Skip to main content
Erschienen in: BMC Medical Genetics 1/2019

Open Access 01.12.2019 | Case report

A novel homozygous frame-shift mutation in the SLC29A3 gene: a new case report and review of literature

verfasst von: Sadaf Noavar, Samira Behroozi, Taraneh Tatarcheh, Farshid Parvini, Majid Foroutan, Hossein Fahimi

Erschienen in: BMC Medical Genetics | Ausgabe 1/2019

Abstract

Background

The SLC29A3 gene, encoding a nucleoside transporter protein, is found in intracellular membranes. Based on the literatures, mutations in this gene cause a wide range of clinical manifestations including H syndrome, pigmented hypertrichosis with insulin dependent diabetes, Faisalabad histiocytosis, and dysosteosclerosis. However, all these disorders with their different names and terminologies are actually the same entity termed H syndrome.

Case presentation

We report four GJB2 and GJB6 negative deaf patients from two Iranian related families who present the associated symptoms of SLC29A3-disorder. Whole Exome Sequencing (WES) using Next Generation Illumina Sequencing was used to enrich all exons of protein-coding genes as well as some other important genomic regions in one of studied patients. A novel homozygous frame-shift mutation c.307-308delTT (p.Phe103fs) in exon 3 of SLC29A3 gene was identified in a 35 years old man with profound hearing loss, camptodactyly, rheumatoid arthritis and delayed puberty without any skin changes, short stature and insulin dependent diabetes mellitus. The mutation found was also confirmed by Sanger sequencing in other studied patients and their healthy parents. In compared to proband, however the clinical manifestations of these patients were different, indicating variable expressivity of mutant SLC29A3 gene as well as possible involvement of other modifier genes.

Conclusion

The present study uncovered a rare novel homozygous frame-shift mutation c.307-308delTT in SLC29A3 gene of four related patients with various manifestation of SLC29A3-disorder. Such studies can help to conduct genetic counseling and subsequently, prenatal diagnosis more accurately for individuals at the high risk of these types of genetic disorders.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ASPCR
Allele Specific PCR
CRP
C reactive protein
DSS
Dysosteosclerosis
ENT3
Equilibrative nucleoside transporter 3
ESR
Erythrocyte sedimentation rate
FHC
Faisalabad histiocytosis
GJB2
Gap junction beta-2 protein
GJB6
Gap Junction Beta 6 protein
IDDM
Insulin dependent diabetes mellitus
NGS
Next generation sequencing
PHID
Pigmented hypertrichosis with insulin dependent diabetes
SHML
Histiocytosis with massive lymphadenopathy
SLC29A3
Solute Carrier Family 29 Member 3
WES
Whole Exome Sequencing

Background

The SLC29A3 gene, also called ENT3 (equilibrative nucleoside transporter 3), encodes a nucleoside transporter protein. The ENT3 protein is found in intracellular membranes, especially in lysosomal and mitochondrial membranes [1, 2]. Because of wide spread biochemical roles of nucleoside molecules, any disruption in metabolism and trafficking of nucleosides could result in various abnormal phenotypes.
Based on the literatures, the mutations in the SLC29A3 gene cause a wide range of different clinical manifestations including H syndrome (defined by cardiac anomalies, camptodactyly, short stature, hypergonadotropic hypogonadism, hepatosplenomegaly, scrotal masses, growth retardation, and sensorineural hearing loss [37]), pigmented hypertrichosis with insulin dependent diabetes (PHID) (characterized by hyperpigmented and hypertrichotic skin, hepatosplenomegaly, hypogonadism, diabetes mellitus, comptodactyly, clinodactyly, chronic inflammatory syndrome, and growth retardation [8]), Faisalabad histiocytosis (FHC) (defined by deafness, growth retardation, hypogonadism, camptodactyly, and Rosai Dorfman disease [9, 10]), and dysosteosclerosis (DSS) [8, 11, 12] (characterized by hyperpigmented skin, frontal bossing, mid-face hypoplasia, short stature, sclerotic platyspondyly, otosclerosis, and compression of CNS and cranial nerves [11]). However, it is worth mentioning that all these disorders, with their different names and terminologies, are actually the same entity termed H syndrome. For this reason, their description as different entities is misleading. As outlined above, a number of phenotypes including patches and plaques of hyperpigmented and hypertrichotic skin, short stature, camptodactyly, hearing problems, myelofibrosis, hepatosplenomegaly, pulmonary stenosis, pericarditis, diabetes, and hypogonadism [7, 11, 13] are common among H syndrome affected patients but not all phenotypes are seen in all patients. In this report, we describe a novel homozygous frame-shift mutation in SLC29A3 gene and its related phenotypes in two Iranian related families.

Case presentation

The studied families are of Iranian origin located in Semnan province (central Iran). As shown in Fig. 1, four affected individuals are included in two related families (a brother and a sister from each family), with two common ancestors. All four patients were born at term, with normal health indicators and metrics, following an uneventful pregnancy. Their ages ranged between 35 and 46 years. The clinical and demographic data of the patients are summarized in Table 1. In addition, camptodactyly phenotype in patient III-5 is depicted in Fig. 2.
Table 1
Summary of clinical and demographic findings
 
Family 1
Family 2
 
Patient III-2
Patient III-5
Patient III-8
Patient III-11a
Sex
Female
Male
Female
Male
Age (years)
46
35
37
41
Height (cm)
170
178
167
180
Age of onset (years)b
15
6
7–8
5
Hearing loss
Mild
Profound
Profound
Profound
Camptodactyly
+
+
+
+
ESR (mm/h)
70–110
45
No data
No data
Polyclonal gammopathy
Severe
Mild
Mild
Skin changes
No
No
No
No
IDDM
+
+
Arthropathy
+
+
+
+
Delayed puberty
Yes
Yes
No
No
CRP mg/dL
30–90
34
No data
No data
Rheumatoid arthritis
+
+
+
+
Lymph nodes
Generalized Lymphoadenopathy
No
No
No
ESR erythrocyte sedimentation rate, IDDM  Insulin dependent diabetes mellitus, CRP C reactive protein
a The patient III-11 died due to the diabetes
b For clinical manifestations
After obtaining informed consent, the peripheral blood samples were collected from patients and their family members. Genomic DNA was extracted from blood samples by QIAamp DNA Blood Mini Kit (Germany) according to the manufacturer’s instructions. At first, the patients were investigated for mutations of GJB2 gene using ASPCR (Allele Specific PCR) and sequencing followed by screening of two known deletions del(D13S1830) and del(D13S1854) of GJB6 gene. Since the studied patients were negative for GJB2 and GJB6 genes mutations, the patient III-5 was more investigated by Next Generation Sequencing. For this purpose, Whole Exome Sequencing (WES) was used to enrich all exons of protein-coding genes as well as some other important genomic regions. The WES was performed to sequence close to 100 million reads on Illumina HiSeq2000 Sequencer. Generally, the test platform examined > 95% of the targeted regions with sensitivity of above 99%. In this test, point mutations and micro-insertions/deletions as well as duplications (< 20 bp) can be simultaneously detected. Analysis of the sequencing results was performed using BWA aligner [14], annovar [15] and GATK [16] open access software as well as public databases ExAC, gnomAD, Kaviar (~Known VARiants) and GME. Additionally, ACMG guidelines and local population database with more than 1500 unrelated individuals (BayanGene) were used. Standard bioinformatics tools used were as follows: CADD_phred, REVEL, MCAP, SIFT, Polyphen, LRT, MutationTaster, and MutationAssessor. Furthermore, to confirm the novel mutation found in the SLC29A3 gene, PCR and sequencing were performed for patients and their normal family members using following primers: F-5′ CAGTCCATGGGCAGAAGTGT 3′ and R- 5′ TCGCCTACCTGTTGACAAGC 3′ (PCR product: 401 bp). Finally, the Sanger sequencing data was analyzed by Chromas software.
Based on the results, all four analyzed patients were negative for GJB2 mutations and two common large deletions of GJB6 gene. Subsequently, the NGS analysis of proband (patient III-5) revealed one deleterious homozygous mutation in SLC29A3 gene. This homozygous deletion includes two thymidine nucleotides (c.307-308delTT) of exon 3 causing a frame-shift mutation that immediately, makes a stop codon (TGA) instead of Phe103 codon (TTT) (SLC29A3:NM_001174098:exon3:c.307-308delTT:p.Phe103fs) (Fig. 3). The mutation found, validated by Sanger sequencing.
At the next step, segregation analysis of novel homozygous mutation found was performed for other family members including another three patients (patients III-2, III-8, and III-11). As expected, parents of proband were heterozygote for reported mutation. A similar result was achieved from the parents of patient III-8 (data not shown). Furthermore, the obtained results showed that patients III-2, III-8, and III-11 have the found deleterious mutation. Furthermore, three normal siblings of the patients were also checked for the deleterious mutation 307-308delTT of SLC29A3 gene. The results clearly revealed that two of them (III-1 and III-6) are heterozygote and one (III-9) is normal homozygote. The corresponding family pedigree with the results of sequence analyses are illustrated in Fig. 1.

Discussion and conclusion

As previously mentioned, the mutation in SLC29A3 gene causes a wide range of mild to severe different medical manifestations which are known as the same entity termed H syndrome. To date, more than 22 pathogenic or likely pathogenic mutations have been reported in the SLC29A3 gene (Table 2). As shown in Table 2, the preliminary report for 308-309delTT (OMIM: 612373.0009) frame-shift mutation, which is resulted in truncated protein has been published in 2010 [10]. However, they found a heterozygous form of this mutation in combination with heterozygous missense mutation Gly437Arg as compound heterozygote, which is led to H syndrome. Here, we have explained the first report of novel homozygous form of this deleterious mutation from two Iranian related families. All four reported patients were results of consanguineous marriages. As previously pointed out, this frame-shift mutation is resulted in premature protein truncation. The truncated ENT3 protein loses its 10 out of 11 transmembrane domains (Fig. 3). Therefore, it seems that this homozygous mutation, as a loss of function mutation, leads to elimination of ENT3 protein function.
Table 2
A summary of the reported pathogenic/likely pathogenic mutations of SLC29A3 gene
Nucleotide change
Genotype
Consequence
(Clinical significance) Main Phenotypes
Family origin
Reference/ (variation ID in ClinVar)
c.243delA
Homo
p.Lys81Asnfs
Pathogenic
Nasal infiltration, Pancreatic exocrine deficiency, Insulin-dependent diabetes, Contractures of the fingers, Contractures of the toes, Retroperitoneal fibrosis
Moroccan
[17]
c.308-309delTT
Hetero
p.Phe103Terfs
(Pathogenic)
Histiocytosis, Rosai-Dorfman disease
Turkish and Palestinian
[10]
c.300 + 1G > A (IVS2 + 1G > A)a
Homo
Splice site
Pakistani
c.73C > T
p.Arg25Ter
(Likely pathogenic)
Histiocytosis-lymphadenopathy plus syndrome
(ID: 212200)
c.300 + 1G > C
Splice site
(Pathogenic)
not provided
(ID: 427021)
c.347 T > G
Homo
p.Met116Arg
(Pathogenic)
insulin-dependent diabetes mellitus and pigmented hypertrichotic skin lesions
Australian Lebanese
[8]
c.940delT
p.Tyr314ThrfsTer91
Indian
c.1309G > A
p.Gly437Arg
Pakistani
c.1330G > T
p.Glu444Ter
North American Caucasian
c.1346C > G
p.Thr449Arg
Australian Lebanese
c.479G > A
p.Trp160Ter
(Pathogenic)
Histiocytosis-lymphadenopathy plus syndrome
(ID: 573984)
c.607 T > C
Hetero
p.Ser203Pro
(Pathogenic)
Dysosteosclerosis
[11]
c.1157G > A
p.Arg386Gln
c.1346C > G
Homo
p.Thr449Arg
c.714_715invTG
p.Val239Ile
(Likely pathogenic)
Histiocytosis-lymphadenopathy plus syndrome
(ID: 300363)
c.1001A > G
p.Asn334Ser
(Likely pathogenic)
Histiocytosis-lymphadenopathy plus syndrome
(ID: 300368)
c.1045delC
Homo
p.Leu349Serfs
(Pathogenic)
Hyperpigmentation, Fixed flexion contractures of proximal interphalangeal joints, Hallux valgus, Prominent gynecomastia, histiocytic and dendritic infiltrate
Bulgarian
[18]
c.1087C > T
p.Arg363Trp
Spanish
[4]
c.1088G > A
p.Arg363Gln
Arab
c.1228C > T
p.Gln410Ter
(Pathogenic)
Histiocytosis-lymphadenopathy plus syndrome
(ID: 130338)
c.1279G > A
Hetero
p.Gly427Ser
(Pathogenic)
seronegative polyarthritis, hypogonadotropic hypogonadism, hearing
loss, Proptosis, Arthropathy, Camptodactyly, Delayed puberty, Polyclonal gammopathy
Arab
[7]
c.307-308delTT
Homo
p.Phe103Terfs
(Pathogenic)
Hearing loss, Camptodactyly, Polyclonal gammopathy, Arthropathy, Delayed puberty, Rheumatoid arthritis
Iranian
This report
aIVS InterVening Sequence (i.e. an intron)
As shown in Table 1, some of the common manifestations have been seen in our patients. Nonetheless, some of the phenotypes have not seen or were different among patients of two studied families. For example, although the short stature is reported in mostly reports for H syndrome [7, 11, 13], all four patients in this report have normal height (167–180 cm), in accordance with two previously reported cases from Morocco [17]. Similarly, and in accordance with some reports [8, 12], all patients in this study have moderate (patient III-2) to profound deafness (patients III-5, III-8, and III-11). Deafness has also been reported in other previous reports [4, 18], which is in accordance with our observations. Although these reported patients showed other manifestations including, hyperpigmentation, hypertrichosis, fixed flexion contractures of proximal interphalangeal joints, Hallux valgus and fixed flexion contractures of toe joints. However, the hearing loss has not been reported in originally reported PHID (other form of H syndrome) patients [19, 20]. Delayed puberty was observed in patients of family 1 (Patients III-2 and III-5). This observation is in line with those reported by Cliffe et al. (2009) and Hussain et al. (2009) [8, 19].
In accordance with other reports [7, 11], all four studied patients were suffered from camptodactyly. While the normal ESR range is 0–22 mm/h for men and 0–29 mm/h for women, a high ESR was observed in two patients III-2 and III-5. On the other words, it seems that the high levels of CRP in patients III-2 and III-5 could because of rheumatoid arthritis in these patients (normal CRP level is below 3.0 mg/L). Although, there was no data for ESR and CRP levels of patients III-8 and III-11, clinical symptoms of the rheumatoid arthritis were also confirmed by a rheumatologist in these patients. This result is in agreement with clinical symptoms of a previously reported patient [7]. While, the severe polyclonal gammopathy was observed in patient III-2, a mild representation was observed in patients III-5 and III-8 indicating variable expressivity of mutant SLC29A3 gene. Polyclonal gammopathy could be due to the elevated production of immunoglobulins by plasma cells [21]. Unlike the monoclonal gammopathy, polyclonal gammopathy is associated with a nonmalignant condition. In the present study, patient III-2 has been shown generalized lymphadenopathy. This observation is similar to previously reported lymphadenopathy in mutated SLC29A3 patients [5, 7, 8, 10, 11, 20]. Corresponding to the previously report from Morocco [17], despite the hyperpigmented/hypertrichotic skin is the common feature in SLC29A3 mutants [11], none of the patients reported in this paper have skin changes. Similarly, although mutation in SLC29A3 gene mostly leads to diabetes [7, 11, 22], we observed this phenotype only in the patients of the family 2 (patients III-8 and III-11) showing the variable expressivity and eventually involvement of other genes. Similar observation has been conducted by Spiegel et al. (2010) [7]. All four patients were suffered from arthropathy and rheumatoid arthritis. This is in accordance with several previously reported cases [7, 23].
In conclusion, this report shows that a homozygous frame-shift mutation c.307-308delTT results in a complex of clinical manifestations of H syndrome. In addition to allelic heterogeneity, it seems that these various clinical symptoms clearly confirm the pleiotropic effects and variable expressivity of SLC29A3 gene as well as possible involvement of other genes. On the other words, the obtained results showed that whole-exome sequencing is a useful method discovering genes involved in human genetic diseases.

Acknowledgments

We are grateful to patients and their families who kindly consented to join the study. We thank Dr. Masoumeh Heshmati and Dr. Nooshin Barikrow (from Pharmaceutical Sciences Research Center, Tehran medical Sciences, Islamic Azad University, Tehran, Iran) for technical collaborations, as well.
This study had been approved by the ethics committee of the pharmaceutical sciences branch of Islamic Azad University, Tehran, Iran. Written informed consent was obtained from the parents to take part in this study and for possible publication. A copy of the written consent is available for review by the Editor of this journal.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Endo Y, Obata T, Murata D, Ito M, Sakamoto K, Fukushima M, et al. Cellular localization and functional characterization of the equilibrative nucleoside transporters of antitumor nucleosides. Cancer Sci. 2007;98(10):1633–7.PubMedCrossRef Endo Y, Obata T, Murata D, Ito M, Sakamoto K, Fukushima M, et al. Cellular localization and functional characterization of the equilibrative nucleoside transporters of antitumor nucleosides. Cancer Sci. 2007;98(10):1633–7.PubMedCrossRef
2.
Zurück zum Zitat Elwi AN, Damaraju VL, Baldwin SA, Young JD, Sawyer MB, Cass CE. Renal nucleoside transporters: physiological and clinical implications. Biochem Cell Biol. 2006;84(6):844–58.PubMedCrossRef Elwi AN, Damaraju VL, Baldwin SA, Young JD, Sawyer MB, Cass CE. Renal nucleoside transporters: physiological and clinical implications. Biochem Cell Biol. 2006;84(6):844–58.PubMedCrossRef
3.
Zurück zum Zitat Molho-Pessach V, Agha Z, Aamar S, Glaser B, Doviner V, Hiller N, et al. The H syndrome: a genodermatosis characterized by indurated, hyperpigmented, and hypertrichotic skin with systemic manifestations. J Am Acad Dermatol. 2008;59(1):79–85.PubMedCrossRef Molho-Pessach V, Agha Z, Aamar S, Glaser B, Doviner V, Hiller N, et al. The H syndrome: a genodermatosis characterized by indurated, hyperpigmented, and hypertrichotic skin with systemic manifestations. J Am Acad Dermatol. 2008;59(1):79–85.PubMedCrossRef
4.
Zurück zum Zitat Molho-Pessach V, Suarez J, Perrin C, Chiaverini C, Doviner V, Tristan-Clavijo E, et al. The H syndrome: two novel mutations affecting the same amino acid residue of hENT3. J Dermatol Sci. 2010;57(1):59–61.PubMedCrossRef Molho-Pessach V, Suarez J, Perrin C, Chiaverini C, Doviner V, Tristan-Clavijo E, et al. The H syndrome: two novel mutations affecting the same amino acid residue of hENT3. J Dermatol Sci. 2010;57(1):59–61.PubMedCrossRef
5.
Zurück zum Zitat Kismet E, KöseoĞlu V, Atay AA, Devecİ S, Demİrkaya E, Tuncer K. Sinus histiocytosis with massive lymphadenopathy in three brothers. Pediatr Int. 2005;47(4):473–6.PubMedCrossRef Kismet E, KöseoĞlu V, Atay AA, Devecİ S, Demİrkaya E, Tuncer K. Sinus histiocytosis with massive lymphadenopathy in three brothers. Pediatr Int. 2005;47(4):473–6.PubMedCrossRef
6.
Zurück zum Zitat Broshtilova V, Ramot Y, Molho-Pessach V, Zlotogorski A. Diabetes mellitus may be the earliest and sole manifestation of the H syndrome. Diabet Med. 2009;26(11):1179–80.PubMedCrossRef Broshtilova V, Ramot Y, Molho-Pessach V, Zlotogorski A. Diabetes mellitus may be the earliest and sole manifestation of the H syndrome. Diabet Med. 2009;26(11):1179–80.PubMedCrossRef
7.
Zurück zum Zitat Spiegel R, Cliffe ST, Buckley MF, Crow YJ, Urquhart J, Horovitz Y, et al. Expanding the clinical spectrum of SLC29A3 gene defects. Eur J Med Genetics. 2010;53(5):309–13.CrossRef Spiegel R, Cliffe ST, Buckley MF, Crow YJ, Urquhart J, Horovitz Y, et al. Expanding the clinical spectrum of SLC29A3 gene defects. Eur J Med Genetics. 2010;53(5):309–13.CrossRef
8.
Zurück zum Zitat Cliffe ST, Kramer JM, Hussain K, Robben JH, de Jong EK, de Brouwer AP, et al. SLC29A3 gene is mutated in pigmented hypertrichosis with insulin-dependent diabetes mellitus syndrome and interacts with the insulin signaling pathway. Hum Mol Genet. 2009;18(12):2257–65.PubMedCrossRef Cliffe ST, Kramer JM, Hussain K, Robben JH, de Jong EK, de Brouwer AP, et al. SLC29A3 gene is mutated in pigmented hypertrichosis with insulin-dependent diabetes mellitus syndrome and interacts with the insulin signaling pathway. Hum Mol Genet. 2009;18(12):2257–65.PubMedCrossRef
9.
Zurück zum Zitat Rossbach HC, Dalence C, Wynn T, Tebbi C. Faisalabad histiocytosis mimics Rosai-Dorfman disease: brothers with lymphadenopathy, intrauterine fractures, short stature, and sensorineural deafness. Pediatr Blood Cancer. 2006;47(5):629–32.PubMedCrossRef Rossbach HC, Dalence C, Wynn T, Tebbi C. Faisalabad histiocytosis mimics Rosai-Dorfman disease: brothers with lymphadenopathy, intrauterine fractures, short stature, and sensorineural deafness. Pediatr Blood Cancer. 2006;47(5):629–32.PubMedCrossRef
10.
Zurück zum Zitat Morgan NV, Morris MR, Cangul H, Gleeson D, Straatman-Iwanowska A, Davies N, et al. Mutations in SLC29A3, encoding an equilibrative nucleoside transporter ENT3, cause a familial histiocytosis syndrome (Faisalabad histiocytosis) and familial Rosai-Dorfman disease. PLoS Genet. 2010;6(2):e1000833.PubMedPubMedCentralCrossRef Morgan NV, Morris MR, Cangul H, Gleeson D, Straatman-Iwanowska A, Davies N, et al. Mutations in SLC29A3, encoding an equilibrative nucleoside transporter ENT3, cause a familial histiocytosis syndrome (Faisalabad histiocytosis) and familial Rosai-Dorfman disease. PLoS Genet. 2010;6(2):e1000833.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Campeau PM, Lu JT, Sule G, Jiang M-M, Bae Y, Madan S, et al. Whole-exome sequencing identifies mutations in the nucleoside transporter gene SLC29A3 in dysosteosclerosis, a form of osteopetrosis. Hum Mol Genet. 2012;21(22):4904–9.PubMedPubMedCentralCrossRef Campeau PM, Lu JT, Sule G, Jiang M-M, Bae Y, Madan S, et al. Whole-exome sequencing identifies mutations in the nucleoside transporter gene SLC29A3 in dysosteosclerosis, a form of osteopetrosis. Hum Mol Genet. 2012;21(22):4904–9.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Jonard L, Couloigner V, Pierrot S, Louha M, Gherbi S, Denoyelle F, et al. Progressive hearing loss associated with a unique cervical node due to a homozygous SLC29A3 mutation: a very mild phenotype. Eur J Med Genetics. 2012;55(1):56–8.CrossRef Jonard L, Couloigner V, Pierrot S, Louha M, Gherbi S, Denoyelle F, et al. Progressive hearing loss associated with a unique cervical node due to a homozygous SLC29A3 mutation: a very mild phenotype. Eur J Med Genetics. 2012;55(1):56–8.CrossRef
13.
Zurück zum Zitat Melki I, Lambot K, Jonard L, Couloigner V, Quartier P, Neven B, et al. Mutation in the SLC29A3 gene: a new cause of a monogenic, autoinflammatory condition. Pediatrics. 2013;131(4):1308–13.PubMedCrossRef Melki I, Lambot K, Jonard L, Couloigner V, Quartier P, Neven B, et al. Mutation in the SLC29A3 gene: a new cause of a monogenic, autoinflammatory condition. Pediatrics. 2013;131(4):1308–13.PubMedCrossRef
15.
Zurück zum Zitat Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38(16):e164–e.PubMedPubMedCentralCrossRef Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38(16):e164–e.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, et al. The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20(9):1297–303.PubMedPubMedCentralCrossRef McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, et al. The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20(9):1297–303.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Bolze A, Abhyankar A, Grant AV, Patel B, Yadav R, Byun M, et al. A mild form of SLC29A3 disorder: a frameshift deletion leads to the paradoxical translation of an otherwise noncoding mRNA splice variant. PLoS One. 2012;7(1):e29708.PubMedPubMedCentralCrossRef Bolze A, Abhyankar A, Grant AV, Patel B, Yadav R, Byun M, et al. A mild form of SLC29A3 disorder: a frameshift deletion leads to the paradoxical translation of an otherwise noncoding mRNA splice variant. PLoS One. 2012;7(1):e29708.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Molho-Pessach V, Lerer I, Abeliovich D, Agha Z, Libdeh AA, Broshtilova V, et al. The H syndrome is caused by mutations in the nucleoside transporter hENT3. Am J Hum Genet. 2008;83(4):529–34.PubMedPubMedCentralCrossRef Molho-Pessach V, Lerer I, Abeliovich D, Agha Z, Libdeh AA, Broshtilova V, et al. The H syndrome is caused by mutations in the nucleoside transporter hENT3. Am J Hum Genet. 2008;83(4):529–34.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Hussain K, Padidela R, Kapoor RR, James C, Banerjee K, Harper J, et al. Diabetes mellitus, exocrine pancreatic deficiency, hypertrichosis, hyperpigmentation, and chronic inflammation: confirmation of a syndrome. Pediatr Diabetes. 2009;10(3):193–7.PubMedCrossRef Hussain K, Padidela R, Kapoor RR, James C, Banerjee K, Harper J, et al. Diabetes mellitus, exocrine pancreatic deficiency, hypertrichosis, hyperpigmentation, and chronic inflammation: confirmation of a syndrome. Pediatr Diabetes. 2009;10(3):193–7.PubMedCrossRef
20.
Zurück zum Zitat Prendiville J, Rogers M, Kan A, Fd C, Wong M, Junker A, et al. Pigmented hypertrichotic dermatosis and insulin dependent diabetes: manifestations of a unique genetic disorder? Pediatr Dermatol. 2007;24(2):101–7.PubMedCrossRef Prendiville J, Rogers M, Kan A, Fd C, Wong M, Junker A, et al. Pigmented hypertrichotic dermatosis and insulin dependent diabetes: manifestations of a unique genetic disorder? Pediatr Dermatol. 2007;24(2):101–7.PubMedCrossRef
21.
Zurück zum Zitat Dispenzieri A, Gertz MA, Therneau TM, Kyle RA, editors. Retrospective cohort study of 148 patients with polyclonal gammopathy. Mayo Clin Proc. 2001;76(5):476–87. Dispenzieri A, Gertz MA, Therneau TM, Kyle RA, editors. Retrospective cohort study of 148 patients with polyclonal gammopathy. Mayo Clin Proc. 2001;76(5):476–87.
22.
Zurück zum Zitat Molho-Pessach V, Ramot Y, Camille F, Doviner V, Babay S, Luis SJ, et al. H syndrome: the first 79 patients. J Am Acad Dermatol. 2014;70(1):80–8.PubMedCrossRef Molho-Pessach V, Ramot Y, Camille F, Doviner V, Babay S, Luis SJ, et al. H syndrome: the first 79 patients. J Am Acad Dermatol. 2014;70(1):80–8.PubMedCrossRef
23.
Zurück zum Zitat Bloom JL, Lin C, Imundo L, Guthery S, Stepenaskie S, Galambos C, et al. H syndrome: 5 new cases from the United States with novel features and responses to therapy. Pediatr Rheumatol. 2017;15(1):76.CrossRef Bloom JL, Lin C, Imundo L, Guthery S, Stepenaskie S, Galambos C, et al. H syndrome: 5 new cases from the United States with novel features and responses to therapy. Pediatr Rheumatol. 2017;15(1):76.CrossRef
Metadaten
Titel
A novel homozygous frame-shift mutation in the SLC29A3 gene: a new case report and review of literature
verfasst von
Sadaf Noavar
Samira Behroozi
Taraneh Tatarcheh
Farshid Parvini
Majid Foroutan
Hossein Fahimi
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Medical Genetics / Ausgabe 1/2019
Elektronische ISSN: 1471-2350
DOI
https://doi.org/10.1186/s12881-019-0879-7

Weitere Artikel der Ausgabe 1/2019

BMC Medical Genetics 1/2019 Zur Ausgabe