic department at the School of Stomatology, Wuhan University. Intraoral examination and panoramic radiographs were taken to verify the number and location of missing teeth of each patient. Pedigree construction was achieved using clinical examinations and verbal interviews with the available family members. Blood samples were collected from the probands, their available family members and 300 unrelated healthy controls. All procedures were approved by the National Natural Science Foundation of the Medical Ethics Committee of the Stomatological Hospital of Wuhan University, China (Ethics Approval Identifier: A 42, date of approval: March 4, 2019). All participants in this study gave their written informed consent to publish these case details.

Identification of mutations

Genomic DNA was extracted from the peripheral blood samples of all consenting family members and controls according to standard salt extraction procedures [31]. Screening of pathogenic mutations was performed using polymerase chain reaction (PCR) amplification and sequencing the complete exons and exon–intron boundaries of these five genes: MSX1 (ENSG00000163132), PAX9 (ENSG00000198807), EDA (ENSG00000158813), AXIN2 (ENSG00000168646) and WNT10A (ENSG00000135925).

Multiple sequence alignments

Multi-species amino acid sequence alignment of the MSX1 protein sequence (NP_002439.2) was performed using Clustal X (https://www.ebi.ac.uk/Tools/msa/). MSX1 sequences from dolphin to human were obtained from ENSEMBL.

3D modeling of MSX1 variants

On the basis of the crystal structure of the MSX1 homeodomain complex with DNA [32], the three-dimensional structure of the wild-type MSX1 homeodomain was derived using SWISS-MODEL (https://www.swissmodel.expasy.org) with PDB: 1ig7.1.C as a template. The 3D model was constructed using Swiss Pdb Viewer V 4.1 [33]. Visualization of the three-dimensional (3D) structures was performed with PyMOL (The PyMOL Molecular Graphics System, Version 0.99rc6, Schrödinger, LLC., Cambridge, MA, USA).

Statistical analysis

Differences between the average number of missing teeth caused by missense mutations in the MSX1 homeodomain with or without alterations in hydrogen bonding were analyzed using an unpaired, two-tailed t-test with GraphPad Prism 5 (GraphPad Software Inc., La Jolla, CA, USA). P-value < 0.05 was considered significant.

Results

Pedigree and phenotype analysis

Pedigree analysis of the two families revealed that the proband (Ⅱ2) in family 1 was a sporadic patient and oligodontia in family 2 was exhibited in an autosomal dominant manner (Fig 1A). The proband (Ⅱ2) in family 1 was a 16-year-old female with the congenital absence of ten permanent teeth excluding third molars. The second upper deciduous teeth were retained (Fig 1B and 1C), and both of her parents and her elder brother had normal dentition. The proband (Ⅲ1) in family 2 was a 21-year-old male with more severe tooth agenesis, characterized by the absence of twenty permanent teeth excluding the third molars. The shape of 25 was conic, and most deciduous teeth were retained (Fig 1B and 1C). The mother of the proband (Ⅱ2) was also diagnosed as tooth agenesis through oral examination. However, she could not recall or confirm extraction of some teeth. No orofacial cleft or other craniofacial abnormalities were noted in the affected members of the two families. Additionally, all reported subjects had normal primary dentition, nails, skin, and hair.

Identification of two Chinese families with non-syndromic tooth agenesis.
Fig. 1. Identification of two Chinese families with non-syndromic tooth agenesis.
(A) Pedigrees. Filled and unfilled symbols indicate affected and unaffected individuals, respectively. Squares and circles represent males and females, respectively. (B) Panoramic radiograph of the probands. The white stars indicate missing permanent teeth. Left: The proband of family I. Right: The proband of family II. (C) Tooth phenotypes of the probands with oligodontia. Stars indicate congenital missing tooth and triangle denotes cone-shaped teeth.

Mutation analysis

Mutation analysis of candidate genes detected two novel heterozygous mutations in MSX1 in two separate families. The proband of family 1 demonstrated a missense mutation at c.572T>C (Fig 2A). Her parents were wild-type at c.572, indicating a de novo mutation. A cross-species alignment of the MSX1 protein showed that p. Phe191 is highly conserved in the homeodomain (Fig 2B). This mutation replaced the hydrophobic phenylalanine with a hydrophilic serine at amino acid position 191. Screening of candidate genes in the proband of family 2 showed a frameshift insertion c.590_594 dup TGTCC in MSX1 (Fig 2C). His mother also carried the mutation. This mutation is occurred in the homeodomain of MSX1, introducing 20 novel amino acids following the first 198 amino acids, while deleting the normal protein sequence from Ile199 through The303. Neither mutation was detected in the control group and had not been reported by 1000 Genomes, dbSNP, Human Gene Mutation Database (professional version) or PubMed. No pathogenic mutations were found in the PAX9, AXIN2, EDA and WNT10A genes of these two patients.

<i>MSX1</i> mutations isolated from tooth agenesis patients.
Fig. 2. MSX1 mutations isolated from tooth agenesis patients.
(A) DNA sequence chromatograms presenting a de novo heterozygous missense mutation c.572T>G in MSX1 identified in family I, compared with wild-type control. (B) Conservation analysis shows that the Phe residue at 191 in Msx1 is conserved across human, gorilla, rhesus, mouse, horse, bovin, pteropus alecto, dophin, pteropus vampyrus. (C) DNA sequence chromatograms showing a heterozygous c.590_594 dup TGTCC mutation identified in family II, compared with wild-type.

Bioinformatics analyses

Secondary structural analysis showed that the homeodomain of the wild-type MSX1 protein consisting of 60 amino acids (No.172-231) is composed of the N-terminal arm (NT arm, No.172-180) and three α-helices: helix I (No.181-193), helix II (No.199-209) and helix III (No.213-231). The mutation p. Phe191Ser was located at the end of the helix I (Fig 3A and 3B). In the wild-type MSX1 protein, the phenyl ring of the p. Phe191 was larger and overlapped with p. Gln195 (Fig 3A). In the p. Phe191Ser-mutated MSX1 protein, the phenyl ring disappeared and resulted in a large gap between p.191Ser and p. Gln195 (Fig 3B). Therefore, the p.Phe191Ser mutation may result in an abnormal structure of the MSX1 protein. In the wild type homeodomain, modelling predicted one hydrogen bond between Phe191 and Leu187 (Fig 3C), while in the mutated protein, there are two additional hydrogen bonds to Glu188 (Fig 3D).

Structural modeling of the wide-type and the mutated homeodomain of MSX1 protein.
Fig. 3. Structural modeling of the wide-type and the mutated homeodomain of MSX1 protein.
(A, B) Structural modeling of the wide-type and the p. Phe191Ser mutated homeodomain of MSX1 protein show that the mutation is located at the end of the helix I. (A) In the wild-type MSX1 protein, the phenyl ring of the p. Phe191 is large and overlapped with p. Gln195. (B) In the p. Phe191Ser-mutated MSX1 protein, the phenyl ring disappears, a large gap between p.191Ser and p.Gln195 arise. (C) Phe191 forms a hydrogen bond with Leu187. (D) The Phe191Ser mutation creates another two new hydrogen bonds with Glu188. (E) Structural modeling of the c.590_594 dup TGTCC mutated homeodomain of MSX1protein by Pymol. The mutant structural shows that helix I is conserved, but helixes II and III are disrupted.

The frameshift insertion (c.590_594 dup TGTCC, p.L197SfsX22) is located at the end of the loop domain between helix I and II, and generates a premature stop codon after an unrelated polypeptide sequence consisting of 22 amino acid residues. The shortened mutant-MSX1 protein is composed of two α-helices: helix I (No.181-193), and helix II (No.200-204), only the first helical region (helix I) was conserved and the other two helical regions (helixes II and III) are disrupted (Fig 3E).

Discussion

MSX1 is the first gene identified causing NSTA, which encodes a transcription factor [19]. It is widely expressed in many organs, particularly during the bud and cap stages of tooth development where epithelial–mesenchymal interactions occur in odontogenesis [34]. MSX1 consists of two exons, the second of which includes the highly conserved homeodomain (HD). The homeodomain is comprised of an extended N-terminal arm and three α-helices [35]. The residues in helices I and II are thought to play an important role in structural stability and binding activity while the residues in helix III combined with those in the N-terminal arm are important for DNA binding specificity[36]. It has been demonstrated that the homeodomain is important for the successful interaction of MSX1 with other proteins such as TATA box binding protein (TBP) and distal-less homeobox (DLX) [37, 38]. However, the role of MSX1 in human craniofacial and dental development has not been fully elucidated [39].

As we know, proteins fold into specific configurations mainly through a large number of non-covalent interactions, including hydrogen bonds, ionic bonds, van der Waals forces and hydrophobic interactions [40]. Hydrogen bonding plays a critical role in secondary structure formation and the integrity of three-dimensional structures [41]. In the homeodomain of MSX1, the stable secondary helical structure is maintained by hydrogen bonds between C = O and N-H groups of different amino acids. In this study, we show that the missense mutation Phe191Ser may lead to two additional hydrogen bonds with Glu188. Moreover, the mutated side chain becomes smaller, resulting in an increased distance between p.191Ser and p.Gln195. This alteration may force the neighboring residues to rearrange their positions, compromising the stability of the homeodomain and affecting the binding ability of MSX1 to DNA.

In order to elucidate the relationship between the hydrogen bond modifications induced by MSX1 mutations and hypodontia phenotypes, we performed homology modeling to analyze the distribution of mutation sites in the homeodomain. We included 7 MSX1 missense mutations from all missense/nonsense MSX1 mutations in HGMD (Human Gene Mutation Database, professional version 2019.3) [42], which are believed to cause non-syndromic tooth agenesis (Table 1)(S2 Table) (S2 Fig). Large deletions and insertions, small in-frame deletions, and nonsense mutations were not included because these mutations would damage the overall structure of MSX1 protein.

Tab. 1. MSX1 homeodomain missense variants in subphenotypes of increasing severity of non-syndromic tooth agenesis (NSTA) and alterations in hydrogen bonds.
MSX1 homeodomain missense variants in subphenotypes of increasing severity of non-syndromic tooth agenesis (NSTA) and alterations in hydrogen bonds.

Our structural modeling predicts a correlation between observed phenotypes and the hydrogen bond alterations. MSX1 mutations that altered hydrogen bonding tend to cause a more severe NSTA phenotype with more missing teeth (the average number of missing teeth were 3.8 vs. 8.8) (S3 Table) (Fig 4). Although hydrogen bonds are not the tractive force of protein or peptide folding, these bonds significantly contribute to the maintenance of the peptide folding [47]. Missense mutations that affect hydrogen bonds may have a significant effect on protein stability, DNA binding specificity, protein expression and interactions. Therefore, it is likely that the mutations we report in this study resulted in more severe selective tooth agenesis. Further functional analysis of these mutations will help to reveal the molecular mechanism of their action.

Statistical analysis.
Fig. 4. Statistical analysis.
Comparison of the average missing teeth number caused by missense mutations in MSX1 homeodomain with and without hydrogen bonds alteration obtained from different non-syndromic tooth agenesis patients (patients caused by missense mutations that had no hydrogen bond alteration, n = 6, vs. patients caused by missense mutations that had hydrogen bonds alteration, n = 19). Data are shown as mean ± SEM. An unpaired t test with two-tailed p-values was performed using GraphPad Prism 5 software. P value = 0.0086 obtained by analyzing data. **denotes a P-value <0.01.

In our study, a novel frameshift insertion of 5 bp (TGTCC) in the homeodomain of the MSX1 gene (NM_002448) was also identified in a Chinese family with autosomal dominant non-syndromic tooth agenesis, affecting the amino acid sequence of the homeodomain at p. 197Leu, changing the amino acid sequence from position 199. This alteration changes the spiral shape of helix II and eliminates the helix III, which is important for DNA-binding and protein-protein interactions including the TBP and DLX families [48]. Previous studies have shown that consensus residues that interact with DNA are located in helix III and the N-terminal arm [35]. We speculate that this novel frameshift mutation has a significant impact on the structure of the truncated protein, leading to severe consequences in protein interactions and DNA binding.

This frameshift mutation likely has considerable significant downstream effects as it demonstrates a substantial impact on primary protein structure. The MSX1 homeodomain contacts its target DNA regions in the major and minor groove at helix III and the N-terminal arm, respectively. The insertion variant found in the present study is predicted to be harmful through structural modeling and bioinformatics analyses, and is predicted to affect the DNA-binding functions of the homeodomain. Further studies are required to investigate the subcellular localization of this mutant MSX1 protein in vitro, to verify whether this is a functionally null variant.

Conclusions

In our current study, we described two novel variants of the MSX1 gene identified in two Chinese patients with isolated tooth agenesis: c.572T>C and c.590_594 dup TGTCC. These two novel mutations were proven to be pathogenic with respect to amino acid structure. These results expanded the mutational spectrum of the MSX1 gene and the options to be considered for their precision treatment later. Further expression and functional studies are required to assess the effect of the identified mutations at the protein level.

Supporting information

S1 Fig [tif]
Comparison of wild type and c. 590_594 dup TGTCC protein sequences of the MSX1 homeodomain underlined.

S2 Fig [a]
Predicted mutational impacts on the MSX1 homeodomain structure.

S1 Table [docx]
Primers for candidate genes.

S2 Table [docx]
Missense /nonsense MSX1 mutations from HGMD (2019.3).

S3 Table [docx]
The (average) number of patients and missing teeth in mutations with and without hydrogen-bonding alteration.


Zdroje

1. Polder BJ, Van't Hof MA, Van der Linden FP, Kuijpers-Jagtman AM. A meta-analysis of the prevalence of dental agenesis of permanent teeth. Community Dent Oral Epidemiol. 2004;32(3):217–26. doi: 10.1111/j.1600-0528.2004.00158.x 15151692.

2. Nieminen P. Genetic basis of tooth agenesis. J Exp Zool B Mol Dev Evol. 2009;312B(4):320–42. Epub 2009/02/17. doi: 10.1002/jez.b.21277 19219933.

3. Jumlongras D, Bei M, Stimson JM, Wang WF, DePalma SR, Seidman CE, et al. A nonsense mutation in MSX1 causes Witkop syndrome. Am J Hum Genet. 2001;69(1):67–74. Epub 2001/05/23. S0002-9297(07)61446-3 [pii] doi: 10.1086/321271 11369996; PubMed Central PMCID: PMC1226049.

4. Ye XQ, Jin HX, Shi LS, Fan MW, Song GT, Fan HL, et al. Identification of novel mutations of IRF6 gene in Chinese families with Van der Woude syndrome. Int J Mol Med. 2005;16(5):851–6. 16211254.

5. Modesto A, Moreno LM, Krahn K, King S, Lidral AC. MSX1 and orofacial clefting with and without tooth agenesis. J Dent Res. 2006;85(6):542–6. doi: 10.1177/154405910608500612 16723652; PubMed Central PMCID: PMC2241923.

6. van den Boogaard MJ, Dorland M, Beemer FA, van Amstel HK. MSX1 mutation is associated with orofacial clefting and tooth agenesis in humans. Nat Genet. 2000;24(4):342–3. doi: 10.1038/74155 10742093.

7. Bailleul-Forestier I, Berdal A, Vinckier F, de Ravel T, Fryns JP, Verloes A. The genetic basis of inherited anomalies of the teeth. Part 2: syndromes with significant dental involvement. Eur J Med Genet. 2008;51(5):383–408. doi: 10.1016/j.ejmg.2008.05.003 18599376.

8. Liang J, Zhu L, Meng L, Chen D, Bian Z. Novel nonsense mutation in MSX1 causes tooth agenesis with cleft lip in a Chinese family. Eur J Oral Sci. 2012;120(4):278–82. doi: 10.1111/j.1600-0722.2012.00965.x 22813217.

9. Goldenberg M, Das P, Messersmith M, Stockton DW, Patel PI, D'Souza RN. Clinical, radiographic, and genetic evaluation of a novel form of autosomal-dominant oligodontia. J Dent Res. 2000;79(7):1469–75. doi: 10.1177/00220345000790070701 11005730.

10. Pirinen S, Kentala A, Nieminen P, Varilo T, Thesleff I, Arte S. Recessively inherited lower incisor hypodontia. J Med Genet. 2001;38(8):551–6. doi: 10.1136/jmg.38.8.551 11494968; PubMed Central PMCID: PMC1734917.

11. Burzynski NJ, Escobar VH. Classification and genetics of numeric anomalies of dentition. Birth Defects Orig Artic Ser. 1983;19(1):95–106. 6362744.

12. Maguire A, Craft AW, Evans RG, Amineddine H, Kernahan J, Macleod RI, et al. The long-term effects of treatment on the dental condition of children surviving malignant disease. Cancer. 1987;60(10):2570–5. doi: 10.1002/1097-0142(19871115)60:10<2570::aid-cncr2820601037>3.0.co;2-q 3664436.

13. Nasman M, Forsberg CM, Dahllof G. Long-term dental development in children after treatment for malignant disease. Eur J Orthod. 1997;19(2):151–9. doi: 10.1093/ejo/19.2.151 9183064.

14. Yu P, Yang W, Han D, Wang X, Guo S, Li J, et al. Mutations in WNT10B Are Identified in Individuals with Oligodontia. Am J Hum Genet. 2016;99(1):195–201. doi: 10.1016/j.ajhg.2016.05.012 27321946; PubMed Central PMCID: PMC5005437.

15. Massink MP, Creton MA, Spanevello F, Fennis WM, Cune MS, Savelberg SM, et al. Loss-of-Function Mutations in the WNT Co-receptor LRP6 Cause Autosomal-Dominant Oligodontia. Am J Hum Genet. 2015;97(4):621–6. PubMed Central PMCID: PMC4596913. doi: 10.1016/j.ajhg.2015.08.014 26387593

16. van den Boogaard MJ, Creton M, Bronkhorst Y, van der Hout A, Hennekam E, Lindhout D, et al. Mutations in WNT10A are present in more than half of isolated hypodontia cases. J Med Genet. 2012;49(5):327–31. doi: 10.1136/jmedgenet-2012-100750 22581971.

17. Stockton DW, Das P, Goldenberg M, D'Souza RN, Patel PI. Mutation of PAX9 is associated with oligodontia. Nat Genet. 2000;24(1):18–9. Epub 1999/12/30. doi: 10.1038/71634 10615120.

18. Tao R, Jin B, Guo SZ, Qing W, Feng GY, Brooks DG, et al. A novel missense mutation of the EDA gene in a Mongolian family with congenital hypodontia. J Hum Genet. 2006;51(5):498–502. Epub 2006/04/04. doi: 10.1007/s10038-006-0389-2 16583127.

19. Vastardis H, Karimbux N, Guthua SW, Seidman JG, Seidman CE. A human MSX1 homeodomain missense mutation causes selective tooth agenesis. Nat Genet. 1996;13(4):417–21. Epub 1996/08/01. doi: 10.1038/ng0896-417 8696335.

20. Lammi L, Arte S, Somer M, Jarvinen H, Lahermo P, Thesleff I, et al. Mutations in AXIN2 cause familial tooth agenesis and predispose to colorectal cancer. Am J Hum Genet. 2004;74(5):1043–50. Epub 2004/03/26. doi: 10.1086/386293 S0002-9297(07)64368-7 [pii]. 15042511; PubMed Central PMCID: PMC1181967.

21. Chassaing N, Bourthoumieu S, Cossee M, Calvas P, Vincent MC. Mutations in EDAR account for one-quarter of non-ED1-related hypohidrotic ectodermal dysplasia. Hum Mutat. 2006;27(3):255–9. doi: 10.1002/humu.20295 16435307.

22. Bergendal B, Klar J, Stecksen-Blicks C, Norderyd J, Dahl N. Isolated oligodontia associated with mutations in EDARADD, AXIN2, MSX1, and PAX9 genes. Am J Med Genet A. 2011;155A(7):1616–22. doi: 10.1002/ajmg.a.34045 21626677.

23. Matalova E, Fleischmannova J, Sharpe PT, Tucker AS. Tooth agenesis: from molecular genetics to molecular dentistry. J Dent Res. 2008;87(7):617–23. doi: 10.1177/154405910808700715 18573979.

24. Arte S, Parmanen S, Pirinen S, Alaluusua S, Nieminen P. Candidate gene analysis of tooth agenesis identifies novel mutations in six genes and suggests significant role for WNT and EDA signaling and allele combinations. PLoS One. 2013;8(8):e73705. doi: 10.1371/journal.pone.0073705 23991204; PubMed Central PMCID: PMC3750013.

25. Hlouskova A, Bielik P, Bonczek O, Balcar VJ, Sery O. Mutations in AXIN2 gene as a risk factor for tooth agenesis and cancer: A review. Neuro Endocrinol Lett. 2017;38(3):131–7. 28759178.

26. Haddaji Mastouri M, De Coster P, Zaghabani A, Jammali F, Raouahi N, Ben Salem A, et al. Genetic study of non-syndromic tooth agenesis through the screening of paired box 9, msh homeobox 1, axin 2, and Wnt family member 10A genes: a case-series. Eur J Oral Sci. 2018;126(1):24–32. doi: 10.1111/eos.12391 29114927.

27. Liu F, Chu EY, Watt B, Zhang Y, Gallant NM, Andl T, et al. Wnt/beta-catenin signaling directs multiple stages of tooth morphogenesis. Dev Biol. 2008;313(1):210–24. Epub 2007/11/21. S0012-1606(07)01460-1 [pii] doi: 10.1016/j.ydbio.2007.10.016 18022614; PubMed Central PMCID: PMC2843623.

28. Fournier BP, Bruneau MH, Toupenay S, Kerner S, Berdal A, Cormier-Daire V, et al. Patterns of Dental Agenesis Highlight the Nature of the Causative Mutated Genes. J Dent Res. 2018;97(12):1306–16. doi: 10.1177/0022034518777460 29879364.

29. Koskinen S, Keski-Filppula R, Alapulli H, Nieminen P, Anttonen V. Familial oligodontia and regional odontodysplasia associated with a PAX9 initiation codon mutation. Clin Oral Investig. 2019;23(11):4107–11. doi: 10.1007/s00784-019-02849-5 30809714.

30. Zhang XX, Wong SW, Han D, Feng HL. Simultaneous Occurence of an Autosomal Dominant Inherited MSX1 Mutation and an X-linked Recessive Inherited EDA Mutation in One Chinese Family with Non-syndromic Oligodontia. Chin J Dent Res. 2015;18(4):229–34. doi: 10.3290/j.cjdr.a35147 26629556.

31. Fan H, Ye X, Shi L, Yin W, Hua B, Song G, et al. Mutations in the EDA gene are responsible for X-linked hypohidrotic ectodermal dysplasia and hypodontia in Chinese kindreds. Eur J Oral Sci. 2008;116(5):412–7. doi: 10.1111/j.1600-0722.2008.00555.x 18821982.

32. Hovde S, Abate-Shen C, Geiger JH. Crystal structure of the Msx-1 homeodomain/DNA complex. Biochemistry. 2001;40(40):12013–21. doi: 10.1021/bi0108148 11580277.

33. Guex N, Peitsch MC. SWISS-MODEL and the Swiss-PdbViewer: an environment for comparative protein modeling. Electrophoresis. 1997;18(15):2714–23. doi: 10.1002/elps.1150181505 9504803.

34. Thesleff I. The genetic basis of tooth development and dental defects. Am J Med Genet A. 2006;140(23):2530–5. doi: 10.1002/ajmg.a.31360 16838332.

35. Shang Z, Isaac VE, Li H, Patel L, Catron KM, Curran T, et al. Design of a "minimAl" homeodomain: the N-terminal arm modulates DNA binding affinity and stabilizes homeodomain structure. Proc Natl Acad Sci U S A. 1994;91(18):8373–7. doi: 10.1073/pnas.91.18.8373 7915838; PubMed Central PMCID: PMC44608.

36. Isaac VE, Sciavolino P, Abate C. Multiple amino acids determine the DNA binding specificity of the Msx-1 homeodomain. Biochemistry. 1995;34(21):7127–34. doi: 10.1021/bi00021a026 7766623.

37. Catron KM, Zhang H, Marshall SC, Inostroza JA, Wilson JM, Abate C. Transcriptional repression by Msx-1 does not require homeodomain DNA-binding sites. Mol Cell Biol. 1995;15(2):861–71. doi: 10.1128/mcb.15.2.861 7823952; PubMed Central PMCID: PMC231967.

38. Zhang H, Catron KM, Abate-Shen C. A role for the Msx-1 homeodomain in transcriptional regulation: residues in the N-terminal arm mediate TATA binding protein interaction and transcriptional repression. Proc Natl Acad Sci U S A. 1996;93(5):1764–9. doi: 10.1073/pnas.93.5.1764 8700832; PubMed Central PMCID: PMC39855.

39. Xin T, Zhang T, Li Q, Yu T, Zhu Y, Yang R, et al. A novel mutation of MSX1 in oligodontia inhibits odontogenesis of dental pulp stem cells via the ERK pathway. Stem Cell Res Ther. 2018;9(1):221. doi: 10.1186/s13287-018-0965-3 30134957; PubMed Central PMCID: PMC6106924.

40. Munoz V, Cronet P, Lopez-Hernandez E, Serrano L. Analysis of the effect of local interactions on protein stability. Fold Des. 1996;1(3):167–78. doi: 10.1016/s1359-0278(96)00029-6 9079379.

41. Wang M, Wales TE, Fitzgerald MC. Conserved thermodynamic contributions of backbone hydrogen bonds in a protein fold. Proc Natl Acad Sci U S A. 2006;103(8):2600–4. doi: 10.1073/pnas.0508121103 16473949; PubMed Central PMCID: PMC1413781.

42. Stenson PD, Mort M, Ball EV, Evans K, Hayden M, Heywood S, et al. The Human Gene Mutation Database: towards a comprehensive repository of inherited mutation data for medical research, genetic diagnosis and next-generation sequencing studies. Hum Genet. 2017;136(6):665–77. doi: 10.1007/s00439-017-1779-6 28349240; PubMed Central PMCID: PMC5429360.

43. Yamaguchi S, Machida J, Kamamoto M, Kimura M, Shibata A, Tatematsu T, et al. Characterization of novel MSX1 mutations identified in Japanese patients with nonsyndromic tooth agenesis. PLoS One. 2014;9(8):e102944. doi: 10.1371/journal.pone.0102944 25101640; PubMed Central PMCID: PMC4125152.

44. Mostowska A, Biedziak B, Jagodzinski PP. Novel MSX1 mutation in a family with autosomal-dominant hypodontia of second premolars and third molars. Arch Oral Biol. 2012;57(6):790–5. Epub 2012/02/03. doi: 10.1016/j.archoralbio.2012.01.003 22297032.

45. Chishti MS, Muhammad D, Haider M, Ahmad W. A novel missense mutation in MSX1 underlies autosomal recessive oligodontia with associated dental anomalies in Pakistani families. J Hum Genet. 2006;51(10):872–8. Epub 2006/08/26. doi: 10.1007/s10038-006-0037-x 16932841.

46. Xuan K, Jin F, Liu YL, Yuan LT, Wen LY, Yang FS, et al. Identification of a novel missense mutation of MSX1 gene in Chinese family with autosomal-dominant oligodontia. Arch Oral Biol. 2008;53(8):773–9. Epub 2008/04/01. doi: 10.1016/j.archoralbio.2008.02.012 18374898.

47. Scheike JA, Baldauf C, Spengler J, Albericio F, Pisabarro MT, Koksch B. Amide-to-ester substitution in coiled coils: the effect of removing hydrogen bonds on protein structure. Angew Chem Int Ed Engl. 2007;46(41):7766–9. doi: 10.1002/anie.200702218 17876795.

48. Zhang H, Hu G, Wang H, Sciavolino P, Iler N, Shen MM, et al. Heterodimerization of Msx and Dlx homeoproteins results in functional antagonism. Mol Cell Biol. 1997;17(5):2920–32. doi: 10.1128/mcb.17.5.2920 9111364; PubMed Central PMCID: PMC232144.


Článek vyšel v časopise

PLOS One


2020 Číslo 1
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Svět praktické medicíny 1/2024 (znalostní test z časopisu)
nový kurz

Koncepce osteologické péče pro gynekology a praktické lékaře
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Význam metforminu pro „udržitelnou“ terapii diabetu
Autoři: prof. MUDr. Milan Kvapil, CSc., MBA

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#