Next Article in Journal
Enhancement of Nitric Oxide Bioavailability by Modulation of Cutaneous Nitric Oxide Stores
Previous Article in Journal
In Vitro Evidence of Statins’ Protective Role against COVID-19 Hallmarks
Previous Article in Special Issue
Clinical and Molecular Spectrum of Sporadic Vascular Malformations: A Single-Center Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Revertant Mosaicism in Genodermatoses: Natural Gene Therapy Right before Your Eyes

by
Peter C. van den Akker
1,2,3,*,†,
Maria C. Bolling
3,† and
Anna M. G. Pasmooij
3,†
1
Groningen Center for Blistering Diseases, Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
2
Division of Biological Chemistry and Drug Development, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
3
Groningen Center for Blistering Diseases, Department of Dermatology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2022, 10(9), 2118; https://doi.org/10.3390/biomedicines10092118
Submission received: 1 July 2022 / Revised: 23 August 2022 / Accepted: 23 August 2022 / Published: 29 August 2022
(This article belongs to the Special Issue Somatic Mosaicism in Skin Disorders)

Abstract

:
Revertant mosaicism (RM) is the intriguing phenomenon in which nature itself has successfully done what medical science is so eagerly trying to achieve: correcting the effect of disease-causing germline variants and thereby reversing the disease phenotype back to normal. RM was molecularly confirmed for the first time in a genodermatosis in 1997, the genetic skin condition junctional epidermolysis bullosa (EB). At that time, RM was considered an extraordinary phenomenon. However, several important discoveries have changed this conception in the past few decades. First, RM has now been identified in all major subtypes of EB. Second, RM has also been identified in many other genodermatoses. Third, a theoretical mathematical exercise concluded that reverse mutations should be expected in all patients with a recessive subtype of EB or any other genodermatosis. This has shifted the paradigm from RM being an extraordinary phenomenon to it being something that every physician working in the field of genodermatoses should be looking for in every patient. It has also raised hope for new treatment options in patients with genodermatoses. In this review, we summarize the current knowledge on RM and discuss the perspectives of RM for the future treatment of patients with genodermatoses.

1. Genodermatoses

Genodermatoses are a group of genetic skin conditions caused by pathogenic genomic variants that affect genes expressed in one of the compartments of the skin. Their (birth) prevalence varies widely, with most being rare to extremely rare. Depending on the definition used, more than 500 different types of genodermatoses can be distinguished, for which several hundred causal genes are known (see the Online Mendelian Inheritance in Man database, www.omim.org, accessed 1 June 2022). The phenotypic spectrum of genodermatoses, as well as their severity, covers a broad continuum. On the most severe end are the lethal neonatal or early childhood phenotypes like acantholytic epidermolysis bullosa (EB) (in the group of erosive skin fragility disorders [1]), which is caused by biallelic carboxyl-terminally truncating mutations in the desmosomal protein desmoplakin encoded by the DSP gene [2,3], autosomal recessive congenital ichthyosis (ARCI)—harlequin type, which is caused by biallelic null-variants in the ABCA12 gene coding for the ATP-binding cassette (ABC) A12-transporter protein [4,5], and restrictive dermopathy caused by biallelic null-variants in the ZMPSTE24 gene, which encodes the zinc metalloproteinase STE24 that is crucial for the processing and maturation of the lamin A protein [6,7]. At the other end of the genodermatosis spectrum are relatively mild and/or late-onset conditions like mild forms of ectodermal dysplasia (ED), which is caused by either homozygous or heterozygous missense variants in the wingless family member 10A gene WNT10A [8], and the nails-only type of localized dystrophic EB (DEB) that is caused by glycine substitutions in the type VII collagen encoding gene COL7A1 [9]. However, the precise phenotype of the multiple genodermatoses varies greatly and depends on the gene involved and the nature of the causative pathogenic variants. Consequently, there is large clinical heterogeneity and variable expression among the genodermatoses, and most disease groups within the spectrum of genodermatoses also have broad clinical spectra. For instance, while biallelic null-variants in ABCA12 cause the very severe harlequin type of ARCI, ‘milder’ variants, usually missense or splice-site variants, cause a type of ARCI with or without a collodion membrane and erythroderma that is very similar to other forms of ARCI that do not affect life expectancy in most cases [10]. In addition, while heterozygous or even homozygous missense variants in WNT10A cause mild forms of ED or isolated hypo- or oligodontia, biallelic truncating variants can cause more severe types of ED like hypohidrotic ED (HED), odonto-onycho-dermal dysplasia, or Schöpf–Schulz–Passarge syndrome [8]. Finally, whereas certain heterozygous glycine substitutions in COL7A1 only affect the halluces [9], biallelic null-variants in COL7A1 are associated with a very severe type of recessive DEB (RDEB) with severe morbidity and early mortality [11]. In addition to clinical heterogeneity, there is also genetic heterogeneity among the genodermatoses. For instance, variants in 13 genes are now known to be implicated in one type of ARCI (www.omim.org, accessed on 1 June 2022), with largely overlapping features [12].

2. Mosaicism in Genodermatoses

Another important disease-modifying phenomenon is somatic mosaicism of the causal gene variants. Somatic mosaicism is the coexistence, in one individual, of cell lines that are (epi)genetically different yet originated from a single zygote [13]. The proportions of the cell lines within the body determine the extent and severity of the associated disease phenotype and, in genodermatoses, often result in skin areas with different disease expression [14]. Mosaicism is usually due to a somatic genetic or epigenetic change that occurred post-zygotically in one of the cells of the otherwise genetically homogeneous developing embryo [13]. A recent review proposed classifying mosaicism according to six different attributes (A–F): Affected tissue (somatic and/or germinal), Body pattern (segmental vs. non-segmental), Change of direction (‘forward’ vs. ‘revertant’), Developmental mechanism (type I vs. type II segmental mosaicism, functional X-linked mosaicism, disorders only manifesting as mosaics), Etiology (type of genomic alteration), and the Fraction of affected tissue (mild–severe involvement) [15]. In this review, we will only discuss mosaicism based on the direction of a somatic genetic alteration, i.e., ‘revertant mosaicism’ (RM), and only briefly discuss ‘forward mosaicism’ as a comparison (Figure 1).

3. Forward Mosaicism in Genodermatoses

In forward mosaicism, a spontaneous somatic variant mutates a wildtype allele, thereby inducing a disease phenotype in the cell population where it occurs. For a recent review on forward mosaicism and the challenges in properly diagnosing it at the molecular level, we refer the reader to Cheraghlou et al. [14]. In cases where the somatic mutation occurs in a cell containing two wildtype alleles and the gene involved is associated with a dominantly inherited disease, the heterozygous cell populations will express a mosaic phenotype of the dominant genodermatosis. Many examples of this ‘type 1 segmental’ mosaicism have been described, e.g., in segmental neurofibromatosis type 1 or mosaic RASopathy-associated epidermal nevi [16,17]. This group also includes the disorders that can only manifest in a mosaic state because the autosomal dominant, and usually activating, variants are lethal if present in an entire body, e.g., McCune–Albright syndrome due to the somatic GNAS variants p.(Arg201His) or p.(Arg201Cys) [18], and PIK3CA-related overgrowth spectrum [19,20].
A similar event can occur in cells that already carry a mutant allele of a gene involved in an autosomal recessive genodermatosis. The affected cell populations will then carry biallelic gene variants and express a disease phenotype in a mosaic fashion. Remarkably, while there is a large number of autosomal recessive genodermatoses and there are millions of carriers of the gene variants in the associated genes, segmental type 1 mosaicism for such recessive genodermatoses is only rarely reported. One of the few examples is a case of blaschkoid congenital ichthyosiform erythroderma due to somatic mosaicism for a second ABCA12 variant [21]. While this has not been thoroughly investigated, this lack of observations may reflect that, for many disorders, cells lacking the protein encoded by the gene involved have a developmental disadvantage.
Forward mosaicism can also occur on the wildtype allele in cells that already carry a heterozygous mutation for an autosomal dominant or X-linked (in women) genodermatosis. This loss of heterozygosity for the wildtype allele either increases the severity of the disease in the affected cell populations or changes the nature of the disease, leading to type 2 segmental mosaicism [22]. Type 2 segmental mosaicism has been reported in many genodermatoses, e.g., Darier disease, which is caused by variants in the ATP2A2 gene, or Gorlin syndrome, caused by variants in the PTCH1 gene [23,24]. Irrespective of the type of mosaicism or the disorder involved, the expression of the somatic phenotype is always determined by the cell type and proportion of cells that carry the somatic variant, the timing of its occurrence, and the nature and effect of the somatic variants [13,14,15].

4. Revertant Mosaicism in Genodermatoses

In RM, an additional somatic change mitigates the effect of the germline variant(s) and the direction of mosaicism is towards improvement rather than the induction or worsening of the disease phenotype [15]. In 1995, Jonkman et al. described a patient with the type XVII collagen-deficient intermediate type of junctional EB (JEB) due to germline COL17A1 mutations who exhibited normal-looking, non-affected skin areas in which blistering could not be induced by rubbing [25]. Skin biopsy sections showed focal areas of positive type XVII collagen-staining, indicating RM in a micro-mosaic pattern. In 1997, the presence of RM in this patient was proven at the DNA level, making it the first confirmed case of RM in a genodermatosis [26], although RM had already been described in other genetic conditions (Supplementary Table S1) [27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61]. In fact, RM had been demonstrated as early as 1977 in patients with Bloom’s syndrome through the observation of coexistence of cells with a greatly increased number of sister chromatid exchanges next to cells with a normal number [62]. However, RM could not be proven at the DNA level at that time due to the lack of appropriate techniques. The first case of RM proven at the DNA level was a patient with the X-linked recessive condition, Lesch–Nyhan syndrome [61]. Despite carrying a germline deleterious intragenic duplication of exons 2 and 3 of the HPRT gene, this patient had an unusually mild presentation as he did not have the severe intellectual disability that is a common feature of the syndrome. This unusually mild presentation was likely explained by the presence of revertant clones that had lost the duplicated genomic region due to a postzygotic gene rearrangement.
At the time of the initial description of RM in a genodermatosis in 1997, RM was considered an extraordinary phenomenon. However, since that initial description, RM has been identified in all major types of EB (Table 1) [63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78], and, in 2012, we were able to confirm revertant skin patches in all ten Dutch patients with the intermediate type of JEB due to pathogenic COL17A1 variants [79]. Around the same time, Choate et al. demonstrated that each of the multiple healthy ‘confetti-like’ spots in patients with ichthyosis with confetti (IWC) due to germline variants in KRT10 (IWC-I) or KRT1 (IWC-II) represent a separate occurrence of RM in a single keratinocyte clone [80,81]. Moreover, RM was identified in a patient with keratitis-ichthyosis-deafness (KID) syndrome due to a heterozygous dominant-negative variant in the GJB2 gene [82]. Suzuki et al. reported that RM also frequently occurs in loricrin keratoderma (LK) [83], and Miyauchi et al. recently reported multiple revertant skin spots in two patients with pityriasis rubra pilaris (PRP) due to gain-of-function missense variants in CARD14 [84]. Figure 2 provides an overview of the revertant skin phenotype in these genodermatoses. Finally, by applying a mathematical developmental model to the occurrence of RM in patients with recessive types of EB, we concluded that reverse mutations should occur multiple times in the skin of all patients with recessive types of EB [85]. This conclusion corroborates the clinical findings that, contrary to previous conceptions, RM is to be expected in all patients with EB and other genodermatoses. The reason why RM is frequently seen in skin likely has to do with the facts that skin can easily be explored visually and is a rapidly self-renewing organ in which a large number of cell divisions take place, which means it is analogous to bone marrow and the frequent occurrence of RM observed in hematologic and immunologic disorders (Supplementary Table S1).
Our mathematical developmental model also revealed that, to be able to grow to the size of a clinically recognizable patch, RM should occur very early in embryogenesis [85]. However, the chance that a reverse mutation occurs at such an early embryonic stage was calculated to be so low that a revertant patch would be expected in only 1 in 10,000 patients, which clearly underestimates the clinical situation. The only likely explanation was that reverse mutations do occur later during (embryonic) life but impose a significant selective growth advantage on revertant cells during a particular time-window, the “late-but-fitter revertant cell” hypothesis. In KID syndrome, the revertant variants identified in cis with the germline variant were also thought to provide the host cells with a selective growth advantage [82]. The UV-signature of the revertant patches in KID syndrome underscores their occurrence later in life and the need for a selective growth advantage. This finding, together with the frequent occurrence of revertant confetti-like spots in IWC and LK during childhood or adolescence that have a specific period of growth followed by stabilization at a maximal size [80,81,83,88,89,90,91] seems to support the “late-but-fitter revertant cell” hypothesis.
Table 1. Genetic correction mechanisms of revertant mosaicism reported in genodermatoses.
Table 1. Genetic correction mechanisms of revertant mosaicism reported in genodermatoses.
Disorder Corrected GeneCorrection
Mechanisms
Corrected Germline
Mutations
Skin LayerReferences
Subtype (MIM)InheritanceName (MIM)Major GroupSpecific TypeConsequence
RNA-Level
Consequence
Protein-Level
Epidermolysis bullosa
EBS, severe
(131760)
ADKRT14 (148066)Second-site mutationNucleotide insertion (1 Nt) Disruption of reading frame
Silencing dominant negative allele
Loss of expression of mutant proteinc.373C>T; p.Arg125CysEKCs[76]
EBS, recessive
(601001)
ARKRT14
(148066)
UnknownUnknownSplice-modulating Generation of in-frame splice variant Introduction of protein lacking 2 AA and carrying
1 missense AA. Non-functional.
c.526-2A>C (SS)EKCs[77]
JEB, intermediate
(226650)
ARCOL17A1 (113811)Gene conversionN/ALoss of heterozygosity for one mutant alleleIntroduction of full-length protein, wildtypec.1601delA; p.Asp534fsEKCs[26,75]
Back-mutation/
mitotic recombination
Nucleotide changeNonsense to wildtype reversionIntroduction of full-length protein, wildtypec.3676C>T; p.Arg1226*EKCs[75]
Second-site mutationNucleotide changeNonsense to missense changeIntroduction of full-length protein carrying 1 missense AAc.3676C>T; p.Arg1226*EKCs[75]
Splice-modulating Restoration of reading frameIntroduction of full-length protein with 13 incorrect AAc.4319dup; p.Gly1441fsEKCs[75]
In-frame skipping of mutant exonIntroduction of shorter protein lacking AA of mutant exonc.2237del; p.Gly746fsEKCs[73,79]
c.3487G>T; p.Glu1163*EKCs[66]
Nucleotide insertion (2 Nt)Restoration of reading frameIntroduction of full-length protein with 25 incorrect AAc.3899_3900del; p.Ser1300fsEKCs[78]
Intragenic genomic deletionIn-frame skipping of mutant exonIntroduction of shorter protein lacking AA of mutant exonc.2237del; p.Gly746fsEKCs[73,79]
In-frame deletion of multiple exonsIntroduction of shorter protein lacking AA of mutant exonc.2237del; p.Gly746fsEKCs[73,79]
Intragenic genomic deletion and insertionIn-frame deletion of mutant exonIntroduction of shorter protein lacking AA of mutant exonc.2237del; p.Gly746fsEKCs[73,79]
LAMB3
(150310)
Second-site mutationNucleotide changeSplice-modulating. Increase in wild-type splicing patternIncreased expression of full-length protein carrying 1 missense mutationc.628G>A; p.Glu210LysEKCs[74]
Splice modulating. Introduction of new, in-frame splice variantIntroduction of protein elongated by 22 AAc.628G>A; p.Glu210LysEKCs[74]
Splice modulating. Increased expression of alternative in-frame splice variantIncreased expression of protein lacking 22 AAc.628G>A; p.Glu210LysEKCs[74]
RDEB, severe
(226600)
ARCOL7A1
(120120)
Intragenic cross-over (mitotic recombination)N/ALoss of heterozygosity of one mutant alleleIntroduction of full-length protein, wildtypec.7786del; p.Gly2596fsEKCs[72]
Second-site mutationNucleotide deletion (1 bp)Restoration of reading frameIntroduction of full-length protein, wildtypec.6527dup; p.Gly2177fsEKCs[71]
Nucleotide changeSplice-modulating. Restoration of splicing pattern towards wild-typeIncreased expression of full-length protein, wildtypec.2142A>G (SS)EKCs[86]
c.425A>G (SS)EKCs[86]
Back mutation/mitotic recombinationN/ALoss of heterozygosity of one mutant alleleIntroduction of full-length protein, wildtypec.884del; p.Gly296fsEKCs[86]
Introduction of full-length protein, wildtype, loss of expression of mutant proteinc.6176A>G; p.Glu2059GlyEKCs[86]
Mitotic recombinationN/ABoth variants on one alleleIntroduction of full-length protein, wildtype425A>G (SS)EKCs[86]
c.1837C>T; p.Arg613*EKCs[86]
RDEB, intermediate
(226600)
ARCOL7A1
(120120)
Mitotic recombinationN/ALoss of heterozygosity of one mutant alleleIntroduction of full-length protein, wildtypec.425A>GEKCs[86]
Second-site mutationNucleotide changeNonsense to missense changeIntroduction of full-length protein with 1 missense AAc.6508C>T; p.Gln2170*EKCs[68]
Intragenic cross-over (mitotic recombination)N/ALoss of heterozygosity of one mutant alleleIntroduction of full-length protein, wildtype, loss of expression of mutant proteinc.6091G>A; p.Gly2031SerDFBs[65]
Both variants on one alleleIntroduction of full-length protein, wildtype, loss of expression of mutant proteinc.5932C>T; p.Arg1978*/
c.8029G>A; p.Gly2677Ser
EKCs[64]
DDEB
(131750)
ADCOL7A1
(120120)
Back mutation/mitotic recombinationNucleotide changeLoss of heterozygosity of mutant allele Loss of expression of mutant proteinc.6127C>A; p.Gly2043ArgEKCs[86]
Kindler EB
(173650)
ARFERMT1
(607900)
Slipped mispairingNucleotide deletion (1 bp)Loss of one mutant alleleIntroduction of full-length protein, wildtypec.676dup; p.Gln226fsEKCs[69,70]
c.456dup; p.Asp153fsEKCs[69,70]
Disorders of cornification
Ichthyosis with confetti type 1, KRT10
(609165)
ADKRT10
(148080)
Mitotic recombinationN/ALoss of heterozygosity of mutant alleleLoss of expression of mutant proteinc.1369G>T (SS)EKCs[81]
c.1373del; p.Ser458fsEKCs[90]
c.1373+1G>A (SS)EKCs[81]
c.1374-2del (SS)EKCs[81]
c.1374-2A>G (SS)EKCs[81,88]
c.1374-1G>A (SS)EKCs[81]
c.1449dup; p.Gly484fsEKCs[81]
c.1546_1551delinsT; p.Gly516fsEKCs[91]
c.1560_1561del;
p.Gly521fs
EKCs[81]
Ichthyosis with confetti type 2, KRT1
(609165)
ADKRT1
(139350)
Mitotic recombinationN/ALoss of heterozygosity of mutant alleleLoss of expression of mutant proteinc.1865dup; p.Val623fsEKCs[80]
c.1759dup; p.Tyr587fs [87]
c.591+332_1129-34del; p.197_375del [89]
Loricrin keratoderma
(604117)
ADLOR
(152445)
Mitotic recombinationN/ALoss of heterozygosity of mutant alleleLoss of expression of mutant proteinc.545dup; p.Gly183fsEKCs[83]
Keratitis-ichthyosis-deafness syndrome
(148210)
ADGJB2
(121011)
Second-site mutationNucleotide changeSilencing dominant negative allele Loss of expression of mutant proteinc.148G>A, p.Asp50AsnEKCs[82]
Pityriasis rubra pilaris
(173200)
ADCARD14
(607211)
Mitotic recombinationN/ALoss of heterozygosity of mutant alleleLoss of expression of mutant proteinc.356T>C; p.Met119ThrEKCs[84]
c.407A>T; p.Gln136LeuEKCs[84]
MIM, online Mendelian Inheritance in Man ID (www.omim.org, accessed on 30 June 2022); EB, epidermolysis bullosa; EBS, epidermolysis bullosa simplex; JEB, junctional epidermolysis bullosa; DEB, dystrophic epidermolysis bullosa; RDEB, recessive DEB; DDEB, dominant DEB; AD, autosomal dominant; AR, autosomal recessive; EKCs, epidermal keratinocytes; DFBs, dermal fibroblasts; SS, splice-site; Nt, nucleotide; AA, amino acid.

5. Molecular Mechanisms of Revertant Mosaicism in Genodermatoses

Over the years, multiple differing mechanisms of somatic correction events have been uncovered (Table 1, Figure 3). The first JEB patient described with RM had a non-affected skin patch in which one of the pathogenic COL17A1 variants was lost due to a gene conversion [26]. This led to loss of heterozygosity for the COL17A1 region where one of the pathogenic variants resided and, consequently, the loss of one mutated allele with the restoration of type XVII collagen production and a non-affected skin phenotype.
In subsequent years, various reversion mechanisms have been uncovered for mutations in EB-related genes. Of the different reversion mechanisms described in EB, most involve correction through a somatic, small intragenic mutational event (referred to as second-site mutation) such as somatic point mutations in a germline nonsense codon that revert it to the wildtype or to a different but functional codon [68,75], or alter the splicing of a mutated exon [66,73,74,75,77,79,86]. A special class of additional point mutations is ‘back-mutation’ in which the somatic event reverts the mutant nucleotide to wildtype. Of note, in the cases with presumed back-mutations, it was not always possible to distinguish a back-mutation from a mitotic recombination event [75,86]. Other somatic, small intragenic mutational events that have been reported are deletions or duplications of one or a few nucleotides that either directly restore a disturbed mutant reading frame or modify mutant splicing patterns and thereby restore mutant reading frames [69,70,71,73,78,79]. Such correction events were observed frequently in Kindler EB as certain repetitive nucleotide sequences in FERMT1 appear to be particularly prone to the duplication or deletion of single nucleotides due to slipped mispairing [69,70]. One revertant nucleotide insertion exerted its corrective effect by disrupting rather than restoring the reading frame [76]. In this case, a nucleotide insertion had occurred on the same allele that carried the dominant germline KRT14 point mutation, which disrupted the reading frame, thereby silencing the expression of the mutant allele and its dominant-negative effect. A similar phenomenon was observed in the revertant patches of a patient with KID syndrome [82]: five different somatic variants in cis with the germline point mutation were found to inhibit the dominant-negative effect of the germline mutations.
Another class of reversion mechanisms involves chromosomal recombination events during mitosis, i.e., gene conversion, intragenic cross-overs, and mitotic recombination. Only a few correction mechanisms identified in EB involve such recombination events [26,64,65,70,72,82,86], probably because most EB patients in which RM has been identified have biallelic gene mutations where recombination events are less likely to create cells without mutations. In contrast, mitotic recombination is the only reversion mechanism so far identified in the autosomal dominant disorders IWC-I, IWC-II, LK, and PRP [80,81,83,84,88,89,90,91]. KID-syndrome is the only autosomal dominant genodermatosis in which a reversion mechanism other than mitotic recombination has been reported (second-site mutation) [82].
The question remains whether there are biological processes that drive the occurrence of RM in genodermatoses. For EB, it is still mostly considered a stochastic process. In contrast, several RM-driving mechanisms are being uncovered for the autosomal dominant genodermatoses. IWC-I, IWC-II, and LK are caused by frameshift mutations in the carboxyl-terminal ends of the KRT10, KRT1, and LOR genes, respectively [80,81,92,93]. These frameshift mutations create arginine-rich tails that subsequently act as nuclear localization signals, causing the mutant proteins to aberrantly mis-localize to the nuclei instead of the cytoplasm and homing to the intermediate filaments or cornified envelope [83,94,95]. The role of this nuclear mis-localization in the pathogenesis of the disturbed keratinocyte differentiation seen in IWC-I, IWC-II, and LK has not been fully elucidated, but it has been suggested that it causes an increased mitotic recombination rate that drives the occurrence of RM [81]. This would explain the high frequency of revertant confetti-like skin patches in patients with these disorders. This hypothesis seems to be supported by the observation of many fewer revertant spots than expected in a patient with a typical IWC-I KRT10 frameshift mutation, who also carried a heterozygous non-pathogenic pericentric inversion at chromosome 17 (inv(17)(p13q12)) that dramatically reduced the options for therapeutic mitotic recombination events in the KRT10 chromosomal region (17q21.2) [88]. In PRP, it was shown that mutant CARD14 protein alters the replication stress response in mutant keratinocytes, which preferentially drives break-induced replication as a DNA-repair pathway, resulting in an increased frequency of mitotic recombinations and, consequently, revertant skin spots [84].

6. Clinical Recognition of Revertant Mosaicism in Genodermatoses

In genodermatoses, RM appears as non-affected skin patches standing out amidst affected skin (Figure 2). Revertant, non-affected patches are therefore best visible in generalized phenotypes that involve the entire skin, like recessive types of EB or IWC [68,74,75,80,81]. In addition, confirming RM at the tissue level is most reliable in recessive disorders in which the germline mutations cause the complete absence of a structural protein [96]. In such diseases, the re-expression of the structural protein in the revertant patches can be unequivocally demonstrated using immunofluorescence or immunohistochemical staining assays on skin biopsy sections. It can be much more challenging to demonstrate RM in autosomal dominant genodermatoses like dominant DEB (DDEB), in which the expression of the protein involved is normal or only reduced to some extent [86]. In other autosomal dominant conditions, differences in staining patterns between mutant and revertant skin or cultured keratinocytes, rather than differences in expression levels, can help identify RM. For instance, in a case of severe autosomal dominant EB simplex (EBS-severe), RM only became apparent incidentally because a proportion of cultured keratinocytes from an unaffected skin area showed a normal keratin filament network when only mutant cells with filament aggregates were expected [76]. In the autosomal dominant disorders IWC-I, IWC-II, and LK, the mutant keratin 10, keratin 1, and loricrin proteins localize to the nuclei, whereas the revertant proteins integrate normally into the cytoplasmic filament network or cornified envelope [80,81,83].
In addition, patient age plays an important role in the clinical detection of RM. In young children, the skin is usually not yet as much affected, meaning it is difficult to clinically recognize revertant patches if they are present. In IWC, many revertant patches occur and grow with age, which explains why they are easier to recognize with ageing. In EB, however, the gradual skin changes due to cumulative damage with ageing make it easier to recognize revertant patches as patients grow older. Interestingly, we have also studied several clearly unaffected skin patches in EB patients with severe and generalized subtypes later in their lives. We expected these patches to be revertant, but they turned out not to be using either the ‘skin rub test’ or molecular analysis. The skin rub test, which examines the resistance of the skin against mechanical stress by rubbing with the tip of a pen, can discriminate revertant from affected skin (See www.youtube.com/watch?v=fz8nW3z51Gw, accessed 1 June 2022, video courtesy of the late Prof. M.F. Jonkman). However, in patients with milder forms of EB, mutant skin can also be resistant to direct mechanical stress, which makes recognition of revertant patches is even more challenging and sometimes impossible.
The revertant areas do vary in size and shape between the different genodermatoses (Figure 2). For instance, in IWC, the confetti-like white, greenish, or brown revertant areas stand out against the red affected skin [93]. In IWC-I, they appear in childhood and over time increase in number to hundreds and in size from 2–10 mm up to 4 cm [81,88,90,91,93]. In IWC-II, the revertant patches usually do not become apparent until early adulthood and remain smaller in size, 4–15 mm [80,87,89]. The size of the spots is limited by the expansion of the single revertant clone, but they can increase until later in life. This is exemplified by a case reported by Suzuki et al. in which a 56-year-old patient with IWC-II initially presented with non-affected white spots of 1–2 mm that increased up to 15 mm 8 years later [87]. In LK, patients with RM have dozens of well-demarcated, whitish, round, or oval-shaped, non-affected skin patches of up to 10 mm in diameter [83]. In PRP, numerous non-affected skin patches were identified that were up to several centimeters in size [84].
In contrast to IWC, LK, and PRP, the revertant areas in EB are usually larger and irregular in shape. In addition, changes in size and shape during adulthood have not been witnessed in patients with intermediate JEB due to COL17A1 mutations [79]. The exact size of the revertant area that originates from a single clone is not clear, as different revertant populations of keratinocytes, each originating from a different reversion event, have sometimes been found in a single revertant area. In intermediate JEB, the patches are especially recognizable after sun exposure because of differences in pigmentation, as type XVII collagen is also involved in melanocyte supply to the epidermis [97]. In the affected areas there is a decreased melanocyte density compared to the revertant areas. In addition, the skin texture of the revertant skin patches was shown to be smooth when compared to the coarse affected skin in a COL17A1-associated JEB-intermediate patient [63]. Only two patients with intermediate JEB due to LAMB3 mutations and RM have been described in the literature, and both had multiple revertant areas with at least one patch carrying two separate reversion events [74]. One of the patients claimed that the revertant area on his lower leg healed to clinically unaffected skin later in life, but there was no photo material available to confirm this. The revertant areas in RDEB are usually more difficult to recognize than those in patients with intermediate JEB, especially compared to patients with mutations in COL17A1. The revertant areas in DEB can vary in size from 3 × 3 cm to 10 × 5 cm, and their pigmentation is usually normal [65,68,71,72,86]. Lastly, in Kindler EB, the revertant areas are numerous and smaller compared to the other major EB types, varying from 0.5 cm2 to 3 cm2 [48,69,70]. Larger areas up to 15 cm2 in size were noted in some patients, but it could not be excluded that these originated from overlapping areas arising from separate reversion events. In one of the patients, the revertant area did not change in size over a 3-year period. One possible explanation for the high number of revertant areas is that these cells have a significant growth advantage compared to their affected counterparts, as mutant kindlin-1 severely impacts keratinocyte proliferation [70].

7. Revertant Mosaicism as Treatment for Genodermatoses

Currently, no truly curative gene-therapy treatments for genodermatoses have been approved, and treatment is usually symptomatic. Nonetheless, there has been much progress with disease-modifying treatments, some of which have led to dramatic improvements in the severity of certain genodermatoses [98,99,100,101,102,103,104,105]. The holy grail for curing genodermatoses is likely a genome-editing method capable of effectively and safely correcting the causative gene variants in the entire body. However, genome-editing poses several challenges including off-target editing effects, the need to target enormous numbers of stem cells on many different body locations, and the difficulty of reaching the genomic DNA in all these cells, which requires vehicle methods that might carry their own risks [106]. In addition, there are ethical issues around genome-editing [107]. Hence, although the first clinical studies have commenced [108], these issues may prevent genome-editing from entering the clinic as a routine systemic treatment for genodermatoses in the short run.
Most approaches under study for EB therefore focus on topical treatment [109], either by topical compound delivery (e.g., in vivo gene-addition by viral [110] or minicircle non-viral vectors [111], antisense oligonucleotides [112]), ex vivo gene-replacement or gene-correction followed by injection of corrected fibroblasts [113], or the transplantation of large epidermal sheets cultured from ex vivo-corrected keratinocytes [114,115,116,117], analogous to the treatment of full-thickness burn-wounds [118]. In 2017, Hirsch et al. reported a 7-year-old boy with laminin 332-deficient JEB in whom almost the entire epidermis (~0.85 m2) was replaced using exogenously corrected, autologous skin grafts [119,120]. Keratinocyte stem cells obtained from a 4 cm2 skin biopsy were first transduced with a retroviral vector expressing the full-length LAMB3 cDNA, then cultured into large transgenic epidermal grafts that were transplanted onto properly prepared dermal wound beds.
It is in this context that it is especially interesting to look more closely at the phenomenon of RM. In RM, nature has successfully achieved what science so eagerly strives for: the complete cure of a genodermatosis in a specific skin area by safely repairing the effect of the germline mutation(s). Therefore, ever since the first discovery of RM in EB in 1997, our group has been looking for ways to acquire revertant skin cells from revertant patches and to expand them and use them to treat affected skin areas. Using naturally corrected, patient-own skin cells would have three major advantages: (i) no need for potentially dangerous gene-correction technologies, (ii) the potential for permanent correction, and (iii) no risk of immuno-rejection of revertant skin transplants or grafts. Our group showed a proof-of-concept for the therapeutic potential of revertant keratinocytes in 2014. Seventy-three 3 mm punch biopsies from a revertant, laminin 332-positive skin patch were transplanted as mini-grafts onto seven persistent ulcers of a patient with laminin 332-deficient JEB [121]. In 18-month follow-up, no blisters were observed and laminin 332-expression was increased in the treated areas, while the revertant donor site also healed with revertant keratinocytes, indicating that the revertant skin cells were able to settle and expand in vivo. Despite these promising results, taking so many biopsies from the revertant skin areas can still only treat relatively small affected areas. Following the full-body transplantation method introduced by Hirsch et al. [120], an abstract to the 2021 Society of Investigative Dermatology Meeting reported the replacement of ~80% of the epidermis of an RDEB patient by epidermal autografts generated from cultured revertant keratinocytes obtained from a revertant skin patch [122]. This provides evidence that revertant keratinocyte stem cells have sufficient proliferative potential to regenerate large areas of epidermis in vitro. Although undoubtedly highly beneficial to the respective patients, the question remains how sustainable this invasive procedure would be in routine practice, as it requires repeated and multiple operations usually under general anesthesia [120]. In addition, the durability of the transplanted revertant skin is another unknown factor.
Induced pluripotent stem cells (iPSCs) have been proposed as a therapeutic route for genodermatoses and have been generated from patients with EBS [123], JEB [124], and RDEB [123,125], among others. When revertant cells are used to create iPSCs, genome-editing to correct the mutations is not required, which is a major advantage. Two groups have reported successful generation of iPSCs from revertant keratinocytes. Umegaki-Arao et al. generated iPSCs from revertant keratinocytes from a patient with JEB due to COL17A1 mutations [126]. The revertant iPSCs were trans-differentiated into keratinocytes that expressed type XVII collagen. The authors showed that they were able to generate three-dimensional epidermal equivalents in vitro and could reconstitute human epidermis from these iPSC-derived keratinocytes in vivo in mice, and that type XVII collagen localized to the basement membrane zone. Tolar et al. generated iPSCs from revertant keratinocytes of a patient with RDEB due to COL7A1 mutations [67] that they were able to trans-differentiate into epidermal and hematopoietic cell populations. These studies showed that revertant keratinocytes can be a viable source for developing iPSC-based therapeutic approaches in genodermatoses. iPSC-derived revertant keratinocytes could serve as an unlimited source of cells for the generation of autografts for transplantation, while iPSC-derived revertant hematopoietic cells could be envisioned in combination with bone marrow transplantation strategies [127]. Whether iPSC-derived revertant keratinocytes possess the growth potential to be cultured into large grafts to treat larger skin areas remains to be seen. While the use of revertant cells in such approaches may circumvent some of the limitations of genome-editing methods, the safety of iPSC-based treatments has been debated. Obstacles that were mentioned for clinical use were carcinogenicity, a lack of in situ integration, genomic instability, and/or immunologic rejection [128]. Progress is, however, being made in the stem cell research community to produce safe methods for the generation and expansion of human pluripotent stem cells. Kim et al. showed that in 2022, there were 19 ongoing therapeutic clinical trials with iPSC strategies, most of them being conducted in Japan (10 trials), but none were yet focused on skin diseases [129]. In addition, the iPSCs by Umegaki-Arao et al. [126] and Tolar et al. [67] were generated using retroviral vectors and therefore cannot be used in a clinical situation, as this strategy carries a risk of uncontrolled genomic integration [130]. Figure 4 summarizes the therapeutic approaches using revertant skin cells that have already been performed and the ones that may be pursued in the future.

8. Concluding Remarks and Future Perspectives

RM is a form of natural gene therapy that occurs more often than previously thought in the skin of patients with genodermatoses. Multiple genetic mechanisms have been found to underlie the reversions to ‘normal’ phenotypes, the nature of which depends on the mode of inheritance and the type of germline mutation. As revertant patches may provide a source for future revertant cell therapy, it is becoming increasingly important to identify these revertant patches on a patient’s body. Clinicians should therefore be on the lookout for patches of skin that are not affected by the phenotype of the respective genodermatosis. The great advantage of using revertant skin cells in therapeutic approaches is that these autologous cells do not require additional genetic modifications. Several in vitro and even in vivo pilot studies have already indicated that revertant cells may provide a source for therapy for the severe genetic skin blistering disease EB. However, several crucial questions need to be addressed before revertant skin cells can be used as an efficient and safe therapy in patients. How can we identify revertant patches in vivo in young patients, where affected skin is much more difficult to differentiate from revertant skin? How can we efficiently identify and isolate revertant stem cells from a revertant skin patch? What are the characteristics of these revertant stem cells? Additionally, do they differ from mutant stem cells from the same patients and healthy controls? Finally, how can we introduce these revertant stem cells back into the patients in the most efficient and least burdensome way? These will be topics for future studies that will hopefully lead to RM as a therapeutic avenue for patients with rare, currently incurable genodermatoses.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/biomedicines10092118/s1, Supplementary Table S1: Genetic conditions in which revertant mosaicism has been reported in tissues other than skin.

Author Contributions

All authors contributed equally to the conceptualization, data acquisition and literature studies, and writing—original draft preparation and editing of this manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank our collaborators who kindly provided high resolution images for Figure 2. We thank Kate McIntyre for carefully editing this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Has, C.; Bauer, J.W.; Bodemer, C.; Bolling, M.C.; Bruckner-Tuderman, L.; Diem, A.; Fine, J.D.; Heagerty, A.; Hovnanian, A.; Marinkovich, M.P.; et al. Consensus reclassification of inherited epidermolysis bullosa and other disorders with skin fragility. Br. J. Dermatol. 2020, 183, 614–627. [Google Scholar] [CrossRef] [PubMed]
  2. Hobbs, R.P.; Han, S.Y.; van der Zwaag, P.A.; Bolling, M.C.; Jongbloed, J.D.; Jonkman, M.F.; Getsios, S.; Paller, A.S.; Green, K.J. Insights from a desmoplakin mutation identified in lethal acantholytic epidermolysis bullosa. J. Investig. Dermatol. 2010, 130, 2680–2683. [Google Scholar] [CrossRef] [PubMed]
  3. Jonkman, M.F.; Pasmooij, A.M.; Pasmans, S.G.; van den Berg, M.P.; Ter Horst, H.J.; Timmer, A.; Pas, H.H. Loss of desmoplakin tail causes lethal acantholytic epidermolysis bullosa. Am. J. Hum. Genet. 2005, 77, 653–660. [Google Scholar] [CrossRef]
  4. Kelsell, D.P.; Norgett, E.E.; Unsworth, H.; Teh, M.T.; Cullup, T.; Mein, C.A.; Dopping-Hepenstal, P.J.; Dale, B.A.; Tadini, G.; Fleckman, P.; et al. Mutations in ABCA12 underlie the severe congenital skin disease harlequin ichthyosis. Am. J. Hum. Genet. 2005, 76, 794–803. [Google Scholar] [CrossRef]
  5. Akiyama, M.; Sugiyama-Nakagiri, Y.; Sakai, K.; McMillan, J.R.; Goto, M.; Arita, K.; Tsuji-Abe, Y.; Tabata, N.; Matsuoka, K.; Sasaki, R.; et al. Mutations in lipid transporter ABCA12 in harlequin ichthyosis and functional recovery by corrective gene transfer. J. Clin. Investig. 2005, 115, 1777–1784. [Google Scholar] [CrossRef] [PubMed]
  6. Moulson, C.L.; Go, G.; Gardner, J.M.; van der Wal, A.C.; Smitt, J.H.; van Hagen, J.M.; Miner, J.H. Homozygous and compound heterozygous mutations in ZMPSTE24 cause the laminopathy restrictive dermopathy. J. Investig. Dermatol. 2005, 125, 913–919. [Google Scholar] [CrossRef]
  7. Navarro, C.L.; De Sandre-Giovannoli, A.; Bernard, R.; Boccaccio, I.; Boyer, A.; Geneviève, D.; Hadj-Rabia, S.; Gaudy-Marqueste, C.; Smitt, H.S.; Vabres, P.; et al. Lamin A and ZMPSTE24 (FACE-1) defects cause nuclear disorganization and identify restrictive dermopathy as a lethal neonatal laminopathy. Hum. Mol. Genet. 2004, 13, 2493–2503. [Google Scholar] [CrossRef]
  8. Bohring, A.; Stamm, T.; Spaich, C.; Haase, C.; Spree, K.; Hehr, U.; Hoffmann, M.; Ledig, S.; Sel, S.; Wieacker, P.; et al. WNT10A mutations are a frequent cause of a broad spectrum of ectodermal dysplasias with sex-biased manifestation pattern in heterozygotes. Am. J. Hum. Genet. 2009, 85, 97–105. [Google Scholar] [CrossRef]
  9. Dharma, B.; Moss, C.; McGrath, J.A.; Mellerio, J.E.; Ilchyshyn, A. Dominant dystrophic epidermolysis bullosa presenting as familial nail dystrophy. Clin. Exp. Dermatol. 2001, 26, 93–96. [Google Scholar]
  10. Akiyama, M. ABCA12 mutations and autosomal recessive congenital ichthyosis: A review of genotype/phenotype correlations and of pathogenetic concepts. Hum. Mutat. 2010, 31, 1090–1096. [Google Scholar] [CrossRef]
  11. van den Akker, P.C.; van Essen, A.J.; Kraak, M.M.; Meijer, R.; Nijenhuis, M.; Meijer, G.; Hofstra, R.M.; Pas, H.H.; Scheffer, H.; Jonkman, M.F. Long-term follow-up of patients with recessive dystrophic epidermolysis bullosa in the Netherlands: Expansion of the mutation database and unusual phenotype-genotype correlations. J. Dermatol. Sci. 2009, 56, 9–18. [Google Scholar] [CrossRef]
  12. Oji, V.; Preil, M.L.; Kleinow, B.; Wehr, G.; Fischer, J.; Hennies, H.C.; Hausser, I.; Breitkreutz, D.; Aufenvenne, K.; Stieler, K.; et al. S1 guidelines for the diagnosis and treatment of ichthyoses-update. J. Dtsch. Dermatol. Ges. 2017, 15, 1053–1065. [Google Scholar] [CrossRef] [PubMed]
  13. Happle, R. Mosaicism in Human Skin: Understanding Nevi, Nevoid Skin Disorders, and Cutaneous Neoplasia, 1st ed.; SpringerLink; Imprint; Springer: Berlin/Heidelberg, Germany, 2014. [Google Scholar]
  14. Cheraghlou, S.; Lim, Y.; Choate, K.A. Mosaicism in genodermatoses. Clin. Dermatol. 2020, 38, 408–420. [Google Scholar] [CrossRef]
  15. Martínez-Glez, V.; Tenorio, J.; Nevado, J.; Gordo, G.; Rodríguez-Laguna, L.; Feito, M.; de Lucas, R.; Pérez-Jurado, L.A.; Ruiz Pérez, V.L.; Torrelo, A.; et al. A six-attribute classification of genetic mosaicism. Genet. Med. 2020, 22, 1743–1757. [Google Scholar] [CrossRef] [PubMed]
  16. Jafry, M.; Sidbury, R. RASopathies. Clin. Dermatol. 2020, 38, 455–461. [Google Scholar] [CrossRef]
  17. Ko, Y.; Lee, C.; Lee, H.; Lee, M.; Lee, J.S. Clinical application of next-generation sequencing for the diagnosis of segmental neurofibromatosis. J. Dermatol. Sci. 2017, 88, 370–372. [Google Scholar] [CrossRef]
  18. Weinstein, L.S.; Shenker, A.; Gejman, P.V.; Merino, M.J.; Friedman, E.; Spiegel, A.M. Activating mutations of the stimulatory G protein in the McCune-Albright syndrome. N. Engl. J. Med. 1991, 325, 1688–1695. [Google Scholar] [CrossRef] [PubMed]
  19. Mirzaa, G.; Timms, A.E.; Conti, V.; Boyle, E.A.; Girisha, K.M.; Martin, B.; Kircher, M.; Olds, C.; Juusola, J.; Collins, S.; et al. PIK3CA-associated developmental disorders exhibit distinct classes of mutations with variable expression and tissue distribution. JCI Insight. 2016, 1, e87623. [Google Scholar] [CrossRef]
  20. Kurek, K.C.; Luks, V.L.; Ayturk, U.M.; Alomari, A.I.; Fishman, S.J.; Spencer, S.A.; Mulliken, J.B.; Bowen, M.E.; Yamamoto, G.L.; Kozakewich, H.P.; et al. Somatic mosaic activating mutations in PIK3CA cause CLOVES syndrome. Am. J. Hum. Genet. 2012, 90, 1108–1115. [Google Scholar] [CrossRef] [PubMed]
  21. van Leersum, F.S.; Seyger, M.M.B.; Theunissen, T.E.J.; Bongers, E.; Steijlen, P.M.; van Geel, M. Recessive mosaicism in ABCA12 causes blaschkoid congenital ichthyosiform erythroderma. Br. J. Dermatol. 2020, 182, 208–211. [Google Scholar] [CrossRef]
  22. Happle, R. The concept of type 2 segmental mosaicism, expanding from dermatology to general medicine. J. Eur. Acad. Dermatol. Venereol. 2018, 32, 1075–1088. [Google Scholar] [CrossRef] [PubMed]
  23. Torrelo, A.; Hernández-Martín, A.; Bueno, E.; Colmenero, I.; Rivera, I.; Requena, L.; Happle, R.; González-Sarmiento, R. Molecular evidence of type 2 mosaicism in Gorlin syndrome. Br. J. Dermatol. 2013, 169, 1342–1345. [Google Scholar] [CrossRef] [PubMed]
  24. Fölster-Holst, R.; Nellen, R.G.; Jensen, J.M.; Poblete-Gutiérrez, P.; Steijlen, P.M.; Schwarz, T.; Happle, R.; Van Geel, M.; Frank, J. Molecular genetic support for the rule of dichotomy in type 2 segmental Darier disease. Br. J. Dermatol. 2012, 166, 464–466. [Google Scholar] [CrossRef]
  25. Jonkman, M.F.; de Jong, M.C.; Heeres, K.; Pas, H.H.; van der Meer, J.B.; Owaribe, K.; Martinez de Velasco, A.M.; Niessen, C.M.; Sonnenberg, A. 180-kD bullous pemphigoid antigen (BP180) is deficient in generalized atrophic benign epidermolysis bullosa. J. Clin. Investig. 1995, 95, 1345–1352. [Google Scholar] [CrossRef]
  26. Jonkman, M.F.; Scheffer, H.; Stulp, R.; Pas, H.H.; Nijenhuis, M.; Heeres, K.; Owaribe, K.; Pulkkinen, L.; Uitto, J. Revertant mosaicism in epidermolysis bullosa caused by mitotic gene conversion. Cell 1997, 88, 543–551. [Google Scholar] [CrossRef]
  27. Tan, S.; Kermasson, L.; Hilcenko, C.; Kargas, V.; Traynor, D.; Boukerrou, A.Z.; Escudero-Urquijo, N.; Faille, A.; Bertrand, A.; Rossmann, M.; et al. Somatic genetic rescue of a germline ribosome assembly defect. Nat. Commun. 2021, 12, 5044. [Google Scholar] [CrossRef]
  28. Hoshino, A.; Yang, X.; Tanita, K.; Yoshida, K.; Ono, T.; Nishida, N.; Okuno, Y.; Kanzaki, T.; Goi, K.; Fujino, H.; et al. Modification of cellular and humoral immunity by somatically reverted T cells in X-linked lymphoproliferative syndrome type 1. J. Allergy Clin. Immunol. 2019, 143, 421–424.e411. [Google Scholar] [CrossRef]
  29. Garelli, E.; Quarello, P.; Giorgio, E.; Carando, A.; Menegatti, E.; Mancini, C.; Di Gregorio, E.; Crescenzio, N.; Palumbo, O.; Carella, M.; et al. Spontaneous remission in a Diamond-Blackfan anaemia patient due to a revertant uniparental disomy ablating a de novo RPS19 mutation. Br. J. Haematol. 2019, 185, 994–998. [Google Scholar] [CrossRef]
  30. Csillag, B.; Ilencikova, D.; Meissl, M.; Webersinke, G.; Laccone, F.; Narumi, S.; Haas, O.; Duba, H.C. Somatic mosaic monosomy 7 and UPD7q in a child with MIRAGE syndrome caused by a novel SAMD9 mutation. Pediatr. Blood Cancer 2019, 66, e27589. [Google Scholar] [CrossRef]
  31. Brigida, I.; Zoccolillo, M.; Cicalese, M.P.; Pfajfer, L.; Barzaghi, F.; Scala, S.; Oleaga-Quintas, C.; Álvarez-Álvarez, J.A.; Sereni, L.; Giannelli, S.; et al. T-cell defects in patients with ARPC1B germline mutations account for combined immunodeficiency. Blood 2018, 132, 2362–2374. [Google Scholar] [CrossRef]
  32. Asur, R.S.; Kimble, D.C.; Lach, F.P.; Jung, M.; Donovan, F.X.; Kamat, A.; Noonan, R.J.; Thomas, J.W.; Park, M.; Chines, P.; et al. Somatic mosaicism of an intragenic FANCB duplication in both fibroblast and peripheral blood cells observed in a Fanconi anemia patient leads to milder phenotype. Mol. Genet. Genomic. Med. 2018, 6, 77–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Venugopal, P.; Moore, S.; Lawrence, D.M.; George, A.J.; Hannan, R.D.; Bray, S.C.; To, L.B.; D’Andrea, R.J.; Feng, J.; Tirimacco, A.; et al. Self-reverting mutations partially correct the blood phenotype in a Diamond Blackfan anemia patient. Haematologica 2017, 102, e506–e509. [Google Scholar] [CrossRef] [PubMed]
  34. Tesi, B.; Davidsson, J.; Voss, M.; Rahikkala, E.; Holmes, T.D.; Chiang, S.C.C.; Komulainen-Ebrahim, J.; Gorcenco, S.; Rundberg Nilsson, A.; Ripperger, T.; et al. Gain-of-function SAMD9L mutations cause a syndrome of cytopenia, immunodeficiency, MDS, and neurological symptoms. Blood 2017, 129, 2266–2279. [Google Scholar] [CrossRef]
  35. Marin, A.V.; Jiménez-Reinoso, A.; Briones, A.C.; Muñoz-Ruiz, M.; Aydogmus, C.; Pasick, L.J.; Couso, J.; Mazariegos, M.S.; Alvarez-Prado, A.F.; Blázquez-Moreno, A.; et al. Primary T-cell immunodeficiency with functional revertant somatic mosaicism in CD247. J. Allergy Clin. Immunol. 2017, 139, 347–349.e348. [Google Scholar] [CrossRef] [PubMed]
  36. Federico, C.; Dugo, K.; Bruno, F.; Longo, A.M.; Grillo, A.; Saccone, S. Somatic mosaicism with reversion to normality of a mutated transthyretin allele related to a familial amyloidotic polyneuropathy. Hum. Genet. 2017, 136, 867–873. [Google Scholar] [CrossRef] [PubMed]
  37. Chen, D.H.; Below, J.E.; Shimamura, A.; Keel, S.B.; Matsushita, M.; Wolff, J.; Sul, Y.; Bonkowski, E.; Castella, M.; Taniguchi, T.; et al. Ataxia-Pancytopenia Syndrome Is Caused by Missense Mutations in SAMD9L. Am. J. Hum. Genet. 2016, 98, 1146–1158. [Google Scholar] [CrossRef]
  38. McDermott, D.H.; Gao, J.L.; Liu, Q.; Siwicki, M.; Martens, C.; Jacobs, P.; Velez, D.; Yim, E.; Bryke, C.R.; Hsu, N.; et al. Chromothriptic cure of WHIM syndrome. Cell 2015, 160, 686–699. [Google Scholar] [CrossRef]
  39. Le Guen, T.; Touzot, F.; André-Schmutz, I.; Lagresle-Peyrou, C.; France, B.; Kermasson, L.; Lambert, N.; Picard, C.; Nitschke, P.; Carpentier, W.; et al. An in vivo genetic reversion highlights the crucial role of Myb-Like, SWIRM, and MPN domains 1 (MYSM1) in human hematopoiesis and lymphocyte differentiation. J. Allergy Clin. Immunol. 2015, 136, 1619–1626.e1615. [Google Scholar] [CrossRef]
  40. Fuchs, S.; Rensing-Ehl, A.; Pannicke, U.; Lorenz, M.R.; Fisch, P.; Jeelall, Y.; Rohr, J.; Speckmann, C.; Vraetz, T.; Farmand, S.; et al. Omenn syndrome associated with a functional reversion due to a somatic second-site mutation in CARD11 deficiency. Blood 2015, 126, 1658–1669. [Google Scholar] [CrossRef]
  41. Jing, H.; Zhang, Q.; Zhang, Y.; Hill, B.J.; Dove, C.G.; Gelfand, E.W.; Atkinson, T.P.; Uzel, G.; Matthews, H.F.; Mustillo, P.J.; et al. Somatic reversion in dedicator of cytokinesis 8 immunodeficiency modulates disease phenotype. J. Allergy Clin. Immunol. 2014, 133, 1667–1675. [Google Scholar] [CrossRef]
  42. Bayer, D.K.; Martinez, C.A.; Sorte, H.S.; Forbes, L.R.; Demmler-Harrison, G.J.; Hanson, I.C.; Pearson, N.M.; Noroski, L.M.; Zaki, S.R.; Bellini, W.J.; et al. Vaccine-associated varicella and rubella infections in severe combined immunodeficiency with isolated CD4 lymphocytopenia and mutations in IL7R detected by tandem whole exome sequencing and chromosomal microarray. Clin. Exp. Immunol. 2014, 178, 459–469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ban, S.A.; Salzer, E.; Eibl, M.M.; Linder, A.; Geier, C.B.; Santos-Valente, E.; Garncarz, W.; Lion, T.; Ott, R.; Seelbach, C.; et al. Combined immunodeficiency evolving into predominant CD4+ lymphopenia caused by somatic chimerism in JAK3. J. Clin. Immunol. 2014, 34, 941–953. [Google Scholar] [CrossRef] [PubMed]
  44. Jongmans, M.C.; Verwiel, E.T.; Heijdra, Y.; Vulliamy, T.; Kamping, E.J.; Hehir-Kwa, J.Y.; Bongers, E.M.; Pfundt, R.; van Emst, L.; van Leeuwen, F.N.; et al. Revertant somatic mosaicism by mitotic recombination in dyskeratosis congenita. Am. J. Hum. Genet. 2012, 90, 426–433. [Google Scholar] [CrossRef]
  45. Xia, B.; Dorsman, J.C.; Ameziane, N.; de Vries, Y.; Rooimans, M.A.; Sheng, Q.; Pals, G.; Errami, A.; Gluckman, E.; Llera, J.; et al. Fanconi anemia is associated with a defect in the BRCA2 partner PALB2. Nat. Genet. 2007, 39, 159–161. [Google Scholar] [CrossRef]
  46. Tone, Y.; Wada, T.; Shibata, F.; Toma, T.; Hashida, Y.; Kasahara, Y.; Koizumi, S.; Yachie, A. Somatic revertant mosaicism in a patient with leukocyte adhesion deficiency type 1. Blood 2007, 109, 1182–1184. [Google Scholar] [CrossRef] [PubMed]
  47. Dorsman, J.C.; Levitus, M.; Rockx, D.; Rooimans, M.A.; Oostra, A.B.; Haitjema, A.; Bakker, S.T.; Steltenpool, J.; Schuler, D.; Mohan, S.; et al. Identification of the Fanconi anemia complementation group I gene, FANCI. Cell Oncol. 2007, 29, 211–218. [Google Scholar] [CrossRef] [PubMed]
  48. Rieux-Laucat, F.; Hivroz, C.; Lim, A.; Mateo, V.; Pellier, I.; Selz, F.; Fischer, A.; Le Deist, F. Inherited and somatic CD3zeta mutations in a patient with T-cell deficiency. N. Engl. J. Med. 2006, 354, 1913–1921. [Google Scholar] [CrossRef] [PubMed]
  49. Wada, T.; Toma, T.; Okamoto, H.; Kasahara, Y.; Koizumi, S.; Agematsu, K.; Kimura, H.; Shimada, A.; Hayashi, Y.; Kato, M.; et al. Oligoclonal expansion of T lymphocytes with multiple second-site mutations leads to Omenn syndrome in a patient with RAG1-deficient severe combined immunodeficiency. Blood 2005, 106, 2099–2101. [Google Scholar] [CrossRef]
  50. Nishikomori, R.; Akutagawa, H.; Maruyama, K.; Nakata-Hizume, M.; Ohmori, K.; Mizuno, K.; Yachie, A.; Yasumi, T.; Kusunoki, T.; Heike, T.; et al. X-linked ectodermal dysplasia and immunodeficiency caused by reversion mosaicism of NEMO reveals a critical role for NEMO in human T-cell development and/or survival. Blood 2004, 103, 4565–4572. [Google Scholar] [CrossRef]
  51. Waisfisz, Q.; Morgan, N.V.; Savino, M.; de Winter, J.P.; van Berkel, C.G.; Hoatlin, M.E.; Ianzano, L.; Gibson, R.A.; Arwert, F.; Savoia, A.; et al. Spontaneous functional correction of homozygous fanconi anaemia alleles reveals novel mechanistic basis for reverse mosaicism. Nat. Genet. 1999, 22, 379–383. [Google Scholar] [CrossRef]
  52. Ariga, T.; Yamada, M.; Sakiyama, Y.; Tatsuzawa, O. A case of Wiskott-Aldrich syndrome with dual mutations in exon 10 of the WASP gene: An additional de novo one-base insertion, which restores frame shift due to an inherent one-base deletion, detected in the major population of the patient’s peripheral blood lymphocytes. Blood 1998, 92, 699–701. [Google Scholar] [PubMed]
  53. Lo Ten Foe, J.R.; Kwee, M.L.; Rooimans, M.A.; Oostra, A.B.; Veerman, A.J.; van Weel, M.; Pauli, R.M.; Shahidi, N.T.; Dokal, I.; Roberts, I.; et al. Somatic mosaicism in Fanconi anemia: Molecular basis and clinical significance. Eur. J. Hum. Genet. 1997, 5, 137–148. [Google Scholar] [CrossRef] [PubMed]
  54. Stephan, V.; Wahn, V.; Le Deist, F.; Dirksen, U.; Broker, B.; Müller-Fleckenstein, I.; Horneff, G.; Schroten, H.; Fischer, A.; de Saint Basile, G. Atypical X-linked severe combined immunodeficiency due to possible spontaneous reversion of the genetic defect in T cells. N. Engl. J. Med. 1996, 335, 1563–1567. [Google Scholar] [CrossRef] [PubMed]
  55. Liehr, T.; Rautenstrauss, B.; Grehl, H.; Bathke, K.D.; Ekici, A.; Rauch, A.; Rott, H.D. Mosaicism for the Charcot-Marie-Tooth disease type 1A duplication suggests somatic reversion. Hum. Genet. 1996, 98, 22–28. [Google Scholar] [CrossRef]
  56. Hirschhorn, R.; Yang, D.R.; Puck, J.M.; Huie, M.L.; Jiang, C.K.; Kurlandsky, L.E. Spontaneous in vivo reversion to normal of an inherited mutation in a patient with adenosine deaminase deficiency. Nat. Genet. 1996, 13, 290–295. [Google Scholar] [CrossRef]
  57. Ellis, N.A.; Lennon, D.J.; Proytcheva, M.; Alhadeff, B.; Henderson, E.E.; German, J. Somatic intragenic recombination within the mutated locus BLM can correct the high sister-chromatid exchange phenotype of Bloom syndrome cells. Am. J. Hum. Genet. 1995, 57, 1019–1027. [Google Scholar]
  58. Kvittingen, E.A.; Rootwelt, H.; Brandtzaeg, P.; Bergan, A.; Berger, R. Hereditary tyrosinemia type I. Self-induced correction of the fumarylacetoacetase defect. J. Clin. Investig. 1993, 91, 1816–1821. [Google Scholar] [CrossRef]
  59. Brunner, H.G.; Jansen, G.; Nillesen, W.; Nelen, M.R.; de Die, C.E.; Höweler, C.J.; van Oost, B.A.; Wieringa, B.; Ropers, H.H.; Smeets, H.J. Brief report: Reverse mutation in myotonic dystrophy. N. Engl. J. Med. 1993, 328, 476–480. [Google Scholar] [CrossRef]
  60. Burrow, K.L.; Coovert, D.D.; Klein, C.J.; Bulman, D.E.; Kissel, J.T.; Rammohan, K.W.; Burghes, A.H.; Mendell, J.R. Dystrophin expression and somatic reversion in prednisone-treated and untreated Duchenne dystrophy. CIDD Study Group. Neurology 1991, 41, 661–666. [Google Scholar] [CrossRef]
  61. Yang, T.P.; Stout, J.T.; Konecki, D.S.; Patel, P.I.; Alford, R.L.; Caskey, C.T. Spontaneous reversion of novel Lesch-Nyhan mutation by HPRT gene rearrangement. Somat. Cell Mol. Genet. 1988, 14, 293–303. [Google Scholar] [CrossRef]
  62. German, J.; Schonberg, S.; Louie, E.; Chaganti, R.S. Bloom’s syndrome. IV. Sister-chromatid exchanges in lymphocytes. Am. J. Hum. Genet. 1977, 29, 248–255. [Google Scholar]
  63. Wang, Y.; Kitahata, H.; Kosumi, H.; Watanabe, M.; Fujimura, Y.; Takashima, S.; Osada, S.I.; Hirose, T.; Nishie, W.; Nagayama, M.; et al. Collagen XVII deficiency alters epidermal patterning. Lab. Investig. 2022, 102, 581–588. [Google Scholar] [CrossRef] [PubMed]
  64. Natsuga, K.; Furuta, Y.; Takashima, S.; Nohara, T.; Kosumi, H.; Mai, Y.; Higashi, H.; Ujiie, H. Detection of revertant mosaicism in epidermolysis bullosa through Cas9-targeted long-read sequencing. Hum. Mutat. 2022, 43, 529–536. [Google Scholar] [CrossRef]
  65. Twaroski, K.; Eide, C.; Riddle, M.J.; Xia, L.; Lees, C.J.; Chen, W.; Mathews, W.; Keene, D.R.; McGrath, J.A.; Tolar, J. Revertant mosaic fibroblasts in recessive dystrophic epidermolysis bullosa. Br. J. Dermatol. 2019, 181, 1247–1253. [Google Scholar] [CrossRef] [PubMed]
  66. Kowalewski, C.; Bremer, J.; Gostynski, A.; Wertheim-Tysarowska, K.; Wozniak, K.; Bal, J.; Jonkman, M.F.; Pasmooij, A.M. Amelioration of junctional epidermolysis bullosa due to exon skipping. Br. J. Dermatol. 2016, 174, 1375–1379. [Google Scholar] [CrossRef] [PubMed]
  67. Tolar, J.; McGrath, J.A.; Xia, L.; Riddle, M.J.; Lees, C.J.; Eide, C.; Keene, D.R.; Liu, L.; Osborn, M.J.; Lund, T.C.; et al. Patient-specific naturally gene-reverted induced pluripotent stem cells in recessive dystrophic epidermolysis bullosa. J. Investig. Dermatol. 2014, 134, 1246–1254. [Google Scholar] [CrossRef]
  68. van den Akker, P.C.; Nijenhuis, M.; Meijer, G.; Hofstra, R.M.; Jonkman, M.F.; Pasmooij, A.M. Natural gene therapy in dystrophic epidermolysis bullosa. Arch. Dermatol. 2012, 148, 213–216. [Google Scholar] [CrossRef]
  69. Lai-Cheong, J.E.; Moss, C.; Parsons, M.; Almaani, N.; McGrath, J.A. Revertant mosaicism in Kindler syndrome. J. Investig. Dermatol. 2012, 132, 730–732. [Google Scholar] [CrossRef]
  70. Kiritsi, D.; He, Y.; Pasmooij, A.M.; Onder, M.; Happle, R.; Jonkman, M.F.; Bruckner-Tuderman, L.; Has, C. Revertant mosaicism in a human skin fragility disorder results from slipped mispairing and mitotic recombination. J. Clin. Investig. 2012, 122, 1742–1746. [Google Scholar] [CrossRef]
  71. Pasmooij, A.M.; Garcia, M.; Escamez, M.J.; Nijenhuis, A.M.; Azon, A.; Cuadrado-Corrales, N.; Jonkman, M.F.; Del Rio, M. Revertant mosaicism due to a second-site mutation in COL7A1 in a patient with recessive dystrophic epidermolysis bullosa. J. Investig. Dermatol. 2010, 130, 2407–2411. [Google Scholar] [CrossRef]
  72. Almaani, N.; Nagy, N.; Liu, L.; Dopping-Hepenstal, P.J.; Lai-Cheong, J.E.; Clements, S.E.; Techanukul, T.; Tanaka, A.; Mellerio, J.E.; McGrath, J.A. Revertant mosaicism in recessive dystrophic epidermolysis bullosa. J. Investig. Dermatol. 2010, 130, 1937–1940. [Google Scholar] [CrossRef]
  73. Jonkman, M.F.; Pasmooij, A.M. Revertant mosaicism-patchwork in the skin. N. Engl. J. Med. 2009, 360, 1680–1682. [Google Scholar] [CrossRef] [PubMed]
  74. Pasmooij, A.M.; Pas, H.H.; Bolling, M.C.; Jonkman, M.F. Revertant mosaicism in junctional epidermolysis bullosa due to multiple correcting second-site mutations in LAMB3. J. Clin. Investig. 2007, 117, 1240–1248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Pasmooij, A.M.; Pas, H.H.; Deviaene, F.C.; Nijenhuis, M.; Jonkman, M.F. Multiple correcting COL17A1 mutations in patients with revertant mosaicism of epidermolysis bullosa. Am. J. Hum. Genet. 2005, 77, 727–740. [Google Scholar] [CrossRef] [PubMed]
  76. Smith, F.J.; Morley, S.M.; McLean, W.H. Novel mechanism of revertant mosaicism in Dowling-Meara epidermolysis bullosa simplex. J. Investig. Dermatol. 2004, 122, 73–77. [Google Scholar] [CrossRef] [PubMed]
  77. Schuilenga-Hut, P.H.; Scheffer, H.; Pas, H.H.; Nijenhuis, M.; Buys, C.H.; Jonkman, M.F. Partial revertant mosaicism of keratin 14 in a patient with recessive epidermolysis bullosa simplex. J. Investig. Dermatol. 2002, 118, 626–630. [Google Scholar] [CrossRef] [PubMed]
  78. Darling, T.N.; Yee, C.; Bauer, J.W.; Hintner, H.; Yancey, K.B. Revertant mosaicism: Partial correction of a germ-line mutation in COL17A1 by a frame-restoring mutation. J. Clin. Investig. 1999, 103, 1371–1377. [Google Scholar] [CrossRef]
  79. Pasmooij, A.M.; Nijenhuis, M.; Brander, R.; Jonkman, M.F. Natural gene therapy may occur in all patients with generalized non-Herlitz junctional epidermolysis bullosa with COL17A1 mutations. J. Investig. Dermatol. 2012, 132, 1374–1383. [Google Scholar] [CrossRef]
  80. Choate, K.A.; Lu, Y.; Zhou, J.; Elias, P.M.; Zaidi, S.; Paller, A.S.; Farhi, A.; Nelson-Williams, C.; Crumrine, D.; Milstone, L.M.; et al. Frequent somatic reversion of KRT1 mutations in ichthyosis with confetti. J. Clin. Investig. 2015, 125, 1703–1707. [Google Scholar] [CrossRef]
  81. Choate, K.A.; Lu, Y.; Zhou, J.; Choi, M.; Elias, P.M.; Farhi, A.; Nelson-Williams, C.; Crumrine, D.; Williams, M.L.; Nopper, A.J.; et al. Mitotic recombination in patients with ichthyosis causes reversion of dominant mutations in KRT10. Science 2010, 330, 94–97. [Google Scholar] [CrossRef]
  82. Gudmundsson, S.; Wilbe, M.; Ekvall, S.; Ameur, A.; Cahill, N.; Alexandrov, L.B.; Virtanen, M.; Hellström Pigg, M.; Vahlquist, A.; Törmä, H.; et al. Revertant mosaicism repairs skin lesions in a patient with keratitis-ichthyosis-deafness syndrome by second-site mutations in connexin 26. Hum. Mol. Genet. 2017, 26, 1070–1077. [Google Scholar] [CrossRef] [PubMed]
  83. Suzuki, S.; Nomura, T.; Miyauchi, T.; Takeda, M.; Fujita, Y.; Nishie, W.; Akiyama, M.; Ishida-Yamamoto, A.; Shimizu, H. Somatic recombination underlies frequent revertant mosaicism in loricrin keratoderma. Life Sci. Alliance 2019, 2, e201800284. [Google Scholar] [CrossRef] [PubMed]
  84. Miyauchi, T.; Suzuki, S.; Takeda, M.; Peh, J.T.; Aiba, M.; Natsuga, K.; Fujita, Y.; Takeichi, T.; Sakamoto, T.; Akiyama, M.; et al. Altered replication stress response due to CARD14 mutations promotes recombination-induced revertant mosaicism. Am. J. Hum. Genet. 2021, 108, 1026–1039. [Google Scholar] [CrossRef] [PubMed]
  85. van den Akker, P.C.; Pasmooij, A.M.G.; Joenje, H.; Hofstra, R.M.W.; Te Meerman, G.J.; Jonkman, M.F. A “late-but-fitter revertant cell” explains the high frequency of revertant mosaicism in epidermolysis bullosa. PLoS ONE 2018, 13, e0192994. [Google Scholar] [CrossRef]
  86. Kiritsi, D.; Garcia, M.; Brander, R.; Has, C.; Meijer, R.; Jose Escámez, M.; Kohlhase, J.; van den Akker, P.C.; Scheffer, H.; Jonkman, M.F.; et al. Mechanisms of natural gene therapy in dystrophic epidermolysis bullosa. J. Investig. Dermatol. 2014, 134, 2097–2104. [Google Scholar] [CrossRef] [PubMed]
  87. Suzuki, S.; Nomura, T.; Miyauchi, T.; Takeda, M.; Nakamura, H.; Shinkuma, S.; Fujita, Y.; Akiyama, M.; Shimizu, H. Revertant Mosaicism in Ichthyosis with Confetti Caused by a Frameshift Mutation in KRT1. J. Investig. Dermatol. 2016, 136, 2093–2095. [Google Scholar] [CrossRef]
  88. Nomura, T.; Suzuki, S.; Miyauchi, T.; Takeda, M.; Shinkuma, S.; Fujita, Y.; Nishie, W.; Akiyama, M.; Shimizu, H. Chromosomal inversions as a hidden disease-modifying factor for somatic recombination phenotypes. JCI Insight 2018, 3, 97595. [Google Scholar] [CrossRef]
  89. Takeichi, T.; Liu, L.; Abdul-Wahab, A.; McMillan, J.R.; Stone, K.L.; Akiyama, M.; Simpson, M.A.; Parsons, M.; Mellerio, J.E.; McGrath, J.A. Large Intragenic KRT1 Deletion Underlying Atypical Autosomal Dominant Keratinopathic Ichthyosis. J. Investig. Dermatol. 2016, 136, 2095–2098. [Google Scholar] [CrossRef]
  90. Lim, Y.H.; Qiu, J.; Saraceni, C.; Burrall, B.A.; Choate, K.A. Genetic Reversion via Mitotic Recombination in Ichthyosis with Confetti due to a KRT10 Polyalanine Frameshift Mutation. J. Investig. Dermatol. 2016, 136, 1725–1728. [Google Scholar] [CrossRef]
  91. Burger, B.; Spoerri, I.; Schubert, M.; Has, C.; Itin, P.H. Description of the natural course and clinical manifestations of ichthyosis with confetti caused by a novel KRT10 mutation. Br. J. Dermatol. 2012, 166, 434–439. [Google Scholar] [CrossRef]
  92. Montero-Vilchez, T.; Martinez-Lopez, A.; Rodriguez-Tejero, A.; Salvador-Rodriguez, L.; García-Durá, E.; Molina-Leyva, A.; Arias-Santiago, S. Loricrin keratoderma: Description of a novel mutation, systematic review and meta-analysis between genotypic and phenotypic features. J. Dtsch. Dermatol. Ges. 2020, 18, 1316–1321. [Google Scholar] [CrossRef] [PubMed]
  93. Guerra, L.; Diociaiuti, A.; El Hachem, M.; Castiglia, D.; Zambruno, G. Ichthyosis with confetti: Clinics, molecular genetics and management. Orphanet. J. Rare Dis. 2015, 10, 115. [Google Scholar] [CrossRef] [PubMed]
  94. Renz, P.; Imahorn, E.; Spoerri, I.; Aushev, M.; March, O.P.; Wariwoda, H.; Von Arb, S.; Volz, A.; Itin, P.H.; Reichelt, J.; et al. Arginine- but not alanine-rich carboxy-termini trigger nuclear translocation of mutant keratin 10 in ichthyosis with confetti. J. Cell Mol. Med. 2019, 23, 8442–8452. [Google Scholar] [CrossRef] [PubMed]
  95. Ishida-Yamamoto, A. Loricrin keratoderma: A novel disease entity characterized by nuclear accumulation of mutant loricrin. J. Dermatol. Sci. 2003, 31, 3–8. [Google Scholar] [CrossRef]
  96. Pasmooij, A.M.; Jonkman, M.F.; Uitto, J. Revertant mosaicism in heritable skin diseases: Mechanisms of natural gene therapy. Discov. Med. 2012, 14, 167–179. [Google Scholar]
  97. Gostyński, A.; Pasmooij, A.M.; Del Rio, M.; Diercks, G.F.; Pas, H.H.; Jonkman, M.F. Pigmentation and melanocyte supply to the epidermis depend on type XVII collagen. Exp. Dermatol. 2014, 23, 130–132. [Google Scholar] [CrossRef]
  98. Zhang, A.; Duchatelet, S.; Lakdawala, N.; Tower, R.L.; Diamond, C.; Marathe, K.; Hill, I.; Richard, G.; Diab, Y.; Kirkorian, A.Y.; et al. Targeted Inhibition of the Epidermal Growth Factor Receptor and Mammalian Target of Rapamycin Signaling Pathways in Olmsted Syndrome. JAMA Dermatol. 2020, 156, 196–200. [Google Scholar] [CrossRef]
  99. Luchsinger, I.; Knöpfel, N.; Theiler, M.; Bonnet des Claustres, M.; Barbieux, C.; Schwieger-Briel, A.; Brunner, C.; Donghi, D.; Buettcher, M.; Meier-Schiesser, B.; et al. Secukinumab Therapy for Netherton Syndrome. JAMA Dermatol. 2020, 156, 907–911. [Google Scholar] [CrossRef]
  100. Greco, C.; Leclerc-Mercier, S.; Chaumon, S.; Doz, F.; Hadj-Rabia, S.; Molina, T.; Boucheix, C.; Bodemer, C. Use of Epidermal Growth Factor Receptor Inhibitor Erlotinib to Treat Palmoplantar Keratoderma in Patients With Olmsted Syndrome Caused by TRPV3 Mutations. JAMA Dermatol. 2020, 156, 191–195. [Google Scholar] [CrossRef]
  101. Hernández-Martín, A.; Kennedy-Batalla, R.; Cañedo, E.; Bernaldo-de-Quirós, E.; Carazo-Gallego, B.; Vera, A.; Torrelo, A.; Noguera-Morel, L.; González-Sarmiento, R.; Bolling, M.; et al. Imbalance in T-Helper 17 Cells and Targeted Therapy in an Infant with SAM-like Syndrome. N. Engl. J. Med. 2019, 381, 2176–2178. [Google Scholar] [CrossRef]
  102. Venot, Q.; Blanc, T.; Rabia, S.H.; Berteloot, L.; Ladraa, S.; Duong, J.P.; Blanc, E.; Johnson, S.C.; Hoguin, C.; Boccara, O.; et al. Targeted therapy in patients with PIK3CA-related overgrowth syndrome. Nature 2018, 558, 540–546. [Google Scholar] [CrossRef] [PubMed]
  103. Paller, A.S.; Czarnowicki, T.; Renert-Yuval, Y.; Holland, K.; Huynh, T.; Sadlier, M.; McAleer, M.A.; Tran, G.; Geddes, G.C.; Irvine, A.D.; et al. The spectrum of manifestations in desmoplakin gene (DSP) spectrin repeat 6 domain mutations: Immunophenotyping and response to ustekinumab. J. Am. Acad. Dermatol. 2018, 78, 498–505.e492. [Google Scholar] [CrossRef] [PubMed]
  104. Czarnowicki, T.; He, H.; Leonard, A.; Malik, K.; Magidi, S.; Rangel, S.; Patel, K.; Ramsey, K.; Murphrey, M.; Song, T.; et al. The Major Orphan Forms of Ichthyosis Are Characterized by Systemic T-Cell Activation and Th-17/Tc-17/Th-22/Tc-22 Polarization in Blood. J. Investig. Dermatol. 2018, 138, 2157–2167. [Google Scholar] [CrossRef]
  105. Paller, A.S.; Renert-Yuval, Y.; Suprun, M.; Esaki, H.; Oliva, M.; Huynh, T.N.; Ungar, B.; Kunjravia, N.; Friedland, R.; Peng, X.; et al. An IL-17-dominant immune profile is shared across the major orphan forms of ichthyosis. J. Allergy Clin. Immunol. 2017, 139, 152–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Davies, B. The technical risks of human gene editing. Hum. Reprod. 2019, 34, 2104–2111. [Google Scholar] [CrossRef] [PubMed]
  107. Xafis, V.; Schaefer, G.O.; Labude, M.K.; Zhu, Y.; Holm, S.; Foo, R.S.; Lai, P.S.; Chadwick, R. Germline genome modification through novel political, ethical, and social lenses. PLoS Genet. 2021, 17, e1009741. [Google Scholar] [CrossRef] [PubMed]
  108. Gillmore, J.D.; Gane, E.; Taubel, J.; Kao, J.; Fontana, M.; Maitland, M.L.; Seitzer, J.; O’Connell, D.; Walsh, K.R.; Wood, K.; et al. CRISPR-Cas9 In Vivo Gene Editing for Transthyretin Amyloidosis. N. Engl. J. Med. 2021, 385, 493–502. [Google Scholar] [CrossRef]
  109. Has, C.; South, A.; Uitto, J. Molecular Therapeutics in Development for Epidermolysis Bullosa: Update 2020. Mol. Diagn. Ther. 2020, 24, 299–309. [Google Scholar] [CrossRef]
  110. Gurevich, I.; Agarwal, P.; Zhang, P.; Dolorito, J.A.; Oliver, S.; Liu, H.; Reitze, N.; Sarma, N.; Bagci, I.S.; Sridhar, K.; et al. In vivo topical gene therapy for recessive dystrophic epidermolysis bullosa: A phase 1 and 2 trial. Nat. Med. 2022, 28, 780–788. [Google Scholar] [CrossRef]
  111. Wang, X.; Alshehri, F.; Manzanares, D.; Li, Y.; He, Z.; Qiu, B.; Zeng, M.; Sigen, A.; Lara-Sáez, I.; Wang, W. Development of Minicircle Vectors Encoding COL7A1 Gene with Human Promoters for Non-Viral Gene Therapy for Recessive Dystrophic Epidermolysis Bullosa. Int. J. Mol. Sci. 2021, 22, 12774. [Google Scholar] [CrossRef]
  112. Bornert, O.; Hogervorst, M.; Nauroy, P.; Bischof, J.; Swildens, J.; Athanasiou, I.; Tufa, S.F.; Keene, D.R.; Kiritsi, D.; Hainzl, S.; et al. QR-313, an Antisense Oligonucleotide, Shows Therapeutic Efficacy for Treatment of Dominant and Recessive Dystrophic Epidermolysis Bullosa: A Preclinical Study. J. Investig. Dermatol. 2021, 141, 883–893.e886. [Google Scholar] [CrossRef] [PubMed]
  113. Lwin, S.M.; Syed, F.; Di, W.L.; Kadiyirire, T.; Liu, L.; Guy, A.; Petrova, A.; Abdul-Wahab, A.; Reid, F.; Phillips, R.; et al. Safety and early efficacy outcomes for lentiviral fibroblast gene therapy in recessive dystrophic epidermolysis bullosa. JCI Insight 2019, 4, e126243. [Google Scholar] [CrossRef] [PubMed]
  114. Marinkovich, M.P.; Tang, J.Y. Gene Therapy for Epidermolysis Bullosa. J. Investig. Dermatol. 2019, 139, 1221–1226. [Google Scholar] [CrossRef] [PubMed]
  115. Bauer, J.W.; Koller, J.; Murauer, E.M.; De Rosa, L.; Enzo, E.; Carulli, S.; Bondanza, S.; Recchia, A.; Muss, W.; Diem, A.; et al. Closure of a Large Chronic Wound through Transplantation of Gene-Corrected Epidermal Stem Cells. J. Investig. Dermatol. 2017, 137, 778–781. [Google Scholar] [CrossRef] [Green Version]
  116. Siprashvili, Z.; Nguyen, N.T.; Gorell, E.S.; Loutit, K.; Khuu, P.; Furukawa, L.K.; Lorenz, H.P.; Leung, T.H.; Keene, D.R.; Rieger, K.E.; et al. Safety and Wound Outcomes Following Genetically Corrected Autologous Epidermal Grafts in Patients With Recessive Dystrophic Epidermolysis Bullosa. JAMA 2016, 316, 1808–1817. [Google Scholar] [CrossRef]
  117. Mavilio, F.; Pellegrini, G.; Ferrari, S.; Di Nunzio, F.; Di Iorio, E.; Recchia, A.; Maruggi, G.; Ferrari, G.; Provasi, E.; Bonini, C.; et al. Correction of junctional epidermolysis bullosa by transplantation of genetically modified epidermal stem cells. Nat. Med. 2006, 12, 1397–1402. [Google Scholar] [CrossRef]
  118. Hickerson, W.L.; Remmers, A.E.; Recker, D.P. Twenty-Five Years’ Experience and Beyond with Cultured Epidermal Autografts for Coverage of Large Burn Wounds in Adult and Pediatric Patients, 1989-2015. J. Burn Care Res. 2019, 40, 157–165. [Google Scholar] [CrossRef]
  119. Kueckelhaus, M.; Rothoeft, T.; De Rosa, L.; Yeni, B.; Ohmann, T.; Maier, C.; Eitner, L.; Metze, D.; Losi, L.; Secone Seconetti, A.; et al. Transgenic Epidermal Cultures for Junctional Epidermolysis Bullosa—5-Year Outcomes. N. Engl. J. Med. 2021, 385, 2264–2270. [Google Scholar] [CrossRef]
  120. Hirsch, T.; Rothoeft, T.; Teig, N.; Bauer, J.W.; Pellegrini, G.; De Rosa, L.; Scaglione, D.; Reichelt, J.; Klausegger, A.; Kneisz, D.; et al. Regeneration of the entire human epidermis using transgenic stem cells. Nature 2017, 551, 327–332. [Google Scholar] [CrossRef]
  121. Gostyński, A.; Pasmooij, A.M.; Jonkman, M.F. Successful therapeutic transplantation of revertant skin in epidermolysis bullosa. J. Am. Acad. Dermatol. 2014, 70, 98–101. [Google Scholar] [CrossRef]
  122. Fusumae, T.T.M.; Yashiro, K.; Kitahara, H.; Amagai, M.; Kubo, A. Replacing COL7A1-deficient epidermis over the entire body by autografting cultured revertant keratinocytes in severe recessive dystrophic epidermolysis bullosa. Society for Investigative Dermatology (SID) 2021 Virtual Meeting. J. Invest. Dermatol. 2021, 141, S28 (abstract 160). [Google Scholar]
  123. Kogut, I.; McCarthy, S.M.; Pavlova, M.; Astling, D.P.; Chen, X.; Jakimenko, A.; Jones, K.L.; Getahun, A.; Cambier, J.C.; Pasmooij, A.M.G.; et al. High-efficiency RNA-based reprogramming of human primary fibroblasts. Nat. Commun. 2018, 9, 745. [Google Scholar] [CrossRef] [PubMed]
  124. Tolar, J.; Xia, L.; Lees, C.J.; Riddle, M.; McElroy, A.; Keene, D.R.; Lund, T.C.; Osborn, M.J.; Marinkovich, M.P.; Blazar, B.R.; et al. Keratinocytes from induced pluripotent stem cells in junctional epidermolysis bullosa. J. Investig. Dermatol. 2013, 133, 562–565. [Google Scholar] [CrossRef] [PubMed]
  125. Tolar, J.; Xia, L.; Riddle, M.J.; Lees, C.J.; Eide, C.R.; McElmurry, R.T.; Titeux, M.; Osborn, M.J.; Lund, T.C.; Hovnanian, A.; et al. Induced pluripotent stem cells from individuals with recessive dystrophic epidermolysis bullosa. J. Investig. Dermatol. 2011, 131, 848–856. [Google Scholar] [CrossRef]
  126. Umegaki-Arao, N.; Pasmooij, A.M.; Itoh, M.; Cerise, J.E.; Guo, Z.; Levy, B.; Gostyński, A.; Rothman, L.R.; Jonkman, M.F.; Christiano, A.M. Induced pluripotent stem cells from human revertant keratinocytes for the treatment of epidermolysis bullosa. Sci. Transl. Med. 2014, 6, 264ra164. [Google Scholar] [CrossRef]
  127. Wagner, J.E.; Ishida-Yamamoto, A.; McGrath, J.A.; Hordinsky, M.; Keene, D.R.; Woodley, D.T.; Chen, M.; Riddle, M.J.; Osborn, M.J.; Lund, T.; et al. Bone marrow transplantation for recessive dystrophic epidermolysis bullosa. N. Engl. J. Med. 2010, 363, 629–639. [Google Scholar] [CrossRef]
  128. Deinsberger, J.; Reisinger, D.; Weber, B. Global trends in clinical trials involving pluripotent stem cells: A systematic multi-database analysis. NPJ Regen. Med. 2020, 5, 15. [Google Scholar] [CrossRef]
  129. Kim, J.Y.; Nam, Y.; Rim, Y.A.; Ju, J.H. Review of the Current Trends in Clinical Trials Involving Induced Pluripotent Stem Cells. Stem Cell Rev. Rep. 2022, 18, 142–154. [Google Scholar] [CrossRef]
  130. Zhao, Z.; Anselmo, A.C.; Mitragotri, S. Viral vector-based gene therapies in the clinic. Bioeng. Transl. Med. 2022, 7, e10258. [Google Scholar] [CrossRef]
Figure 1. Comparison of forward and revertant mosaicism in genodermatoses. (a) In forward mosaicism, a somatic variant occurring on a wildtype allele induces a mosaic disease phenotype amidst otherwise healthy skin. (b) In revertant mosaicism, a somatic variant corrects the effect of a germline variant leading to the correction of the disease phenotype in a mosaic distribution. Light pink: healthy skin. Red: affected skin.
Figure 1. Comparison of forward and revertant mosaicism in genodermatoses. (a) In forward mosaicism, a somatic variant occurring on a wildtype allele induces a mosaic disease phenotype amidst otherwise healthy skin. (b) In revertant mosaicism, a somatic variant corrects the effect of a germline variant leading to the correction of the disease phenotype in a mosaic distribution. Light pink: healthy skin. Red: affected skin.
Biomedicines 10 02118 g001
Figure 2. Overview of the revertant skin phenotypes in different genodermatoses. Revertant skin patches in (a) the COL17A1-associated intermediate type of junctional epidermolysis bullosa (JEB-intermed) [75], (b) the LAMB3-associated intermediate type of junctional epidermolysis bullosa (JEB-intermed) [74], (c) the generalized type of dominant dystrophic epidermolysis bullosa (DDEB-gen, COL7A1) [86], (d) the intermediate type of generalized recessive dystrophic epidermolysis bullosa (RDEB-intermed, COL7A1) [86], (e) the severe type of generalized recessive dystrophic epidermolysis bullosa (RDEB-gen, COL7A1) [68], (f) Kindler epidermolysis bullosa (Kindler EB, FERMT1) [70], (g) ichthyosis with confetti type 1 (IWC-I, KRT10) [81], (h) ichthyosis with confetti type 2 (IWC-II, KRT1) [87], (i) loricrin keratoderma (LK, LOR) [83], (j) keratitis-ichthyosis-deafness syndrome (KID, GJB2), and (k) pityriasis rubra pilaris (PRP, CARD14) [84]. Solid or dashed lines indicate revertant patches amidst affected skin (note that on several panels more revertant patches are visible than highlighted). Note that the revertant patches in the different types of recessive EB are generally larger than in the autosomal dominant ichthyotic disorders, whereas multiple revertant spots are usually identified simultaneously in the latter. All images re-used with permission and the original articles are cited.
Figure 2. Overview of the revertant skin phenotypes in different genodermatoses. Revertant skin patches in (a) the COL17A1-associated intermediate type of junctional epidermolysis bullosa (JEB-intermed) [75], (b) the LAMB3-associated intermediate type of junctional epidermolysis bullosa (JEB-intermed) [74], (c) the generalized type of dominant dystrophic epidermolysis bullosa (DDEB-gen, COL7A1) [86], (d) the intermediate type of generalized recessive dystrophic epidermolysis bullosa (RDEB-intermed, COL7A1) [86], (e) the severe type of generalized recessive dystrophic epidermolysis bullosa (RDEB-gen, COL7A1) [68], (f) Kindler epidermolysis bullosa (Kindler EB, FERMT1) [70], (g) ichthyosis with confetti type 1 (IWC-I, KRT10) [81], (h) ichthyosis with confetti type 2 (IWC-II, KRT1) [87], (i) loricrin keratoderma (LK, LOR) [83], (j) keratitis-ichthyosis-deafness syndrome (KID, GJB2), and (k) pityriasis rubra pilaris (PRP, CARD14) [84]. Solid or dashed lines indicate revertant patches amidst affected skin (note that on several panels more revertant patches are visible than highlighted). Note that the revertant patches in the different types of recessive EB are generally larger than in the autosomal dominant ichthyotic disorders, whereas multiple revertant spots are usually identified simultaneously in the latter. All images re-used with permission and the original articles are cited.
Biomedicines 10 02118 g002
Figure 3. Mechanisms of revertant mosaicism identified in genodermatoses. Different reversion mechanisms can correct pathogenic germline variants. The mechanisms reported in genodermatoses are shown with the diseases indicated below. For every cell (black boxes), one chromosome pair is shown (vertical blue and pink bars); one from the mother, the other from the father. Horizontal black bars on the chromosomes indicate pathogenic germline variants. During mitosis, the DNA of each chromosome is first duplicated to yield two identical sister chromatids. With normal segregation, each daughter cell obtains one sister chromatid from the maternal chromosome and one from the paternal chromosome. (a) Mechanisms of reversion reported in autosomal recessive genodermatoses. Gene conversion is the non-reciprocal transfer of DNA from one chromosome to the other. When this occurs in the region where the inherited mutation is located, the mutation is lost in one of the daughter cells, which then only carries one of the recessive mutations and will produce protein (green shading). Intragenic cross-over (a form of mitotic recombination) results in one daughter cell with three inherited mutations, and a revertant daughter cell with only one of the inherited mutations. Second-site mutations indicate additional mutation events (horizontal white bars) on one of the mutant alleles that correct the effect of the pathogenic germline variant. Many different types of second-site mutations have been reported (see Table 1) and the protein that is produced may be similar to the wildtype protein or may be slightly aberrant but still functional (green/white shading). Back-mutation is a form of second-site mutation in which the additional event changes the germline variant back to wildtype. (b) In autosomal dominant genodermatoses, where a heterozygous variant causes the disease phenotype, the most prominent reversion mechanism is mitotic recombination. Mitotic recombination is a normal process during DNA replication, but it can lead to cells that carry both mutant alleles and to revertant cells that carry both wildtype alleles. Boxes with red shading indicate mutant cells. Genodermatosis names in italics indicate that it was not possible to distinguish between mitotic recombination and back-mutation as the reversion mechanism. JEB, junctional epidermolysis bullosa; RDEB, recessive dystrophic epidermolysis bullosa; KEB, Kindler epidermolysis bullosa; IWC, ichthyosis with confetti; LK, loricrin keratoderma; PRP, pityriasis rubra pilaris; DDEB, dominant dystrophic epidermolysis bullosa; EBS, epidermolysis bullosa simplex; KID, keratitis-ichthyosis-deafness syndrome.
Figure 3. Mechanisms of revertant mosaicism identified in genodermatoses. Different reversion mechanisms can correct pathogenic germline variants. The mechanisms reported in genodermatoses are shown with the diseases indicated below. For every cell (black boxes), one chromosome pair is shown (vertical blue and pink bars); one from the mother, the other from the father. Horizontal black bars on the chromosomes indicate pathogenic germline variants. During mitosis, the DNA of each chromosome is first duplicated to yield two identical sister chromatids. With normal segregation, each daughter cell obtains one sister chromatid from the maternal chromosome and one from the paternal chromosome. (a) Mechanisms of reversion reported in autosomal recessive genodermatoses. Gene conversion is the non-reciprocal transfer of DNA from one chromosome to the other. When this occurs in the region where the inherited mutation is located, the mutation is lost in one of the daughter cells, which then only carries one of the recessive mutations and will produce protein (green shading). Intragenic cross-over (a form of mitotic recombination) results in one daughter cell with three inherited mutations, and a revertant daughter cell with only one of the inherited mutations. Second-site mutations indicate additional mutation events (horizontal white bars) on one of the mutant alleles that correct the effect of the pathogenic germline variant. Many different types of second-site mutations have been reported (see Table 1) and the protein that is produced may be similar to the wildtype protein or may be slightly aberrant but still functional (green/white shading). Back-mutation is a form of second-site mutation in which the additional event changes the germline variant back to wildtype. (b) In autosomal dominant genodermatoses, where a heterozygous variant causes the disease phenotype, the most prominent reversion mechanism is mitotic recombination. Mitotic recombination is a normal process during DNA replication, but it can lead to cells that carry both mutant alleles and to revertant cells that carry both wildtype alleles. Boxes with red shading indicate mutant cells. Genodermatosis names in italics indicate that it was not possible to distinguish between mitotic recombination and back-mutation as the reversion mechanism. JEB, junctional epidermolysis bullosa; RDEB, recessive dystrophic epidermolysis bullosa; KEB, Kindler epidermolysis bullosa; IWC, ichthyosis with confetti; LK, loricrin keratoderma; PRP, pityriasis rubra pilaris; DDEB, dominant dystrophic epidermolysis bullosa; EBS, epidermolysis bullosa simplex; KID, keratitis-ichthyosis-deafness syndrome.
Biomedicines 10 02118 g003
Figure 4. Therapeutic prospects of revertant mosaicism. (a) The ultimate goal of revertant cell therapy is to use the patient’s own naturally corrected cells to cure the entire skin. (b) Several attempts have already been made to use revertant skin cells for treatment, such as direct transplantation of small revertant biopsies to an affected skin area (grey arrow) [121]. Others have focused on first expanding the revertant cells to produce larger revertant epidermal grafts that were then transplanted onto larger areas (pathway indicated by blue arrows) [122]. Lastly, induced pluripotent stem cells (iPSCs) can be generated from revertant cells to provide an unlimited supply of revertant cells (iPSC pathways indicated by red arrows). These iPSCs can be subsequently differentiated into keratinocytes to form revertant epidermal autografts [126]. Another possibility is to differentiate these iPSCs into hematopoietic stem cells (HSCs) or mesenchymal stromal cells (MSCs) that could be used for bone marrow transplantation [67] or subcutaneous or intravenous injection, respectively. The question mark indicates that these possibilities have not yet been pursued on patients and the outcome of such approaches is thus unknown.
Figure 4. Therapeutic prospects of revertant mosaicism. (a) The ultimate goal of revertant cell therapy is to use the patient’s own naturally corrected cells to cure the entire skin. (b) Several attempts have already been made to use revertant skin cells for treatment, such as direct transplantation of small revertant biopsies to an affected skin area (grey arrow) [121]. Others have focused on first expanding the revertant cells to produce larger revertant epidermal grafts that were then transplanted onto larger areas (pathway indicated by blue arrows) [122]. Lastly, induced pluripotent stem cells (iPSCs) can be generated from revertant cells to provide an unlimited supply of revertant cells (iPSC pathways indicated by red arrows). These iPSCs can be subsequently differentiated into keratinocytes to form revertant epidermal autografts [126]. Another possibility is to differentiate these iPSCs into hematopoietic stem cells (HSCs) or mesenchymal stromal cells (MSCs) that could be used for bone marrow transplantation [67] or subcutaneous or intravenous injection, respectively. The question mark indicates that these possibilities have not yet been pursued on patients and the outcome of such approaches is thus unknown.
Biomedicines 10 02118 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

van den Akker, P.C.; Bolling, M.C.; Pasmooij, A.M.G. Revertant Mosaicism in Genodermatoses: Natural Gene Therapy Right before Your Eyes. Biomedicines 2022, 10, 2118. https://doi.org/10.3390/biomedicines10092118

AMA Style

van den Akker PC, Bolling MC, Pasmooij AMG. Revertant Mosaicism in Genodermatoses: Natural Gene Therapy Right before Your Eyes. Biomedicines. 2022; 10(9):2118. https://doi.org/10.3390/biomedicines10092118

Chicago/Turabian Style

van den Akker, Peter C., Maria C. Bolling, and Anna M. G. Pasmooij. 2022. "Revertant Mosaicism in Genodermatoses: Natural Gene Therapy Right before Your Eyes" Biomedicines 10, no. 9: 2118. https://doi.org/10.3390/biomedicines10092118

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