[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (39)

Search Parameters:
Keywords = DFNB77

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 6380 KiB  
Article
Novel Cases of Non-Syndromic Hearing Impairment Caused by Pathogenic Variants in Genes Encoding Mitochondrial Aminoacyl-tRNA Synthetases
by María Domínguez-Ruiz, Margarita Olarte, Esther Onecha, Irene García-Vaquero, Nancy Gelvez, Greizy López, Manuela Villamar, Matías Morín, Miguel A. Moreno-Pelayo, Carmelo Morales-Angulo, Rubén Polo, Martha L. Tamayo and Ignacio del Castillo
Genes 2024, 15(7), 951; https://doi.org/10.3390/genes15070951 - 19 Jul 2024
Viewed by 394
Abstract
Dysfunction of some mitochondrial aminoacyl-tRNA synthetases (encoded by the KARS1, HARS2, LARS2 and NARS2 genes) results in a great variety of phenotypes ranging from non-syndromic hearing impairment (NSHI) to very complex syndromes, with a predominance of neurological signs. The diversity of [...] Read more.
Dysfunction of some mitochondrial aminoacyl-tRNA synthetases (encoded by the KARS1, HARS2, LARS2 and NARS2 genes) results in a great variety of phenotypes ranging from non-syndromic hearing impairment (NSHI) to very complex syndromes, with a predominance of neurological signs. The diversity of roles that are played by these moonlighting enzymes and the fact that most pathogenic variants are missense and affect different domains of these proteins in diverse compound heterozygous combinations make it difficult to establish genotype–phenotype correlations. We used a targeted gene-sequencing panel to investigate the presence of pathogenic variants in those four genes in cohorts of 175 Spanish and 18 Colombian familial cases with non-DFNB1 autosomal recessive NSHI. Disease-associated variants were found in five cases. Five mutations were novel as follows: c.766C>T in KARS1, c.475C>T, c.728A>C and c.1012G>A in HARS2, and c.795A>G in LARS2. We provide audiograms from patients at different ages to document the evolution of the hearing loss, which is mostly prelingual and progresses from moderate/severe to profound, the middle frequencies being more severely affected. No additional clinical sign was observed in any affected subject. Our results confirm the involvement of KARS1 in DFNB89 NSHI, for which until now there was limited evidence. Full article
(This article belongs to the Section Cytogenomics)
Show Figures

Figure 1

Figure 1
<p>Pathogenic variants that were found in <span class="html-italic">KARS1</span> in this study. (<b>a</b>) Pedigrees showing the segregation of variants. Arrow indicates propositus. (<b>b</b>) Electropherograms from subject HRC19 II:1 (left panel) and from subject HRC20 II:3 (right panel). (<b>c</b>) Alignment of KARS1 protein orthologous sequences from human and seven other animal species. Asterisks indicate identical residues across all sequences; colons, conserved positions (residues of strongly similar properties); periods, semi-conserved positions (residues of weakly similar properties). Sequence accession numbers: <span class="html-italic">Homo sapiens</span> (NP_001123561.1); <span class="html-italic">Mus musculus</span> (NP_001124340.1); <span class="html-italic">Bos taurus</span> (XP_010812603.1); <span class="html-italic">Gallus gallus</span> (NP_001025754); <span class="html-italic">Xenopus tropicalis</span> (XP_012816251.2); <span class="html-italic">Danio rerio</span> (NP_001002386.1); <span class="html-italic">Drosophila melanogaster</span> (NP_572573.1); <span class="html-italic">Caenorhabditis elegans</span> (NP_495454.1).</p>
Full article ">Figure 2
<p>Audiograms from subjects with <span class="html-italic">KARS1</span> variants causing sensorineural hearing loss. Only results for air conduction are shown. Red line and circles, right ear. Blue line and crosses, left ear. (<b>a</b>) Affected subjects from family HRC19. Audiograms at different ages, to illustrate mild progression of the hearing loss. (<b>b</b>) Affected subjects from family HRC20.</p>
Full article ">Figure 3
<p>Pathogenic variants that were found in <span class="html-italic">HARS2</span> in this study. (<b>a</b>) Pedigrees showing the segregation of variants. Arrow indicates propositus. (<b>b</b>) Electropherograms from subject HRC21 II:2 (left panel) and from subject 491NS II:1 (right panel). (<b>c</b>) Alignment of HARS2 protein orthologous sequences from human and seven other animal species. Asterisks indicate identical residues across all sequences; colons, conserved positions (residues of strongly similar properties); periods, semi-conserved positions (residues of weakly similar properties). Sequence accession numbers: <span class="html-italic">H. sapiens</span> (NP_036340.1); <span class="html-italic">M. musculus</span> (NP_542367.1); <span class="html-italic">B. taurus</span> (XP_010805666.1); <span class="html-italic">G. gallus</span> (XP_040538754.1); <span class="html-italic">X. tropicalis</span> (XP_031754386.1); <span class="html-italic">D. rerio</span> (NP_001289185); <span class="html-italic">D. melanogaster</span> (NP_728180.1); <span class="html-italic">C. elegans</span> (NP_001023374.1).</p>
Full article ">Figure 4
<p>Audiograms from subjects with <span class="html-italic">HARS2</span> variants causing sensorineural hearing loss. Only results for air conduction are shown. Red line and circles, right ear. Blue line and crosses, left ear. (<b>a</b>) Affected subjects from family HRC21. Audiograms at different ages, to illustrate the evolution of the hearing loss. (<b>b</b>) Affected subject from family 491NS.</p>
Full article ">Figure 5
<p>Pathogenic variants that were found in <span class="html-italic">LARS2</span> in this study. (<b>a</b>) Pedigree showing the segregation of variants. Arrow indicates propositus. (<b>b</b>) Electropherograms from subject HRC22 II:1. (<b>c</b>) Alignment of LARS2 protein orthologous sequences from human and seven other animal species. Asterisks indicate identical residues across all sequences; colons, conserved positions (residues of strongly similar properties); periods, semi-conserved positions (residues of weakly similar properties). Sequence accession numbers: <span class="html-italic">H. sapiens</span> (NP_056155.1); <span class="html-italic">M. musculus</span> (NP_001335096.1); <span class="html-italic">B. taurus</span> (XP_059735480.1); <span class="html-italic">G. gallus</span> (XP_040521344.1); <span class="html-italic">X. tropicalis</span> (XP_031759687.1); <span class="html-italic">D. rerio</span> (NP_001099171.1); <span class="html-italic">D. melanogaster</span> (NP_647932.1); <span class="html-italic">C. elegans</span> (NP_001021875.1).</p>
Full article ">Figure 6
<p>Audiograms from affected subjects from family HRC22 who carry <span class="html-italic">LARS2</span> variants causing sensorineural hearing loss. Only results for air conduction are shown. Red line and circles, right ear. Blue line and crosses, left ear. Audiograms at different ages, to illustrate the evolution of the hearing loss.</p>
Full article ">
15 pages, 1980 KiB  
Article
Deafness DFNB128 Associated with a Recessive Variant of Human MAP3K1 Recapitulates Hearing Loss of Map3k1-Deficient Mice
by Rabia Faridi, Rizwan Yousaf, Sayaka Inagaki, Rafal Olszewski, Shoujun Gu, Robert J. Morell, Elizabeth Wilson, Ying Xia, Tanveer Ahmed Qaiser, Muhammad Rashid, Cristina Fenollar-Ferrer, Michael Hoa, Sheikh Riazuddin and Thomas B. Friedman
Genes 2024, 15(7), 845; https://doi.org/10.3390/genes15070845 - 27 Jun 2024
Viewed by 765
Abstract
Deafness in vertebrates is associated with variants of hundreds of genes. Yet, many mutant genes causing rare forms of deafness remain to be discovered. A consanguineous Pakistani family segregating nonsyndromic deafness in two sibships were studied using microarrays and exome sequencing. A 1.2 [...] Read more.
Deafness in vertebrates is associated with variants of hundreds of genes. Yet, many mutant genes causing rare forms of deafness remain to be discovered. A consanguineous Pakistani family segregating nonsyndromic deafness in two sibships were studied using microarrays and exome sequencing. A 1.2 Mb locus (DFNB128) on chromosome 5q11.2 encompassing six genes was identified. In one of the two sibships of this family, a novel homozygous recessive variant NM_005921.2:c.4460G>A p.(Arg1487His) in the kinase domain of MAP3K1 co-segregated with nonsyndromic deafness. There are two previously reported Map3k1-kinase-deficient mouse models that are associated with recessively inherited syndromic deafness. MAP3K1 phosphorylates serine and threonine and functions in a signaling pathway where pathogenic variants of HGF, MET, and GAB1 were previously reported to be associated with human deafness DFNB39, DFNB97, and DFNB26, respectively. Our single-cell transcriptome data of mouse cochlea mRNA show expression of Map3k1 and its signaling partners in several inner ear cell types suggesting a requirement of wild-type MAP3K1 for normal hearing. In contrast to dominant variants of MAP3K1 associated with Disorders of Sex Development 46,XY sex-reversal, our computational modeling of the recessive substitution p.(Arg1487His) predicts a subtle structural alteration in MAP3K1, consistent with the limited phenotype of nonsyndromic deafness. Full article
(This article belongs to the Special Issue Molecular Basis of Rare Genetic Diseases)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Protein structure of human <span class="html-italic">MAP3K1</span> modified from [<a href="#B17-genes-15-00845" class="html-bibr">17</a>]. GEF, putative Guanine Exchange Factor domain; SWIM, SWI2/SNF2 and MuDR domain; RING/PHD, RING-CH-C4HC3_ZSWM2 with Plant Homeodomain motif; TOG, tumor overexpressed gene; ARM, armadillo repeats; Kinase, kinase domain. Reported pathogenic variants of <span class="html-italic">MAP3K1</span> and their associated phenotypes are listed in <a href="#app1-genes-15-00845" class="html-app">Table S3</a>. The light green regions are not recognized as amino acid sequences belonging to reported domains.</p>
Full article ">Figure 2
<p>Hearing loss associated with biallelic variants of human <span class="html-italic">MAP3K1</span>. (<b>A</b>) Pedigree of a six-generation family with genotypic data from fourteen individuals. Circles and squares illustrate female and male individuals, respectively. Filled circles and squares refer to deaf individuals, open circles and squares represent individuals with normal hearing thresholds, and * indicates individuals with exome sequencing data. For <span class="html-italic">MAP3K1</span>, G is the wild-type allele, while A is the mutant allele. For CABP2, G is the wild-type allele, and T is the mutant allele. (<b>B</b>) Audiograms of individual VI:2, VI:3, VI:4, and VI:8. Individual VI:2 shows hearing thresholds within normal range. VI:3 has bilateral profound SNHL, and VI:4 has bilateral, severe-to-profound SNHL, whereas individual VI:8 has a moderate-to-severe degree of HL. Arrows indicate no response to the auditory stimulus at the indicated levels.</p>
Full article ">Figure 3
<p>Refinement of the <span class="html-italic">DFNB128</span> region of homozygosity (ROH), chromatograms, and sequence alignments. (<b>A</b>) The thick vertical bar represents human chromosome 5q. The ROH for deafness segregating in Family PKDF1419 is indicated by a thin vertical bar. The gray shaded region is the DFNB128 ROH with the position of the <span class="html-italic">MAP3K1</span> gene in bold. (<b>B</b>) Representative chromatograms of the gDNA sequences obtained from individuals V:4 and VI:3 who are heterozygous (G/A) and homozygous (A/A) for the p.(Arg1487His) variant, respectively. The affected codon is shaded in gray. Each color line refers to a base. Green refers to adenine, Red to thymine, Blue to cytosine and Black to guanine. (<b>C</b>) Conservation of human Arg1487 residue in <span class="html-italic">MAP3K1</span> orthologs shows that Arg1487 residue is conserved among a variety of species. (RefSeq IDs: human; NP_005912.1; mouse NP_036075.1; rat; NP_446339.2; rhesus, XP_002804414.2; cattle, NP_001192835.1; frog, XP_012822348.1; zebrafish; XP_005155564.1.). * indicates identical in all sequences in the alignment; : indicates conserved substitutions have been observed.</p>
Full article ">Figure 4
<p>Structural model of the kinase domain of human <span class="html-italic">MAP3K1</span> wild type (<b>A</b>) and close-up views of residue at position 1487 for the wild type (Arg at the position of 1487) and p.(Arg1487His) variant (His at residue 1487). (<b>B</b>) The activating loop of the domain is colored in yellow, while the residues taking part in the interacting network in which Arg1487 participates are colored in red. Blue color indicates nitrogen atom. The magnesium ion is shown as a green sphere in (<b>A</b>). Distances between heteroatoms (N, O, C) are shown in Angstroms and represented as dashed lines in (<b>B</b>).</p>
Full article ">Figure 5
<p>Single-nucleus RNA-Seq data on the stria vascularis extracted from our previous work [<a href="#B41-genes-15-00845" class="html-bibr">41</a>] and single-cell RNA-Seq data on the spiral ganglion neurons (SGNs) [<a href="#B42-genes-15-00845" class="html-bibr">42</a>] and organ of Corti [<a href="#B43-genes-15-00845" class="html-bibr">43</a>] demonstrate the expression of <span class="html-italic">Map3k1</span> in regions of the cochlea. (<b>A</b>) In the stria vascularis (1st violin plot on left), <span class="html-italic">Map3k1</span> is expressed in marginal cells of the stria vascularis, spindle cells, root cells, and Reissner’s membrane from P30 CBA/J mice. (<b>B</b>) Amongst SGNs (2nd violin plot in middle), <span class="html-italic">Map3k1</span> expression is observed across all SGN subtypes from P25 to 27 of CBA/CaJ mice. (<b>C</b>) Minimal expression of <span class="html-italic">Map3k1</span> is detected in inner hair cells (IHCs), outer hair cells (OHCs), pillar, or Deiters’ cells in the organ of Corti from P7 CD1 mice (3rd violin plot on right).</p>
Full article ">
10 pages, 837 KiB  
Article
Towards Comprehensive Newborn Hearing and Genetic Screening in Russia: Perspectives of Implementation
by Svetlana Chibisova, Tatiana Markova, Evgenia Tsigankova and George Tavartkiladze
J. Otorhinolaryngol. Hear. Balance Med. 2024, 5(1), 6; https://doi.org/10.3390/ohbm5010006 - 15 May 2024
Viewed by 722
Abstract
The universal newborn hearing screening (NHS) program was implemented in Russia in 2008 to replace the high-risk newborn hearing screening. More than 95% coverage and significant improvement in early detection and intervention is achieved. Meanwhile, it was shown that current OAE-based hearing screening [...] Read more.
The universal newborn hearing screening (NHS) program was implemented in Russia in 2008 to replace the high-risk newborn hearing screening. More than 95% coverage and significant improvement in early detection and intervention is achieved. Meanwhile, it was shown that current OAE-based hearing screening missed 13% of newborns with genetically ascertained hereditary sensorineural hearing loss (SNHL). The aim of the study is to assess the results of genetic investigation and NHS in a large cohort of Russian children with bilateral SNHL and to study the feasibility of implementation of combined hearing and genetic screening in Russia. Genetic, audiological and NHS data of 1292 pediatric patients with bilateral SNHL born in 2008–2021 were analyzed. GJB2 sequencing was performed for all subjects, 644 patients had pathological GJB2 genotype, 406 of them were homozygous for c.35delG variant. The group of 155 GJB2-negative patients were searched for other SNHL genes, The pathological genotypes were identified at 87 patients. The most frequent genes were STRC (21.8%), USH2A (16.1%), OTOF (8%) and SLC26A4 (6.9%). Children with confirmed genetic etiology passed NHS in 21% of cases. The perspectives of implementation of national comprehensive newborn hearing and genetic screening including whole exome sequencing technologies are discussed. Full article
Show Figures

Figure 1

Figure 1
<p>The results of genetic investigation in the study sample. SNHL—sensorineural hearing loss.</p>
Full article ">Figure 2
<p>Distribution of patients by hearing loss severity and genotype. (<b>a</b>) comparison between <span class="html-italic">GJB2</span>-positive group, other genes-positive group and gene-negative group; (<b>b</b>) comparison between different <span class="html-italic">GJB2</span> genotypes groups—c.35delG homozygotes (c.35delGx2), other T/T genotypes, T/NT and NT/NT genotypes. T—truncating variant, NT—non-truncating variant.</p>
Full article ">
13 pages, 1330 KiB  
Article
Detailed Clinical Features of PTPRQ-Associated Hearing Loss Identified in a Large Japanese Hearing Loss Cohort
by Naoko Sakuma, Shin-ya Nishio, Shin-ichi Goto, Yohei Honkura, Kiyoshi Oda, Hidehiko Takeda, Marina Kobayashi, Kozo Kumakawa, Satoshi Iwasaki, Masahiro Takahashi, Taku Ito, Yasuhiro Arai, Yasuhiro Isono, Natsuko Obara, Takeshi Matsunobu, Kimihiro Okubo and Shin-ichi Usami
Genes 2024, 15(4), 489; https://doi.org/10.3390/genes15040489 - 12 Apr 2024
Viewed by 1020
Abstract
The PTPRQ gene has been identified as one of the genes responsible for non-syndromic sensorineural hearing loss (SNHL), and assigned as DFNA73 and DFNB84. To date, about 30 causative PTPRQ variants have been reported to cause SNHL. However, the detailed clinical features of [...] Read more.
The PTPRQ gene has been identified as one of the genes responsible for non-syndromic sensorineural hearing loss (SNHL), and assigned as DFNA73 and DFNB84. To date, about 30 causative PTPRQ variants have been reported to cause SNHL. However, the detailed clinical features of PTPRQ-associated hearing loss (HL) remain unclear. In this study, 15,684 patients with SNHL were enrolled and genetic analysis was performed using massively parallel DNA sequencing (MPS) for 63 target deafness genes. We identified 17 possibly disease-causing PTPRQ variants in 13 Japanese patients, with 15 of the 17 variants regarded as novel. The majority of variants identified in this study were loss of function. Patients with PTPRQ-associated HL mostly showed congenital or childhood onset. Their hearing levels at high frequency deteriorated earlier than that at low frequency. The severity of HL progressed from moderate to severe or profound HL. Five patients with profound or severe HL received cochlear implantation, and the postoperative sound field threshold levels and discrimination scores were favorable. These findings will contribute to a greater understanding of the clinical features of PTPRQ-associated HL and may be relevant in clinical practice. Full article
(This article belongs to the Special Issue Next Generation Sequencing in Human Disease)
Show Figures

Figure 1

Figure 1
<p>Pedigree and audiograms for each family for the <span class="html-italic">PTPRQ</span>-associated hearing loss patients in this study. Arrows show the probands in each family. The variants identified in this study are indicated on the figure.</p>
Full article ">Figure 2
<p>Serial audiograms of two patients with <span class="html-italic">PTPRQ</span> variants (family numbers 1 and 6).</p>
Full article ">Figure 3
<p>Audiograms and the hearing thresholds with hearing aid or cochlear implant. HA; hearing aid, CI; cochlear implant.</p>
Full article ">
18 pages, 3913 KiB  
Article
Functional Studies of Deafness-Associated Pendrin and Prestin Variants
by Satoe Takahashi, Takashi Kojima, Koichiro Wasano and Kazuaki Homma
Int. J. Mol. Sci. 2024, 25(5), 2759; https://doi.org/10.3390/ijms25052759 - 27 Feb 2024
Cited by 1 | Viewed by 969
Abstract
Pendrin and prestin are evolutionary-conserved membrane proteins that are essential for normal hearing. Dysfunction of these proteins results in hearing loss in humans, and numerous deafness-associated pendrin and prestin variants have been identified in patients. However, the pathogenic impacts of many of these [...] Read more.
Pendrin and prestin are evolutionary-conserved membrane proteins that are essential for normal hearing. Dysfunction of these proteins results in hearing loss in humans, and numerous deafness-associated pendrin and prestin variants have been identified in patients. However, the pathogenic impacts of many of these variants are ambiguous. Here, we report results from our ongoing efforts to experimentally characterize pendrin and prestin variants using in vitro functional assays. With previously established fluorometric anion transport assays, we determined that many of the pendrin variants identified on transmembrane (TM) 10, which contains the essential anion binding site, and on the neighboring TM9 within the core domain resulted in impaired anion transport activity. We also determined the range of functional impairment in three deafness-associated prestin variants by measuring nonlinear capacitance (NLC), a proxy for motor function. Using the results from our functional analyses, we also evaluated the performance of AlphaMissense (AM), a computational tool for predicting the pathogenicity of missense variants. AM prediction scores correlated well with our experimental results; however, some variants were misclassified, underscoring the necessity of experimentally assessing the effects of variants. Together, our experimental efforts provide invaluable information regarding the pathogenicity of deafness-associated pendrin and prestin variants. Full article
Show Figures

Figure 1

Figure 1
<p>TM9-10 of pendrin. (<b>A</b>) The homodimeric structure of mouse pendrin (PDB: 7WK1). Protomers are shown in green and gray. Transmembrane and cytosolic domains are indicated in the lateral view (left). TM9 and TM10 are highlighted in cyan and blue, respectively, with connecting residues highlighted in yellow. The bound chlorides are indicated by red spheres. In the extracellular view (right), the core domain of one of the protomers is shown in bright orange. (<b>B</b>) TM9 and TM10 region of the structure (residues 376–420), extracted from the right protomer in (<b>A</b>). TM9 (381–398) is shown in cyan, linker region (399–405) is in yellow, and TM10 (406–416) is in blue. Bound chloride is shown as a red sphere. (<b>C</b>) Partial amino acid sequences of human and mouse pendrin (A4) and prestin (A5) showing the TM9 and TM10 region. Numbers in parentheses indicate the residue numbers at the N- and C-terminal ends of the partial amino acid sequences. TM9 and TM10 are highlighted in cyan and blue, respectively, with connecting residues highlighted in yellow as in (<b>A</b>). The residues with asterisks (*) on top and gray shades indicate the locations of missense changes evaluated in <a href="#ijms-25-02759-f002" class="html-fig">Figure 2</a>.</p>
Full article ">Figure 2
<p>HCO<sub>3</sub><sup>−</sup>/Cl<sup>−</sup> and I<sup>−</sup>/Cl<sup>−</sup> antiport assays on pendrin variants. HCO<sub>3</sub><sup>−</sup>/Cl<sup>−</sup> (<b>A</b>) and I<sup>−</sup>/Cl<sup>−</sup> (<b>B</b>) antiport rates were plotted against doxycycline (Dox) concentration (0.1–10 µg/mL) for each mTq2-tagged pendrin variant alongside WT as indicated. Horizontal dotted lines indicate transport rates of uninduced cells. Error bars indicate SD. Solid lines indicate linear regressions (log<sub>10</sub> [Dox] vs. transport rates). Sample size information and statistics are provided in <a href="#ijms-25-02759-t001" class="html-table">Table 1</a>.</p>
Full article ">Figure 3
<p>Ala<sup>100</sup>, Pro<sup>119</sup>, and Ser<sup>441</sup> sites in prestin. (<b>A</b>) The homodimeric structure of human prestin (PDB: 7LGU). Protomers are shown in green and gray. In right panel, the core domain of one of the protomers is shown in bright orange. The Ala<sup>100</sup>, Pro<sup>119</sup>, and Ser<sup>441</sup> sites and bound chlorides are indicated by cyan, blue, purple, and red spheres, respectively. (<b>B</b>) Partial amino acid sequences of human and mouse pendrin (A4) and prestin (A5). Numbers in parentheses indicate the residue numbers at the N- and C-terminal ends. The residues with asterisks indicate Ala<sup>100</sup>, Pro<sup>119</sup>, and Ser<sup>441</sup> in human prestin (hA5) and equivalents in others.</p>
Full article ">Figure 4
<p>NLC measurements. (<b>A</b>) Examples of NLC recorded in HEK293T cells expressing WT, p.A100T, p.P119S, p.S441L, or p.S441A prestin. Different colors indicate individual recordings. (<b>B</b>) Summaries of the NLC parameters (α, V<sub>pk</sub>, and CD). Error bars indicate SD. ns, <span class="html-italic">p</span> ≥ 0.05; ** 0.001 &lt; <span class="html-italic">p</span> ≤ 0.01; *** 0.0001 &lt; <span class="html-italic">p</span> ≤ 0.001; **** <span class="html-italic">p</span> ≤ 0.0001.</p>
Full article ">Figure 5
<p>The effects of missense changes at Ala<sup>104</sup> and Ala<sup>451</sup> on the anion transport function of pendrin. HCO<sub>3</sub><sup>−</sup>/Cl<sup>−</sup> antiport assay conducted for p.A104V (<b>left</b>), p.A104T (<b>left</b>), p.A451G (<b>right</b>), p.A451S (<b>right</b>), and p.A451L (<b>right</b>) pendrin alongside WT control. Error bars indicate SD. Horizontal dotted lines indicate transport rates of uninduced cells. Solid lines indicate linear regressions (log<sub>10</sub> [Dox] vs. transport rates). Sample size information and statistics are provided in <a href="#ijms-25-02759-t001" class="html-table">Table 1</a>.</p>
Full article ">Figure 6
<p>Correlation with AlphaMissense pathogenicity scores. HCO<sub>3</sub><sup>−</sup>/Cl<sup>−</sup> transport rates of pendrin WT and variants from this study and the previous study [<a href="#B10-ijms-25-02759" class="html-bibr">10</a>] were normalized to the WT value and plotted against AlphaMissense (AM) pathogenicity scores [<a href="#B16-ijms-25-02759" class="html-bibr">16</a>]. Light blue shades indicate HCO<sub>3</sub><sup>−</sup>/Cl<sup>−</sup> rate of WT with errors (propagated errors). Gray shade between AM scores 0.34–0.56 marks the “ambiguous” category, with scores lower being “benign” and higher being “pathogenic” as indicated. Red line indicates the linear fit between the HCO<sub>3</sub><sup>−</sup>/Cl<sup>−</sup> antiport activity and the AM scores. Inset: Region indicated by the broken lines in left are enlarged to visualize variants with little or no HCO<sub>3</sub><sup>−</sup>/Cl<sup>−</sup> transport activity.</p>
Full article ">
15 pages, 454 KiB  
Review
Autosomal Recessive Non-Syndromic Deafness: Is AAV Gene Therapy a Real Chance?
by Davide Brotto, Marco Greggio, Cosimo De Filippis and Patrizia Trevisi
Audiol. Res. 2024, 14(2), 239-253; https://doi.org/10.3390/audiolres14020022 - 22 Feb 2024
Viewed by 1799
Abstract
The etiology of sensorineural hearing loss is heavily influenced by genetic mutations, with approximately 80% of cases attributed to genetic causes and only 20% to environmental factors. Over 100 non-syndromic deafness genes have been identified in humans thus far. In non-syndromic sensorineural hearing [...] Read more.
The etiology of sensorineural hearing loss is heavily influenced by genetic mutations, with approximately 80% of cases attributed to genetic causes and only 20% to environmental factors. Over 100 non-syndromic deafness genes have been identified in humans thus far. In non-syndromic sensorineural hearing impairment, around 75–85% of cases follow an autosomal recessive inheritance pattern. In recent years, groundbreaking advancements in molecular gene therapy for inner-ear disorders have shown promising results. Experimental studies have demonstrated improvements in hearing following a single local injection of adeno-associated virus-derived vectors carrying an additional normal gene or using ribozymes to modify the genome. These pioneering approaches have opened new possibilities for potential therapeutic interventions. Following the PRISMA criteria, we summarized the AAV gene therapy experiments showing hearing improvement in the preclinical phases of development in different animal models of DFNB deafness and the AAV gene therapy programs currently in clinical phases targeting autosomal recessive non syndromic hearing loss. A total of 17 preclinical studies and 3 clinical studies were found and listed. Despite the hurdles, there have been significant breakthroughs in the path of HL gene therapy, holding great potential for providing patients with novel and effective treatment. Full article
(This article belongs to the Special Issue Genetics of Hearing Loss—Volume II)
Show Figures

Figure 1

Figure 1
<p>Flow diagram. * databases and registers (they are reported after the colon). ** all records excluded from databases and/or registers after the screening processes.</p>
Full article ">
10 pages, 1296 KiB  
Article
Novel Pathogenic Variants in the Gene Encoding Stereocilin (STRC) Causing Non-Syndromic Moderate Hearing Loss in Spanish and Argentinean Subjects
by María Domínguez-Ruiz, Laura Ruiz-Palmero, Paula I. Buonfiglio, Irene García-Vaquero, Elena Gómez-Rosas, Marina Goñi, Manuela Villamar, Matías Morín, Miguel A. Moreno-Pelayo, Ana B. Elgoyhen, Francisco J. del Castillo, Viviana Dalamón and Ignacio del Castillo
Biomedicines 2023, 11(11), 2943; https://doi.org/10.3390/biomedicines11112943 - 31 Oct 2023
Viewed by 1536
Abstract
Non-syndromic hearing impairment (NSHI) is a very heterogeneous genetic condition, involving over 130 genes. Mutations in GJB2, encoding connexin-26, are a major cause of NSHI (the DFNB1 type), but few other genes have significant epidemiological contributions. Mutations in the STRC gene result [...] Read more.
Non-syndromic hearing impairment (NSHI) is a very heterogeneous genetic condition, involving over 130 genes. Mutations in GJB2, encoding connexin-26, are a major cause of NSHI (the DFNB1 type), but few other genes have significant epidemiological contributions. Mutations in the STRC gene result in the DFNB16 type of autosomal recessive NSHI, a common cause of moderate hearing loss. STRC is located in a tandem duplicated region that includes the STRCP1 pseudogene, and so it is prone to rearrangements causing structural variations. Firstly, we screened a cohort of 122 Spanish familial cases of non-DFNB1 NSHI with at least two affected siblings and unaffected parents, and with different degrees of hearing loss (mild to profound). Secondly, we screened a cohort of 64 Spanish sporadic non-DFNB1 cases, and a cohort of 35 Argentinean non-DFNB1 cases, all of them with moderate hearing loss. Amplification of marker D15S784, massively parallel DNA sequencing, multiplex ligation-dependent probe amplification and long-range gene-specific PCR followed by Sanger sequencing were used to search and confirm single-nucleotide variants (SNVs) and deletions involving STRC. Causative variants were found in 13 Spanish familial cases (10.7%), 5 Spanish simplex cases (7.8%) and 2 Argentinean cases (5.7%). In all, 34 deleted alleles and 6 SNVs, 5 of which are novel. All affected subjects had moderate hearing impairment. Our results further support this strong genotype–phenotype correlation and highlight the significant contribution of STRC mutations to moderate NSHI in the Spanish population. Full article
(This article belongs to the Special Issue Genetic Research on Hearing Loss 2.0)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Tandemly duplicated region that contains the <span class="html-italic">STRC</span> gene on 15q15.3. Above: the number of MLPA probes is indicated within the red arrowhead above each gene. Below: exon–intron structure of the <span class="html-italic">STRC</span> gene, red arrowheads indicating the position of the MLPA probes on different exons.</p>
Full article ">Figure 2
<p>Accumulated audiograms of 27 subjects with biallelic <span class="html-italic">STRC</span> pathogenic variants. Only results for air conduction are shown. As the hearing loss was bilateral and symmetrical, mean values of the two ears are shown for each patient.</p>
Full article ">
34 pages, 2562 KiB  
Review
Functional Consequences of Pathogenic Variants of the GJB2 Gene (Cx26) Localized in Different Cx26 Domains
by Olga L. Posukh, Ekaterina A. Maslova, Valeriia Yu. Danilchenko, Marina V. Zytsar and Konstantin E. Orishchenko
Biomolecules 2023, 13(10), 1521; https://doi.org/10.3390/biom13101521 - 13 Oct 2023
Cited by 2 | Viewed by 1929
Abstract
One of the most common forms of genetic deafness has been predominantly associated with pathogenic variants in the GJB2 gene, encoding transmembrane protein connexin 26 (Cx26). The Cx26 molecule consists of an N-terminal domain (NT), four transmembrane domains (TM1–TM4), two extracellular loops (EL1 [...] Read more.
One of the most common forms of genetic deafness has been predominantly associated with pathogenic variants in the GJB2 gene, encoding transmembrane protein connexin 26 (Cx26). The Cx26 molecule consists of an N-terminal domain (NT), four transmembrane domains (TM1–TM4), two extracellular loops (EL1 and EL2), a cytoplasmic loop, and a C-terminus (CT). Pathogenic variants in the GJB2 gene, resulting in amino acid substitutions scattered across the Cx26 domains, lead to a variety of clinical outcomes, including the most common non-syndromic autosomal recessive deafness (DFNB1A), autosomal dominant deafness (DFNA3A), as well as syndromic forms combining hearing loss and skin disorders. However, for rare and poorly documented variants, information on the mode of inheritance is often lacking. Numerous in vitro studies have been conducted to elucidate the functional consequences of pathogenic GJB2 variants leading to amino acid substitutions in different domains of Cx26 protein. In this work, we summarized all available data on a mode of inheritance of pathogenic GJB2 variants leading to amino acid substitutions and reviewed published information on their functional effects, with an emphasis on their localization in certain Cx26 domains. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

Figure 1
<p>The proportions of different <span class="html-italic">GJB2</span> variants and the distribution of PLP variants according to their molecular consequences.</p>
Full article ">Figure 2
<p>The schematic topology of the Cx26 protein protomer. Amino acids affected by known PLP missense variants leading to non-synonymous substitutions are colored (dark blue—analyzed by functional studies, light blue—without functional studies). NT—N-terminus, including intramembrane (2–13 a.a.) and cytoplasmic (14–20 a.a.) parts; TM1 (21–40 a.a.), TM2 (74–94 a.a.), TM3 (136–156 a.a.), and TM4 (190–210 a.a.)—transmembrane domains; CL (95–135 a.a.)—cytoplasmic loop; EL1 (41–73 a.a.) and EL2 (157–189 a.a.)—extracellular loops; CT—C-terminus (cytoplasmic, 211–226 a.a.). Amino acids (a.a.) positions for each domain are indicated in brackets.</p>
Full article ">Figure 3
<p>The rate of amino acid substitutions across Cx26 domains. Four Cx26 domains (N-terminus, TM1, EL1, and TM2) with a rate above the mean (median value = 1.09) are colored.</p>
Full article ">Figure 4
<p>Distribution of the PLP missense variants across Cx26 domains according to their mode of inheritance. AR—non-syndromic autosomal recessive, AD—non-syndromic dominant, S (AD)—syndromic dominant, ?—uncertain type of inheritance.</p>
Full article ">
14 pages, 1750 KiB  
Article
A Novel 13q12 Microdeletion Associated with Familial Syndromic Corneal Opacification
by Jasmine Y. Serpen, William Presley, Adelyn Beil, Stephen T. Armenti, Kayla Johnson, Shahzad I. Mian, Jeffrey W. Innis and Lev Prasov
Genes 2023, 14(5), 1034; https://doi.org/10.3390/genes14051034 - 1 May 2023
Viewed by 2003
Abstract
Progressive corneal opacification can result from multiple etiologies, including corneal dystrophies or systemic and genetic diseases. We describe a novel syndrome featuring progressive epithelial and anterior stromal opacification in a brother and sister and their mildly affected father, with all three family members [...] Read more.
Progressive corneal opacification can result from multiple etiologies, including corneal dystrophies or systemic and genetic diseases. We describe a novel syndrome featuring progressive epithelial and anterior stromal opacification in a brother and sister and their mildly affected father, with all three family members having sensorineural hearing loss and two also with tracheomalacia/laryngomalacia. All carried a 1.2 Mb deletion at chromosome 13q12.11, with no other noteworthy co-segregating variants identified on clinical exome or chromosomal microarray. RNAseq analysis from an affected corneal epithelial sample from the proband’s brother revealed downregulation of XPO4, IFT88, ZDHHC20, LATS2, SAP18, and EEF1AKMT1 within the microdeletion interval, with no notable effect on the expression of nearby genes. Pathway analysis showed upregulation of collagen metabolism and extracellular matrix (ECM) formation/maintenance, with no significantly down-regulated pathways. Analysis of overlapping deletions/variants demonstrated that deleterious variants in XPO4 were found in patients with laryngomalacia and sensorineural hearing loss, with the latter phenotype also being a feature of variants in the partially overlapping DFNB1 locus, yet none of these had reported corneal phenotypes. Together, these data define a novel microdeletion-associated syndromic progressive corneal opacification and suggest that a combination of genes within the microdeletion may contribute to ECM dysregulation leading to pathogenesis. Full article
(This article belongs to the Special Issue Genetics of Eye Development and Disease)
Show Figures

Figure 1

Figure 1
<p>Pedigree depicting 13q12.11 deletion in affected family members and their corresponding phenotypes.</p>
Full article ">Figure 2
<p>Clinical features of syndromic corneal opacification family. II-1: Slit lamp (<b>A</b>) and external (<b>B</b>) photos at presentation (age 5 years) and external photos at follow-up after superficial keratectomy (age 13 years) (<b>C</b>) II-2: Slit lamp (<b>D</b>) and external (<b>E</b>) photos at presentation (age 8 years) and external photos at follow-up after superficial keratectomy (age 9 years) (<b>F</b>). The current age of the proband (II-1) is 15, and the proband’s brother (II-2) is 12.</p>
Full article ">Figure 3
<p>Chromosome 13q12 microdeletion interval showing genes and overlapping microdeletions. UCSC genome browser region depicting region and involved genes of family’s microdeletion as well as overlapping microdeletions identified by literature review and the DECIPHER database. Browser coordinates use the hg19 genome build. Phenotypes present for each microdeletion are highlighted [<a href="#B13-genes-14-01034" class="html-bibr">13</a>,<a href="#B14-genes-14-01034" class="html-bibr">14</a>,<a href="#B15-genes-14-01034" class="html-bibr">15</a>].</p>
Full article ">Figure 4
<p>Region-focused DEGs analysis. A Heatmap showing the normalized Z-scores and log<sub>2</sub> fold changes (log<sub>2</sub>fc) for the genes in/around the microdeletion, as well as their average corneal expression in normalized counts (AvgExp) across the samples.</p>
Full article ">Figure 5
<p>RNAseq analysis from corneal epithelium. (<b>A</b>) A volcano plot showing the DESeq2 comparison of global gene expression between a corneal epithelial sample from our case and controls (healthy, mildly myopic patients). Green is log<sub>2</sub> fold change (FC) &gt;|2| and <span class="html-italic">p</span>-value &gt; 10<sup>−6</sup>; blue is log<sub>2</sub> fold change &lt;|2| and <span class="html-italic">p</span>-value &lt; 10<sup>−6</sup>; red is log<sub>2</sub> fold change &gt;|2| and <span class="html-italic">p</span>-value &lt; 10<sup>−6</sup>. (<b>B</b>) A scatter plot showing the fold enrichment of the top ten most significantly upregulated Gene Ontology (GO) processes from our case sample as determined by the lowest false discovery rate (FDR). TM, tube morphogenesis; BVM, blood vessel morphogenesis; CMP, collagen metabolic processes; TD, tube development; VD, vasculature development; BVD, blood vessel development; EESO, external encapsulating structure organization; EMO, extracellular matrix organization; CFO, collagen fibril organization.</p>
Full article ">Figure 6
<p>Model for dysregulated gene expression leading to corneal opacification phenotype. EGF and TGFβ signaling work synergistically to promote ECM protein formation/deposition in the cornea, a process negatively regulated by four of the genes lost in the microdeletion: <span class="html-italic">XPO4</span>, <span class="html-italic">LATS2</span>, <span class="html-italic">ZDHHC20</span>, and <span class="html-italic">IFT88</span>. <span class="html-italic">LATS2</span> is thought to inhibit TGFβ signaling through its role in the YAZ/TAP pathway, whereas <span class="html-italic">XPO4</span> inhibits the function/localization of key TGFβ effector SMAD3. Additionally, <span class="html-italic">ZDHHC20</span> inhibits EGF signaling via palmitoylation of EGF receptors (EGFR). <span class="html-italic">IFT88</span> works downstream of both pathways in regulating the ECM through its roles in collagen production/fiber organization, Hedgehog signaling, ciliary calcium response, and protease endocytosis. The loss of one or more of these genes could thus result in the inappropriate accumulation of ECM proteins in the anterior cornea, triggering a fibrotic response and resulting in a progressive haze.</p>
Full article ">
14 pages, 2177 KiB  
Article
The GJB2 (Cx26) Gene Variants in Patients with Hearing Impairment in the Baikal Lake Region (Russia)
by Vera G. Pshennikova, Fedor M. Teryutin, Alexandra M. Cherdonova, Tuyara V. Borisova, Aisen V. Solovyev, Georgii P. Romanov, Igor V. Morozov, Alexander A. Bondar, Olga L. Posukh, Sardana A. Fedorova and Nikolay A. Barashkov
Genes 2023, 14(5), 1001; https://doi.org/10.3390/genes14051001 - 28 Apr 2023
Cited by 2 | Viewed by 1838
Abstract
The GJB2 (Cx26) gene pathogenic variants are associated with autosomal recessive deafness type 1A (DFNB1A, OMIM #220290). Direct sequencing of the GJB2 gene among 165 hearing-impaired individuals living in the Baikal Lake region of Russia identified 14 allelic variants: pathogenic/likely pathogenic—nine variants, benign—three [...] Read more.
The GJB2 (Cx26) gene pathogenic variants are associated with autosomal recessive deafness type 1A (DFNB1A, OMIM #220290). Direct sequencing of the GJB2 gene among 165 hearing-impaired individuals living in the Baikal Lake region of Russia identified 14 allelic variants: pathogenic/likely pathogenic—nine variants, benign—three variants, unclassified—one variant, and one novel variant. The contribution of the GJB2 gene variants to the etiology of hearing impairment (HI) in the total sample of patients was 15.8% (26 out of 165) and significantly differed in patients of different ethnicity (5.1% in Buryat patients and 28.9% in Russian patients). In patients with DFNB1A (n = 26), HIs were congenital/early onset (92.3%), symmetric (88.5%), sensorineural (100.0%), and variable in severity (moderate—11.6%, severe—26.9% or profound—61.5%). The reconstruction of the SNP haplotypes with three frequent GJB2 pathogenic variants (c.-23+1G>A, c.35delG or c.235delC), in comparison with previously published data, supports a major role of the founder effect in the expansion of the c.-23+1G>A and c.35delG variants around the world. Comparative analysis of the haplotypes with c.235delC revealed one major haplotype G A C T (97.5%) in Eastern Asians (Chinese, Japanese and Korean patients) and two haplotypes, G A C T (71.4%) and G A C C (28.6%), in Northern Asians (Altaians, Buryats and Mongols). The variable structure of the c.235delC-haplotypes in Northern Asians requires more studies to expand our knowledge about the origin of this pathogenic variant. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

Figure 1
<p>Identified allelic variants of the <span class="html-italic">GJB2</span> gene among 165 patients with HI in the Republic of Buryatia. Schematic structure of the <span class="html-italic">GJB2</span> gene is based on NC_000013.11 reference sequence (<a href="https://www.ncbi.nlm.nih.gov/gene/2706" target="_blank">https://www.ncbi.nlm.nih.gov/gene/2706</a> accessed on 25 January 2023); Pathogenic/likely pathogenic variants are shown in red, benign variants are shown in green and variants with uncertain significance (including the novel c.-49G&gt;A variant) are shown in black.</p>
Full article ">Figure 2
<p>Contribution of the <span class="html-italic">GJB2</span> variants to the etiology of HI in studied patients. (<b>A</b>)—Contribution of the <span class="html-italic">GJB2</span> variants to the etiology of HI in Buryat patients. (<b>B</b>)—Contribution of the <span class="html-italic">GJB2</span> variants to the etiology of HI in Russian patients. Proportions of <span class="html-italic">GJB2</span>-negative and <span class="html-italic">GJB2</span>-positive patients are shown in blue and red, accordingly. The allele frequency of the <span class="html-italic">GJB2</span> variants was calculated in unrelated patients.</p>
Full article ">Figure 3
<p>Audiological profiles in patients with the <span class="html-italic">GJB2</span> gene variants. The reference group (shown in blue)—patients with genotype c.[35delG];[35delG] (16 individuals, 32 ears).</p>
Full article ">Figure 4
<p>The PTA<sub>0.5,1.0,2.0,4.0kHz</sub> hearing thresholds in patients with HI caused by the <span class="html-italic">GJB2</span> gene variants. Reference group (shown in blue)—patients with the c.[35delG];[35delG] genotype (16 individuals, 32 ears). The red lines—the median hearing thresholds in the PTA<sub>0.5,1.0,2.0,4.0kHz.</sub> The blue line—the median hearing threshold in the PTA<sub>0.5,1.0,2.0,4.0kHz</sub> of the reference group. Statistically significant differences (<span class="html-italic">p</span> &lt; 0.05) are shown in bold.</p>
Full article ">Figure 5
<p>The genotypes of 12 SNPs, flanking the <span class="html-italic">GJB2</span> gene in 16 patients homozygous for c.-23+1G&gt;A (n = 1), c.35delG (n = 14), and c.235delC (n = 1). Positions of c.-23+1G&gt;A, c.35delG, c.235delC and 12 SNPs were defined according to GRCh38.p13 Genome Assembly (<a href="https://www.ncbi.nlm.nih.gov/assembl" target="_blank">https://www.ncbi.nlm.nih.gov/assembl</a>, accessed on 25 January 2023).</p>
Full article ">Figure 6
<p>The diversity of the haplotypes with three pathogenic <span class="html-italic">GJB2</span> variants. (<b>A</b>) The haplotypes with c.35delG. (<b>B</b>) The haplotypes with c.-23+1G&gt;A. (<b>C</b>) The haplotypes with c.235delC.</p>
Full article ">
Article
Insight into the Natural History of Pathogenic Variant c.919-2A>G in the SLC26A4 Gene Involved in Hearing Loss: The Evidence for Its Common Origin in Southern Siberia (Russia)
by Valeriia Yu. Danilchenko, Marina V. Zytsar, Ekaterina A. Maslova, Konstantin E. Orishchenko and Olga L. Posukh
Genes 2023, 14(4), 928; https://doi.org/10.3390/genes14040928 - 17 Apr 2023
Viewed by 1688
Abstract
Pathogenic variants in the SLC26A4 gene leading to nonsyndromic recessive deafness (DFNB4), or Pendred syndrome, are some of the most common causes of hearing loss worldwide. Earlier, we found a high proportion of SLC26A4-related hearing loss with prevailing pathogenic variant c.919-2A>G (69.3% [...] Read more.
Pathogenic variants in the SLC26A4 gene leading to nonsyndromic recessive deafness (DFNB4), or Pendred syndrome, are some of the most common causes of hearing loss worldwide. Earlier, we found a high proportion of SLC26A4-related hearing loss with prevailing pathogenic variant c.919-2A>G (69.3% among all mutated SLC26A4 alleles that have been identified) in Tuvinian patients belonging to the indigenous Turkic-speaking Siberian people living in the Tyva Republic (Southern Siberia, Russia), which implies a founder effect in the accumulation of c.919-2A>G in Tuvinians. To evaluate a possible common origin of c.919-2A>G, we genotyped polymorphic STR and SNP markers, intragenic and flanking SLC26A4, in patients homozygous for c.919-2A>G and in healthy controls. The common STR and SNP haplotypes carrying c.919-2A>G were revealed, which convincingly indicates the origin of c.919-2A>G from a single ancestor, supporting a crucial role of the founder effect in the c.919-2A>G prevalence in Tuvinians. Comparison analysis with previously published data revealed the identity of the small SNP haplotype (~4.5 kb) in Tuvinian and Han Chinese carriers of c.919-2A>G, which suggests their common origin from founder chromosomes. We assume that c.919-2A>G could have originated in the geographically close territories of China or Tuva and subsequently spread to other regions of Asia. In addition, the time intervals of the c.919-2A>G occurrence in Tuvinians were roughly estimated. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

Figure 1
<p>Schematic structure of the <span class="html-italic">SLC26A4</span> gene and the location of genetic markers (five STRs and nine SNPs) that were used to reconstruct the c.919-2A&gt;G haplotypes. Location of <span class="html-italic">SLC26A4</span> gene is shown by red square. The c.919-2A&gt;G variant is marked by red color. Four of SNP markers from the study by Wu et al. [<a href="#B12-genes-14-00928" class="html-bibr">12</a>] are marked by blue color.</p>
Full article ">Figure 2
<p>The hot map demonstrating the proportion of c.919-2A&gt;G among all mutated <span class="html-italic">SLC26A4</span> alleles revealed in patients with <span class="html-italic">SLC26A4</span>-related hearing loss in the territory of Eurasia. The geographic regions for which no data are available are marked by gray color.</p>
Full article ">
11 pages, 1547 KiB  
Case Report
Temperature-Sensitive Auditory Neuropathy: Report of a Novel Variant of OTOF Gene and Review of Current Literature
by Francesca Forli, Silvia Capobianco, Stefano Berrettini, Luca Bruschini, Silvia Romano, Antonella Fogli, Veronica Bertini and Francesco Lazzerini
Medicina 2023, 59(2), 352; https://doi.org/10.3390/medicina59020352 - 13 Feb 2023
Viewed by 2385
Abstract
Background and objectives: Otoferlin is a multi-C2 domain protein implicated in neurotransmitter-containing vesicle release and replenishment of the cochlear inner hair cell (IHC) synapses. Mutations in the OTOF gene have been associated with two different clinical phenotypes: a prelingual severe-to-profound sensorineural hearing [...] Read more.
Background and objectives: Otoferlin is a multi-C2 domain protein implicated in neurotransmitter-containing vesicle release and replenishment of the cochlear inner hair cell (IHC) synapses. Mutations in the OTOF gene have been associated with two different clinical phenotypes: a prelingual severe-to-profound sensorineural hearing loss (ANSD-DFNB9); and the peculiar temperature-sensitive auditory neuropathy (TS-ANSD), characterized by a baseline mild-to-moderate hearing threshold that worsens to severe-to-profound when the body temperature rises that returns to a baseline a few hours after the temperature has fallen again. The latter clinical phenotype has been described only with a few OTOF variants with an autosomal recessive biallelic pattern of inheritance. Case report: A 7-year-old boy presented a picture compatible with TS-ANSD exacerbated by febrile states or physical exercise with mild-to-moderate hearing loss at low and medium frequencies and a decrease in speech discrimination that worsened with an unfavorable speech-to-noise ratio. Otoacoustic emissions (OAEs) were present whereas auditory brainstem responses (ABRs) evoked by a click or tone-burst were generally absent. No inner ear malformations were described from the CT scan or MRI. Next-generation sequencing (NGS) of the known deafness genes and multi-phasic bioinformatic analyses of the data detected in OTOF a c.2521G>A missense variant and the deletion of 7.4 Kb, which was confirmed by array-comparative genomic hybridization (array-CGH). The proband’s parents, who were asymptomatic, were tested by Sanger sequencing and the father presented the c.2521G>A missense variant. Conclusions: The picture presented by the patient was compatible with OTOF-induced TS-ANSD. OTOF has been generally associated with an autosomal recessive biallelic pattern of inheritance; in this clinical report, two pathogenic variants never previously associated with TS-ANSD were described. Full article
(This article belongs to the Special Issue Metabolic Disorders and Sensorineural Hearing Loss)
Show Figures

Figure 1

Figure 1
<p>Pure−tone audiogram at 7 years of age in afebrile state. Red line: right ear. Blue line: left ear.</p>
Full article ">Figure 2
<p>Pure−tone audiogram at 8 years of age during an episode of fever (T = 38.7 °C). Red line: right ear. Blue line: left ear.</p>
Full article ">Figure 3
<p>Variants in the OTOF gene in the proband. On the left are the electropherograms showing the wild-type sequence and the heterozygous missense variant (c.2521G&gt;A). On the right are the IGV alignments showing a halved coverage from exon 10 to 14, indicating a genomic deletion (GRCh37/hg19; RefSeq: NM_194248.2). Reads that are colored red have larger than expected inferred sizes, and therefore indicate possible deletions. However, red reads are randomly present also in the wild-type control, so taken <span class="html-italic">per se</span> they have no clinical significance. Blue rectangles represent exons, connected by blue lines which represent introns.</p>
Full article ">Figure 4
<p>Mutations in the OTOF gene carried by TS-ANSD patients. The numbers were based on the amino acid sequence of human otoferlin and C2 domains (C2A to F) and the transmembrane domain (TMD) is depicted according to an in silico analysis [<a href="#B2-medicina-59-00352" class="html-bibr">2</a>]. The point mutation reported in the present study is indicated in red. The protein area affected by the deletion is indicated in yellow.</p>
Full article ">
17 pages, 2158 KiB  
Article
Analysis of SLC26A4, FOXI1, and KCNJ10 Gene Variants in Patients with Incomplete Partition of the Cochlea and Enlarged Vestibular Aqueduct (EVA) Anomalies
by Leonid A. Klarov, Vera G. Pshennikova, Georgii P. Romanov, Aleksandra M. Cherdonova, Aisen V. Solovyev, Fedor M. Teryutin, Nikolay V. Luginov, Petr M. Kotlyarov and Nikolay A. Barashkov
Int. J. Mol. Sci. 2022, 23(23), 15372; https://doi.org/10.3390/ijms232315372 - 6 Dec 2022
Cited by 3 | Viewed by 2470
Abstract
Pathogenic variants in the SLC26A4, FOXI1, and KCNJ10 genes are associated with hearing loss (HL) and specific inner ear abnormalities (DFNB4). In the present study, phenotype analyses, including clinical data collection, computed tomography (CT), and audiometric examination, were performed on deaf [...] Read more.
Pathogenic variants in the SLC26A4, FOXI1, and KCNJ10 genes are associated with hearing loss (HL) and specific inner ear abnormalities (DFNB4). In the present study, phenotype analyses, including clinical data collection, computed tomography (CT), and audiometric examination, were performed on deaf individuals from the Sakha Republic of Russia (Eastern Siberia). In cases with cochleovestibular malformations, molecular genetic analysis of the coding regions of the SLC26A4, FOXI1, and KCNJ10 genes associated with DFNB4 was completed. In six of the 165 patients (3.6%), CT scans revealed an incomplete partition of the cochlea (IP-1 and IP-2), in isolation or combined with an enlarged vestibular aqueduct (EVA) anomaly. Sequencing of the SLC26A4, FOXI1, and KCNJ10 genes was performed in these six patients. In the SLC26A4 gene, we identified four variants, namely c.85G>C p.(Glu29Gln), c.757A>G p.(Ile253Val), c.2027T>A p.(Leu676Gln), and c.2089+1G>A (IVS18+1G>A), which are known as pathogenic, as well as c.441G>A p.(Met147Ile), reported previously as a variant with uncertain significance. Using the AlphaFold algorithm, we found in silico evidence of the pathogenicity of this variant. We did not find any causative variants in the FOXI1 and KCNJ10 genes, nor did we find any evidence of digenic inheritance associated with double heterozygosity for these genes with monoallelic SLC26A4 variants. The contribution of biallelic SLC26A4 variants in patients with IP-1, IP-2, IP-2+EVA, and isolated EVA was 66.7% (DFNB4 in three patients, Pendred syndrome in one patient). Seventy-five percent of SLC26A4-biallelic patients had severe or profound HL. The morphology of the inner ear anomalies demonstrated that, among SLC26A4-biallelic patients, all types of incomplete partition of the cochlea are possible, from IP-1 and IP-2, to a normal cochlea. However, the dominant type of anomaly was IP-2+EVA (50.0%). This finding is very important for cochlear implantation, since the IP-2 anomaly does not have an increased risk of “gushers” and recurrent meningitis. Full article
Show Figures

Figure 1

Figure 1
<p>Identification of the c.441G&gt;A p.(Met147Ile) variant in the <span class="html-italic">SLC26A4</span> gene in a homozygous state in a patient with bilateral IP-1 and unilateral EVA. <b>Note.</b> (<b>a</b>) Pedigree of the patient (an arrow indicates the proband). (<b>b</b>) Computed tomography (CT) of the temporal bones in the axial projection. Upper panel: Patient II:3 with an IP-1 anomaly (black filled arrows indicate the cystic cochlea), vestibule dilatation (black open arrows), and unilateral EVA (the triangle indicates the enlarged vestibular aqueduct); lower panel, a patient without anomalies (white arrows). (<b>c</b>) Upper panel: a fragment of the chromatogram of the <span class="html-italic">SLC26A4</span> gene sequence with-the identified c.441G&gt;A p.(Met147Thr) variant in a homozygous state; lower panel: the normal sequence.</p>
Full article ">Figure 2
<p>Identification of the c.85G&gt;C p.(Glu29Gln) and c.2089+1G&gt;A (IVS18+1G&gt;A) variants in the <span class="html-italic">SLC26A4</span> gene in the compound heterozygous state in siblings with bilateral IP-2 and EVA anomalies. Note. (<b>a</b>) Pedigree of the patients (individuals with HL are highlighted in black; arrows indicate probands). (<b>b</b>) Computed tomography (CT) of the temporal bones in the axial projection of Patients II:3 and II:4 with IP-2 and EVA (black filled arrows indicate the cystic cochlea; triangles indicate the enlarged vestibular aqueduct); (<b>c</b>) The fragment of the chromatogram of the <span class="html-italic">SLC26A4</span> gene sequence with the c.85G&gt;C p.(Glu29Gln) and c.2089+1G&gt;A (IVS18+1G&gt;A) pathogenic variants identified in the compound heterozygous state.</p>
Full article ">Figure 3
<p>Identification of the c.2027T&gt;A p.(Leu676Gln) variant in the <span class="html-italic">SLC26A4</span> gene in a homozygous state in a patient with bilateral EVA anomalies. <b>Note.</b> (<b>a</b>) Pedigree of the patient (the arrow indicates the proband). (<b>b</b>) Computed tomography (CT) of the temporal bones in the axial projection. Upper panel: Patient II:3 with a preserved cochlea (white arrows), vestibule dilatation, and bilateral EVA anomalies (triangles indicate the enlarged vestibular aqueduct); lower panel, a patient without anomalies (white arrows). (<b>c</b>) Upper panel: a fragment of the chromatogram of the <span class="html-italic">SLC26A4</span> gene sequence with the c.2027T&gt;A p.(Leu676Gln) variant in a homozygous state; lower panel: the normal sequence.</p>
Full article ">Figure 4
<p>Morphology of the inner ear anomalies in biallelic <span class="html-italic">SLC26A4</span> and monoallelic <span class="html-italic">SLC26A4</span> patients. <b>Note.</b> IP-1, incomplete partition Type 1; IP-2, incomplete partition Type 2; EVA, enlargement of the vestibular aqueduct.</p>
Full article ">Figure 5
<p>A 3D reconstruction of the incomplete partitions of the cochlea and enlarged vestibular aqueduct anomalies in biallelic <span class="html-italic">SLC26A4</span> patients. <b>Note.</b> (<b>a</b>) Biallelic <span class="html-italic">SLC26A4</span> patient with an incomplete partition Type 1 (IP-1) cystic cochlear anomaly; (<b>b</b>) biallelic <span class="html-italic">SLC26A4</span> patient with an incomplete partition Type 2 (IP-2) cystic cochlear anomaly combined with EVA; (<b>c</b>) biallelic <span class="html-italic">SLC26A4</span> patient with a normal cochlea and an isolated EVA anomaly.</p>
Full article ">
17 pages, 2348 KiB  
Article
SLC26A4 Phenotypic Variability Influences Intra- and Inter-Familial Diagnosis and Management
by Mohamed Tawalbeh, Dunia Aburizeg, Bayan O. Abu Alragheb, Wala Sami Alaqrabawi, Zain Dardas, Luma Srour, Baraah Hatem Altarayra, Ayman A. Zayed, Zaid El Omari and Bilal Azab
Genes 2022, 13(12), 2192; https://doi.org/10.3390/genes13122192 - 23 Nov 2022
Cited by 1 | Viewed by 2406
Abstract
SLC26A4 is one of the most common genes causing autosomal recessive non-syndromic sensorineural hearing loss (SNHL). It has been reported to cause Pendred Syndrome (PDS) and DFNB4 which is deafness with enlarged vestibular aqueduct (EVA). However, mutated SLC26A4 is not conclusive for having [...] Read more.
SLC26A4 is one of the most common genes causing autosomal recessive non-syndromic sensorineural hearing loss (SNHL). It has been reported to cause Pendred Syndrome (PDS) and DFNB4 which is deafness with enlarged vestibular aqueduct (EVA). However, mutated SLC26A4 is not conclusive for having either DFNB4 or PDS. Three unrelated Jordanian families consisting of eight affected individuals with congenital bilateral hearing loss (HL) participated in this study. Whole-exome and Sanger sequencing were performed to investigate the underlying molecular etiology of HL. Further clinical investigations, including laboratory blood workup for the thyroid gland, CT scan for the temporal bone, and thyroid ultrasound were performed. Three disease-causing variants were identified in SLC26A4 in the three families, two of which were novel. Two families had a novel pathogenic homozygous splice-site accepter variant (c.165-1G>C), while the third family had compound heterozygous pathogenic variants (c.1446G>A; p.Trp482* and c.304G>A; p.Gly102Arg). Our approach helped in redirecting the diagnosis of several affected members of three different families from non-syndromic HL to syndromic HL. Two of the affected individuals had typical PDS, one had DFNB4, while the rest had atypical PDS. Our work emphasized the intra- and inter-familial variability of SLC26A4-related phenotypes. In addition, we highlighted the variable phenotypic impact of SLC26A4 on tailoring a personalized healthcare management. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

Figure 1
<p>Pedigrees of the three investigated Jordanian families (F1, F2, and F3) with variants in <span class="html-italic">SLC26A4</span>. Females are represented by circles and males are represented by squares. Filled symbols indicate affected individuals with hearing loss while empty symbols represent unaffected individuals. Arrows point to the proband of each family. The zygosity of the identified genotypes, validated using Sanger sequencing, was presented under the symbol of affected individuals and first-degree family members. Abbreviations: W: wild type, M1: mutation c.165-1G&gt;C, M2: mutation (c.304G&gt;A; p. Gly102Arg), and M3: mutation (c.1446G&gt;A; p.Trp482*).</p>
Full article ">Figure 2
<p>Audiograms of selected family members. Audiograms of F1: IV3 (c.165−1G&gt;C) are shown in (<b>A</b>–<b>C</b>) at the age of 1 year and 7 months, 6 years, and 17 years, respectively. (<b>D</b>) for F1: IV-2 at the age of 3 years. Audiograms of F1: IV−5 are depicted in (<b>E</b>,<b>F</b>) before and after cochlear implant at the age of 4 and 5 years, respectively. (<b>G</b>) for F2 (p.Gly102Arg and p.Trp482*): IV−1 at the age of 7 years. (<b>H</b>) for F2: IV-3 at the age of 5 years. (<b>I</b>) for F3 (c.165−1G&gt;C): V−4 at the age of 20 years. The symbols of the red circles and the blue crosses represent the readings of earphones unmasked air conduction of the right and left ear, respectively. Blue triangles for the masked air conduction readings of the right ear. The (&lt;) and (&gt;) symbols are used to represent the reading of the mastoid unmasked air conduction of the right and left ears, respectively. Arrows on any of the symbols represent no response.</p>
Full article ">Figure 3
<p>Schematic representation of the location of the variants in SLC26A6. The upper pane represents the corresponding chromatograms of the identified variants. Variant locus is highlighted in yellow. Black peak, G base; blue peak, C base; red peak, T base; green peak, A base. Abbreviations: Hom: homozygous, Het: Heterozygous.</p>
Full article ">Figure 4
<p>Temporal CT scan findings for F1: IV-3 and F1: IV-4. (<b>A</b>,<b>B</b>) Axial view of bilateral temporal bone CT scan for F1: IV-4 at the age of 17 years old post cochlear implantation where the blue lines represent the length of the EVA. The red circle shows the electrodes of the cochlear implant and the green rectangle shows the implanted cochlear device (<b>A</b>) for the right side and (<b>B</b>) for the left side. (<b>B</b>,<b>C</b>) Axial view of bilateral temporal bone CT scan for F1: IV-3 at the age of 25 years old showing features of incomplete partition II (Mondini Deformity) indicated by the blue circle (<b>C</b>) for the left side and (<b>D</b>) for the right side.</p>
Full article ">
15 pages, 2925 KiB  
Article
Molecular Features of SLC26A4 Common Variant p.L117F
by Arnoldas Matulevičius, Emanuele Bernardinelli, Zippora Brownstein, Sebastian Roesch, Karen B. Avraham and Silvia Dossena
J. Clin. Med. 2022, 11(19), 5549; https://doi.org/10.3390/jcm11195549 - 22 Sep 2022
Viewed by 1846
Abstract
The SLC26A4 gene, which encodes the anion exchanger pendrin, is involved in determining syndromic (Pendred syndrome) and non-syndromic (DFNB4) autosomal recessive hearing loss. SLC26A4 c.349C>T, p.L117F is a relatively common allele in the Ashkenazi Jewish community, where its minor allele frequency is increased [...] Read more.
The SLC26A4 gene, which encodes the anion exchanger pendrin, is involved in determining syndromic (Pendred syndrome) and non-syndromic (DFNB4) autosomal recessive hearing loss. SLC26A4 c.349C>T, p.L117F is a relatively common allele in the Ashkenazi Jewish community, where its minor allele frequency is increased compared to other populations. Although segregation and allelic data support the pathogenicity of this variant, former functional tests showed characteristics that were indistinguishable from those of the wild-type protein. Here, we applied a triad of cell-based assays, i.e., measurement of the ion transport activity by a fluorometric method, determination of the subcellular localization by confocal microscopy, and assessment of protein expression levels, to conclusively assign or exclude the pathogenicity of SLC26A4 p.L117F. This protein variant showed a moderate, but significant, reduction in ion transport function, a partial retention in the endoplasmic reticulum, and a strong reduction in expression levels as a consequence of an accelerated degradation by the Ubiquitin Proteasome System, all supporting pathogenicity. The functional and molecular features of human pendrin p.L117F were recapitulated by the mouse ortholog, thus indicating that a mouse carrying this variant might represent a good model of Pendred syndrome/DFNB4. Full article
Show Figures

Figure 1

Figure 1
<p>Ion transport activity of human (<b>a</b>) and <span class="html-italic">Mus musculus</span> (<b>b</b>) wild-type pendrin and pendrin variant p.L117F. HEK 293 Phoenix cells were co-transfected with wild-type (WT) or p.L117F pendrin, and the iodide sensor enhanced yellow fluorescent protein (EYFP) H148Q;I152L or EYFP H148Q;I152L alone (empty vector) and bathed in chloride- or iodide-containing solutions. The arrow indicates the addition of the iodide-containing solution to the bath. Left panels show the average fluorescence intensity over time normalized for the average fluorescence intensity measured in the chloride-containing solution. Right panels represent the % decrease in fluorescence intensity determined over the experimental period (19 s). <span class="html-italic">n</span> = 60 measurements from 5 independent experiments. *** <span class="html-italic">p</span> &lt; 0.0001, one-way ANOVA with Bonferroni’s multiple comparison post-test.</p>
Full article ">Figure 2
<p>Transcript and protein levels of human wild type pendrin and pendrin variant p.L117F. (<b>a</b>) HEK 293 Phoenix or HeLa cells were left untransfected (native) or transfected with the same plasmid constructs used for the determination of protein levels, and the <span class="html-italic">SLC26A4</span> transcript levels were quantified by qPCR. Human thyroid was tested in parallel as a reference. <span class="html-italic">n</span> refers to the number of biological replicates. (<b>b</b>) Representative images of fixed Hela cells transfected with wild type (WT) or p.L117F SLC26A4-EYFP (yellow) and counterstained with DAPI (cyan). The corresponding merge images are shown. Scale bar: 100 μm. (<b>c</b>) Pendrin protein expression levels expressed as fluorescence intensity (levels of gray) normalized for the cell density. <span class="html-italic">n</span> = 24 imaging fields from 4 independent subcultures. (<b>d</b>) Representative western blot on HEK 293 Phoenix cells transfected with wild type (WT) or p.L117F SLC26A4 or with an empty vector (mock). (<b>e</b>) Densitometry of <span class="html-italic">n</span> = 6 samples from 3 independent subcultures. n.s., not significant, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.0001, unpaired, two-tailed Student´s <span class="html-italic">t</span> test.</p>
Full article ">Figure 2 Cont.
<p>Transcript and protein levels of human wild type pendrin and pendrin variant p.L117F. (<b>a</b>) HEK 293 Phoenix or HeLa cells were left untransfected (native) or transfected with the same plasmid constructs used for the determination of protein levels, and the <span class="html-italic">SLC26A4</span> transcript levels were quantified by qPCR. Human thyroid was tested in parallel as a reference. <span class="html-italic">n</span> refers to the number of biological replicates. (<b>b</b>) Representative images of fixed Hela cells transfected with wild type (WT) or p.L117F SLC26A4-EYFP (yellow) and counterstained with DAPI (cyan). The corresponding merge images are shown. Scale bar: 100 μm. (<b>c</b>) Pendrin protein expression levels expressed as fluorescence intensity (levels of gray) normalized for the cell density. <span class="html-italic">n</span> = 24 imaging fields from 4 independent subcultures. (<b>d</b>) Representative western blot on HEK 293 Phoenix cells transfected with wild type (WT) or p.L117F SLC26A4 or with an empty vector (mock). (<b>e</b>) Densitometry of <span class="html-italic">n</span> = 6 samples from 3 independent subcultures. n.s., not significant, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.0001, unpaired, two-tailed Student´s <span class="html-italic">t</span> test.</p>
Full article ">Figure 3
<p>Colocalization of human wild-type pendrin and pendrin variant p.L117F with the plasma membrane. (<b>a</b>) Representative images of living Hela cells transfected with wild type (WT) or p.L117F SLC26A4-EYFP (green) and stained with the plasma membrane (PM) marker CellMask<sup>TM</sup> Deep Red (magenta). The corresponding merge images and scatter plots are shown. Scale bar: 25 μm. (<b>b</b>) Average Pearson’s correlation coefficient, overlap coefficient, and colocalization rate. <span class="html-italic">n</span> refers to the number of cells from 3 independent subcultures. n.s., not significant, unpaired, two-tailed Student´s <span class="html-italic">t</span> test.</p>
Full article ">Figure 4
<p>Colocalization of human wild-type pendrin and pendrin variant p.L117F with the endoplasmic reticulum. (<b>a</b>) Representative images of living Hela cells transfected with wild type (WT) or p.L117F SLC26A4-EYFP (green) and stained with the endoplasmic reticulum (ER) marker ER-Tracker<sup>TM</sup> Red (red). The corresponding merge images and scatter plots are shown. Scale bar: 25 μm. (<b>b</b>) Average Pearson´s correlation coefficient, overlap coefficient, and colocalization rate. <span class="html-italic">n</span> refers to the number of cells from 3 independent subcultures. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, unpaired, two-tailed Student´s <span class="html-italic">t</span> test.</p>
Full article ">Figure 5
<p>Expression and function of <span class="html-italic">Mus musculus</span> wild type pendrin and pendrin variant p.L117F in the presence of carfilzomib or its vehicle in HEK 293 Phoenix cells. (<b>a</b>) Cells were transfected with wild type (WT) or p.L117F SLC26A4-EYFP for 48 h and incubated with 1 μM carfilzomib or the vehicle (0.01% DMSO) for 16 h. Protein expression levels are expressed as fluorescence intensity (levels of gray) normalized for the cell density. <span class="html-italic">n</span> = 20 imaging fields from 4 independent subcultures. (<b>b</b>) Cells were co-transfected with wild type (WT) or p.L117F SLC26A4 and the iodide sensor enhanced yellow fluorescent protein (EYFP) H148Q;I152L or EYFP H148Q;I152L alone (empty vector) for 48 h and incubated with carfilzomib or the vehicle for 16 h. The % decrease in fluorescence intensity indicates an iodide influx from the bath solution towards the intracellular environment. <span class="html-italic">n</span> = 24 measurements from 4 independent experiments. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, one-way ANOVA with Bonferroni’s multiple comparison post-test.</p>
Full article ">Figure 6
<p>Multiple sequence alignment of the SLC26A4 protein sequence with its orthologues (top) and human paralogues (bottom). Amino acid residues conserved in all proteins are shaded in gray. The putative position of the second transmembrane (TM 2) α-helix according to [<a href="#B45-jcm-11-05549" class="html-bibr">45</a>] is indicated above the sequence. The position of the amino acid Leu117 (L117) is also given. (a) <span class="html-italic">H. sapiens</span> (NP_000432.1), (b) <span class="html-italic">P. Anubis</span> (XP_021791848.1), (c) <span class="html-italic">S. scrofa</span> (XP_003357559.1), (d) <span class="html-italic">C. familiaris</span> (XP_022260905.1), (e) <span class="html-italic">F. catus</span> (XP_003982698.1), (f) <span class="html-italic">M. musculus</span> (NP_035997.1), (g) <span class="html-italic">R. norvegicus</span> (NP_062087.1), (h) <span class="html-italic">X. laevis</span> (AAI69726.1), (i) <span class="html-italic">X. tropicalis</span> (NP_001107135.1), (l) <span class="html-italic">D. rerio</span> (NP_001159387.1), SCL26A4 (NP_000432.1), SLC26A1 (NP_998778.1), SLC26A2 (NP_000103.2), SLC26A3 (NP_000102.1), SLC26A5 (NP_945350.1), SLC26A6 (NP_075062.2), SLC26A7 (NP_001269285.1), SLC26A8 (NP_001180405.1), SLC26A9 (NP_443166.1).</p>
Full article ">
Back to TopTop