Mehrjardi M. Y. V, Kalantar S. M, Jaafarinia M, Dehghani M. The Effect of HOXB1 Gene Expression in HCFP Patient Using Real Time PCR Assay in Iranian Family. Biomed Pharmacol J 2017;10(2).
Manuscript received on :March 14, 2017
Manuscript accepted on :March 23, 2017
Published online on: --
Plagiarism Check: Yes
How to Cite    |   Publication History
Views Views: (Visited 519 times, 1 visits today)   Downloads PDF Downloads: 628

Mohammad Yayha Vahidi Mehrjardi1, Seyed Mehdi Kalantar2, Mojtaba Jaafariniaand Mohammadreza Dehghani3

1Department of Genetic, Marvdasht branch, Islamic Azad University, Marvdasht, Iran.

2Reproductive and Genetic Unit, Yazd Research and Clinical center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.

3Medical genetics research center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.

Corresponding Author E-mail: mrezdehghani@gmail.com

DOI : https://dx.doi.org/10.13005/bpj/1173

Abstract

Hereditary Congenital Facial Paresis (HCFP) is a rare syndrome of isolated facial nerve palsy causing facial asymmetry and ptosis. Many described cases showed an autosomal dominant pattern of inheritance. HOXB1 is the first identified gene in HCFP. This study was aimed to evaluation the effect of a deletion mutation on HOXB1 gene expression in Iranian HCFP patient using Real Time PCR. A large Iranian kindred with overall 5 affected individuals along with their unaffected siblings and parents were recruited. The candidate gene HOXB1 was screened and analyzed. After RNA extraction from blood, cDNA were synthesized according to protocol and Real Time PCR carried out using SYBR®Premix Ex TaqTM II kit. HOXB1 expression level was analyzed by ΔΔCT method. The results of this study demonstrates that expression level of HOXB1 gene in homo and hetero had similar expression. However, no differences expression level of HOXB1 gene were found. Masked-like faces, bilateral facial palsy with variable sensorineural hearing loss, as well as low-set ears and some dysmorphic features were the most remarkable findings in the affected members of the family. Our findings expand the mutational spectrum of HOXB1 involved in HCFP, due to the number of patient that had deletion mutation did not show any expression change in HOXB1 gene. Investigation of HOXB1 gene expression in larger family might have different results.

Keywords

hereditary congenital facial paresis; HOXB1; Real Time PCR; Iran

Download this article as: 
Copy the following to cite this article:

Mehrjardi M. Y. V, Kalantar S. M, Jaafarinia M, Dehghani M. The Effect of HOXB1 Gene Expression in HCFP Patient Using Real Time PCR Assay in Iranian Family. Biomed Pharmacol J 2017;10(2).

Copy the following to cite this URL:

Mehrjardi M. Y. V, Kalantar S. M, Jaafarinia M, Dehghani M. The Effect of HOXB1 Gene Expression in HCFP Patient Using Real Time PCR Assay in Iranian Family. Biomed Pharmacol J 2017;10(2). Available from: http://biomedpharmajournal.org/?p=14928

Introduction

Hereditary congenital facial paresis (HCFP) due to congenital cranial dysinnervation disorders is characterized by isolated dysfunction of the facial nerve (Cranial Nerve VII). HCFP be the property of the congenital cranial dysinnervation disorders. HCFP is characterized by the isolated dysfunction of the seventh cranial nerve and in addition, strabismus, hearing loss, feeding difficulties and some recognizable dysmorphic features limited to the orofacial region can be detected (Vogel et al., 2016).  Linkage analysis in large families with autosomal dominant HCFP revealed one locus mapped to chromosome 3q21-q22 (HCFP1; MIM 601471) and a second to 10q21.3-q22.1 (HCFP2; MIM 604185) and HOXB1 (17q21; HCFP3; MIM 614744) (van der Zwaag et al., 2005; Webb et al., 2012).  HCFP has genetic heterogeneity and HOXB1 (17q21; HCFP3; MIM 614744) is the first identified gene (Sahin et al., 2016) and the only known causative gene for HCFP, encoding one of 39 homeodomain containing transcription factor of the HOX gene family under the HOXL subgroup of ANTP-class (Boncinelli et al., 1997), which regulates early developmental morphogenetic processes especially the anterior–posterior patterning of the developing embryo (Mallo and Alonso, 2013). The previously reported HOXB1 mutations change arginine 207 to another residue in the homeodomain and alter binding capacity of HOXB1 for transcriptional co-regulators and DNA (Vogel et al., 2016). HOXB1 has a characteristic helix-turnhelix DNA binding motif with three alpha helical regions (a1, a2, a3) where the specificity may be contemplated by heterodimerization with PBX1 (Piper et al., 1999). NMR studies showed that the conserved hexapeptide of HOXB1 (TFDWMK) stabilizes binding of PBX1 and HOXB1 to DNA (Slupsky et al., 2001).

To date, mutations in ten HOX genes have been found to cause different human disorders (Quinonez and Innis, 2014). Molecular modeling and in vitro functional analysis predicted the arginine-to-cysteine change at position 207 in the homeodomain to diminish binding of HOXB1 to transcriptional co-regulators and DNA, thereby altering transcriptional activity of HOXB1 (Webb et al., 2012).

The analysis of gene expression for mRNA sequences requires precise, sensitive, and reproducible measurements. Gene expression levels were commonly determined using northern blot analysis. However, this technique requires a large quantity of RNA and is timeconsuming (Dean et al., 2002). At present Real Time PCR is the most sensitive method for the detection of low abundance mRNAs, and can be used for different applications, such as clinical diagnostic, for the gene expression analysis of tissue-specific, and for plant studies (Gachon et al., 2004). Real Time PCR is typically referenced to an internal control gene. The conditions of the experiment should not influence the expression of this internal control gene (Schmittgen and Zakrajsek, 2000). However, many studies showed that internal standards, mainly reference genes used for the quantification of mRNA expression, could vary with the experimental conditions. A lot of reference genes are well described for the normalization of expression signals (Stürzenbaum and Kille, 2001). The most common are actin, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), ribosomal genes, cyclophilin, 18s rRNA and elongation factor 1-a (ef1a) (Dean et al., 2002). Adenine phosphoribosyl transferase (aprt) and tubulin may also be used. Many studies on housekeeping gene expression mainly deal with human tissues, bacteria and viruses (Volkov et al., 2003). Consequently, choosing an internal control is too important to gene expression quantify (Langer et al., 2002). The use of GAPDH as the internal standard could be a valuable alternative to quantify genes of interest, keeping in mind that it could reduce the variations of expression (Nicot et al., 2005).

The aim of present study was to evaluation the effect of a deletion mutation on HOXB1 gene expression in HCFP patient using Real Time PCR in a large Iranian family.

Materials and Methods

Sample Collection

36 blood samples were obtained from patients randomly. Clinical and pathological data of patients were collected. The experiment protocols and informed consent forms were approved by the Human Studies Committee at the Affiliated the Medical Science University.

RNA Extraction and cDNA Synthesis

Total RNA was extracted from blood samples using the RNXTM-Plus solution (SinaClon, IRAN) according to the manufacturer’s instructions, except for an extended 1-h treatment with DNase I. RNA was analyzed by Thermo Scientific NanoDrop™ 1000 Spectrophotometer to check its purity and concentration, and electrophoresd on 2% agarose gel to confirm its integrity. One microgram of RNA was used for complementary DNA (cDNA) synthesis by using random hexamer priming and PrimeScriptTM-RT reagent kit (TaKaRa, Japan). Synthesized cDNA was then checked spectrophotometrically to estimate its concentration.

Real Time PCR

All samples were carried out on a rotor gene 6000 corbett detection system and Real Time PCR using SYBR®Premix Ex TaqTM II kit (TaKaRa, Japan) according to the manufacturer’s instructions. Thermal cycling conditions were an initial activation step for 5 min at 95 °C followed by 40 cycles at 95 °C for 15 s and 65 °C for 1 min. No template control (NTC) consisting of H2O was included in each run. Forward and reverse primers sequences have shown in table 1. Melting curve analysis was performed to verify specificity of PCR products. Besides, PCR products were electrophoresed on 2 % agarose gel to verify product sizes and specificity. For Real Time PCR analysis, all samples were normalized to GAPDH. The mean value in each triplicate was used to calculate (ΔCt = Ct mean lncRNAs-Ct mean GAPDH). Expression fold changes were calculated using 2-ΔΔCt methods. The qPCR assays were performed in triplicate and the data were presented as the mean ± standard error of the mean (SEM).

Statistical Analysis

The pairwise fixed reallocation randomization test with 2000 iterations in the REST 2009 software was used to determine the significances. The level of statistical significance was set at P<0.05. Statistical analyses of demographic and clinical data were performed using SPSSv.15.0.1 (SPSS Inc., Chicago, IL). Chi-square and independent t tests were used for testing the relationship between categorical variables. Significance was defined as P<0.05.

Results

General Statistical Information

Data have been analyzed based on the information taken from questionnaires, interviews, and clinical and laboratory tests.

Expression of HOXB1 gene

The results of this study demonstrates that expression level of HOXB1 gene in hemo and hetero had similar expression. 22 and 9 out of 36 were hetero and hemo respectively for this mutation. However, no differences expression level of HOXB1 gene were found. Expression level of HOXB1 gene have shown in figure 1.

Figure 1a: Pedigree of the Iranian family with five individuals affected by HCFP (indicated as solid black symbols).

Figure 1a: Pedigree of the Iranian family with five individuals affected by HCFP (indicated as solid black symbols).

Click here to View figure

Segregation of the LOF mutation with the family with mutant allele shown by ‘+’ and wild-type allele shown by ‘−’. B) A section of Sanger sequencing Chromatograms for the homozygous mutation (below) and wild type (top) are depicted and demonstrate the insertion (in the box). C) Expression level of HOXB1 gene.

Discussion

Facial paresis is a rare, hereditary congenital and nonprogressive. From 1995 till now only 3 cases of the disease have been reported in the world therefore there is no definition specific prevalence of this disease and its hereditary have been reported as autosomal recessive (Uyguner et al., 2015). Facial paresis due to the similarity of symptoms named Moebius syndrome. Three gene loci associated with HCFP have been reported HCFP1, HCFP2 and HCFP3 respectively (Alrashdi et al., 2010). HOXB1 gene have been introduced the most likely gene related to HCFP. In 2015 a study on 56 family involved with HCFP indicated a mutation in HOXB1 gene which had occurred in the same location (Miller, 2007) .

The first report was done by Goddard et al in 1996 showed a nonsense mutation in HOXB1 gene in mice caused impaired development of facial nerve but no similar cases were reported in human (Goddard et al., 1996). In 1999 Piper et al investigated the existence of a binding motif in HOXB1 gene (Piper et al., 1999). The role of HOXB1 gene and its relationship with other developmental genes had been evaluated by Carolyn et al in 2001 (Slupsky et al., 2001). The results of Carolyn et al indicated that the structure of the protein product has been quite stable.

177 out of 272 families with MBS or HCFP previously reported by Webb et al. (2012), along with 95 samples (56 from Netherlands and 39 from Turkey), 3 cases were found to carry mutations in HOXB1, establishing the frequency of HOXB1 mutations to be 1.1% in patients afflicted with congenital facial paralysis (Webb et al., 2012). Sahin et al. 2016 showed case reported has p.Arg230Trp mutation in the HOXB1 gene which causes HCFP3 in a large family of Turkish origin. This mutation brings the total number of HOXB1 mutations identified in HCFP3 to four within five families. This mutation lies in the DNA-binding homeodomain of HOXB1 located in between 203 and 262 residues, which is highly conserved among several species (Sahin et al., 2016).

Michielse et al in 2006 using linkage analysis method find HCFP 3q21 in a Pakistani family and did not find result of on search for other candidate genes (Michielse et al., 2006). Another homozygous missense mutation in HOXB1, also affecting arginine 207 [c.620G>A/p. (Arg207His)] was found in a Turkish girl with bilateral facial weakness, left esotropia, left ptosis, and midface retrusion. Auditory brainstem response test revealed normal results and MRI scans showed no structural anomalies (MacKinnon et al., 2014). Webb et al. reported that another missense mutation in the critical homeodomain, would reduce binding of HOXB1 to transcriptional co-regulators and DNA, hence altering transcriptional activity of HOXB1 (Webb et al., 2012). Clinical features of all affected individuals including hearing loss, midface retrusion, lagophthalmos, oral dysfunction, swallowing difficulties, dysarthria, and speech delay suggested the diagnosis of HCFP (Vogel et al., 2016). The results of Vogel et al. 2016 in Germany indicated in a family with two consanguineous marriages and four individuals affected by HCFP over two generations, we identified a novel homozygous one-base-pair substitution in the HOXB1 gene leading to introduction of a premature stop codon. This germline mutation most probably represents the first loss-of-function HOXB1 allele associated with HCFP (Verzijl et al., 2003).

Conclusion

HOXB1 gene Identify was an effective step in HCFP. Accordingly comparative study of the disease using new techniques such as SNP array, NGS and Real Time PCR can be a milestone in the recognition of HCFP. Our findings expand the expression of HOXB1 involved in HCFP, and consolidate the role of the gene in development of autosomal recessive type of HCFP. The number of patient that had 7bp mutation did not show any expression change in HOXB1 gene. Investigation of HOXB1 gene expression in larger family might have different results.

Acknowledgement

We gratefully thank the Medical genetics research center of the Shahid Sadoughi University of Medical Sciences and the patients and their families for cooperation in this study.

Conflict of interest

The authors declare no conflict of interest.

References

  1. Alrashdi, I. S., Rich, P., & Patton, M. A. A family with hereditary congenital facial paresis and a brief review of the literature. Clinical Dysmorphology, 2010; 19(4), 198–201.
    CrossRef
  2. Boncinelli, E., Booth, H. A. F., Bruford, E. A., Bürglin, T., Bürglin, T., Boncinelli, E., et al. Homeobox genes and disease. Current Opinion in Genetics & Development, 1997; 7(3), 331–337.
    CrossRef
  3. Dean, J. D., Goodwin, P. H., & Hsiang, T. Comparison of relative RT-PCR and northern blot analyses to measure expression of β-1,3-glucanase inNicotiana benthamiana infected withColltotrichum destructivum. Plant Molecular Biology Reporter, 2002; 20(4), 347–356.
    CrossRef
  4. Gachon, C., Mingam, A., & Charrier, B. Real-time PCR: what relevance to plant studies? Journal of experimental botany, 2004; 55(402), 1445–54.
    CrossRef
  5. Goddard, J. M., Rossel, M., Manley, N. R., & Capecchi, M. R. Mice with targeted disruption of Hoxb-1 fail to form the motor nucleus of the VIIth nerve. Development (Cambridge, England), 1996; 122(10), 3217–28.
  6. Langer, K., Ache, P., Geiger, D., Stinzing, A., Arend, M., Wind, C., et al. Poplar potassium transporters capable of controlling K+ homeostasis and K+-dependent xylogenesis. The Plant journal : for cell and molecular biology, 2002; 32(6), 997–1009.
  7. MacKinnon, S., Oystreck, D. T., Andrews, C., Chan, W.-M., Hunter, D. G., & Engle, E. C. Diagnostic Distinctions and Genetic Analysis of Patients Diagnosed with Moebius Syndrome. Ophthalmology, 2014; 121(7), 1461–1468.
    CrossRef
  8. Mallo, M., & Alonso, C. R. The regulation of Hox gene expression during animal development. Development, 2013; 140(19), 3951–3963.
    CrossRef
  9. Michielse, C. B., Bhat, M., Brady, A., Jafrid, H., van den Hurk, J. A. J. M., Raashid, Y., et al. Refinement of the locus for hereditary congenital facial palsy on chromosome 3q21 in two unrelated families and screening of positional candidate genes. European Journal of Human Genetics, 2006; 14(12), 1306–1312.
    CrossRef
  10. Miller, G. Neurological disorders: The Mystery of the Missing Smile. Science, 2007; 316(5826), 826–827.
    CrossRef
  11. Nicot, N., Hausman, J.-F., Hoffmann, L., & Evers, D. Housekeeping gene selection for real-time RT-PCR normalization in potato during biotic and abiotic stress. Journal of Experimental Botany, 2005; 56(421), 2907–2914.
    CrossRef
  12. Piper, D. E., Batchelor, A. H., Chang, C. P., Cleary, M. L., & Wolberger, C. Structure of a HoxB1-Pbx1 heterodimer bound to DNA: role of the hexapeptide and a fourth homeodomain helix in complex formation. Cell, 1999; 96(4), 587–97.
    CrossRef
  13. Quinonez, S. C., & Innis, J. W. Human HOX gene disorders. Molecular Genetics and Metabolism, 2014; 111(1), 4–15.
    CrossRef
  14. Sahin, Y., Güngör, O., Ayaz, A., Gülay, G., Bedia, S., Kursad, Y., & Serdar, C. A novel homozygous HOXB1 mutation in a Turkish family with hereditary congenital facial paresis. Brain  development, 2016; 39(2), 166–170.
    CrossRef
  15. Schmittgen, T. D., & Zakrajsek, B. A. Effect of experimental treatment on housekeeping gene expression: validation by real-time, quantitative RT-PCR. Journal of biochemical and biophysical methods, 2000; 46(1–2), 69–81.
    CrossRef
  16. Slupsky, C. M., Sykes, D. B., Gay, G. L., & Sykes, B. D. The HoxB1 hexapeptide is a prefolded domain: Implications for the Pbx1/Hox interaction. Protein Science, 2001; 10(6), 1244–1253.
    CrossRef
  17. Stürzenbaum, S. R., & Kille, P. Control genes in quantitative molecular biological techniques: the variability of invariance. Comparative Biochemistry and Physiology Part B: Biochemistry and Molecular Biology, 2001; 130(3), 281–289.
    CrossRef
  18. Uyguner, Z. O., Toksoy, G., Altunoglu, U., Ozgur, H., Basaran, S., & Kayserili, H. A new hereditary congenital facial palsy case supports arg5 in HOX-DNA binding domain as possible hot spot for mutations. European Journal of Medical Genetics, 2015; 58(6), 358–363.
    CrossRef
  19. van der Zwaag, B., Burbach, J. P. H., Scharfe, C., Oefner, P. J., Brunner, H. G., Padberg, G. W., & van Bokhoven, H. Identifying new candidate genes for hereditary facial paresis on chromosome 3q21–q22 by RNA in situ hybridization in mouse. Genomics, 2005; 86(1), 55–67.
    CrossRef
  20. Verzijl, H. T. F. M., van der Zwaag, B., Cruysberg, J. R. M., & Padberg, G. W. Möbius syndrome redefined: a syndrome of rhombencephalic maldevelopment. Neurology, 2003; 61(3), 327–33.
    CrossRef
  21. Vogel, M., Velleuer, E., Schmidt-Jiménez, L. F., Mayatepek, E., Borkhardt, A., Alawi, M., et al. Homozygous HOXB1 loss-of-function mutation in a large family with hereditary congenital facial paresis. American Journal of Medical Genetics Part A, 2016; 170(7), 1813–1819.
    CrossRef
  22. Volkov, R. A., Panchuk, I. I., & Schöffl, F. Heat-stress-dependency and developmental modulation of gene expression: the potential of house-keeping genes as internal standards in mRNA expression profiling using real-time RT-PCR. Journal of Experimental Botany, 2003; 54(391), 2343–2349.
    CrossRef
  23. Webb, B. D., Shaaban, S., Gaspar, H., Cunha, L. F., Schubert, C. R., Hao, K., et al. HOXB1 Founder Mutation in Humans Recapitulates the Phenotype of Hoxb1−/− Mice. The American Journal of Human Genetics, 2012; 91(1), 171–179.
    CrossRef
Share Button
(Visited 519 times, 1 visits today)

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.