Next Article in Journal
The Many Faces of Lipids in Genome Stability (and How to Unmask Them)
Next Article in Special Issue
Natural Autoimmunity to Selenoprotein P Impairs Selenium Transport in Hashimoto’s Thyroiditis
Previous Article in Journal
Cryptochrome 2 from Lilium × formolongi Regulates Photoperiodic Flowering in Transgenic Arabidopsis thaliana
Previous Article in Special Issue
Loss of SELENOF Induces the Transformed Phenotype in Human Immortalized Prostate Epithelial Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Human Genetic Disorders Resulting in Systemic Selenoprotein Deficiency

by
Erik Schoenmakers
and
Krishna Chatterjee
*
Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(23), 12927; https://doi.org/10.3390/ijms222312927
Submission received: 9 November 2021 / Revised: 26 November 2021 / Accepted: 27 November 2021 / Published: 29 November 2021
(This article belongs to the Special Issue Molecular Biology of Selenium in Health and Disease)

Abstract

:
Selenium, a trace element fundamental to human health, is incorporated as the amino acid selenocysteine (Sec) into more than 25 proteins, referred to as selenoproteins. Human mutations in SECISBP2, SEPSECS and TRU-TCA1-1, three genes essential in the selenocysteine incorporation pathway, affect the expression of most if not all selenoproteins. Systemic selenoprotein deficiency results in a complex, multifactorial disorder, reflecting loss of selenoprotein function in specific tissues and/or long-term impaired selenoenzyme-mediated defence against oxidative and endoplasmic reticulum stress. SEPSECS mutations are associated with a predominantly neurological phenotype with progressive cerebello-cerebral atrophy. Selenoprotein deficiency due to SECISBP2 and TRU-TCA1-1 defects are characterized by abnormal circulating thyroid hormones due to lack of Sec-containing deiodinases, low serum selenium levels (low SELENOP, GPX3), with additional features (myopathy due to low SELENON; photosensitivity, hearing loss, increased adipose mass and function due to reduced antioxidant and endoplasmic reticulum stress defence) in SECISBP2 cases. Antioxidant therapy ameliorates oxidative damage in cells and tissues of patients, but its longer term benefits remain undefined. Ongoing surveillance of patients enables ascertainment of additional phenotypes which may provide further insights into the role of selenoproteins in human biological processes.

1. Introduction

Dietary selenium (Se) is absorbed as inorganic Se (e.g., selenate; selenite) or organic Se (e.g., Se-methionine; selenocysteine) and metabolized to hydrogen selenide (H2Se) before incorporation into the amino acid selenocysteine (Sec) [1]. Selenocysteine is different from other amino acids in that, uniquely, it is synthesized on its own tRNA, (Sec-tRNA[Ser]Sec; encoded by TRU-TCA1-1), via a well described pathway including O-phosphoserine-tRNA:selenocysteine tRNA synthase (SEPSECS) (Figure 1) [2,3]. Selenocysteine is incorporated into selenoproteins, at the position of a UGA codon in the mRNA, which ordinarily encodes a termination codon that dictates the cessation of protein synthesis. Unique Sec-insertion machinery, involving a cis-acting SEleniumCysteine Insertion Sequence (SECIS) stem-loop structure located in the 3′-UTR of all selenoprotein mRNAs and the UGA codon, interacting with trans-acting factors (SECIS binding protein 2 (SECISBP2), Sec-tRNA specific eukaryotic elongation factor (EEFSEC) and Sec-tRNA[Ser]Sec) (Figure 1), recodes UGA as a codon mediating Sec incorporation rather than termination of protein translation [3,4,5].
At least 25 human selenoproteins are described and recognized functions include maintenance of redox potential, regulating redox sensitive biochemical pathways, protection of genetic material, proteins and membranes from oxidative damage, metabolism of thyroid hormones, regulation of gene expression and control of protein folding (Table 1) [3,6]. The importance of selenoproteins is illustrated by the embryonic lethal phenotype of Trsp (mouse Sec-tRNA[Ser]Sec) and Secisbp2 knockout mice [7,8]. It is well known that dietary Se intake affects systemic Se-status and selenoprotein expression, but not all selenoproteins are affected to the same extent. Thus, expression of housekeeping selenoproteins (e.g., TXNRD1, TXNRD3, GPX4) is less affected by low circulating Se-levels compared to stress-related selenoproteins (e.g., GPX1, GPX3, SELENOW). Such differential preservation of selenoprotein expression is attributed to the existence of a “hierarchy of selenoprotein synthesis”, whose underlying molecular basis is unclear [3,9]. With this knowledge, it is no surprise that mutations in key components of the Sec-insertion pathway (SEPSECS, SECISBP2, TRU-TCA1-1) result in generalized deficiency of selenoproteins associated with a complex, multisystem phenotypes. Here, we describe the clinical consequences of mutations in these three human genes and suggest possible links with loss-of-function of known selenoproteins.

2. SECISBP2 Mutations

SECISBP2 is an essential and limiting factor for biosynthesis of selenoproteins [4,10] and functions as a scaffold, recruiting ribosomes, EEFSEC, and Sec-tRNA[Ser]Sec to the UGA codon by binding to SECIS-elements in selenoprotein mRNAs, generating a dynamic ribosome-Sec-incorporation complex (Figure 1). The first 400 amino (N-)terminal residues of SECISBP2 have no clear function; in contrast the carboxy (C-)terminal region (amino acids 399–784) is both necessary and sufficient for Sec-incorporation (Sec incorporation domain: SID) and binding to the SECIS element (RNA-binding domain: RBD) in vitro (Figure 2). The RBD, contains a L7Ae-type RNA interaction module and a lysine-rich domain, mediating specific recognition of ‘‘stem-loop’’ structures adopted by SECIS elements and other regulatory RNA motifs [11,12,13]. The C-terminal region also contains motifs (nuclear localization signal; nuclear export signal) involved in cellular localization of SECISBP2 and a cysteine rich domain (Figure 2) [14]. In the N-terminal region, alternative splicing events and ATG start codons have been described, generating different SECISBP2 isoforms [14], but all containing the essential C-terminal region. These events, together with regulatory domains in the C-terminal region, are thought to control SECISBP2-dependent Sec incorporation and the hierarchy of selenoprotein expression in vivo.
Homozygous or compound heterozygous mutations in SECISBP2 have been described in individuals from 11 families from diverse ethnic backgrounds [15] (Table 2, Figure 2); hitherto no phenotypes have been described in heterozygous individuals. Most SECISBP2 mutations identified to date result in premature stops in the N-terminal region upstream of an alternative start codon (Met 300), permitting synthesis of the shorter, C-terminal, minimal functional domain of SECISBP2 (Figure 2) [14,16,17,18,19,20,21,22,23]. Conversely, stop mutations (e.g., R770X, Q782X) [18,23], distal to the minimal functional domain might generate C-terminally truncated proteins with residual but altered function. In one patient with an intronic mutation (IVS8ds + 29G > A) leading to a stop in the SID-domain, correct mRNA splicing was only reduced by 50% [16], a mechanism preserving some SECISBP2 synthesis that may operate in other splice site mutation contexts.
Only three missense SECISBP2 mutations, situated in the RBD (R540Q, E679D and C691R) are described. The R540Q mutation, in the lysine-rich domain, fails to bind a specific subset of SECIS-elements in vitro and a mouse model revealed an abnormal pattern of Secisbp2 and selenoprotein expression in tissues [16,24,25]. The E679D and C691R mutations are situated in the L7Ae RNA-binding module and part of the CRD. C691R mutant SECISBP2 undergoes enhanced proteasomal degradation, with loss of RNA-binding [19,25]. The E679D mutation has not been investigated but is predicted to be deleterious (PolyPhen-2 algorithm score of 0.998), possibly affecting RNA-binding [23].
Complete knockout of Secisbp2 in mice is embryonic lethal [8], suggesting some functional protein, or an alternative rescue mechanism, is present in humans with SECISBP2 mutations. Studies suggest that most combinations of SECISBP2 mutations in patients hitherto are hypomorphic, with at least one allele directing synthesis of protein at either reduced levels or that is partially functional (Table 2). Because it is rate limiting for Sec incorporation, decreased SECISBP2 function will affect most if not all selenoprotein synthesis, as confirmed by available selenoprotein expression data in the patients [16,19].
Hitherto, only a small number of patients are described, from different ethnic and geographical backgrounds, often with compound heterozygous mutations and with limited information of their phenotypes. Some clinical phenotypes are attributable to deficiencies of particular selenoproteins in specific tissues whilst other features have a complex, multifactorial, basis possible linked to unbalanced antioxidant defence or protein folding pathways or loss of selenoproteins of unknown function. Increased cellular oxidative stress, readily measurable in most cells and tissues from patients, results in cumulative membrane and DNA damage. A common biochemical signature in all patients consists of low circulating selenium (reflecting low plasma SELENOP and GPX3) and abnormal thyroid hormone levels due to diminished activity of deiodinases resulting in raised FT4, normal to low FT3, raised reverse T3 and normal or high TSH concentrations [15,16,19]. Most cases were diagnosed in childhood with growth retardation (e.g., failure to thrive, short stature, delayed bone age) and developmental delay (e.g., delayed speech, intellectual- and motor coordination deficits) as common features, due not only to abnormal thyroid hormone metabolism [26,27] but also effects of specific selenoproteins deficiency in tissues (e.g., neuronal [8] or skeletal [28]). Fatigue and muscle weakness is a recognized feature in several patients and is attributable at least in part to a progressive muscular dystrophy affecting axial and proximal limb muscles, and very similar to the phenotype of myopathy due to selenoprotein N-deficiency [29]. Mild, bilateral, high-frequency sensorineural hearing loss is observed in some patients and is possibly due to ROS-mediated damage in the auditory system [30,31] as it is progressive in nature, worsening in older patients. An adult male patient was azoospermic, with marked deficiency of testis-expressed selenoproteins (GPX4, TXNRD3, SELENOV) [32,33,34,35,36]. Several other recorded phenotypes (increased whole body, subcutaneous fat mass, increased systemic insulin sensitivity, cutaneous photosensitivity) probably have a multifactorial basis which includes loss of antioxidant and endoplasmic reticulum stress defence. Studies of mouse models and in humans provide a substantial body of evidence to suggest a link between selenoproteins and most of these phenotypes [19,37,38,39,40,41].
Clinical management of these patients is mostly limited to correcting abnormal thyroid hormone metabolism with liothyronine supplementation if necessary. No specific therapies exist for other phenotypes (e.g., myopathy), but their progressive nature can require supportive intervention (e.g., nocturnal assisted ventilation for respiratory muscle weakness, aid for hearing loss). Oral selenium supplementation did raise total serum Se levels in some SECISBP2-deficient patients, but without clinical [17,18,21] or biochemical (circulating GPX’s, SELENOP, thyroid hormone metabolism) effect [42]. Antioxidant (alpha tocopherol) treatment was beneficial in one patient, reducing circulating levels of products of lipid peroxidation with reversal of these changes after treatment withdrawal [40]. These observations suggest that treatment with antioxidants is a rational therapeutic approach, but the longterm consequences in this multisystem disorder are unpredictable.

3. TRU-TCA1-1 Mutations

Selenocysteine is different from other amino acids in that it is synthesized uniquely on its own tRNA, encoded by TRU-TCA1-1, via a well described pathway including SEPSECS (Figure 1) [2,3]. Two major isoforms of the mature Sec-tRNA[Ser]Sec have been identified, containing either 5-methoxycarbonyl-methyluridine (mcm5U) or its methylated form 5-methoxycarbonylmethyl-2′-O-methyluridine (mcm5Um) at position 34, with the level of methylation being dependent on selenium status (Figure 3). The methylation state of uridine 34, located in the anticodon loop, is thought to contribute to stabilization of the codon–anticodon interaction and to play a role in mediating the hierarchy of selenoprotein expression. Thus, expression of essential, cellular housekeeping selenoproteins (e.g., TXNRDs, GPX4) is dependent on the mcm5U isoform, whilst synthesis of cellular, stress-related selenoproteins (e.g., GPX1, GPX3) synthesis require the mcm5Um isoform [43,44].
The first patient with a homozygous nucleotide change at position 65 (C > G) in TRU-TCA1-1 (Figure 3) [45], presented with a similar clinical and biochemical phenotype (fatigue and muscle weakness, raised FT4, normal T3, raised rT3 and TSH, low plasma selenium concentrations) to that seen in patients with SECISBP2 deficiency. However, the pattern of selenoprotein expression in his cells differed, with preservation housekeeping selenoproteins (e.g., TXNRDs, GPX4), but not stress-related selenoproteins (e.g., GPX1, GPX3) in cells from the TRU-TCA1-1 mutation patient. This pattern is similar to the differential preservation of selenoprotein synthesis described in murine Sec-tRNA[Ser]Sec mutant models [3,44]. Recently, a second, unrelated patient with the same, homozygous TRU-TCA1-1 mutation (C65G) with raised FT4 and low plasma GPX3 levels has been described [46].
The mechanism for such differential selenoprotein expression is unresolved, but a possible explanation is the observation that the TRU-TCA1-1 C65G mutation results in lower total Sec-tRNA[Ser]Sec expression in patients cells, with disproportionately greater diminution in Sec-tRNA[Ser]Sec mcm5Um levels. This suggest that the low Sec-tRNA[Ser]Sec levels in the proband are still sufficient for normal synthesis of housekeeping selenoproteins, whereas diminution of Sec-tRNA[Ser]Sec mcm5Um levels accounts for reduced synthesis of stress-related selenoproteins.
Clinical management of the patient is limited to alleviating clinical symptoms. However, with the knowledge that changing systemic selenium status can alter the relative proportions of the Sec-RNA[Ser]Sec isoforms [44,47,48], selenium supplementation of this patient, aiming to restore specific selenoprotein deficiencies, may be a rational therapeutic approach.

4. SEPSECS Mutations

The human SEPSECS protein was first characterized as an autoantigen (soluble liver antigen/liver pancreas, SLA) in autoimmune hepatitis [49]. The observation that it was present in a ribonucleoprotein complex with Sec-tRNA[Ser]Sec, led to its identification as the enzyme that catalyzes the final step of Sec formation by converting O-phosphoserine-tRNA[Ser]Sec into Sec-tRNA[Ser]Sec using selenophosphate as substrate donor [50,51] (Figure 1).
Homozygous and compound heterozygous mutations in SEPSECS have been identified in 20 patients (Table 3, Figure 4). The availability of the crystal structures of the archaeal and murine SEPSECS apo-enzymes as well as human wild type and mutant SECSEPS (A239T, Y334C, T325S and Y429X) complexed with Sec-tRNA[Ser]Sec provides functional information [52,53,54,55]. The four premature stop mutants are predicted to be insoluble and inactive, as documented for the Y429X mutant. Mutants at Tyrosine 334 are predicted to fold like wild type SEPSECS and retain binding to Sec-tRNA[Ser]Sec, but with reduced enzyme activity. The A239T mutant failed to form stable tetramers, possible as result of a steric clash destabilizing the enzyme’s core, rendering it inactive [55]. The other mutants for which no structure is available have been analyzed in silico and are predicted to be deleterious to varying degrees [15].
Patients with mutations in SEPSECS have profound intellectual disability, global developmental delay, spasticity, epilepsy, axonal neuropathy, optic atrophy and hypotonia with progressive microcephaly due to cortical and cerebellar atrophy [56,58,61,63]. The disorder is classified as autosomal recessive pontocerebellar hypoplasia type 2D (PCH2D, OMIM#613811), also referred to as progressive cerebellocerebral atrophy (PCCA) [56,67]. SEPSECS is required for generation of Sec-tRNA[Ser]Sec, which is essential for survival as demonstrated by the Trsp (mouse tRNA[Ser]Sec) knockout mouse model [44]. The Y334C-Sepsecs mouse model exhibits a phenotype similar to features described in patients [68]. However, there is some variation in impact of SEPSECS mutations and specific phenotypes, with three patients (homozygous for G441R; compound heterozygous for R26Pfs*42/N119S or N119S/R156Q), presenting with late-onset PCH2D and progressive but milder degree of CNS atrophy [62,64]. In silico analyses suggest that these mutations have a less deleterious effect on SEPSECS function [15], although environmental factors or patients’ genetic background modulating phenotype cannot be excluded.
The young age and severity of neurological problems in SEPSECS patients has precluded detailed investigation of selenoprotein expression and associated phenotypes. Studies of brain tissue from some patients showed decreased selenoprotein expression, correlating with increased cellular oxidative stress, but selenoprotein expression in other cell types (fibroblasts, muscle cells) was not significantly affected [58]. Serum selenium concentrations and thyroid status has been partially investigated in five patients, documenting either normal levels [61,65] or normal T4 but elevated TSH levels [58]. This suggests that the biochemical hallmarks of selenoprotein deficiency in SECISBP2 and TRU-TCA1-1 disorders (low circulating selenium and abnormal thyroid hormone levels) are not a significant feature in patients with SEPSECS mutations. Myopathic features with raised CK levels, abnormal mitochondria, cytoplasmic bodies and increased lipid accumulation in muscle are documented in one SEPSECS mutation case [61], with broad-based gait and postural instability suggesting muscle weakness in another patient [64]. These findings are similar to observations in selenoprotein N-deficient patients with SECISBP2 mutations [19]. Overall, limited studies to date suggest that SEPSECS patients exhibit phenotypes associated with selenoprotein deficiency, but that these features can be mutation and tissue dependent.

5. Conclusions

In humans, 25 genes, encoding different selenoproteins containing the amino acid selenocysteine (Sec), have been identified. In selenoprotein mRNAs the amino acid Sec is encoded by the triplet UGA which usually constitutes a stop codon, requiring its recoding by a complex, multiprotein mechanism. Failure of selenoprotein synthesis due to SECISBP2, TRU-TCA1-1 or SEPSECS defects, essential components of the selenoprotein biosynthesis pathway, results in complex disorders.
Individuals with SECISBP2 defects exhibit a multisystem phenotype including growth retardation, fatigue and muscle weakness, sensorineural hearing loss, increased whole body fat mass, azoospermia and cutaneous photosensitivity. Most patients were identified due to a characteristic biochemical signature with raised FT4, normal to low FT3, raised rT3 and normal/slightly high TSH and low plasma selenium levels. A similar biochemical phenotype and clinical features are described in one individual with a TRU-TCA1-1 mutation, although with relative preservation of essential housekeeping versus stress-related selenoprotein expression in his cells. Individuals with SEPSECS defects, essential for Sec-tRNA[Ser]Sec synthesis, present with a disorder characterized by cerebello-cerebral atrophy. Due to the young age and severe phenotype of patients, the effect of SEPSECS mutations on selenoprotein expression has not been studied in detail. In contrast, it is noteworthy that an early-onset central nervous system phenotype is not a feature in patients with SECISBP2 or TRU-TCA1-1 mutations.
As the function of many selenoproteins is unknown, or simultaneous deficiency of several selenoproteins exerts additive, synergistic or antagonistic effects culminating in complex dysregulation, linking disease phenotypes with altered expression of specific selenoproteins is challenging. Nevertheless, some causal links between specific selenoprotein deficiencies and phenotypes (e.g., abnormal thyroid function and deiodinase enzymes; low plasma Se and SELENOP, GPX3; azoospermia and SELENOV, GPX4, TXRND3; myopathy and SELENON) can be made. Other, progressive, phenotypes (e.g., photosensitivity, age-dependent hearing loss, neurodegeneration) may reflect absence of selenoenzymes mediating defence against oxidative and endoplasmic reticulum stress, resulting in cumulative tissue damage.
Triiodothyronine supplementation can correct abnormal thyroid hormone metabolism, with other medical intervention being mainly supportive. Selenium supplementation is of no proven benefit in SECISBP2 mutation patients, but needs evaluation in the TRU-TCA1-1 mutation case. Antioxidants, targeting the imbalance in oxidoredox and protein folding control pathways, could be beneficial in many selenoprotein deficient patients, but due to the complex interplay between different selenoproteins and their role in diverse biological processes, such treatment will require careful evaluation.

Author Contributions

E.S., writing—original draft preparation; K.C., writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Wellcome Trust Investigator Award, grant number 210755/Z/18/Z and the NIHR Cambridge Biomedical Research Centre.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Ferreira, R.; Sena-Evangelista, K.; de Azevedo, E.P.; Pinheiro, F.I.; Cobucci, R.N.; Pedrosa, L. Selenium in Human Health and Gut Microflora: Bioavailability of Selenocompounds and Relationship With Diseases. Frontiers in nutrition 2021, 8, 685317. [Google Scholar] [CrossRef]
  2. Turanov, A.A.; Xu, X.M.; Carlson, B.A.; Yoo, M.H.; Gladyshev, V.N.; Hatfield, D.L. Biosynthesis of selenocysteine, the 21st amino acid in the genetic code, and a novel pathway for cysteine biosynthesis. Ad. Nutr. 2011, 2, 122–128. [Google Scholar] [CrossRef] [Green Version]
  3. Labunskyy, V.M.; Hatfield, D.L.; Gladyshev, V.N. Selenoproteins: Molecular pathways and physiological roles. Physiol. Rev. 2014, 94, 739–777. [Google Scholar] [CrossRef] [Green Version]
  4. Copeland, P.R.; Fletcher, J.E.; Carlson, B.A.; Hatfield, D.L.; Driscoll, D.M. A novel RNA binding protein, SBP2, is required for the translation of mammalian selenoprotein mRNAs. EMBO J. 2000, 19, 306–314. [Google Scholar] [CrossRef]
  5. Martin, G.W.; Harney, J.W.; Berry, M.J. Selenocysteine incorporation in eukaryotes: Insights into mechanism and efficiency from sequence, structure, and spacing proximity studies of the type 1 deiodinase SECIS element. RNA 1996, 2, 171–182. [Google Scholar]
  6. Zoidis, E.; Seremelis, I.; Kontopoulos, N.; Danezis, G.P. Selenium-Dependent Antioxidant Enzymes: Actions and Properties of Selenoproteins. Antioxidants 2018, 7, 66–91. [Google Scholar] [CrossRef] [Green Version]
  7. Bösl, M.R.; Takaku, K.; Oshima, M.; Nishimura, S.; Taketo, M.M. Early embryonic lethality caused by targeted disruption of the mouse selenocysteine tRNA gene (Trsp). PNAS 1997, 94, 5531–5534. [Google Scholar] [CrossRef] [Green Version]
  8. Seeher, S.; Atassi, T.; Mahdi, Y.; Carlson, B.A.; Braun, D.; Wirth, E.K.; Klein, M.O.; Reix, N.; Miniard, A.C.; Schomburg, L.; et al. Secisbp2 is essential for embryonic development and enhances selenoprotein expression. Antioxid. Redox Signal 2014, 21, 835–849. [Google Scholar] [CrossRef] [Green Version]
  9. Sunde, R.A.; Raines, A.M. Selenium regulation of the selenoprotein and nonselenoprotein transcriptomes in rodents. Ad. Nutr. 2011, 2, 138–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Copeland, P.R.; Driscoll, D.M. Purification, redox sensitivity, and RNA binding properties of SECIS-binding protein 2, a protein involved in selenoprotein biosynthesis. J. Biol. Chem. 1999, 274, 25447–25454. [Google Scholar] [CrossRef] [Green Version]
  11. Caban, K.; Kinzy, S.A.; Copeland, P.R. The L7Ae RNA binding motif is a multifunctional domain required for the ribosome-dependent Sec incorporation activity of Sec insertion sequence binding protein 2. Mol. Cell. Biol. 2007, 27, 6350–6360. [Google Scholar] [CrossRef] [Green Version]
  12. Donovan, J.; Caban, K.; Ranaweera, R.; Gonzalez-Flores, J.N.; Copeland, P.R. A novel protein domain induces high affinity selenocysteine insertion sequence binding and elongation factor recruitment. J. Biol. Chem. 2008, 283, 35129–35139. [Google Scholar] [CrossRef] [Green Version]
  13. Takeuchi, A.; Schmitt, D.; Chapple, C.; Babaylova, E.; Karpova, G.; Guigo, R.; Krol, A.; Allmang, C. A short motif in Drosophila SECIS Binding Protein 2 provides differential binding affinity to SECIS RNA hairpins. Nucleic acids res. 2009, 37, 2126–2141. [Google Scholar] [CrossRef]
  14. Papp, L.V.; Lu, J.; Holmgren, A.; Khanna, K.K. From selenium to selenoproteins: Synthesis, identity, and their role in human health. Antioxid. Redox Signal 2007, 9, 775–806. [Google Scholar] [CrossRef]
  15. Schoenmakers, E.; Chatterjee, K. Human Disorders Affecting the Selenocysteine Incorporation Pathway Cause Systemic Selenoprotein Deficiency. Antioxid Redox Signal. 2020, 33, 481–497. [Google Scholar] [CrossRef]
  16. Dumitrescu, A.M.; Liao, X.H.; Abdullah, M.S.; Lado-Abeal, J.; Majed, F.A.; Moeller, L.C.; Boran, G.; Schomburg, L.; Weiss, R.E.; Refetoff, S. Mutations in SECISBP2 result in abnormal thyroid hormone metabolism. Nat. Genet. 2005, 37, 1247–1252. [Google Scholar] [CrossRef] [PubMed]
  17. Di Cosmo, C.; McLellan, N.; Liao, X.H.; Khanna, K.K.; Weiss, R.E.; Papp, L.; Refetoff, S. Clinical and molecular characterization of a novel selenocysteine insertion sequence-binding protein 2 (SBP2) gene mutation (R128X). J. Clin. Endocrinol. Metab. 2009, 94, 4003–4009. [Google Scholar] [CrossRef] [Green Version]
  18. Azevedo, M.F.; Barra, G.B.; Naves, L.A.; Ribeiro Velasco, L.F.; Godoy Garcia Castro, P.; de Castro, L.C.; Amato, A.A.; Miniard, A.; Driscoll, D.; Schomburg, L.; et al. Selenoprotein-related disease in a young girl caused by nonsense mutations in the SBP2 gene. J. Clin. Endocrinol. Metab. 2010, 95, 4066–4071. [Google Scholar] [CrossRef] [Green Version]
  19. Schoenmakers, E.; Agostini, M.; Mitchell, C.; Schoenmakers, N.; Papp, L.; Rajanayagam, O.; Padidela, R.; Ceron-Gutierrez, L.; Doffinger, R.; Prevosto, C.; et al. Mutations in the selenocysteine insertion sequence-binding protein 2 gene lead to a multisystem selenoprotein deficiency disorder in humans. J. Clin. Invest. 2010, 120, 4220–4235. [Google Scholar] [CrossRef] [Green Version]
  20. Hamajima, T.; Mushimoto, Y.; Kobayashi, H.; Saito, Y.; Onigata, K. Novel compound heterozygous mutations in the SBP2 gene: Characteristic clinical manifestations and the implications of GH and triiodothyronine in longitudinal bone growth and maturation. Eur. J. Endocrinol. 2012, 166, 757–764. [Google Scholar] [CrossRef] [Green Version]
  21. Çatli, G.; Fujisawa, H.; Kirbiyik, Ö.; Mimoto, M.S.; Gençpinar, P.; Özdemir, T.R.; Dündar, B.N.; Dumitrescu, A.M. A Novel Homozygous Selenocysteine Insertion Sequence Binding Protein 2 (SECISBP2, SBP2) Gene Mutation in a Turkish Boy. Thyroid 2018, 28, 1221–1223. [Google Scholar] [CrossRef]
  22. Korwutthikulrangsri, M.; Raimondi, C.; Dumitrescu, A.M. Novel Compound Heterozygous SBP2 Gene Mutations in a Boy with Developmental Delay and Failure to Thrive; 13th IWRTH: Doorn, The Netherlands, 2018; p. 22, Abstract Book. [Google Scholar]
  23. Fu, J.; Korwutthikulrangsri, M.; Gönç, E.N.; Sillers, L.; Liao, X.H.; Alikaşifoğlu, A.; Kandemir, N.; Menucci, M.B.; Burman, K.D.; Weiss, R.E.; et al. Clinical and Molecular Analysis in 2 Families With Novel Compound Heterozygous SBP2 (SECISBP2) Mutations. J. Clin. Endocrinol. Metab. 2020, 105, e6–e11. [Google Scholar] [CrossRef]
  24. Bubenik, J.L.; Driscoll, D.M. Altered RNA binding activity underlies abnormal thyroid hormone metabolism linked to a mutation in selenocysteine insertion sequence-binding protein 2. J. Biol. Chem. 2007, 282, 34653–34662. [Google Scholar] [CrossRef] [Green Version]
  25. Zhao, W.; Bohleber, S.; Schmidt, H.; Seeher, S.; Howard, M.T.; Braun, D.; Arndt, S.; Reuter, U.; Wende, H.; Birchmeier, C.; et al. Ribosome profiling of selenoproteins in vivo reveals consequences of pathogenic Secisbp2 missense mutations. J. Biol. Chem. 2019, 294, 14185–14200. [Google Scholar] [CrossRef] [Green Version]
  26. Ng, L.; Goodyear, R.J.; Woods, C.A.; Schneider, M.J.; Diamond, E.; Richardson, G.P.; Kelley, M.W.; Germain, D.L.; Galton, V.A.; Forrest, D. Hearing loss and retarded cochlear development in mice lacking type 2 iodothyronine deiodinase. Proc. Natl. Acad. Sci. USA 2004, 101, 3474–3479. [Google Scholar] [CrossRef] [Green Version]
  27. Hernandez, A.; Martinez, M.E.; Fiering, S.; Galton, V.A.; St Germain, D. Type 3 deiodinase is critical for the maturation and function of the thyroid axis. J. Clin. Invest. 2006, 116, 476–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Downey, C.M.; Horton, C.R.; Carlson, B.A.; Parsons, T.E.; Hatfield, D.L.; Hallgrímsson, B.; Jirik, F.R. Osteo-chondroprogenitor-specific deletion of the selenocysteine tRNA gene, Trsp, leads to chondronecrosis and abnormal skeletal development: A putative model for Kashin-Beck disease. PLoS Genet. 2009, 5, e1000616. [Google Scholar] [CrossRef] [Green Version]
  29. Silwal, A.; Sarkozy, A.; Scoto, M.; Ridout, D.; Schmidt, A.; Laverty, A.; Henriques, M.; D’Argenzio, L.; Main, M.; Mein, R.; et al. Selenoprotein N-related myopathy: A retrospective natural history study to guide clinical trials. Ann. Clin. Transl. neurol. 2020, 7, 2288–2296. [Google Scholar] [CrossRef] [PubMed]
  30. McFadden, S.L.; Ohlemiller, K.K.; Ding, D.; Shero, M.; Salvi, R.J. The Influence of Superoxide Dismutase and Glutathione Peroxidase Deficiencies on Noise-Induced Hearing Loss in Mice. Noise health 2001, 3, 49–64. [Google Scholar]
  31. Riva, C.; Donadieu, E.; Magnan, J.; Lavieille, J.P. Age-related hearing loss in CD/1 mice is associated to ROS formation and HIF target proteins up-regulation in the cochlea. Exp. Geront. 2007, 42, 327–336. [Google Scholar] [CrossRef]
  32. Ursini, F.; Heim, S.; Kiess, M.; Maiorino, M.; Roveri, A.; Wissing, J.; Flohé, L. Dual function of the selenoprotein PHGPx during sperm maturation. Science 1999, 285, 1393–1396. [Google Scholar] [CrossRef] [Green Version]
  33. Foresta, C.; Flohé, L.; Garolla, A.; Roveri, A.; Ursini, F.; Maiorino, M. Male fertility is linked to the selenoprotein phospholipid hydroperoxide glutathione peroxidase. Biol. reprod. 2002, 67, 967–971. [Google Scholar] [CrossRef] [Green Version]
  34. Kryukov, G.V.; Castellano, S.; Novoselov, S.V.; Lobanov, A.V.; Zehtab, O.; Guigó, R.; Gladyshev, V.N. Characterization of mammalian selenoproteomes. Science 2003, 300, 1439–1443. [Google Scholar] [CrossRef] [Green Version]
  35. Su, D.; Novoselov, S.V.; Sun, Q.A.; Moustafa, M.E.; Zhou, Y.; Oko, R.; Hatfield, D.L.; Gladyshev, V.N. Mammalian selenoprotein thioredoxin-glutathione reductase. Roles in disulfide bond formation and sperm maturation. J. Biol. Chem. 2005, 280, 26491–26498. [Google Scholar] [CrossRef] [Green Version]
  36. Schneider, M.; Förster, H.; Boersma, A.; Seiler, A.; Wehnes, H.; Sinowatz, F.; Neumüller, C.; Deutsch, M.J.; Walch, A.; Hrabé de Angelis, M.; et al. Mitochondrial glutathione peroxidase 4 disruption causes male infertility. FASEB J. 2009, 23, 3233–3242. [Google Scholar] [CrossRef] [Green Version]
  37. Schweikert, K.; Gafner, F.; Dell’Acqua, G. A bioactive complex to protect proteins from UV-induced oxidation in human epidermis. Int. J. Cosm. Sci. 2010, 32, 29–34. [Google Scholar] [CrossRef]
  38. Sengupta, A.; Lichti, U.F.; Carlson, B.A.; Ryscavage, A.O.; Gladyshev, V.N.; Yuspa, S.H.; Hatfield, D.L. Selenoproteins are essential for proper keratinocyte function and skin development. PLoS ONE 2010, 5, e12249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Verma, S.; Hoffmann, F.W.; Kumar, M.; Huang, Z.; Roe, K.; Nguyen-Wu, E.; Hashimoto, A.S.; Hoffmann, P.R. Selenoprotein K knockout mice exhibit deficient calcium flux in immune cells and impaired immune responses. J. Immunol. 2011, 186, 2127–2137. [Google Scholar] [CrossRef] [Green Version]
  40. Saito, Y.; Shichiri, M.; Hamajima, T.; Ishida, N.; Mita, Y.; Nakao, S.; Hagihara, Y.; Yoshida, Y.; Takahashi, K.; Niki, E.; et al. Enhancement of lipid peroxidation and its amelioration by vitamin E in a subject with mutations in the SBP2 gene. Lipid Res. 2015, 56, 2172–2182. [Google Scholar] [CrossRef] [Green Version]
  41. Liao, C.; Carlson, B.A.; Paulson, R.F.; Prabhu, K.S. The intricate role of selenium and selenoproteins in erythropoiesis. Free Radic. Biol. Med. 2018, 127, 165–171. [Google Scholar] [CrossRef]
  42. Schomburg, L.; Dumitrescu, A.M.; Liao, X.H.; Bin-Abbas, B.; Hoeflich, J.; Köhrle, J.; Refetoff, S. Selenium supplementation fails to correct the selenoprotein synthesis defect in subjects with SBP2 gene mutations. Thyroid 2009, 19, 277–281. [Google Scholar] [CrossRef] [Green Version]
  43. Shetty, S.P.; Copeland, P.R. Selenocysteine incorporation: A trump card in the game of mRNA decay. Biochimie 2015, 114, 97–101. [Google Scholar] [CrossRef] [Green Version]
  44. Carlson, B.A.; Yoo, M.H.; Tsuji, P.A.; Gladyshev, V.N.; Hatfield, D.L. Mouse models targeting selenocysteine tRNA expression for elucidating the role of selenoproteins in health and development. Molecules 2009, 14, 3509–3527. [Google Scholar] [CrossRef] [Green Version]
  45. Schoenmakers, E.; Carlson, B.; Agostini, M.; Moran, C.; Rajanayagam, O.; Bochukova, E.; Tobe, R.; Peat, R.; Gevers, E.; Muntoni, F.; et al. Mutation in human selenocysteine transfer RNA selectively disrupts selenoprotein synthesis. J. Clin. Invest. 2016, 126, 992–996. [Google Scholar] [CrossRef] [Green Version]
  46. Geslot, A.; Savagner, F.; Caron, P. Inherited selenocysteine transfer RNA mutation: Clinical and hormonal evaluation of 2 patients. Eur. Thyroid, J. 2021, 10, 542–547. [Google Scholar] [CrossRef]
  47. Hatfield, D.; Lee, B.J.; Hampton, L.; Diamond, A.M. Selenium induces changes in the selenocysteine tRNA[Ser]Sec population in mammalian cells. Nucleic acids res. 1991, 19, 939–943. [Google Scholar] [CrossRef] [Green Version]
  48. Diamond, A.M.; Choi, I.S.; Crain, P.F.; Hashizume, T.; Pomerantz, S.C.; Cruz, R.; Steer, C.J.; Hill, K.E.; Burk, R.F.; McCloskey, J.A.; et al. Dietary selenium affects methylation of the wobble nucleoside in the anticodon of selenocysteine tRNA([Ser]Sec). J. Biol. Chem. 1993, 268, 14215–14223. [Google Scholar] [CrossRef]
  49. Kernebeck, T.; Lohse, A.W.; Grötzinger, J. A bioinformatical approach suggests the function of the autoimmune hepatitis target antigen soluble liver antigen/liver pancreas. Hepatology 2001, 34, 230–233. [Google Scholar] [CrossRef]
  50. Small-Howard, A.; Morozova, N.; Stoytcheva, Z.; Forry, E.P.; Mansell, J.B.; Harney, J.W.; Carlson, B.A.; Xu, X.M.; Hatfield, D.L.; Berry, M.J. Supramolecular complexes mediate selenocysteine incorporation in vivo. Mol. Cell. Biol. 2006, 26, 2337–2346. [Google Scholar] [CrossRef] [Green Version]
  51. Xu, X.M.; Mix, H.; Carlson, B.A.; Grabowski, P.J.; Gladyshev, V.N.; Berry, M.J.; Hatfield, D.L. Evidence for direct roles of two additional factors, SECp43 and soluble liver antigen, in the selenoprotein synthesis machinery. J. Biol. Chem. 2005, 280, 41568–41575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Araiso, Y.; Palioura, S.; Ishitani, R.; Sherrer, R.L.; O’Donoghue, P.; Yuan, J.; Oshikane, H.; Domae, N.; Defranco, J.; Söll, D.; et al. Structural insights into RNA-dependent eukaryal and archaeal selenocysteine formation. Nucleic Acids Res. 2008, 36, 1187–1199. [Google Scholar] [CrossRef] [Green Version]
  53. Ganichkin, O.M.; Xu, X.M.; Carlson, B.A.; Mix, H.; Hatfield, D.L.; Gladyshev, V.N.; Wahl, M.C. Structure and catalytic mechanism of eukaryotic selenocysteine synthase. J. Biol. Chem. 2008, 283, 5849–5865. [Google Scholar] [CrossRef] [Green Version]
  54. Palioura, S.; Sherrer, R.L.; Steitz, T.A.; Söll, D.; Simonovic, M. The human SepSecS-tRNASec complex reveals the mechanism of selenocysteine formation. Science 2009, 325, 321–325. [Google Scholar] [CrossRef] [Green Version]
  55. Puppala, A.K.; French, R.L.; Matthies, D.; Baxa, U.; Subramaniam, S.; Simonović, M. Structural basis for early-onset neurological disorders caused by mutations in human selenocysteine synthase. Sci. Rep. 2016, 6, 32563. [Google Scholar] [CrossRef] [Green Version]
  56. Agamy, O.; Ben Zeev, B.; Lev, D.; Marcus, B.; Fine, D.; Su, D.; Narkis, G.; Ofir, R.; Hoffmann, C.; Leshinsky-Silver, E.; et al. Mutations disrupting selenocysteine formation cause progressive cerebello-cerebral atrophy. Am. J. Hum. Genet. 2010, 87, 538–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Makrythanasis, P.; Nelis, M.; Santoni, F.A.; Guipponi, M.; Vannier, A.; Béna, F.; Gimelli, S.; Stathaki, E.; Temtamy, S.; Mégarbané, A.; et al. Diagnostic exome sequencing to elucidate the genetic basis of likely recessive disorders in consanguineous families. Hum. Mutat. 2014, 35, 1203–1210. [Google Scholar] [CrossRef] [PubMed]
  58. Anttonen, A.K.; Hilander, T.; Linnankivi, T.; Isohanni, P.; French, R.L.; Liu, Y.; Simonović, M.; Söll, D.; Somer, M.; Muth-Pawlak, D.; et al. Selenoprotein biosynthesis defect causes progressive encephalopathy with elevated lactate. Neurology 2015, 85, 306–315. [Google Scholar] [CrossRef] [Green Version]
  59. Zhu, X.; Petrovski, S.; Xie, P.; Ruzzo, E.K.; Lu, Y.F.; McSweeney, K.M.; Ben-Zeev, B.; Nissenkorn, A.; Anikster, Y.; Oz-Levi, D.; et al. Whole-exome sequencing in undiagnosed genetic diseases: Interpreting 119 trios. Genet. Med. 2015, 17, 774–781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Alazami, A.M.; Patel, N.; Shamseldin, H.E.; Anazi, S.; Al-Dosari, M.S.; Alzahrani, F.; Hijazi, H.; Alshammari, M.; Aldahmesh, M.A.; Salih, M.A.; et al. Accelerating novel candidate gene discovery in neurogenetic disorders via whole-exome sequencing of prescreened multiplex consanguineous families. Cell Rep. 2015, 10, 148–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Pavlidou, E.; Salpietro, V.; Phadke, R.; Hargreaves, I.P.; Batten, L.; McElreavy, K.; Pitt, M.; Mankad, K.; Wilson, C.; Cutrupi, M.C.; et al. Pontocerebellar hypoplasia type 2D and optic nerve atrophy further expand the spectrum associated with selenoprotein biosynthesis deficiency. Eur. J. Paediatr. Neurol. 2016, 20, 483–488. [Google Scholar] [CrossRef] [PubMed]
  62. Iwama, K.; Sasaki, M.; Hirabayashi, S.; Ohba, C.; Iwabuchi, E.; Miyatake, S.; Nakashima, M.; Miyake, N.; Ito, S.; Saitsu, H.; et al. Milder progressive cerebellar atrophy caused by biallelic SEPSECS mutations. J. Hum. Genet. 2016, 61, 527–531. [Google Scholar] [CrossRef]
  63. Olson, H.E.; Kelly, M.; LaCoursiere, C.M.; Pinsky, R.; Tambunan, D.; Shain, C.; Ramgopal, S.; Takeoka, M.; Libenson, M.H.; Julich, K.; et al. Genetics and genotype-phenotype correlations in early onset epileptic encephalopathy with burst suppression. Ann. Neurol. 2017, 81, 419–429. [Google Scholar] [CrossRef] [PubMed]
  64. Van Dijk, T.; Vermeij, J.D.; van Koningsbruggen, S.; Lakeman, P.; Baas, F.; Poll-The, B.T. A SEPSECS mutation in a 23-year-old woman with microcephaly and progressive cerebellar ataxia. J. Inherit. Metab. Dis 2018, 41, 897–898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Arrudi-Moreno, M.; Fernández-Gómez, A.; Peña-Segura, J.L. A new mutation in the SEPSECS gene related to pontocerebellar hypoplasia type 2D. Med. Clin. 2021, 156, 94–95. [Google Scholar] [CrossRef] [PubMed]
  66. Nejabat, M.; Inaloo, S.; Sheshdeh, A.T.; Bahramjahan, S.; Sarvestani, F.M.; Katibeh, P.; Nemati, H.; Tabei, S.; Faghihi, M.A. Genetic Testing in Various Neurodevelopmental Disorders Which Manifest as Cerebral Palsy: A Case Study From Iran. Frontiers in Pediatrics 2021, 9, 734946. [Google Scholar] [CrossRef] [PubMed]
  67. Ben-Zeev, B.; Hoffman, C.; Lev, D.; Watemberg, N.; Malinger, G.; Brand, N.; Lerman-Sagie, T. Progressive cerebellocerebral atrophy: A new syndrome with microcephaly, mental retardation, and spastic quadriplegia. J. Med. Genet. 2003, 40, e96. [Google Scholar] [CrossRef] [Green Version]
  68. Fradejas-Villar, N.; Zhao, W.; Reuter, U.; Doengi, M.; Ingold, I.; Bohleber, S.; Conrad, M.; Schweizer, U. Missense mutation in selenocysteine synthase causes cardio-respiratory failure and perinatal death in mice which can be compensated by selenium-independent GPX4. Redox Biol. 2021, 48, 102188. [Google Scholar] [CrossRef]
Figure 1. Biosynthesis of selenocysteine (Sec) and selenoproteins. Dietary sources of selenium exist in inorganic form (e.g., selenate, selenite) and organic form (e.g., Sec, SeMet). Inorganic selenium is reduced to selenide by TXNRD/TRX or GPX/GSH systems and organic selenium is metabolized to Sec, used by SCLY to generate selenide. De novo Sec synthesis takes place on its own tRNA (tRNA[Ser]Sec), which undergoes maturation through sequential modifications (SARS-mediated addition of Ser, PSTK-mediated phosphorylation of Ser), with acceptance of a selenophosphate (generated from selenide by SEPHS2) catalysed by SEPSECS as final step. Expression of selenoproteins requires recoding of an UGA codon as the amino acid Sec instead of a premature stop. The incorporation of Sec is mediated by a multiprotein complex containing SECISBP2, bound to the SECIS element situated in the 3′-untranslated region of selenoproteins, the Sec elongation factor EEFSEC, together with Sec-tRNA[Ser]Sec at the ribosomal acceptor site. The other factors (e.g., ribosomal protein L30, eukaryotic initiation factor eIF4a3, nucleolin) have regulatory roles.
Figure 1. Biosynthesis of selenocysteine (Sec) and selenoproteins. Dietary sources of selenium exist in inorganic form (e.g., selenate, selenite) and organic form (e.g., Sec, SeMet). Inorganic selenium is reduced to selenide by TXNRD/TRX or GPX/GSH systems and organic selenium is metabolized to Sec, used by SCLY to generate selenide. De novo Sec synthesis takes place on its own tRNA (tRNA[Ser]Sec), which undergoes maturation through sequential modifications (SARS-mediated addition of Ser, PSTK-mediated phosphorylation of Ser), with acceptance of a selenophosphate (generated from selenide by SEPHS2) catalysed by SEPSECS as final step. Expression of selenoproteins requires recoding of an UGA codon as the amino acid Sec instead of a premature stop. The incorporation of Sec is mediated by a multiprotein complex containing SECISBP2, bound to the SECIS element situated in the 3′-untranslated region of selenoproteins, the Sec elongation factor EEFSEC, together with Sec-tRNA[Ser]Sec at the ribosomal acceptor site. The other factors (e.g., ribosomal protein L30, eukaryotic initiation factor eIF4a3, nucleolin) have regulatory roles.
Ijms 22 12927 g001
Figure 2. Functional domains of human SECISBP2 with the position of mutations described hitherto. Arrowheads denote the location of ATG codons; NLS: nuclear localisation signal (380–390); NES: nuclear export signals (634–657 and 756–770); SID: Sec incorporation domain; CRD: cysteine rich domain; RBD: minimal RNA-binding domain with the Lysine-rich domain (K-rich) and the L7Ae RNA-binding module; the black bar denotes the minimal protein region required for full functional activity in vitro.
Figure 2. Functional domains of human SECISBP2 with the position of mutations described hitherto. Arrowheads denote the location of ATG codons; NLS: nuclear localisation signal (380–390); NES: nuclear export signals (634–657 and 756–770); SID: Sec incorporation domain; CRD: cysteine rich domain; RBD: minimal RNA-binding domain with the Lysine-rich domain (K-rich) and the L7Ae RNA-binding module; the black bar denotes the minimal protein region required for full functional activity in vitro.
Ijms 22 12927 g002
Figure 3. Sec-tRNA[Ser]Sec showing the position of human mutation. The primary structure of human Sec-tRNA[Ser]Sec is shown in a cloverleaf model, with the location of C65G mutation and posttranscriptional modification at positions U34 (mcm5U or mcm5Um, in the anticodon), A37 (i6A), U55 (⍦) and A58 (m1A).
Figure 3. Sec-tRNA[Ser]Sec showing the position of human mutation. The primary structure of human Sec-tRNA[Ser]Sec is shown in a cloverleaf model, with the location of C65G mutation and posttranscriptional modification at positions U34 (mcm5U or mcm5Um, in the anticodon), A37 (i6A), U55 (⍦) and A58 (m1A).
Ijms 22 12927 g003
Figure 4. Functional domains of SEPSECS with the positions of the human mutations. Schematic of the human SEPSECS protein with key amino acids (above) that are part of the active domain (black bars) or interact with tRNA[ser]sec] (white shaded boxes) and mutations described hitherto below.
Figure 4. Functional domains of SEPSECS with the positions of the human mutations. Schematic of the human SEPSECS protein with key amino acids (above) that are part of the active domain (black bars) or interact with tRNA[ser]sec] (white shaded boxes) and mutations described hitherto below.
Ijms 22 12927 g004
Table 1. Human selenoproteins.
Table 1. Human selenoproteins.
SelenoproteinFunctionExpression
Subcellular Localization
GPX1
glutathione peroxidase 1
Oxidoreductasemost tissues
cytoplasmic
GPX2
glutathione peroxidase 2
Oxidoreductaselimited number of tissues
Nucleus and cytoplasmic
GPX3
glutathione peroxidase 3
Oxidoreductasemost tissues
secreted
GPX4
glutathione peroxidase 4
Oxidoreductasemost tissues
Nucleus and mitochondria
GPX6
glutathione peroxidase 6
Oxidoreductasetestis, epididymis, olfactory system
predicted secreted
TXNRD1
thioredoxin reductase 1
OxidoreductaseUbiquitous
Nucleus and cytoplasmic
TXNRD2
Thioredoxin reductase 2
OxidoreductaseUbiquitous
cytoplasmic and mitochondria
TXNRD3
Thioredoxin reductase 3
Oxidoreductasemost tissues, high in testis
Intracellular
DIO1
Iodothyronine deiodinase 1
Thyroid hormone metabolismkidney, liver, thyroid gland
Intracellular membrane-associated
DIO2
Iodothyronine deiodinase 2
Thyroid hormone metabolismcentral nervous system, pituitary
Intracellular membrane-associated
DIO3
Iodothyronine deiodinase 3
Thyroid hormone metabolismseveral tissues
Intracellular membrane-associated
MSRB1
methionine sulfoxide reductase B1
Met sulfoxide reductionUbiquitous
Nucleus and cytoplasmic
SELENOF
Selenoprotein F
Protein folding controlUbiquitous
endoplasmic reticulum
SELENOH
Selenoprotein H
Unknown oxidoreductaseUbiquitous
Nucleus
SELENOI
Selenoprotein I
Phospholipid biosynthesisUbiquitous
transmembrane
SELENOK
Selenoprotein K
Protein folding controlUbiquitous
ER, plasma membrane
SELENOM
Selenoprotein M
UnknownUbiquitous
Nuclear and perinuclear
SELENON
Selenoprotein N
Redox-calcium homeostasisUbiquitous
endoplasmic reticulum
SELENOO
Selenoprotein O
Protein AMPylation activityUbiquitous
mitochondria
SELENOP
Selenoprotein P
Transport/oxidoreductasemost tissues
secreted, cytoplasmic
SELENOS
Selenoprotein S
Protein folding controlUbiquitous
endoplasmic reticulum
SELENOT
Selenoprotein T
Unknown oxidoreductaseUbiquitous
endoplasmic reticulum
SELENOV
Selenoprotein V
Unknownthyroid, parathyroid, testis, brain
Intracellular
SELENOW
Selenoprotein W
OxidoreductaseUbiquitous
Intracellular
SEPHS2
Selenophosphate synthetase 2
Selenophosphate synthesisUbiquitous, high in liver and kidney
Intracellular
Table 2. Human SECISBP2 mutations.
Table 2. Human SECISBP2 mutations.
Age in Years (Gender)MutationProtein ChangeAlleles
Affected
EthnicityReference
26 (M 1); 19 (M); 19 (F 2)c.1619 G > AR540QhomozygousSaudi
Arabian
[16]
25 (M)c.1312 A > T
c.IVS8ds + 29 G > A
K438 *
fs431 *
compound
heterozygous
Irish[16]
19 (M)c.382 C > TR128 *homozygousGhanaian[17]
18 (F)c.358 C > T
c.2308 C > T
R120 *
R770 *
compound
heterozygous
Brazilian[18]
44 (M)c.668delT
c.IVS7 -155, T > A
F223fs255 *
fs295 * + fs302 *
compound
heterozygous
British[19]
13 (M)c. 2017 T > C
1–5 intronic SNP’s
C691R
fs65 * + fs76 *
compound
heterozygous
British[19]
15 (M)c.1529_1541dup CCAGCGCCCCACT
c.235 C > T
M515fs563 *
Q79 *
compound
heterozygous
Japanese[20]
10 (M)c.800_801insAK267Kfs * 2homozygousTurkish[21]
3.5 (M)c.283delT
c.589 C > T
T95Ifs31 *
R197 *
compound
heterozygous
N/A 3[22]
11 (F)c.2344 C > T
c.2045–2048 delAACA
Q782 *
K682fs683 *
compound
heterozygous
Turkish[23]
5 (F)c.589 C > T
c.2108 G > T or C
R197 *
E679D
compound
heterozygous
Argentinian[23]
1 M: Male; 2 F: Female; 3 N/A: Not available.
Table 3. Human SEPSECS mutations.
Table 3. Human SEPSECS mutations.
Age in Year (Gender)MutationProtein
Change
Alleles
Affected
EthnicityReference
6 (F 1); 7.5 (2)c.1001 A > GY334ChomozygousJewish/Iraq[56]
4 (F); 2.5 (M)c.715 G > A
c.1001 A > G
A239T
Y334C
compound
heterozygous
Iraqi/
Moroccan
[56]
7 (F); 4 (F);
2 (F)
c.1466 A > TD489VhomozygousJordan[57]
0 (M); 0 (F); 0 (F); 0 (F)c.974 C > G
c.1287 C > A
T325S
Y429X
compound
heterozygous
Finnish[58]
14 (F)c.1 A > G
c.388 + 3 A > G
M1V
G130Vfs * 5
compound
heterozygous
N/A 3[59]
N/Ac.1027–1120delE343Lfs * 2HomozygousN/A 3[60]
9 (M)c.1001 A > CY334HhomozygousArabian[61]
10 (F)c.77delG
c.356 A > G
R26Pfs * 42
N119S
compound
heterozygous
Japanese[62]
21 (F)c.356 A > G
c.467 G > A
N119S
R156Q
compound
heterozygous
Japanese[62]
1 (M)c.176 C > TA59VHomozygousN/A 3[63]
23 (F)c.1321 G > AG441RHomozygousN/A 3[64]
4 (F)c.114 + 3 A > GN/A 3HomozygousMoroccan[65]
N/A 1c.877 G > AA293THomozygousN/A 3[66]
1 F: Female; 2 M: Male; 3 N/A: Not available.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Schoenmakers, E.; Chatterjee, K. Human Genetic Disorders Resulting in Systemic Selenoprotein Deficiency. Int. J. Mol. Sci. 2021, 22, 12927. https://doi.org/10.3390/ijms222312927

AMA Style

Schoenmakers E, Chatterjee K. Human Genetic Disorders Resulting in Systemic Selenoprotein Deficiency. International Journal of Molecular Sciences. 2021; 22(23):12927. https://doi.org/10.3390/ijms222312927

Chicago/Turabian Style

Schoenmakers, Erik, and Krishna Chatterjee. 2021. "Human Genetic Disorders Resulting in Systemic Selenoprotein Deficiency" International Journal of Molecular Sciences 22, no. 23: 12927. https://doi.org/10.3390/ijms222312927

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

Article Metrics

Back to TopTop