Next Article in Journal
Selective Breeding for Disease-Resistant PRNP Variants to Manage Chronic Wasting Disease in Farmed Whitetail Deer
Next Article in Special Issue
Chronic Pancreatitis: The True Pathogenic Culprit within the SPINK1 N34S-Containing Haplotype Is No Longer at Large
Previous Article in Journal
Evolutionary Analysis of OAT Gene Family in River and Swamp Buffalo: Potential Role of SLCO3A1 Gene in Milk Performance
Previous Article in Special Issue
Extended Phenotyping and Functional Validation Facilitate Diagnosis of a Complex Patient Harboring Genetic Variants in MCCC1 and GNB5 Causing Overlapping Phenotypes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

A Novel Splicing Variant of COL2A1 in a Fetus with Achondrogenesis Type II: Interpretation of Pathogenicity of In-Frame Deletions

1
Medical Genetics Unit, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
2
Medical Genetics Unit, Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, University of Florence, 50121 Florence, Italy
3
Fetal Medicine Unit, ASL Bari, 70012 Bari, Italy
4
Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
5
Unit of Medical Genetics, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
*
Author to whom correspondence should be addressed.
Genes 2021, 12(9), 1395; https://doi.org/10.3390/genes12091395
Submission received: 19 August 2021 / Revised: 7 September 2021 / Accepted: 8 September 2021 / Published: 10 September 2021

Abstract

:
Achondrogenesis type II (ACG2) is a lethal skeletal dysplasia caused by dominant pathogenic variants in COL2A1. Most of the variants found in patients with ACG2 affect the glycine residue included in the Gly-X-Y tripeptide repeat that characterizes the type II collagen helix. In this study, we reported a case of a novel splicing variant of COL2A1 in a fetus with ACG2. An NGS analysis of fetal DNA revealed a heterozygous variant c.1267-2_1269del located in intron 20/exon 21. The variant occurred de novo since it was not detected in DNA from the blood samples of parents. We generated an appropriate minigene construct to study the effect of the variant detected. The minigene expression resulted in the synthesis of a COL2A1 messenger RNA lacking exon 21, which generated a predicted in-frame deleted protein. Usually, in-frame deletion variants of COL2A1 cause a phenotype such as Kniest dysplasia, which is milder than ACG2. Therefore, we propose that the size and position of an in-frame deletion in COL2A1 may be relevant in determining the phenotype of skeletal dysplasia.

1. Introduction

Achondrogenesis type II (ACG2) is a rare, lethal genetic disease that is part of an uncommon skeletal dysplasia group. ACG2 is characterized by a short neck with macrocephaly; a small thorax; underdeveloped lungs; a prominent abdomen; short arms; short legs; and incomplete ossification of the vertebral column, sacrum, and pubic bones. Moreover, it is marked by distinctive facial features such as a prominent forehead; a small chin; and in some cases, a cleft palate [1].
ACG2, first described by Langer [2] and Saldino [3], is caused by a dominant variant in COL2A1. Pathogenic variants in this gene cause different skeletal disorders, including achondrogenesis type II and hypochondrogenesis (MIM#200610), Kniest dysplasia (MIM#156550), platyspondylic dysplasia Torrance type (MIM#151210), spondyloepiphyseal dysplasia congenita (SEDC) (MIM#183900), spondyloperipheral dysplasia (MIM#271700), SED with metatarsal shortening (formerly Czech dysplasia) (MIM#609162), SED with pronounced metaphyseal changes (including SEMD Strudwick type) (MIM#184250), and Stickler syndrome (MIM#108300) [4,5].
ACG2 is the most severe phenotype, while Kniest and SED dysplasia are milder but still have grave conditions, characterized by skeletal anomalies [6]. Stickler syndrome is the mildest condition, and frequently, ocular findings are the only clinical manifestations. Different types of COL2A1 variants are associated with the various phenotypes [7].
COL2A1 is located at 12q13.11 (MIM#120140) and contains 54 exons. It encodes the polypeptide subunit of the type II collagen, which is a homotrimer formed by three α-1(II) chains [8]. Similar to other collagens, a repeating Gly-X-Y motif, which is fundamental for homotrimer formation, characterizes the COL2A1 protein. In fact, in ACG2, the most common COL2A1 variants affect the glycine residue in Gly-X-Y repeats of the α-1 chain [9,10]. These variants cause a dominant-negative effect that dramatically impairs homotrimer assembly and stability, leading to growth bone alterations and severe phenotypes such as ACG2 and hypochondrogenesis. In contrast, nonsense mutations or out-of-frame deletions can cause a premature stop codon that leads to haploinsufficiency, resulting in a decrease in collagen synthesis, as well as diseases with milder phenotypes than that observed in ACG2 [7,11]. Splice site variants leading to an in-frame deletion of COL2A1 have already been described and are mainly associated with Kniest dysplasia [12,13]. In this study, we reported a case of a novel COL2A1 splicing mutation in a fetus with an ACG2 phenotype, and tentatively explained this uncommon genotype–phenotype association. Moreover, we performed a minigene assay to show that the splicing variant generated an exon skipping, leading to an in-frame deletion of COL2A1.

2. Materials and Methods

2.1. Case Report

A 35-year-old Italian primigravida was referred to our genetic unit at 18-week gestation (WG) due to abnormal ultrasonographic findings. The parents of the fetus were non-consanguineous, and the familial history did not reveal any relevant information. Informed written consent for both diagnostic and research purposes was obtained from the parents.
A three-dimensional ultrasound was performed to examine the fetus. The fetal 3D ultrasound examination showed a brachycephaly, a prominent abdomen, short limbs (<at the third centile), a facial dysmorphism characterized by abnormal implantation of the ears, microretrognathia, frontal bossing, a long philtrum, and congenital megalophthalmos. The three-dimensional rendering of the fetal skeleton showed reduced ossification of the sacrum, iliac spines, and arched femurs (Figure 1A). Amniocentesis performed at 19-WG for fetal karyotyping and array-CGH were negative.
At 20-WG gestation, the parents decided to interrupt the pregnancy. Postmortem radiography confirmed reduced ossification of the sacrum and arched femurs (Figure 1B). An autopsy was also performed (Figure 1C), and a cleft palate was observed. The prenatal and postnatal findings were strongly suggestive of skeletal dysplasia, in particular, of achondrogenesis type 2.

2.2. Gene Sequencing

Whole exome sequencing (WES) and in silico analysis of COL2A1 (NM_001844.5) on the fetal DNA revealed a heterozygous base-pair deletion involving the exon 21 of the COL2A1 gene (c.1267-2_1269del).
DNA from the fetal tissue was enriched with SeqCap EZ Exome v3, prepared according to the manufacturer’s protocol (Nimblegen, Roche, Basel, Switzerland), and 150 × 2 bp end sequenced on NextSeq550 (Illumina Inc., San Diego, CA, USA) with a 100× mean coverage. Reads were aligned with the human genome reference sequence (hg19) using Burrows–Wheeler Aligner (BWA) and then mapped and analyzed by the IGV software (Integrative Genome Viewer, 2013 Broad Institute, Cambridge, MA, USA). The variant call for identifying nucleotide variants was performed using automated in-house pipelines (Genome Analysis ToolKit Unified Genotyper Module, GATK, Cambridge, MA, USA). Variants were classified following the guidelines of the American College of Medical Genetics and Genomics (ACMG) [14]. The data interpretation was made by in silico filtering for COL2A1, and a heterozygous base-pair deletion involving the exon 21 of COL2A1 (c.1267-2_1269del) was detected.
Genomic DNA was also extracted from the blood samples of the patient’s parents. The genomic DNA was amplified by the polymerase chain reaction (PCR) with primers forward 5′-ggttggggttcattctttgc-3′ and reverse 5′-tgatggggtttgactccaga-3′ for exon 21 of COL2A1. The PCR products were then purified and bidirectionally sequenced on a 310 ABI PRISM Genetic Analyzer (Applied Biosystems, Waltham, MA, USA). The c.1267-2_1269del variant of COL2A1 was confirmed in the fetus and excluded in the parents.

2.3. Minigene Assay

A wild-type DNA fragment containing COL2A1 from exon 19 to exon 22 was amplified by PCR using human genomic DNA as a template and oligonucleotides, including restriction sites for XhoI and HindIII: Forward 5′-cagAAGCTTgggttgggtgcatgtgcataat-3′ and reverse 5′-gtaCTCGAGtgaagctgtatctgggccttct-3′. The wild-type fragment was then used as a template to generate a fragment containing the variant c.1267-2_1269del by site-directed mutagenesis (overlap extension PCR) using the following additional oligonucleotides: Forward 5′-tcatgcccacgctcctggcatt-3′ and reverse 5′-aatgccaggagcgtgggcatga-3′. All the PCR amplifications were performed with the Pfu DNA polymerase. The wild-type and mutated amplified products were purified by a gel extraction kit (QIAquick Gel Extraction kit, Qiagen, Hilden, Germany) and then cloned into a pcDNA3 expression vector at the XhoI and HindIII restriction sites to generate two minigene constructs: COL2A1-WT and COL2A1-MUT. Recombinant vectors were checked by a sequence analysis.
Human embryonic kidney fibroblasts (HEK 293T) cells were grown in Dulbecco’s modified Eagle’s medium, DMEM (Sigma-Aldrich, St. Louis, MO, USA), supplemented with 10% fetal bovine serum at 37 °C with 5% CO2 atmosphere. Transient transfection was performed on 70% confluent cells plated in six-well dishes with 2 μg of wild-type or mutant minigenes, using the Lipofectamine 3000 transfection reagent (Thermo Fisher, Waltham, MA, USA), according to the manufacturer’s instructions. The cells were harvested 48 h after the transfection, and total RNA was extracted using the RNeasy Mini Kit (Qiagen, Hilden, Germany) by following the manufacturer’s instructions. Total RNA samples were reverse-transcribed to cDNA using the High-Capacity RNA-to-cDNA Kit (Applied Biosystems). The cDNA was amplified by PCR using the following primers: Forward 5′-tcgcggtgaacctggtact-3′ (sequence located in exon 19 of COL2A1) and reverse 5′-gccttgttcacctttgaagcca-3′ (sequence located in exon 22 of COL2A1). PCR products were visualized by 1.5% agarose gel, and the cDNA amplified fragments were purified by a gel extraction kit (QIAquick Gel Extraction kit, Qiagen, Hilden, Germany) and then bidirectionally sequenced.

3. Results

WES of the fetal DNA revealed a heterozygous variant c.1267-2_1269del (NC_000012.11: g.48380957_48380961del) located in intron 20/exon 21 of COL2A1 (NM_001844.5). This variant has never been reported in control populations (dbSNPs, EVS, GnomAD, and 1000 genomes databases) and, as a pathogenic variant, in ClinVar and LOVD databases. Sanger sequencing across the site of the variant observed in the affected fetus confirmed the presence of the wild-type sequence in the unaffected parents, consistent with a de novo mutation in the fetus (Figure 2).
COL2A1 in-frame deletions have already been reported and are responsible for a milder phenotype. We considered this variant as strongly causative of the observed phenotype since we did not identify additional variants in COL2A1 coding and regulatory regions. In addition, we did not detect rare homozygous or putative compound heterozygous combinations in other genes previously reported in overlapping conditions.
Since the variant does not contain the splice acceptor site of exon 21 of COL2A1, to study the variant’s effect, we generated an appropriate minigene construct, as described in the Methods section (Figure 3A). We transfected mutant and wild-type minigene constructs in HEK293T cells and detected the expression of minigenes by RT-PCR. The band of amplified cDNA corresponding to the spliced transcript in the mutant sample was lower than that detected in the wild-type sample, which is consistent with the skipping of exon 21 in COL2A1 messenger RNA (Figure 3B). Furthermore, the sequence of cDNA-amplified products confirmed that, in the mutant sample, the minigene expression resulted in the skipping of exon 21 (Figure 3C). The skipping of exon 21 generated an in-frame deletion (p.Gly423_Thr455del) in the COL2A1 protein (NP_001835.3). The PROVEAN algorithm predicted this variant to be deleterious [15], with a score of −122.637 recorded (cut-off of −2.5).

4. Discussion

To classify the detected in-frame deletion as pathogenic, we applied the following ACMG criteria: PS2 (de novo variant), PM4_Strong (protein changing length variant), PM2 (absent in population databases), PP3 (computational evidence), and PP4 (patient’s phenotype highly specific for a gene). The minigene assay demonstrated that the splicing variant generated an in-frame deletion, and therefore, we applied the PM4 criterion. In addition, we promoted PM4 to a strong criterion since in-frame deletions in COL2A1 are likely to be pathogenic [7,9].
COL2A1 pathogenic variants are associated with many skeletal dysplasia, with a series of phenotypes ranging from lethal to relatively mild forms often reported [6]. Since a dominant inheritance characterizes the diseases associated with COL2A1 mutations, two mechanisms have been linked with pathogenetic COL2A1 variants: Haploinsufficiency and negative dominance, with the latter being related to a more severe phenotype [9].
The substitution of glycine in the G-X-Y repeat of COL2A1 protein is the typical example of dominant-negative mutation and is mainly associated with ACG2. In contrast, variants leading to a premature stop codon can activate a mechanism of nonsense-mediated decay (NMD), resulting in haploinsufficiency of COL2A1. These variants are mainly associated with the mildest clinical forms of skeletal dysplasia [7,9].
Variants that affect splicing can generate either an out-of-frame protein, possibly activating an NMD mechanism or an in-frame protein with a small deletion. The in-frame deletions of COL2A1 are primarily associated with Kniest dysplasia [12], an intermediate phenotype that is less severe than ACG2. In contrast, a milder phenotype is expected in the case of NMD activation caused by an out-of-frame exon deletion [9]. Functional studies assessing the pathogenicity of the variant are thus required in order to determine whether a new variant in a splice site generates a frameshift [16]. In our case, the detected splice site variant generated an in-frame deletion that showed a substantial expression in a minigene splicing assay. Therefore, the severe phenotype observed was possibly due to a dominant-negative mechanism caused by the resulting protein deletion variant.
To better understand the phenotype generated by the COL2A1 variant detected by us, we listed variants of COL2A1 already reported in the literature to cause in-frame deletions in order to perform a genotype–phenotype correlation. The size of the in-frame deletion seems to be very important for the determination of the phenotype. Variants with deletions of 18 amino acids or fewer are associated with less severe phenotypes (Figure 4). Notably, 18 amino acids correspond to the number of residues in a turn of the extended left-handed helix formed by each collagen chain in the collagen triple helix (Figure 4) [17].
Therefore, a deletion longer than 18 amino acid residues might cause the formation of more extended loops, introducing bulkier structural defects in the heterotrimers, whereas shorter loops might be accommodated more easily, producing less irregular protein assemblies. There are only two exceptions to this rule: the quite long deletion p.Gly1164_Ala1199del at C-termini associated with an intermediate phenotype [26], and the small deletion p.Gly1017_Val1022del associated with a severe phenotype [18]. However, the p.Gly1164_Ala1199del variant occurs at the C-terminal side of the triple-helical region. In this location, it might simply disfavor the initial process of triple helix formation. Hence, producing less aberrant multimers and reducing dominant effects. Therefore, the unique exception remains the p.Gly1017_Val1022del variant [18]. In a large study that attempted to show a genotype-phenotype correlation for COL2A1-related disorders, severe phenotypes were found to be more associated with in-frame deletions than frameshift variants [7].
In vitro studies have shown that the in-frame deletion of COL2A1 generates a loop structure in the trimer of collagen type II molecules that are highly susceptible to trypsin’s proteolytic activity [13]. In addition, heterotrimers composed of mutant and wild-type sequences assemble with local disrupted domains at the deletion site, destabilizing the collagen fiber structure. Therefore, a more extended deletion could more efficiently destabilize the trimer, increasing the proteolytic damage operated by matrix proteases.
In the two studies, transgenic mice were generated with a construct containing a human COL2A1 with an in-frame deletion. In the first study, exon 7 was eliminated, while in the second one, 12 exons were removed [27,28]. These transgenic mice showed severe chondrodysplasia and died shortly after birth. Vandenberg et al. [27] hypothesized a mechanism of collagen suicide induced by the short deleted COL2A1 protein. Furthermore, in both cases, a high expression of the transgene seems also relevant to determine the phenotype. Therefore, the expression of a COL2A1 pathogenic variant could determine the generation of a product that interferes with the wild-type form, inhibiting the correct formation of collagen trimers. In our case, the minigene assay was valuable for defining the expression of the COL2A1 carrying the splicing variant.
In conclusion, our study showed that the size and position of the deletions across the helical regions could be relevant for determining the pathogenicity of COL2A1 variants capable of generating in-frame deletions. More cases involving large in-frame deletions are needed to confirm this hypothesis. Functional studies are critical to demonstrate the generation of an expressed in-frame deletion variant, which could help variant classification or explain variant pathogenicity.

Author Contributions

Medical examination of the fetus, V.B. and C.V.; COL2A1 variant detection, A.L.B. and S.G.; COL2A1 segregation analysis, V.B, E.C., and R.I.; COL2A1 minigene generation and expression, V.B., C.B.S., V.D. and R.I.; data analysis, V.B., E.C., E.B., N.P. and R.I.; writing, V.B., E.B., N.P. and R.I. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Ethics Committee “Sezione Area Centro, Regione Calabria”, protocol number 63, date of approval 20 February 2020.

Informed Consent Statement

Written informed consent was obtained from the subjects involved in the study.

Acknowledgments

We thank Fernanda Fabiani for her technical support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Chen, H.; Liu, C.T.; Yang, S.S.; Opitz, J.M. Achondrogenesis: A review with special consideration of achondrogenesis type II (Langer-Saldino). Am. J. Med. Genet. 1981, 10, 379–394. [Google Scholar] [CrossRef] [PubMed]
  2. Langer, L.O., Jr.; Spranger, J.W.; Greinacher, I.; Herdman, R.C. Thanatophoric dwarfism: A condition confused with achondroplasia in the neonate, with brief comments on achondrogenesis and homozygous achondroplasia. Radiology 1969, 92, 285–294. [Google Scholar] [CrossRef] [PubMed]
  3. Saldino, M.D.; Ronald, M. Lethal short-limbed dwarfism: Achondrogenesis and thanatophoric dwarfism. Am. J. Roentgen. Radium Ther. Nucl. Med. 1971, 112, 185–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Warman, M.L.; Cormier-Daire, V.; Hall, C.; Krakow, D.; Lachman, R.; LeMerrer, M.; Mortier, G.; Mundlos, S.; Nishimura, G.; Rimoin, D.L.; et al. Nosology and classification of genetic skeletal disorders: 2010 revision. Am. J. Med. Genet. Part A 2011, 155, 943–968. [Google Scholar] [CrossRef] [PubMed]
  5. Terhal, P.A.; Nievelstein, R.J.; Verver, E.J.; Topsakal, V.; van Dommelen, P.; Hoornaert, K.; Le Merrer, M.; Zankl, A.; Simon, M.E.; Smithson, S.F.; et al. A study of the clinical and radiological features in a cohort of 93 patients with a COL2A1 mutation causing spondyloepiphyseal dysplasia congenita or a related phenotype. Am. J. Med. Genet. Part A 2015, 167, 461–475. [Google Scholar] [CrossRef] [PubMed]
  6. Francomano, C.A.; McIntosh, I.; Wilkin, D.J. Bone dysplasias in man: Molecular insights. Curr. Opin. Genet. Dev. 1996, 6, 301–308. [Google Scholar] [CrossRef]
  7. Zhang, B.; Zhang, Y.; Wu, N.; Li, J.; Liu, H.; Wang, J. Integrated analysis of COL2A1 variant data and classification of type II collagenopathies. Clin. Genet. 2020, 97, 383–395. [Google Scholar] [CrossRef] [Green Version]
  8. Lovell-Badge, R.H.; Bygrave, A.; Bradley, A.; Robertson, E.; Tilly, R.; Cheah, K.S.E. Tissue-specific expression of the human type II collagen gene in mice. Proc. Nat. Acad. Sci. USA 1987, 84, 2803–2807. [Google Scholar] [CrossRef] [Green Version]
  9. Barat-Houari, M.; Sarrabay, G.; Gatinois, V.; Fabre, A.; Dumont, B.; Genevieve, D.; Touitou, I. Mutation Update for COL2A1 Gene Variants Associated with Type II Collagenopathies. Hum. Mutat. 2016, 37, 7–15. [Google Scholar] [CrossRef]
  10. Barat-Houari, M.; Dumont, B.; Fabre, A.; Them, F.T.; Alembik, Y.; Alessandri, J.L.; Amiel, J.; Audebert, S.; Baumann-Morel, C.; Blanchet, P.; et al. The expanding spectrum of COL2A1 gene variants IN 136 patients with a skeletal dysplasia phenotype. Eur. J. Hum. Genet. 2016, 24, 992–1000. [Google Scholar] [CrossRef] [Green Version]
  11. Huang, L.; Chen, C.; Wang, Z.; Sun, L.; Li, S.; Zhang, T.; Luo, X.; Ding, X. Mutation Spectrum and De Novo Mutation Analysis in Stickler Syndrome Patients with High Myopia or Retinal Detachment. Genes 2020, 11, 882. [Google Scholar] [CrossRef]
  12. Wilkin, D.J.; Artz, A.S.; South, S.; Lachman, R.S.; Rimoin, D.L.; Wilcox, W.R.; McKusick, V.A.; Stratakis, C.A.; Francomano, C.A.; Cohn, D.H. Small deletions in the type II collagen triple helix produce kniest dysplasia. Am. J. Med. Genet. 1999, 85, 105–112. [Google Scholar] [CrossRef]
  13. Weis, M.A.; Wilkin, D.J.; Kim, H.J.; Wilcox, W.R.; Lachman, R.S.; Rimoin, D.L.; Cohn, D.H.; Eyre, D.R. Structurally abnormal type II collagen in a severe form of Kniest dysplasia caused by an exon 24 skipping mutation. J. Biol. Chem. 1998, 273, 4761–4768. [Google Scholar] [CrossRef] [Green Version]
  14. Richards, S.; Aziz, N.; Bale, S.; Bick, D.; Das, S.; Gastier-Foster, J.; Grody, W.W.; Hegde, M.; Lyon, E.; Spector, E.; et al. Standards and guidelines for the interpretation of sequence variants: A joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet. Med. 2015, 17, 405–424. [Google Scholar] [CrossRef]
  15. Choi, Y.; Chan, A.P. PROVEAN web server: A tool to predict the functional effect of amino acid substitutions and indels. Bioinformatics 2015, 31, 2745–2747. [Google Scholar] [CrossRef] [Green Version]
  16. Richards, A.J.; Snead, M.P. The influence of pre-mRNA splicing on phenotypic modification in Stickler’s syndrome and other type II collagenopathies. Eye 2008, 22, 1243–1250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Gelse, K.; Pöschl, E.; Aigner, T. Collagens-structure, function, and biosynthesis. Adv. Drug Deliv. Rev. 2003, 55, 1531–1546. [Google Scholar] [CrossRef] [Green Version]
  18. Körkkö, J.; Cohn, D.H.; Ala-Kokko, L.; Krakow, D.; Prockop, D.J. Widely Distributed Mutations in the COL2A1 Gene Produce Achondrogenesis Type II/Hypochondrogenesis. Am. J. Med. Genet. 2000, 92, 95–100. [Google Scholar] [CrossRef]
  19. Winterpacht, A.; Hilbert, M.; Schwarze, U.; Mundlos, S.; Spranger, J.; Zabel, B.U. Kniest and Stickler dysplasia phenotypes caused by collagen type II gene (COL2A1) defect. Nat. Genet. 1993, 3, 323–326. [Google Scholar] [CrossRef] [PubMed]
  20. Bogaert, R.; Wilkin, D.; Wilcox, W.R.; Lachman, R.; Rimoin, D.; Cohn, D.H.; Eyre, D.R. Expression, in cartilage, of a 7-amino-acid deletion in type II collagen from two unrelated individuals with Kniest dysplasia. Am. J. Hum. Genet. 1994, 55, 1128–1136. [Google Scholar] [PubMed]
  21. Fernandes, R.J.; Wilkin, D.J.; Weis, M.A.; Wilcox, W.R.; Cohn, D.H.; Rimoin, D.L.; Eyre, D.R. Incorporation of structurally defective type II collagen into cartilage matrix in kniest chondrodysplasia. Arch. Biochem. Biophys. 1998, 355, 282–290. [Google Scholar] [CrossRef]
  22. Wada, R.; Sawai, H.; Nishimura, G.; Isono, K.; Minagawa, K.; Takenobu, T.; Harada, K.; Tanaka, H.; Ishikura, R.; Komori, S. Prenatal diagnosis of Kniest dysplasia with three-dimensional helical computed tomography. J. Matern. Fetal Neonatal Med. 2011, 24, 1181–1184. [Google Scholar] [CrossRef]
  23. Spranger, J.; Winterpacht, A.; Zabel, B. Kniest dysplasia: Dr. W. Kniest, his patient, the molecular defect. Am. J. Med. Genet. 1997, 69, 79–84. [Google Scholar] [CrossRef]
  24. Winterpacht, A.; Schwarze, U.; Mundlos, S.; Menger, H.; Spranger, J.; Zabel, B. Alternative splicing as the result of a type II procollagen gene (COL2A1) mutation in a patient with Kniest dysplasia. Hum. Mol. Genet. 1994, 3, 1891–1893. [Google Scholar] [CrossRef] [PubMed]
  25. Mortier, G.R.; Weis, M.; Nuytinck, L.; King, L.M.; Wilkin, D.J.; De Paepe, A.; Lachman, R.S.; Rimoin, D.L.; Eyre, D.R.; Cohn, D.H. Report of five novel and one recurrent COL2A1 mutations with analysis of genotype-phenotype correlation in patients with a lethal type II collagen disorder. J. Med. Genet. 2000, 37, 263–271. [Google Scholar] [CrossRef] [Green Version]
  26. Lee, B.; Vissing, H.; Ramirez, F.; Rogers, D.; Rimoin, D. (Identification of the molecular defect in a family with spondyloepiphyseal dysplasia. Science 1989, 244, 978–980. [Google Scholar] [CrossRef] [PubMed]
  27. Vandenberg, P.; Khillan, J.S.; Prockop, D.J.; Helminen, H.; Kontusaari, S.; Ala-Kokko, L. Expression of a partially deleted gene of human type II procollagen (COL2A1) in transgenic mice produces a chondrodysplasia. Proc. Natl. Acad. Sci. USA 1991, 88, 7640–7644. [Google Scholar] [CrossRef] [Green Version]
  28. Metsäranta, M.; Garofalo, S.; Decker, G.; Rintala, M.; de Crombrugghe, B.; Vuorio, E. Chondrodysplasia in transgenic mice harboring a 15-amino acid deletion in the triple helical domain of pro α 1(II) collagen chain. J. Cell Biol. 1992, 118, 203–212. [Google Scholar] [CrossRef]
Figure 1. (A) Three-dimensional ultrasound image of the fetus showing reduced ossification of the sacrum, iliac spines, and arched femurs. (B) Radiograph of the fetus showing reduced ossification of the sacrum, iliac spines, and arched femurs. (C) Frontal view of the fetus postmortem.
Figure 1. (A) Three-dimensional ultrasound image of the fetus showing reduced ossification of the sacrum, iliac spines, and arched femurs. (B) Radiograph of the fetus showing reduced ossification of the sacrum, iliac spines, and arched femurs. (C) Frontal view of the fetus postmortem.
Genes 12 01395 g001
Figure 2. Genealogic tree of the family with Sanger sequencing of exon 21 of COL2A1.
Figure 2. Genealogic tree of the family with Sanger sequencing of exon 21 of COL2A1.
Genes 12 01395 g002
Figure 3. (A) Schematic representation of minigene construct. Exons 19, 20, 21, and 22 are represented, and expected spliced forms are indicated on the right side. Wild-type and mutated constructs are represented on the top and the bottom, respectively. (B) Electrophoresis of PCR amplification of nucleic acids extracted from HEK293T-transfected cells. RNA samples were reverse-transcribed and subjected to PCR amplification with specific primers located in exons 20 (Forward) and 23 (Reverse) of COL2A1. The DNA band of 810 bp represents the non-spliced DNA of the transfected constructs, while the 232 and 133 bp bands represent the spliced transcripts in the cells transfected with the wild-type and mutated constructs. DNA was purified from the 232 and 133 bp bands and then sequenced (EV: Empty vector; WT: Wild-type minigene construct; MUT: Mutated minigene construct; and NC: Negative control). (C) Electropherograms of amplified products (wild-type and mutated) are shown in panel B.
Figure 3. (A) Schematic representation of minigene construct. Exons 19, 20, 21, and 22 are represented, and expected spliced forms are indicated on the right side. Wild-type and mutated constructs are represented on the top and the bottom, respectively. (B) Electrophoresis of PCR amplification of nucleic acids extracted from HEK293T-transfected cells. RNA samples were reverse-transcribed and subjected to PCR amplification with specific primers located in exons 20 (Forward) and 23 (Reverse) of COL2A1. The DNA band of 810 bp represents the non-spliced DNA of the transfected constructs, while the 232 and 133 bp bands represent the spliced transcripts in the cells transfected with the wild-type and mutated constructs. DNA was purified from the 232 and 133 bp bands and then sequenced (EV: Empty vector; WT: Wild-type minigene construct; MUT: Mutated minigene construct; and NC: Negative control). (C) Electropherograms of amplified products (wild-type and mutated) are shown in panel B.
Genes 12 01395 g003
Figure 4. Schematic view of collagen α-1(II) chain (top) that shows the novel protein deletion position reported in this work and previously published deletions. Deletions, characterized by the loss of 18 amino acids or fewer, causing intermediate (*) and severe (**) effects are shown in light green and grey, respectively. Larger deletions causing intermediate (*) and severe (**) effects are shown in light blue and yellow, respectively. In the severe group, there are cases of ACG2 or hypochondrogenesis. In the intermediate group, there are cases of Kniest dysplasia or SEDC. Periodicity of 18 amino acids (bottom) of the extended left-handed helix of each protein chain in the collagen triple helix are exemplified here by a portion of the Protein Data Bank structure 3HQV (Cα atoms are represented by spheres). Amino acid numbering refers to the protein with NCBI accession code NP_001835.3. p.Gly423_Thr455del (our study); p.Gly732_Arg767del and p.Gly1017_Val1022del (Körkkö et al. 2000) [18]; p.Pro916_Gly1074del (Barat-Houari et al. 2016) [10]; p.Gly291_Lys308del (Winterpacht et al. 1993) [19]; p.Ala302_Lys308del (Bogaert et al. 1994) [20]; p.Gly324_Ala341del, p.Gly357_Gly363del, p.Gly423_Ala428del, and p.Gly456_Pro473del (Wilkin et al. 1999) [12]; p.Gly342_Pro356del (Fernandes et al. 1988; Wada et al. 2011) [21,22]; p.Gly408_Ala422del (Spranger et al. 1997) [23]; p.Gly474_Gln479del (Winterpacht et al. 1994) [24]; p.Gly561_Ser578del (Weis et al. 1998) [13]; p.Gly804_Pro821del (Mortier et al. 2000) [25]; p.Gly1164_Ala1199del (Lee et al. 1989) [26].
Figure 4. Schematic view of collagen α-1(II) chain (top) that shows the novel protein deletion position reported in this work and previously published deletions. Deletions, characterized by the loss of 18 amino acids or fewer, causing intermediate (*) and severe (**) effects are shown in light green and grey, respectively. Larger deletions causing intermediate (*) and severe (**) effects are shown in light blue and yellow, respectively. In the severe group, there are cases of ACG2 or hypochondrogenesis. In the intermediate group, there are cases of Kniest dysplasia or SEDC. Periodicity of 18 amino acids (bottom) of the extended left-handed helix of each protein chain in the collagen triple helix are exemplified here by a portion of the Protein Data Bank structure 3HQV (Cα atoms are represented by spheres). Amino acid numbering refers to the protein with NCBI accession code NP_001835.3. p.Gly423_Thr455del (our study); p.Gly732_Arg767del and p.Gly1017_Val1022del (Körkkö et al. 2000) [18]; p.Pro916_Gly1074del (Barat-Houari et al. 2016) [10]; p.Gly291_Lys308del (Winterpacht et al. 1993) [19]; p.Ala302_Lys308del (Bogaert et al. 1994) [20]; p.Gly324_Ala341del, p.Gly357_Gly363del, p.Gly423_Ala428del, and p.Gly456_Pro473del (Wilkin et al. 1999) [12]; p.Gly342_Pro356del (Fernandes et al. 1988; Wada et al. 2011) [21,22]; p.Gly408_Ala422del (Spranger et al. 1997) [23]; p.Gly474_Gln479del (Winterpacht et al. 1994) [24]; p.Gly561_Ser578del (Weis et al. 1998) [13]; p.Gly804_Pro821del (Mortier et al. 2000) [25]; p.Gly1164_Ala1199del (Lee et al. 1989) [26].
Genes 12 01395 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bruni, V.; Spoleti, C.B.; La Barbera, A.; Dattilo, V.; Colao, E.; Votino, C.; Bellacchio, E.; Perrotti, N.; Giglio, S.; Iuliano, R. A Novel Splicing Variant of COL2A1 in a Fetus with Achondrogenesis Type II: Interpretation of Pathogenicity of In-Frame Deletions. Genes 2021, 12, 1395. https://doi.org/10.3390/genes12091395

AMA Style

Bruni V, Spoleti CB, La Barbera A, Dattilo V, Colao E, Votino C, Bellacchio E, Perrotti N, Giglio S, Iuliano R. A Novel Splicing Variant of COL2A1 in a Fetus with Achondrogenesis Type II: Interpretation of Pathogenicity of In-Frame Deletions. Genes. 2021; 12(9):1395. https://doi.org/10.3390/genes12091395

Chicago/Turabian Style

Bruni, Valentina, Cristina Barbara Spoleti, Andrea La Barbera, Vincenzo Dattilo, Emma Colao, Carmela Votino, Emanuele Bellacchio, Nicola Perrotti, Sabrina Giglio, and Rodolfo Iuliano. 2021. "A Novel Splicing Variant of COL2A1 in a Fetus with Achondrogenesis Type II: Interpretation of Pathogenicity of In-Frame Deletions" Genes 12, no. 9: 1395. https://doi.org/10.3390/genes12091395

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop