44 results on '"REVERTANT MOSAICISM"'
Search Results
2. Revertant Mosaicism
- Author
-
Happle, Rudolf, Torrelo, Antonio, Happle, Rudolf, and Torrelo, Antonio
- Published
- 2023
- Full Text
- View/download PDF
3. Modified Non-Cultured Cell Spray Induced Epithelization in LAMB3 Mutation Epidermolysis Bullosa
- Author
-
Widhiati S, Dewi ST, Yefta, Danarti R, Soebono H, Irmawati YE, Puspitasari M, Trisnowati N, Wibawa T, Purnomosari D, and Wirohadidjojo YW
- Subjects
revertant mosaicism ,silver-sulfadiazine ,plastic wrap ,chronic wound ,junctional epidermolysis bullosa ,Dermatology ,RL1-803 - Abstract
Suci Widhiati,1,2 Shinta Trilaksmi Dewi,3 Yefta,3 Retno Danarti,3 Hardyanto Soebono,3 Yulia Eka Irmawati,3 Monika Puspitasari,3 Niken Trisnowati,3 Tri Wibawa,4 Dewajani Purnomosari,5 Yohanes Widodo Wirohadidjojo3 1Department of Dermatology and Venereology, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia; 2Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia; 3Department of Dermatology and Venereology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia; 4Department of Microbiology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia; 5Department of Histology and Cell Biology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, IndonesiaCorrespondence: Yohanes Widodo Wirohadidjojo, Department of Dermatology and Venereology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Gedung Radiopoetro, lt. 3 Sekip, Yogyakarta, 55281, Indonesia, Tel +62 274 560700, Email yohanes.widodo@ugm.ac.idBackground: Autologous non-cultured cell (ANCC) spray has been used to treat burns, chronic wounds, and vitiligo, but its use in junctional epidermolysis bullosa (JEB) has not been published previously. Chronic wounds in JEB are caused by mutations of laminin 332 (L322), whose function is to attach and act as a glue in the basal membrane. It is proposed that ANCC applications can provide keratinocytes and fibroblasts required to improve epithelization and spontaneously correct revertant keratinocytes in the wound area.Purpose: To develop a modified procedure of ANCC spray and improve epithelization using silver sulfadiazine covered with plastic wrap to treat chronic wounds of JEB.Patients and Methods: Shave excision of the donor site was performed on a 19-year-old girl with JEB. The ANCC spray was prepared and applied to the chronic wound, which was then covered with silver sulfadiazine occluded with plastic wrap.Results: Following the ANCC spray application, epithelization was successfully initiated. Unfortunately, the wounds recurred after four months of follow-up.Conclusion: The modified application method of ANCC spray provides a good alternative to treat chronic wounds in JEB.Keywords: revertant mosaicism, silver-sulfadiazine, plastic wrap, chronic wound, junctional epidermolysis bullosa
- Published
- 2022
4. Integrated Management Strategies for Epidermolysis Bullosa: Current Insights
- Author
-
Sait H, Srivastava S, and Saxena D
- Subjects
epidermolysis bullosa ,blistering skin disorder ,gene replacement ,gene editing ,antisense oligonucleotides ,sirna therapeutics ,spliceosome-mediated rna trans-splicing ,revertant mosaicism ,readthrough therapies ,squamous cell carcinoma ,Medicine (General) ,R5-920 - Abstract
Haseena Sait, Somya Srivastava, Deepti Saxena Department of Medical Genetics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, IndiaCorrespondence: Deepti Saxena, Department of Medical Genetics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India, Email saxenadrdeepti@gmail.comAbstract: Epidermolysis bullosa (EB) is a group of rare genodermatoses that is characterized by skin fragility resulting from minor trauma. There are four major subtypes, namely, EB simplex, junctional EB, dystrophic EB and Kindler EB, depending upon the localization of defective protein and resulting plane of blister formation. The phenotype is heterogeneous in terms of severity and majority of them present at birth or neonatal period. Currently, the treatment is mainly supportive and requires multidisciplinary care. The complex molecular pathology creates difficulty in discovering a unified curative treatment approach. But with arduous efforts, significant progress has been made in the development of treatment strategies in the last decade. The management strategies range from targeting the underlying causative factor to symptom-relieving approaches, and include gene, mRNA, protein, cell and combination therapies. In this review, we enumerate the promising approaches that are currently under various stages of investigation to provide effective treatment for patients with EB.Keywords: epidermolysis bullosa, blistering skin disorder, gene replacement, gene editing, antisense oligonucleotides, siRNA therapeutics, spliceosome-mediated RNA trans-splicing, revertant mosaicism, readthrough therapies, squamous cell carcinoma
- Published
- 2022
5. A case of revertant mosaic-like normal-looking spots in a patient with erythroderma with IL36RN and CARD14 heterozygous mutations.
- Author
-
Matsuo M, Zang X, Miyauchi T, Mizutani Y, Niwa H, Tanaka K, and Iwata H
- Abstract
An 89-year-old Japanese woman presented with erythroderma associated with significant scaling. A histological examination showed acanthosis with hyperkeratosis and hyperkeratinization of the hair follicles. Genetic analyses using DNA from the peripheral blood revealed heterozygous mutations in IL36RN (c.115+6T>C) and CARD14 c.2648G>A (p.Arg883His). Based on these findings, we diagnosed her with erythroderma attributable to autoinflammatory keratinization disease. She then developed more than 30 small, round, well-defined, spots on her back and extremities that appeared histologically normal. We suspected that these spots might be revertant mosaicism. Immunohistochemical staining with p65, which is a component of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), revealed nuclear staining in epidermal keratinocytes in erythematous lesions, but not in the normal-looking spots. However, mutations in IL36RN and CARD14 unexpectedly persisted in the epidermis and dermis of the normal-looking spots., (© 2024 The Author(s). The Journal of Dermatology published by John Wiley & Sons Australia, Ltd on behalf of Japanese Dermatological Association.)
- Published
- 2024
- Full Text
- View/download PDF
6. Revertant Mosaicism in Genodermatoses: Natural Gene Therapy Right before Your Eyes.
- Author
-
van den Akker, Peter C., Bolling, Maria C., and Pasmooij, Anna M. G.
- Subjects
GENE therapy ,NATUROPATHY ,EPIDERMOLYSIS bullosa ,MOSAICISM ,MEDICAL sciences ,PHYSICIANS - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
7. Detection of revertant mosaicism in epidermolysis bullosa through Cas9‐targeted long‐read sequencing.
- Author
-
Natsuga, Ken, Furuta, Yoshikazu, Takashima, Shota, Nohara, Takuma, Kosumi, Hideyuki, Mai, Yosuke, Higashi, Hideaki, and Ujiie, Hideyuki
- Abstract
Revertant mosaicism (RM) is a phenomenon in which inherited mutations are spontaneously corrected in somatic cells. RM occurs in some congenital skin diseases, but genetic validation of RM in clinically revertant skin has been challenging, especially when homologous recombination (HR) is responsible for RM. Here, we introduce nanopore Cas9‐targeted sequencing (nCATS) for identifying HR in clinically revertant skin. We took advantage of compound heterozygous COL7A1 mutations in a patient with recessive dystrophic epidermolysis bullosa who showed revertant skin spots. Cas9‐mediated enrichment of genomic DNA (gDNA) covering the two mutation sites (>8 kb) in COL7A1 and subsequent MinION sequencing successfully detected intragenic crossover in the epidermis of the clinically revertant skin. This method enables the discernment of haplotypes of up to a few tens of kilobases of gDNA. Moreover, it is devoid of polymerase chain reaction amplification, which can technically induce recombination. We, therefore, propose that nCATS is a powerful tool for understanding complicated gene modifications, including RM. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
8. Revertant Mosaicism in Genodermatoses: Natural Gene Therapy Right before Your Eyes
- Author
-
Peter C. van den Akker, Maria C. Bolling, and Anna M. G. Pasmooij
- Subjects
genodermatosis ,revertant mosaicism ,natural gene therapy ,epidermolysis bullosa ,ichthyosis ,gene therapy ,Biology (General) ,QH301-705.5 - 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.
- Published
- 2022
- Full Text
- View/download PDF
9. Altered replication stress response due to CARD14 mutations promotes recombination-induced revertant mosaicism.
- Author
-
Miyauchi, Toshinari, Suzuki, Shotaro, Takeda, Masae, Peh, Jin Teng, Aiba, Masayuki, Natsuga, Ken, Fujita, Yasuyuki, Takeichi, Takuya, Sakamoto, Taiko, Akiyama, Masashi, Shimizu, Hiroshi, and Nomura, Toshifumi
- Subjects
- *
MOSAICISM , *DOUBLE-strand DNA breaks , *GENETIC mutation , *MUTANT proteins , *NATUROPATHY , *DNA damage - Abstract
Revertant mosaicism, or "natural gene therapy," refers to the spontaneous in vivo reversion of an inherited mutation in a somatic cell. Only approximately 50 human genetic disorders exhibit revertant mosaicism, implicating a distinctive role played by mutant proteins in somatic correction of a pathogenic germline mutation. However, the process by which mutant proteins induce somatic genetic reversion in these diseases remains unknown. Here we show that heterozygous pathogenic CARD14 mutations causing autoinflammatory skin diseases, including psoriasis and pityriasis rubra pilaris, are repaired mainly via homologous recombination. Rather than altering the DNA damage response to exogenous stimuli, such as X-irradiation or etoposide treatment, mutant CARD14 increased DNA double-strand breaks under conditions of replication stress. Furthermore, mutant CARD14 suppressed new origin firings without promoting crossover events in the replication stress state. Together, these results suggest that mutant CARD14 alters the replication stress response and preferentially drives break-induced replication (BIR), which is generally suppressed in eukaryotes. Our results highlight the involvement of BIR in reversion events, thus revealing a previously undescribed role of BIR that could potentially be exploited to develop therapeutics for currently intractable genetic diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
10. Revertant somatic mosaicism as a cause of cancer.
- Author
-
Inaba, Toshiya and Nagamachi, Akiko
- Abstract
Revertant (somatic) mosaicism is a spontaneous correction of a causative mutation in patients with congenital diseases. A relatively frequent event, revertant mosaicism may bring favorable outcomes that ameliorate disorders, and is therefore called "natural gene therapy." However, it has been revealed recently that "overcorrection" of inherited bone marrow failure in patients with sterile alpha motif domain containing 9 (SAMD9)/9L syndromes by revertant mosaicism induces myelodysplastic syndrome (MDS) with monosomy 7 that occasionally proceeds to acute myelogenous leukemia (AML). In this review, we interpret very complex mechanisms underlying MDS/AML in patients with SAMD9/9L syndromes. This includes multiple myeloid tumor suppressors on the long arm of chromosome 7, all of which act in a haploinsufficient fashion, and a difference in sensitivity to interferon between cells carrying a mutation and revertants. Overcorrection of mutants by somatic mosaicism is likely a novel mechanism in carcinogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
11. Revertant Mosaicism in Epidermolysis Bullosa
- Author
-
Cameron Meyer-Mueller, Mark J. Osborn, Jakub Tolar, Christina Boull, and Christen L. Ebens
- Subjects
epidermolysis bullosa ,revertant mosaicism ,cellular therapy ,gene therapy ,autograft ,loss of heterozygosity ,Biology (General) ,QH301-705.5 - Abstract
Epidermolysis bullosa (EB) is a group of genetic blistering diseases characterized by mechanically fragile skin and mucocutaneous involvement. Historically, disease management has focused on supportive care. The development of new genetic, cellular, and recombinant protein therapies has shown promise, and this review summarizes a unique gene and cell therapy phenomenon termed revertant mosaicism (RM). RM is the spontaneous correction of a disease-causing mutation. It has been reported in most EB subtypes, some with relatively high frequency, and has been observed in both keratinocytes and fibroblasts. RM manifests as identifiable patches of unaffected, blister-resistant skin and can occur through a variety of molecular mechanisms, including true back mutation, intragenic crossover, mitotic gene conversion, and second-site mutation. RM cells represent a powerful autologous platform for therapy, and leveraging RM cells as a therapeutic substrate may avoid the inherent mutational risks of gene therapy/editing. However, further examination of the genomic integrity and long-term functionality of RM-derived cells, as well in vivo testing of systemic therapies with RM cells, is required to realize the full therapeutic promise of naturally occurring RM in EB.
- Published
- 2022
- Full Text
- View/download PDF
12. Recombination-induced revertant mosaicism in ichthyosis with confetti and loricrin keratoderma.
- Author
-
Nomura, Toshifumi
- Subjects
- *
MOSAICISM , *MUTANT proteins , *SOMATIC cells , *SKIN diseases , *CELL populations - Abstract
• Somatic correction of a germline mutation is referred to as revertant mosaicism. • To date, revertant mosaicism has been documented in ∼50 genetic diseases. • Ichthyosis with confetti and loricrin keratoderma develop numerous revertant spots. • Homologous recombination underlies the mutation reversion in these diseases. Revertant mosaicism refers to a condition in which a pathogenic germline mutation is spontaneously corrected in somatic cells, resulting in the presence of two or more cell populations with different genotypes in an organism arising from a single fertilized egg. If the revertant cells are clonally expanded due to a survival advantage over the surrounding mutant cells, patients benefit from this self-healing phenomenon which leads to the development of milder-than-expected clinical phenotypes; in genetic skin diseases, patients with revertant mosaicism present with small islands of healthy skin. To date, revertant mosaicism has been reported in ∼50 genetic diseases involving the skin, blood, liver, muscle, and brain. In this review, I briefly summarize current knowledge on revertant mosaicism in two particular skin diseases, ichthyosis with confetti (IWC) and loricrin keratoderma (LK), both of which develop numerous revertant skin patches. Notably, homologous recombination (HR) is the only mechanism underlying the reversion of pathogenic mutations in IWC and LK, and this was identified following the analysis of ∼50 revertant epidermis samples. All the samples showed long-tract loss of heterozygosity (LOH) that originated at regions centromeric to pathogenic mutations and extended to the telomere of the mutation-harboring chromosomes. Elucidating the molecular mechanisms underlying revertant mosaicism in IWC and LK—especially how mutant proteins induce long-tract LOH—would potentially expand the possibility of manipulating HR to induce the reversion of disease-causing mutations and help devising novel therapies not only for IWC and LK but also for other intractable genetic diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
13. Primary adrenal insufficiency: New genetic causes and their long‐term consequences.
- Author
-
Buonocore, Federica and Achermann, John C.
- Subjects
- *
ADDISON'S disease , *ADRENAL insufficiency , *GAIN-of-function mutations , *ADRENOGENITAL syndrome , *GENETICS - Abstract
Primary adrenal insufficiency (PAI) is a potentially life‐threatening condition that requires urgent diagnosis and treatment. Whilst the most common causes are congenital adrenal hyperplasia (CAH) in childhood and autoimmune adrenal insufficiency in adolescence and adulthood, more than 30 other physical and genetics cause of PAI have been reported. Reaching a specific diagnosis can have implications for management and for monitoring associated features, as well as for counselling families about recurrence risk in siblings and relatives. Here, we describe some recent insights into the genetics of adrenal insufficiency and associated molecular mechanisms. We discuss (a) the role of the nuclear receptors DAX‐1 (NR0B1) and steroidogenic factor‐1 (SF‐1, NR5A1) in human adrenal and reproductive dysfunction; (b) multisystem growth restriction syndromes due to gain‐of‐function in the growth repressors CDKN1C (IMAGE syndrome) and SAMD9 (MIRAGE syndrome), or loss of POLE1; (c) nonclassic forms of STAR and P450scc/CYP11A1 insufficiency that present with a delayed‐onset adrenal phenotype and represent a surprisingly prevalent cause of undiagnosed PAI; and (d) a new sphingolipidosis causing PAI due to defects in sphingosine‐1‐phosphate lyase‐1 (SGPL1). Reaching a specific diagnosis can have life‐long implications for management. In some situations, milder or nonclassic forms of these conditions can first present in adulthood and may have been labelled, "Addison's disease." [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
14. Spontaneous remission in a Diamond‐Blackfan anaemia patient due to a revertant uniparental disomy ablating a de novo RPS19 mutation.
- Author
-
Garelli, Emanuela, Quarello, Paola, Giorgio, Elisa, Carando, Adriana, Menegatti, Elisa, Mancini, Cecilia, Di Gregorio, Eleonora, Crescenzio, Nicoletta, Palumbo, Orazio, Carella, Massimo, Dimartino, Paola, Pippucci, Tommaso, Dianzani, Irma, Ramenghi, Ugo, and Brusco, Alfredo
- Subjects
- *
MOSAICISM , *RED blood cell transfusion , *ANEMIA - Abstract
The article focuses on a study which applies high density single nucleotide polymorphism (SNP)-arrays and whole exome sequencing (WES) to solve a clinically and molecularly intricate case of remitted sporadic Diamond-Blackfan Anaemia (DBA). The case of a 35-year-old male, the first of two siblings born full term after an uncomplicated pregnancy from healthy unrelated White parents, is presented.
- Published
- 2019
- Full Text
- View/download PDF
15. Detection of revertant mosaicism in epidermolysis bullosa through Cas9-targeted long-read sequencing
- Author
-
Ken Natsuga, Yoshikazu Furuta, Shota Takashima, Takuma Nohara, Hideyuki Kosumi, Yosuke Mai, Hideaki Higashi, and Hideyuki Ujiie
- Subjects
revertant mosaicism ,Collagen Type VII ,Mosaicism ,Epidermolysis Bullosa Dystrophica ,Mutation ,intragenic crossover ,long-read sequencing ,Genetics ,Humans ,epidermolysis bullosa ,CRISPR-Cas Systems ,Genetics (clinical) ,Skin - Abstract
Revertant mosaicism (RM) is a phenomenon in which inherited mutations are spontaneously corrected in somatic cells. RM occurs in some congenital skin diseases, but genetic validation of RM in clinically revertant skin has been challenging, especially when homologous recombination (HR) is responsible for RM. Here, we introduce nanopore Cas9-targeted sequencing (nCATS) for identifying HR in clinically revertant skin. We took advantage of compound heterozygous COL7A1 mutations in a patient with recessive dystrophic epidermolysis bullosa who showed revertant skin spots. Cas9-mediated enrichment of genomic DNA (gDNA) covering the two mutation sites (>8 kb) in COL7A1 and subsequent MinION sequencing successfully detected intragenic crossover in the epidermis of the clinically revertant skin. This method enables the discernment of haplotypes of up to a few tens of kilobases of gDNA. Moreover, it is devoid of polymerase chain reaction amplification, which can technically induce recombination. We, therefore, propose that nCATS is a powerful tool for understanding complicated gene modifications, including RM.
- Published
- 2022
16. Somatic mosaicism of an intragenic <italic>FANCB</italic> duplication in both fibroblast and peripheral blood cells observed in a Fanconi anemia patient leads to milder phenotype.
- Author
-
Asur, Rajalakshmi S., Kimble, Danielle C., Lach, Francis P., Jung, Moonjung, Donovan, Frank X., Kamat, Aparna, Noonan, Raymond J., Thomas, James W., Park, Morgan, Chines, Peter, Vlachos, Adrianna, Auerbach, Arleen D., Smogorzewska, Agata, and Chandrasekharappa, Settara C.
- Subjects
- *
MOSAICISM , *FIBROBLASTS , *FANCONI'S anemia , *VATER syndrome , *ALLELES - Abstract
Abstract: Background: Fanconi anemia (FA) is a rare disorder characterized by congenital malformations, progressive bone marrow failure, and predisposition to cancer. Patients harboring X‐linked
FANCB pathogenic variants usually present with severe congenital malformations resembling VACTERL syndrome with hydrocephalus. Methods: We employed the diepoxybutane (DEB) test for FA diagnosis, arrayCGH for detection of duplication, targeted capture and next‐gen sequencing for defining the duplication breakpoint, PacBio sequencing of full‐length FANCB aberrant transcript, FANCD2 ubiquitination and foci formation assays for the evaluation of FANCB protein function by viral transduction of FANCB‐null cells with lentiviralFANCB WT and mutant expression constructs, and droplet digital PCR for quantitation of the duplication in the genomic DNA and cDNA. Results: We describe here an FA‐B patient with a mild phenotype. The DEB diagnostic test for FA revealed somatic mosaicism. We identified a 9154 bp intragenic duplication inFANCB, covering the first coding exon 3 and the flanking regions. A four bp homology (GTAG) present at both ends of the breakpoint is consistent with microhomology‐mediated duplication mechanism. The duplicated allele gives rise to an aberrant transcript containing exon 3 duplication, predicted to introduce a stop codon in FANCB protein (p.A319*). Duplication levels in the peripheral blood DNA declined from 93% to 7.9% in the span of eleven years. Moreover, the patient fibroblasts have shown 8% of wild‐type (WT) allele and his carrier mother showed higher than expected levels of WT allele (79% vs. 50%) in peripheral blood, suggesting that the duplication was highly unstable. Conclusion: Unlike sequence point variants, intragenic duplications are difficult to precisely define, accurately quantify, and may be very unstable, challenging the proper diagnosis. The reversion of genomic duplication to the WT allele results in somatic mosaicism and may explain the relatively milder phenotype displayed by the FA‐B patient described here. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
17. Detection of revertant mosaicism in epidermolysis bullosa through Cas9-targeted long-read sequencing
- Author
-
1000070645457, Natsuga, Ken, Furuta, Yoshikazu, Takashima, Shota, Nohara, Takuma, Kosumi, Hideyuki, Mai, Yosuke, Higashi, Hideaki, 1000060374435, Ujiie, Hideyuki, 1000070645457, Natsuga, Ken, Furuta, Yoshikazu, Takashima, Shota, Nohara, Takuma, Kosumi, Hideyuki, Mai, Yosuke, Higashi, Hideaki, 1000060374435, and Ujiie, Hideyuki
- Abstract
Revertant mosaicism (RM) is a phenomenon in which inherited mutations are spontaneously corrected in somatic cells. RM occurs in some congenital skin diseases, but genetic validation of RM in clinically revertant skin has been challenging, especially when homologous recombination (HR) is responsible for RM. Here, we introduce nanopore Cas9-targeted sequencing (nCATS) for identifying HR in clinically revertant skin. We took advantage of compound heterozygous COL7A1 mutations in a patient with recessive dystrophic epidermolysis bullosa who showed revertant skin spots. Cas9-mediated enrichment of genomic DNA (gDNA) covering the two mutation sites (>8 kb) in COL7A1 and subsequent MinION sequencing successfully detected intragenic crossover in the epidermis of the clinically revertant skin. This method enables the discernment of haplotypes of up to a few tens of kilobases of gDNA. Moreover, it is devoid of polymerase chain reaction amplification, which can technically induce recombination. We, therefore, propose that nCATS is a powerful tool for understanding complicated gene modifications, including RM.
- Published
- 2022
18. Revertant somatic mosaicism as a cause of cancer
- Author
-
Akiko Nagamachi and Toshiya Inaba
- Subjects
0301 basic medicine ,Cancer Research ,Myeloid ,Carcinogenesis ,Somatic cell ,Review Article ,03 medical and health sciences ,Myelogenous ,0302 clinical medicine ,Germline mutation ,hemic and lymphatic diseases ,Animals ,Humans ,Medicine ,somatic mutation ,Review Articles ,revertant mosaicism ,Chromosome 7 (human) ,SAMD9/9L syndromes ,tumor suppressors ,Mosaicism ,business.industry ,Tumor Suppressor Proteins ,Intracellular Signaling Peptides and Proteins ,Bone marrow failure ,General Medicine ,medicine.disease ,haploinsufficiency ,Leukemia, Myeloid, Acute ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Chromosome Deletion ,business ,Haploinsufficiency ,Chromosomes, Human, Pair 7 - Abstract
Revertant (somatic) mosaicism is a spontaneous correction of a causative mutation in patients with congenital diseases. A relatively frequent event, revertant mosaicism may bring favorable outcomes that ameliorate disorders, and is therefore called “natural gene therapy.” However, it has been revealed recently that “overcorrection” of inherited bone marrow failure in patients with sterile alpha motif domain containing 9 (SAMD9)/9L syndromes by revertant mosaicism induces myelodysplastic syndrome (MDS) with monosomy 7 that occasionally proceeds to acute myelogenous leukemia (AML). In this review, we interpret very complex mechanisms underlying MDS/AML in patients with SAMD9/9L syndromes. This includes multiple myeloid tumor suppressors on the long arm of chromosome 7, all of which act in a haploinsufficient fashion, and a difference in sensitivity to interferon between cells carrying a mutation and revertants. Overcorrection of mutants by somatic mosaicism is likely a novel mechanism in carcinogenesis., A scheme of myelodysplastic syndrome (MDS) carrying ‐7/del(7q). In patients with SAMD9/9L syndromes, bone marrow cells with SAMD9/9L+/− (revertants) show a high sensitivity to growth factors and a low sensitivity to (the suppressive effects of) interferon (IFN). In addition, surrounding bone marrow cells (SAMD9/9L+/mut) have a high sensitivity to IFN. As a result, the rapid expansion of a ‐7/del(7q) clone causes an “overcorrection,” leading to MDS. This mechanism would be partially applied to sporadic MDS patients with ‐7/del(7q) in old age.
- Published
- 2021
- Full Text
- View/download PDF
19. Mosaicism in Cutaneous Disorders.
- Author
-
Lim, Young H., Moscato, Zoe, and Choate, Keith A.
- Abstract
Genetic mosaicism arises when a zygote harbors two or more distinct genotypes, typically due to de novo , somatic mutation during embryogenesis. The clinical manifestations largely depend on the differentiation status of the mutated cell; earlier mutations target pluripotent cells and generate more widespread disease affecting multiple organ systems. If gonadal tissue is spared-as in somatic genomic mosaicism-the mutation and its effects are limited to the proband, whereas mosaicism also affecting the gametes, such as germline or gonosomal mosaicism, is transmissible. Mosaicism is easily appreciated in cutaneous disorders, as phenotypically distinct mutant cells often give rise to lesions in patterns determined by the affected cell type. Genetic investigation of cutaneous mosaic disorders has identified pathways central to disease pathogenesis, revealing novel therapeutic targets. In this review, we discuss examples of cutaneous mosaicism, approaches to gene discovery in these disorders, and insights into molecular pathobiology that have potential for clinical translation. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
20. Revertant mosaicism in genodermatoses.
- Author
-
Lim, Young, Fisher, Jonathan, and Choate, Keith
- Subjects
- *
MOSAICISM , *SKIN disease genetics , *DNA repair , *EPIDERMOLYSIS bullosa , *KERATIN - Abstract
Inherited monogenic skin disorders include blistering disorders, inflammatory disorders, and disorders of differentiation or development. In most cases, the skin is broadly involved throughout the affected individual's lifetime, but rarely, appearance of normal skin clones has been described. In these cases of revertant mosaicism, cells undergo spontaneous correction to ameliorate the effects of genetic mutation. While targeted reversion of genetic mutation would have tremendous therapeutic value, the mechanisms of reversion in the skin are poorly understood. In this review, we provide an overview of genodermatoses that demonstrate widespread reversion and their corrective mechanisms, as well as the current research aimed to understand this 'natural gene therapy'. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
21. Consensus reclassification of inherited epidermolysis bullosa and other disorders with skin fragility
- Subjects
VII COLLAGEN ,GLYCINE SUBSTITUTION ,CYTOPLASMIC DOMAIN ,DISEASE SEVERITY ,EXTRACUTANEOUS MANIFESTATIONS ,COL7A1 ,REVERTANT MOSAICISM ,KINDLER-SYNDROME ,STEM-CELLS ,SPLICE-SITE MUTATION - Abstract
Background: Several new genes and clinical subtypes have been identified since the publication in 2014 of the report of the last International Consensus Meeting on Epidermolysis Bullosa (EB). Objectives: We sought to reclassify disorders with skin fragility, with a focus on EB, based on new clinical and molecular data. Methods: This was a consensus expert review. Results: In this latest consensus report, we introduce the concept of genetic disorders with skin fragility, of which classical EB represents the prototype. Other disorders with skin fragility, where blisters are a minor part of the clinical picture or are not seen because skin cleavage is very superficial, are classified as separate categories. These include peeling skin disorders, erosive disorders, hyperkeratotic disorders, and connective tissue disorders with skin fragility. Because of the common manifestation of skin fragility, these ‘EB-related’ disorders should be considered under the EB umbrella in terms of medical and socioeconomic provision of care. Conclusions: The proposed classification scheme should be of value both to clinicians and researchers, emphasizing both clinical and genetic features of EB. What is already known about this topic?. Epidermolysis bullosa (EB) is a group of genetic disorders with skin blistering. The last updated recommendations on diagnosis and classification were published in 2014. What does this study add?. We introduce the concept of genetic disorders with skin fragility, of which classical EB represents the prototype. Clinical and genetic aspects, genotype–phenotype correlations, disease-modifying factors and natural history of EB are reviewed. Other disorders with skin fragility, e.g. peeling skin disorders, erosive disorders, hyperkeratotic disorders, and connective tissue disorders with skin fragility are classified as separate categories; these ‘EB-related’ disorders should be considered under the EB umbrella in terms of medical and socioeconomic provision of care. Linked Comment: Pope. Br J Dermatol 2020; 183:603.
- Published
- 2020
- Full Text
- View/download PDF
22. Challenges in Treating Genodermatoses: New Therapies at the Horizon
- Author
-
Marie-Anne Morren, Eric Legius, Fabienne Giuliano, Smail Hadj-Rabia, Daniel Hohl, and Christine Bodemer
- Subjects
NETHERTON SYNDROME ,Pharmacology ,RESTORES SKIN INTEGRITY ,VII COLLAGEN ,Science & Technology ,unmet medical needs ,XERODERMA-PIGMENTOSUM ,GENE-THERAPY ,personalised medicine ,REVERTANT MOSAICISM ,RM1-950 ,Review ,reoriented drugs ,PITYRIASIS-RUBRA-PILARIS ,genodermatoses ,FIBROBLAST CELL THERAPY ,Pharmacology (medical) ,Pharmacology & Pharmacy ,DYSTROPHIC EPIDERMOLYSIS-BULLOSA ,Therapeutics. Pharmacology ,genetic engeneering ,cell therapy ,Life Sciences & Biomedicine ,PLURIPOTENT STEM-CELLS - Abstract
Genodermatoses are rare inherited skin diseases that frequently affect other organs. They often have marked effects on wellbeing and may cause early death. Progress in molecular genetics and translational research has unravelled many underlying pathological mechanisms, and in several disorders with high unmet need, has opened the way for the introduction of innovative treatments. One approach is to intervene where cell-signaling pathways are dysregulated, in the case of overactive pathways by the use of selective inhibitors, or when the activity of an essential factor is decreased by augmenting a molecular component to correct disequilibrium in the pathway. Where inflammatory reactions have been induced by a genetically altered protein, another possible approach is to suppress the inflammation directly. Depending on the nature of the genodermatosis, the implicated protein or even on the particular mutation, to correct the consequences or the genetic defect, may require a highly personalised stratagem. Repurposed drugs, can be used to bring about a "read through" strategy especially where the genetic defect induces premature termination codons. Sometimes the defective protein can be replaced by a normal functioning one. Cell therapies with allogeneic normal keratinocytes or fibroblasts may restore the integrity of diseased skin and allogeneic bone marrow or mesenchymal cells may additionally rescue other affected organs. Genetic engineering is expanding rapidly. The insertion of a normal functioning gene into cells of the recipient is since long explored. More recently, genome editing, allows reframing, insertion or deletion of exons or disruption of aberrantly functioning genes. There are now several examples where these stratagems are being explored in the (pre)clinical phase of therapeutic trial programmes. Another stratagem, designed to reduce the severity of a given disease involves the use of RNAi to attenuate expression of a harmful protein by decreasing abundance of the cognate transcript. Most of these strategies are short-lasting and will thus require intermittent life-long administration. In contrast, insertion of healthy copies of the relevant gene or editing the disease locus in the genome to correct harmful mutations in stem cells is more likely to induce a permanent cure. Here we discuss the potential advantages and drawbacks of applying these technologies in patients with these genetic conditions. Given the severity of many genodermatoses, prevention of transmission to future generations remains an important goal including offering reproductive choices, such as preimplantation genetic testing, which can allow selection of an unaffected embryo for transfer to the uterus. ispartof: FRONTIERS IN PHARMACOLOGY vol:12 ispartof: location:Switzerland status: published
- Published
- 2022
23. The categories of cutaneous mosaicism: A proposed classification.
- Author
-
Happle, Rudolf
- Abstract
Mosaic disorders can most easily be studied in the skin. This article presents a comprehensive overview of the different forms of cutaneous mosaicism. Major categories are genomic versus epigenetic mosaicism and nonsegmental versus segmental mosaicism. The class of nonsegmental mosaics includes single point mosaicism as exemplified by solitary benign or malignant skin tumors; disseminated mosaicism as noted in autosomal dominant tumor syndromes such as neurofibromatosis 1; and patchy mosaicism without midline separation as found in giant melanocytic nevus. The class of segmental mosaics includes segmental manifestation of lethal genes surviving by mosaicism as noted in Proteus syndrome; type 1 segmental mosaicism of autosomal dominant skin disorders reflecting heterozygosity for a postzygotic new mutation; type 2 segmental mosaicism of autosomal dominant skin disorders reflecting loss of heterozygosity that occurred at an early developmental stage in a heterozygous embryo; and isolated or superimposed segmental mosaicism of common polygenic skin disorders such as psoriasis or atopic dermatitis. A particular form of genomic mosaicism is didymosis (twin spotting). Revertant mosaicism is recognizable as one or more areas of healthy skin in patients with epidermolysis bullosa or other serious genodermatoses. The category of epigenetic mosaicism includes several X-linked, male lethal disorders such as incontinentia pigmenti, and the patterns of lyonization as noted in X-linked non-lethal disorders such as hypohidrotic ectodermal dysplasia of the Christ-Siemens-Touraine type. An interesting field of future research will be the concept of epigenetic autosomal mosaicism that may explain some unusual cases of autosomal transmission of linear hypo- or hypermelanosis. © 2015 Wiley Periodicals, Inc. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
24. Primary adrenal insufficiency: New genetic causes and their long‐term consequences
- Author
-
Federica Buonocore and John C. Achermann
- Subjects
steroidogenesis ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Review Article ,Disease ,Bioinformatics ,Primary Adrenal Insufficiency ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Addison Disease ,Internal medicine ,Adrenal insufficiency ,medicine ,Humans ,genetics ,Sphingolipidosis ,Congenital adrenal hyperplasia ,IMAGe Syndrome ,Review Articles ,revertant mosaicism ,business.industry ,Cholesterol side-chain cleavage enzyme ,medicine.disease ,3. Good health ,adrenal ,030220 oncology & carcinogenesis ,Addison's disease ,sphingolipidosis ,adrenal insufficiency ,business - Abstract
Primary adrenal insufficiency (PAI) is a potentially life‐threatening condition that requires urgent diagnosis and treatment. Whilst the most common causes are congenital adrenal hyperplasia (CAH) in childhood and autoimmune adrenal insufficiency in adolescence and adulthood, more than 30 other physical and genetics cause of PAI have been reported. Reaching a specific diagnosis can have implications for management and for monitoring associated features, as well as for counselling families about recurrence risk in siblings and relatives. Here, we describe some recent insights into the genetics of adrenal insufficiency and associated molecular mechanisms. We discuss (a) the role of the nuclear receptors DAX‐1 (NR0B1) and steroidogenic factor‐1 (SF‐1, NR5A1) in human adrenal and reproductive dysfunction; (b) multisystem growth restriction syndromes due to gain‐of‐function in the growth repressors CDKN1C (IMAGE syndrome) and SAMD9 (MIRAGE syndrome), or loss of POLE1; (c) nonclassic forms of STAR and P450scc/CYP11A1 insufficiency that present with a delayed‐onset adrenal phenotype and represent a surprisingly prevalent cause of undiagnosed PAI; and (d) a new sphingolipidosis causing PAI due to defects in sphingosine‐1‐phosphate lyase‐1 (SGPL1). Reaching a specific diagnosis can have life‐long implications for management. In some situations, milder or nonclassic forms of these conditions can first present in adulthood and may have been labelled, “Addison's disease.”
- Published
- 2019
- Full Text
- View/download PDF
25. Dystrophic epidermolysis bullosa: a review.
- Author
-
Shinkuma, Satoru
- Subjects
EPIDERMOLYSIS bullosa ,COLLAGEN ,GENETIC mutation ,DISEASE prevalence ,CHROMOSOMES - Abstract
Dystrophic epidermolysis bullosa is a rare inherited blistering disorder caused by mutations in the COL7A1 gene encoding type VII collagen. The deficiency and/or dysfunction of type VII collagen leads to subepidermal blistering immediately below the lamina densa, resulting in mucocutaneous fragility and disease complications such as intractable ulcers, extensive scarring, malnutrition, and malignancy. The disease is usually diagnosed by immunofluorescence mapping and/or transmission electron microscopy and subsequently subclassified into one of 14 subtypes. This review provides practical knowledge on the disease, including new therapeutic strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
26. Vaccine-associated varicella and rubella infections in severe combined immunodeficiency with isolated CD4 lymphocytopenia and mutations in IL 7 R detected by tandem whole exome sequencing and chromosomal microarray.
- Author
-
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., Leduc, M. S., Yang, Y., Eng, C. M., Patel, A., Rodningen, O. K., Muzny, D. M., Gibbs, R. A., Campbell, I. M., Shaw, C. A., and Baker, M. W.
- Subjects
- *
IMMUNODEFICIENCY , *CHICKENPOX vaccines , *DNA , *T cell receptors , *MEDICAL screening - Abstract
In areas without newborn screening for severe combined immunodeficiency ( SCID), disease-defining infections may lead to diagnosis, and in some cases, may not be identified prior to the first year of life. We describe a female infant who presented with disseminated vaccine-acquired varicella ( VZV) and vaccine-acquired rubella infections at 13 months of age. Immunological evaluations demonstrated neutropenia, isolated CD4 lymphocytopenia, the presence of CD8+ T cells, poor lymphocyte proliferation, hypergammaglobulinaemia and poor specific antibody production to VZV infection and routine immunizations. A combination of whole exome sequencing and custom-designed chromosomal microarray with exon coverage of primary immunodeficiency genes detected compound heterozygous mutations (one single nucleotide variant and one intragenic copy number variant involving one exon) within the IL7 R gene. Mosaicism for wild-type allele (20-30%) was detected in pretransplant blood and buccal DNA and maternal engraftment (5-10%) demonstrated in pretransplant blood DNA. This may be responsible for the patient's unusual immunological phenotype compared to classical interleukin ( IL)-7 Rα deficiency. Disseminated VZV was controlled with anti-viral and immune-based therapy, and umbilical cord blood stem cell transplantation was successful. Retrospectively performed T cell receptor excision circle ( TREC) analyses completed on neonatal Guthrie cards identified absent TREC. This case emphasizes the danger of live viral vaccination in severe combined immunodeficiency ( SCID) patients and the importance of newborn screening to identify patients prior to high-risk exposures. It also illustrates the value of aggressive pathogen identification and treatment, the influence newborn screening can have on morbidity and mortality and the significant impact of newer genomic diagnostic tools in identifying the underlying genetic aetiology for SCID patients. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
27. Successful therapeutic transplantation of revertant skin in epidermolysis bullosa.
- Author
-
Gostyński, Antoni, Pasmooij, Anna M.G., and Jonkman, Marcel F.
- Abstract
Background: Epidermolysis bullosa (EB) is a group of genetic blistering diseases. Despite many efforts, treatment for EB remains symptomatic. Revertant mosaicism, coexistence of cells carrying disease-causing mutations with cells in which the inherited mutation is genetically corrected by a spontaneous genetic event (revertant cells) in 1 individual, can be found in EB. The naturally corrected revertant keratinocytes provide an opportunity for autologous cell therapy. Objective: We sought to locally treat EB by transplantation of revertant skin. Methods: Persistent ulcers in a patient with non-Herlitz junctional EB caused by mutations in the LAMB3 gene were treated by transplantation of split-thickness biopsy specimens from one of his revertant patches. Results: All transplanted biopsy specimens were accepted and complete re-epithelialization occurred within 14 days. During 18 months of follow-up, the patient never experienced blisters or wounds in the grafted area, nor in the healed donor site. Immunofluorescence and DNA sequencing showed that acceptor sites healed with transplanted revertant keratinocytes. Limitations: Punch grafting allows only limited expansion of revertant skin. Conclusions: We demonstrate that phenotypical and genotypical correction of skin in patients with revertant mosaicism by expansion of revertant skin might be a promising therapeutic option for cutaneous manifestations of EB. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
28. Consensus reclassification of inherited epidermolysis bullosa and other disorders with skin fragility
- Author
-
Christine Bodemer, Jouni Uitto, Maria C. Bolling, Giovanna Zambruno, Cristina Has, Anna E. Martinez, Adrian Heagerty, Dedee F. Murrell, John A. McGrath, Jemima E. Mellerio, Katsuto Tamai, M.P. Marinkovich, Johann W. Bauer, Francis Palisson, Celia Moss, Jo-David Fine, David T. Woodley, Anja Diem, Agnes Schwieger-Briel, Eli Sprecher, Leena Bruckner-Tuderman, and Alain Hovnanian
- Subjects
medicine.medical_specialty ,VII COLLAGEN ,Consensus ,Classification scheme ,Dermatology ,Kindler syndrome ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,Skin fragility ,CYTOPLASMIC DOMAIN ,Blister ,Disease severity ,Skin blistering ,medicine ,COL7A1 ,Humans ,KINDLER-SYNDROME ,Genetic Association Studies ,Skin ,GLYCINE SUBSTITUTION ,integumentary system ,business.industry ,Inherited epidermolysis bullosa ,REVERTANT MOSAICISM ,medicine.disease ,Natural history ,DISEASE SEVERITY ,EXTRACUTANEOUS MANIFESTATIONS ,Epidermolysis bullosa ,business ,Epidermolysis Bullosa ,STEM-CELLS ,SPLICE-SITE MUTATION - Abstract
Background: Several new genes and clinical subtypes have been identified since the publication in 2014 of the report of the last International Consensus Meeting on Epidermolysis Bullosa (EB). Objectives: We sought to reclassify disorders with skin fragility, with a focus on EB, based on new clinical and molecular data. Methods: This was a consensus expert review. Results: In this latest consensus report, we introduce the concept of genetic disorders with skin fragility, of which classical EB represents the prototype. Other disorders with skin fragility, where blisters are a minor part of the clinical picture or are not seen because skin cleavage is very superficial, are classified as separate categories. These include peeling skin disorders, erosive disorders, hyperkeratotic disorders, and connective tissue disorders with skin fragility. Because of the common manifestation of skin fragility, these ‘EB-related’ disorders should be considered under the EB umbrella in terms of medical and socioeconomic provision of care. Conclusions: The proposed classification scheme should be of value both to clinicians and researchers, emphasizing both clinical and genetic features of EB. What is already known about this topic?. Epidermolysis bullosa (EB) is a group of genetic disorders with skin blistering. The last updated recommendations on diagnosis and classification were published in 2014. What does this study add?. We introduce the concept of genetic disorders with skin fragility, of which classical EB represents the prototype. Clinical and genetic aspects, genotype–phenotype correlations, disease-modifying factors and natural history of EB are reviewed. Other disorders with skin fragility, e.g. peeling skin disorders, erosive disorders, hyperkeratotic disorders, and connective tissue disorders with skin fragility are classified as separate categories; these ‘EB-related’ disorders should be considered under the EB umbrella in terms of medical and socioeconomic provision of care. Linked Comment: Pope. Br J Dermatol 2020; 183:603.
- Published
- 2020
29. Somatic gene mutation and human disease other than cancer: An update
- Author
-
Erickson, Robert P.
- Subjects
- *
MOSAICISM , *GENETIC mutation , *GENES , *GENETIC disorders , *DUCHENNE muscular dystrophy , *GERM cells , *GENOMICS , *GENETICS , *BIOLOGICAL variation - Abstract
Abstract: Somatic mosaicism is well known in disorders where the manifestations are readily seen, e.g. the skin in neurofibromatosis I. In single gene disorders of higher frequency, especially X-linked ones, the frequency of combined germ-line and somatic mosaicism is increasingly being appreciated, e.g. Duchenne Muscular Dystrophy. Cell separation techniques; such as the fluorescence-activated cell sorter (FACS) also detect much somatic mosaicism among blood cells in disorders such as paroxysmal nocturnal hemoglobinuria. Depending on the disorder and the class of mutation, in genes for which there are sufficient numbers of patients studied, 6–20% of cases are due to somatic mutation. This update of my previous review is stimulated by the rapid application of new technologies for the study of DNA variation in disease. The results of these studies implicate somatic mutation in a greater variety of genetic diseases and a wider spectrum of tissues than have previously been shown, including heart and kidney. The classes of mutation have also expanded beyond base pair changes, insertions/deletion (indels), and short tandem repeat mutations to include copy number variants and transposon-mediated mutations. I also briefly discuss previously well-known mosaicism for chromosomal mutations. Genomic sequencing, performed on DNA from blood, shows many mutations which are conclusively somatic in origin. It is still too early to see if there is a different pattern of somatic mutation compared to germ-line mutation. Though the parameters to allow careful quantification are not yet available, it seems that the frequency of gene mutation in embryonic cells is not markedly different than that in the germ line. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
30. Adhesive stripping to remove epidermis in junctional epidermolysis bullosa for revertant cell therapy.
- Author
-
Gostynski, A., Deviaene, F. C. L., Pasmooij, A. M. G., Pas, H. H., and Jonkman, M. F.
- Subjects
- *
EPIDERMOLYSIS bullosa , *KERATINOCYTES , *GENE therapy , *CELLULAR therapy , *EPIDERMIS , *ADHESIVE tape - Abstract
Background Replacing mutant skin in epidermolysis bullosa (EB) by epithelial sheets of transduced autologous keratinocytes is the essential surgical step of ex vivo gene therapy. The same applies for revertant cell therapy in which epithelial sheets of revertant autologous keratinocytes are used. Revertant cells can be found in patches of normal skin in patients with junctional EB (JEB) due to revertant mosaicism caused by in vivo reversions. Objectives To develop a technique of adhesive tape stripping as a method for epidermis removal to prepare the acceptor site for revertant cell therapy in a patient with revertant mosaic JEB. Methods We performed revertant cell therapy on a patient with mosaic type XVII collagen-deficient non-Herlitz JEB. Skin biopsies were taken from revertant skin on the wrist. Graft production took place on a 3T3-J2 feeder layer resulting in two 6 × 7 cm grafts. An innovative method that uses the pathological plane of least resistance of JEB skin was developed to prepare the acceptor site. A polyacrylate adhesive plaster was placed on the skin and then pulled off with the epidermis. Results The epidermis was easily removed with the plaster. The skin separated at the level of the lamina lucida, leaving a bloodless wound bed of naked lamina densa. Transplantation was successful; the acceptor site healed without scarring. However, blistering could be provoked. The functional repair was not achieved due to the low percentage of revertant cells in the graft. Conclusions We conclude that adhesive stripping is a simple, effective and almost painless procedure for removing epidermis for ex vivo cell therapy in EB. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
31. Revertant Mosaicism in Epidermolysis Bullosa.
- Author
-
Meyer-Mueller, Cameron, Osborn, Mark J., Tolar, Jakub, Boull, Christina, and Ebens, Christen L.
- Subjects
MOSAICISM ,GENE conversion ,GENE therapy ,EPIDERMOLYSIS bullosa ,RECOMBINANT proteins ,CELLULAR therapy ,GENETIC transformation ,GENETIC mutation - Abstract
Epidermolysis bullosa (EB) is a group of genetic blistering diseases characterized by mechanically fragile skin and mucocutaneous involvement. Historically, disease management has focused on supportive care. The development of new genetic, cellular, and recombinant protein therapies has shown promise, and this review summarizes a unique gene and cell therapy phenomenon termed revertant mosaicism (RM). RM is the spontaneous correction of a disease-causing mutation. It has been reported in most EB subtypes, some with relatively high frequency, and has been observed in both keratinocytes and fibroblasts. RM manifests as identifiable patches of unaffected, blister-resistant skin and can occur through a variety of molecular mechanisms, including true back mutation, intragenic crossover, mitotic gene conversion, and second-site mutation. RM cells represent a powerful autologous platform for therapy, and leveraging RM cells as a therapeutic substrate may avoid the inherent mutational risks of gene therapy/editing. However, further examination of the genomic integrity and long-term functionality of RM-derived cells, as well in vivo testing of systemic therapies with RM cells, is required to realize the full therapeutic promise of naturally occurring RM in EB. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
32. Somatic mosaicism of an intragenicFANCBduplication in both fibroblast and peripheral blood cells observed in a Fanconi anemia patient leads to milder phenotype
- Author
-
Adrianna Vlachos, Rajalakshmi S. Asur, James W. Thomas, Morgan Park, Frank X. Donovan, Moonjung Jung, Agata Smogorzewska, Arleen D. Auerbach, Danielle C. Kimble, Settara C. Chandrasekharappa, Raymond J. Noonan, Francis P. Lach, Peter S. Chines, and Aparna Kamat
- Subjects
Male ,0301 basic medicine ,milder phenotype ,Adolescent ,Genotype ,intragenic duplication ,Biology ,FANCB ,Homology (biology) ,droplet digital PCR ,03 medical and health sciences ,Exon ,Genes, X-Linked ,Fanconi anemia ,Gene Duplication ,FANCD2 ,Gene duplication ,Genetics ,medicine ,Humans ,Allele ,Molecular Biology ,Alleles ,Genetics (clinical) ,revertant mosaicism ,Blood Cells ,Base Sequence ,Mosaicism ,Breakpoint ,Exons ,Original Articles ,Fibroblasts ,medicine.disease ,Fanconi Anemia Complementation Group Proteins ,3. Good health ,Fanconi Anemia ,Phenotype ,030104 developmental biology ,Original Article - Abstract
Background Fanconi anemia (FA) is a rare disorder characterized by congenital malformations, progressive bone marrow failure, and predisposition to cancer. Patients harboring X‐linked FANCB pathogenic variants usually present with severe congenital malformations resembling VACTERL syndrome with hydrocephalus. Methods We employed the diepoxybutane (DEB) test for FA diagnosis, arrayCGH for detection of duplication, targeted capture and next‐gen sequencing for defining the duplication breakpoint, PacBio sequencing of full‐length FANCB aberrant transcript, FANCD2 ubiquitination and foci formation assays for the evaluation of FANCB protein function by viral transduction of FANCB‐null cells with lentiviral FANCB WT and mutant expression constructs, and droplet digital PCR for quantitation of the duplication in the genomic DNA and cDNA. Results We describe here an FA‐B patient with a mild phenotype. The DEB diagnostic test for FA revealed somatic mosaicism. We identified a 9154 bp intragenic duplication in FANCB, covering the first coding exon 3 and the flanking regions. A four bp homology (GTAG) present at both ends of the breakpoint is consistent with microhomology‐mediated duplication mechanism. The duplicated allele gives rise to an aberrant transcript containing exon 3 duplication, predicted to introduce a stop codon in FANCB protein (p.A319*). Duplication levels in the peripheral blood DNA declined from 93% to 7.9% in the span of eleven years. Moreover, the patient fibroblasts have shown 8% of wild‐type (WT) allele and his carrier mother showed higher than expected levels of WT allele (79% vs. 50%) in peripheral blood, suggesting that the duplication was highly unstable. Conclusion Unlike sequence point variants, intragenic duplications are difficult to precisely define, accurately quantify, and may be very unstable, challenging the proper diagnosis. The reversion of genomic duplication to the WT allele results in somatic mosaicism and may explain the relatively milder phenotype displayed by the FA‐B patient described here.
- Published
- 2017
- Full Text
- View/download PDF
33. Spontaneous remission in a Diamond-Blackfan anaemia patient due to a revertant uniparental disomy ablating a de novo RPS19 mutation
- Author
-
Paola Quarello, Tommaso Pippucci, Irma Dianzani, Cecilia Mancini, Emanuela Garelli, Alfredo Brusco, Elisa Giorgio, Orazio Palumbo, Nicoletta Crescenzio, Ugo Ramenghi, Eleonora Di Gregorio, Paola Dimartino, Adriana Carando, Massimo Carella, and Elisa Menegatti
- Subjects
0301 basic medicine ,Adult ,Male ,Ribosomal Proteins ,medicine.medical_specialty ,RPS19 ,complete remission ,Diamond-Blackfan ,revertant mosaicism ,uniparental disomy ,Hematology ,Revertant ,Spontaneous remission ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Anemia, Diamond-Blackfan ,business.industry ,Complete remission ,A diamond ,Uniparental Disomy ,medicine.disease ,Uniparental disomy ,030104 developmental biology ,Mutation (genetic algorithm) ,Mutation ,Cancer research ,business ,030215 immunology - Published
- 2018
34. Pigmentation and melanocyte supply to the epidermis depend on type XVII collagen.
- Author
-
Gostyński, Antoni, Pasmooij, Anna M.G., Rio, Marcela, Diercks, Gilles F., Pas, Hendri H., and Jonkman, Marcel F.
- Subjects
- *
MELANOCYTES , *EPIDERMOLYSIS bullosa , *MOSAICISM , *SOMATIC mutation , *HYPERPIGMENTATION - Abstract
Genetic deficiency of type XVII collagen (C17), laminin-332 or type VII collagen causes epidermolysis bullosa ( EB). Spontaneous correction of the deficiency, also known as revertant mosaicism, is caused by a second somatic mutation that restores protein expression resulting in clinically healthy (revertant) patches surrounded by fragile (mutant) skin. Interestingly, in some patients, patches of revertant skin show hyperpigmentation. To study the possible role of affected proteins in pigmentation and melanocyte distribution, we investigated clinical documentation and skin biopsy specimens of 13 revertant EB patients having correcting mutations in the COL17A1, LAMB3 or COL7A1 genes. Analysis revealed that lack of C17 led to decreased melanin intensity and melanocyte density in the epidermis when compared with the revertant patches. Reversions of LAMB3 and COL7A1 in keratinocytes did not influence clinical pigmentation or density of melanocytes. We conclude that in human skin, melanocyte supply to the epidermis depends on C17 expression in keratinocytes. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
35. Pigmentation and melanocyte supply to the epidermis depend on type XVII collagen
- Subjects
revertant mosaicism ,melanocyte ,integumentary system ,JUNCTIONAL EPIDERMOLYSIS-BULLOSA ,IL-8 ,NICHE ,melanocyte stem cell ,VITILIGO ,COL17A1 MUTATIONS ,CELLS ,NATURAL GENE-THERAPY ,pigmentation ,epidermolysis bullosa ,type XVII collagen - Abstract
Genetic deficiency of type XVII collagen (C17), laminin-332 or type VII collagen causes epidermolysis bullosa (EB). Spontaneous correction of the deficiency, also known as revertant mosaicism, is caused by a second somatic mutation that restores protein expression resulting in clinically healthy (revertant) patches surrounded by fragile (mutant) skin. Interestingly, in some patients, patches of revertant skin show hyperpigmentation. To study the possible role of affected proteins in pigmentation and melanocyte distribution, we investigated clinical documentation and skin biopsy specimens of 13 revertant EB patients having correcting mutations in the COL17A1, LAMB3 or COL7A1 genes. Analysis revealed that lack of C17 led to decreased melanin intensity and melanocyte density in the epidermis when compared with the revertant patches. Reversions of LAMB3 and COL7A1 in keratinocytes did not influence clinical pigmentation or density of melanocytes. We conclude that in human skin, melanocyte supply to the epidermis depends on C17 expression in keratinocytes.
- Published
- 2014
- Full Text
- View/download PDF
36. Long-term follow-up of patients with recessive dystrophic epidermolysis bullosa in the Netherlands
- Author
-
Hendri H. Pas, Robert M.W. Hofstra, Miranda Nijenhuis, Peter C. van den Akker, Marcel F. Jonkman, Hans Scheffer, Anthonie J. van Essen, Rowdy Meijer, Gonnie Meijer, Marian M.J. Kraak, and Translational Immunology Groningen (TRIGR)
- Subjects
Male ,Pathology ,Recessive dystrophic epidermolysis bullosa ,Neuroinformatics [DCN 3] ,medicine.disease_cause ,Biochemistry ,DOMAIN ,Genotype ,COL7A1 ,Child ,Netherlands ,Mutation ,MESSENGER-RNA DECAY ,Exons ,REVERTANT MOSAICISM ,Middle Aged ,Phenotype ,ANCHORING FIBRILS ,Epidermolysis Bullosa Dystrophica ,Pedigree ,NONSENSE ,Child, Preschool ,Type VII collagen ,DISEASE SEVERITY ,Female ,Mechanobullous skin disease ,Adult ,medicine.medical_specialty ,Collagen Type VII ,Adolescent ,Genes, Recessive ,Phenotype-genotype correlations ,Dermatology ,Biology ,Genomic disorders and inherited multi-system disorders [IGMD 3] ,Young Adult ,COL7A1 MUTATION ,Microscopy, Electron, Transmission ,Anchoring fibrils ,VII COLLAGEN GENE ,medicine ,Humans ,Genetic Predisposition to Disease ,Molecular Biology ,Gene ,Aged ,EXONIC SPLICING ENHANCERS ,Infant ,Immunostaining ,SKIN ,Follow-Up Studies - Abstract
Background: The current classification of recessive dystrophic epidermolysis bullosa (RDEB) comprises two major subtypes: 'severe generalized RDEB'(RDEB-sev gen) with early-onset, extensive, generalized blistering and scarring, complete absence of type VII Collagen, and bi-allelic COL7A1 null mutations; milder 'generalized other RDEB' (RDEB-O) with reduced-to-normal type VII Collagen expression, and non-null genotypes.Objective: To search for previously unrecognized phenotype-genotype correlations in 33 Dutch RDEB families.Methods: We analyzed extensive clinical follow-up data, available for all patients up to 19 years, detailed type VII Collagen immunostaining and genotypes, and correlated clinical phenotype to molecular phenotype and genotype.Results: We identified 20 novel COL7A1 mutations. In 14 of 15 RDEB-sev gen patients type VII Collagen was completely absent, one had strongly reduced type VII Collagen, and all carried bi-allelic null mutations. Five of 11 RDEB-O patients developed pseudosyndactyly of the fingers preceded by skin atrophy and flexion contractures later in childhood and adolescence. All five had esophageal involvement and growth retardation. Type VII collagen immunostaining ranged from strongly reduced to slightly reduced in RDEB-O patients with pseudosyndactyly, whereas RDEB-O patients without pseudosyndactyly had slightly reduced to normal type VII Collagen staining. There was no difference in genotypes between both groups, although we unexpectedly found bi-allelic null mutations in two of five RDEB-O patients with pseudosyndactyly.Conclusion: Pseudosyndactyly occurs in approximately half of RDEB-O patients when type VII Collagen is strongly reduced. The prognosis in RDEB cannot always be simply predicted from the COL7A1 genotype. (C) 2009 Japanese Society for Investigative Dermatology. Published by Elsevier Ireland Ltd. All rights reserved.
- Published
- 2009
- Full Text
- View/download PDF
37. Adhesive stripping to remove epidermis in junctional epidermolysis bullosa for revertant cell therapy
- Author
-
Anna M.G. Pasmooij, A. Gostynski, F. C. L. Deviaene, Hendri H. Pas, M. F. Jonkman, and Translational Immunology Groningen (TRIGR)
- Subjects
medicine.medical_specialty ,Pathology ,Cell Transplantation ,Cosmetic Techniques ,MOSAICISM ,Dermatology ,Junctional epidermolysis bullosa (medicine) ,Cell therapy ,Cell Adhesion ,revertant cell therapy ,medicine ,Humans ,type XVII collagen ,revertant mosaicism ,Wound Healing ,Tissue Scaffolds ,integumentary system ,TRANSPLANTATION ,business.industry ,medicine.disease ,Lamina lucida ,Transplantation ,medicine.anatomical_structure ,Female ,Tissue Adhesives ,Lamina densa ,Epidermolysis bullosa ,Epidermis ,Epidermolysis Bullosa, Junctional ,business ,Ex vivo - Abstract
BackgroundReplacing mutant skin in epidermolysis bullosa (EB) by epithelial sheets of transduced autologous keratinocytes is the essential surgical step of ex vivo gene therapy. The same applies for revertant cell therapy in which epithelial sheets of revertant autologous keratinocytes are used. Revertant cells can be found in patches of normal skin in patients with junctional EB (JEB) due to revertant mosaicism caused by in vivo reversions.ObjectivesTo develop a technique of adhesive tape stripping as a method for epidermis removal to prepare the acceptor site for revertant cell therapy in a patient with revertant mosaic JEB.MethodsWe performed revertant cell therapy on a patient with mosaic type XVII collagen-deficient non-Herlitz JEB. Skin biopsies were taken from revertant skin on the wrist. Graft production took place on a 3T3-J2 feeder layer resulting in two 6 x 7 cm grafts. An innovative method that uses the pathological plane of least resistance of JEB skin was developed to prepare the acceptor site. A polyacrylate adhesive plaster was placed on the skin and then pulled off with the epidermis.ResultsThe epidermis was easily removed with the plaster. The skin separated at the level of the lamina lucida, leaving a bloodless wound bed of naked lamina densa. Transplantation was successful; the acceptor site healed without scarring. However, blistering could be provoked. The functional repair was not achieved due to the low percentage of revertant cells in the graft.ConclusionsWe conclude that adhesive stripping is a simple, effective and almost painless procedure for removing epidermis for ex vivo cell therapy in EB.
- Published
- 2009
- Full Text
- View/download PDF
38. Localized and generalized forms of blistering in junctional epidermolysis bullosa due to COL17A1 mutations in the Netherlands
- Author
-
Hendri H. Pas, G. H. L. Jansen, Anna M.G. Pasmooij, Henny H. Lemmink, M. F. Jonkman, and Translational Immunology Groningen (TRIGR)
- Subjects
Male ,Pathology ,Turkey ,ECTODOMAIN ,BP180 ,medicine.disease_cause ,Junctional epidermolysis bullosa (medicine) ,Autoantigens ,Blister ,DOMAIN ,Genotype ,Amelogenesis imperfecta ,deletion ,Child ,Netherlands ,Mutation ,medicine.diagnostic_test ,integumentary system ,PEMPHIGOID ANTIGEN ,REVERTANT MOSAICISM ,Middle Aged ,Non-Fibrillar Collagens ,Phenotype ,Child, Preschool ,Female ,SQUAMOUS-CELL CARCINOMA ,Epidermolysis Bullosa, Junctional ,genodermatosis ,Adult ,medicine.medical_specialty ,COL17A1 ,Dermatology ,Immunofluorescence ,COLLAGEN GENE COL17A1 ,Antigen ,medicine ,Humans ,Aged ,GLYCINE SUBSTITUTION ,MILD FORM ,business.industry ,nH-JEB ,Genodermatosis ,medicine.disease ,XVII COLLAGEN ,Microscopy, Fluorescence ,business - Abstract
Background Mutations in the gene COL17A1 coding for type XVII collagen cause non-Herlitz junctional epidermolysis bullosa (nH-JEB).Objectives Here we give an overview of the genotype-phenotype correlation in 12 patients from the Netherlands with type XVII collagen-deficient nH-JEB.Patient and methods Family and personal history and clinical presentation were recorded from each patient, and skin biopsies of intact and bullous skin were taken for immunofluorescence and electron microscopy. The mutations were identified by analysing the patient's DNA isolated from peripheral blood cells.Results DNA analysis identified five novel deletions: 1284delA, 1365delC, 3236delT, 3600-3601delCT and 4425delT. Interestingly, we identified a new patient, homozygous for 4425delT, with an exceptionally mild blistering phenotype. All together, three patients had more localized blistering confined to hands, lower legs and face, absent or very mild nail dystrophy, normal primary hair and sparse secondary hair. Nine patients had generalized blistering, nail dystrophy, sparse primary and absent secondary hair. All 12 patients had amelogenesis imperfecta (enamel pitting). Immunofluorescence (IF) antigen mapping with monoclonal antibodies 1A8C and 1D1 that bind to type XVII collagen, but not to its 97-kDa fragment was completely negative in patients with generalized blistering, whereas reduced in patients with localized blistering.Conclusions Our data reveal that in patients with COL17A1 mutations a localized nH-JEB phenotype can be differentiated from a generalized nH-JEB phenotype by IF antigen mapping. The data are important for genetic counselling at early age when the clinical phenotype is not yet clear.
- Published
- 2007
39. Natural repair mechanisms in correcting pathogenic mutations in inherited skin disorders
- Subjects
SPLICING ENHANCER ,JUNCTIONAL EPIDERMOLYSIS-BULLOSA ,DNA OLIGONUCLEOTIDE ,SIMPLEX ,NONSENSE MUTATION ,EXON ,REVERTANT MOSAICISM ,GENE ,SEQUENCE ,PATIENT - Abstract
This review assesses molecular aspects of the rescue of disease-causing mutations in genodermatoses by means of naturally occurring secondary genetic phenomena. Such data have important implications for the design of gene therapy approaches for inherited skin diseases. Reversal of the phenotype depends on three elements: the number of cells involved; the degree of gene reversal; and the specific timing of the reversion. If reversion occurs in somatic cells, revertant mosaicism may occur. This is the situation in which a patient's skin is generally affected by the genodermatosis, but islands of normal skin stand out. These reflect the presence of revertant cells that are sufficient to restore a normal local skin phenotype. Reversion of the original mutation may also be partial, in which case the phenotype may display no, or only limited, improvement. Nevertheless, the phenotype may ameliorate with age if the reverted cells preferentially expand in time or if the time of onset of reversion is after birth. In essence, the complexities of naturally occurring rescue processes are important to understand because the inherent mechanisms may provide clues and insight into optimal therapeutic gene manipulation, and the possibility of mimicking nature in the management of patients with diverse genodermatoses.
- Published
- 2003
40. Mechanisms of Natural Gene Therapy in Dystrophic Epidermolysis Bullosa
- Author
-
Marcela Del Rio, Marcel F. Jonkman, Cristina Has, Marta García, Renske Brander, María José Escámez, Rowdy Meijer, Anna M.G. Pasmooij, J. Kohlhase, Hans Scheffer, Dimitra Kiritsi, Leena Bruckner-Tuderman, Peter C. van den Akker, and Translational Immunology Groningen (TRIGR)
- Subjects
Adult ,Mitotic crossover ,Collagen Type VII ,Adolescent ,Genetic enhancement ,Reversion ,Fluorescent Antibody Technique ,Germline mosaicism ,Dermatology ,Biology ,medicine.disease_cause ,Biochemistry ,CLASSIFICATION ,COL17A1 MUTATIONS ,COL7A1 MUTATION ,medicine ,Missense mutation ,Humans ,COHORT ,Child ,Molecular Biology ,EPIDERMIS ,Genetics ,Mutation ,Mosaicism ,Genetic Therapy ,Cell Biology ,REVERTANT MOSAICISM ,Middle Aged ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,medicine.disease ,Phenotype ,Epidermolysis Bullosa Dystrophica ,MITOTIC RECOMBINATION ,Epidermolysis bullosa ,STEM-CELLS - Abstract
Item does not contain fulltext Revertant mosaicism has been reported in several inherited diseases, including the genetic skin fragility disorder epidermolysis bullosa (EB). Here, we describe the largest cohort of seven patients with revertant mosaicism and dystrophic EB (DEB), associated with mutations in the COL7A1 gene, and determine the underlying molecular mechanisms. We show that revertant mosaicism occurs both in autosomal dominantly and recessively inherited DEB. We found that null mutations resulting in complete loss of collagen VII and severe disease, as well as missense or splice-site mutations associated with some preserved collagen VII function and a milder phenotype, were corrected by revertant mosaicism. The mutation, subtype, and severity of the disease are thus not decisive for the presence of revertant mosaicism. Although collagen VII is synthesized and secreted by both keratinocytes and fibroblasts, evidence for reversion was only found in keratinocytes. The reversion mechanisms included back mutations/mitotic recombinations in 70% of the cases and second-site mutations affecting splicing in 30%. We conclude that revertant mosaicism is more common than previously assumed in patients with DEB, and our findings will have implications for future therapeutic strategies using the patient's naturally corrected cells as a source for cell-based therapies.
- Published
- 2014
41. Applications of Next-Generation Sequencing to Rare Disease
- Author
-
Lee, Catherine
- Subjects
- Adrenoleukodystrophy, Brain Microvascular Endothelial Cells, Rare Disease, Recessive Dystrophic Epidermolysis Bullosa, Revertant Mosaicism, Single-cell RNA-seq
- Abstract
Since the discovery of the structure of DNA in 1953, researchers and clinicians have been painstakingly paving the way for the use of genetic information in the treatment of disease. In order for this to be possible, specific genetic targets must be identified. For this dissertation, I use next generation, single-cell, and third generation RNA-sequencing techniques to identify markers of genetic heterogeneity and potential therapeutic targets in the rare diseases recessive dystrophic epidermolysis bullosa (RDEB) and cerebral childhood adrenoleukodystrophy (ccALD). RDEB is a an inherited blistering disorder caused by mutations in the key structural skin protein, type VII collagen (C7). It can partially treated by hematopoietic stem cell transplant (HSCT), however, how blistered RDEB skin signals to donor cells is unknown. In Chapter 2, to identify potential signals, I performed single-cell RNA-seq (scRNA-seq) on patient fibroblasts and implemented a variance-driven multitask clustering (scVDMC), that utilizes multiple single-cell populations from biological replicates or different samples. scVDMC clusters single cells in multiple scRNA-seq experiments of similar cell types and markers but varying expression patterns such that the scRNA-seq data are better integrated than typical pooled analyses which only increase the sample size. By controlling the variance among the cell clusters within each dataset and across all the datasets, scVDMC detects cell sub-populations in each individual experiment with shared cell-type markers but varying cluster centers among all the experiments. scVDMC was then applied to two previously published scRNA-seq datasets with several replicates and one large-scale Drop-seq dataset on three patient samples. scVDMC more accurately detected cell populations and known cell markers than pooled clustering and other recently proposed scRNA-seq clustering methods. When applied to the scRNA-seq RDEB patient fibroblast data, scVDMC revealed several new cell types and unknown markers that I validated by flow cytometry. ccALD is caused by mutations in the \emph{ABCD1} gene and manifests in early childhood with neuropathological symptoms and hyper-pigmentation, culminating in massive breakdown of the blood-brain barrier (BBB) and death if HSCT is not performed at an early stage. It is difficult to model the BBB of this disease as primary cells do not recapitulate the barrier in culture and the mouse model shows incomplete penetrance. In Chapter 3, I model the blood-brain barrier of ccALD patients and wild-type (WT) controls using directed differentiation of induced pluripotent stem cells (iPSCs) into induced brain microvascular endothelial cells (iBMECs). Immunocytochemistry and PCR confirmed characteristic expression of brain microvascular endothelial cell (BMEC) markers. Barrier properties of iBMECs were measured via trans-endothelial electrical resistance (TEER), sodium fluorescein permeability, and frayed junction analysis. Electron microscopy and RNA-seq were used to further characterize disease-specific differences. Oil-Red-O staining was used to quantify differences in lipid accumulation. To evaluate whether treatment with block copolymers of poly(ethylene oxide) and poly(propylene oxide) (PEO-PPO) could mitigate defective properties, ccALD-iBMECs were treated with PEO-PPO block copolymers and their barrier properties and lipid accumulation levels were quantified. iBMECs from patients with ccALD had significantly decreased TEER (2592 ± 110 $\Omega \cdot cm^2$) compared to WT controls (5001 ± 172 $\Omega \cdot cm^2$). They also accumulated lipid droplets to a greater extent than WT-iBMECs. Upon treatment with a PEO-PPO diblock copolymer during the differentiation process, an increase in TEER and a reduction in lipid accumulation were observed for the polymer treated ccALD-iBMECs compared to untreated controls. The finding that BBB integrity is decreased in ccALD and can be rescued with block copolymers opens the door for the discovery of BBB-specific molecular markers that can indicate the onset of ccALD and has therapeutic implications for preventing the conversion to ccALD. Revertant mosaicism in RDEB patients is seen as patches of skin that have never blistered. At the molecular level, these patches of skin contain detectable amounts of C7, indicating that a reversion of the disease-causing mutation has occurred at the DNA level. One of the limited treatment options available for treating RDEB is the use of C7 expressing stem cells or differentiated skin cells to replace C7 at the dermal-epidermal junction and restore the overall integrity of the skin architecture. However, this typically requires the use of gene therapy or allogeneic cells, which can be costly and cause adverse reactions in the recipient. Mosaic cells could potentially be used for these purposes, however, isolating and purifying them has proven difficult. In Chapter 4, I describe a method utilizing synthetic micro RNA (miR) switches, whereby differences in endogenous miR activity are exploited to purify mosaic cells in culture, which may be useful in generating pure populations of mosaic cells that can then be used in future clinical applications. Chapter 5 of this dissertation uses third generation or long read sequencing to look more closely at the underlying genetic event resulting in mosaic expression in one particular RDEB patient. These studies identify genetic heterogeneity in cell types relevant to the respective rare diseases being examined and give support to developing precision medicine techniques to treat these rare diseases.
- Published
- 2018
42. Natural Gene Therapy May Occur in All Patients with Generalized Non-Herlitz Junctional Epidermolysis Bullosa with COL17A1 Mutations
- Author
-
Anna M.G. Pasmooij, Marcel F. Jonkman, Miranda Nijenhuis, Renske Brander, and Translational Immunology Groningen (TRIGR)
- Subjects
Adult ,Male ,DNA Repair ,DNA repair ,Somatic cell ,Genetic enhancement ,Remission, Spontaneous ,RECOMBINATION ,Locus (genetics) ,Dermatology ,Biology ,Biochemistry ,Autoantigens ,Exon ,Germline mutation ,INDEL Mutation ,SIMPLEX ,LOCUS ,Humans ,Child ,Gene ,Molecular Biology ,Germ-Line Mutation ,Aged ,Genetics ,MILD FORM ,integumentary system ,Mosaicism ,Cell Biology ,Genetic Therapy ,Sequence Analysis, DNA ,REVERTANT MOSAICISM ,Middle Aged ,Non-Fibrillar Collagens ,REVERSION ,Molecular biology ,XVII COLLAGEN ,Female ,AUTOANTIGEN ,Epidermolysis Bullosa ,MESSENGER-RNA ,STEM-CELLS - Abstract
Mutations in the type XVII collagen gene (COL17A1) result in the blistering disorder non-Herlitz junctional epidermolysis bullosa (JEB-nH). The incidence of revertant mosaicism, also called "natural gene therapy", was identified in a cohort of 14 patients with JEB-nH caused by COL17A1 mutations in the Netherlands. Five different in vivo reversions, all correcting the germ-line COL17A1 mutation c.2237delG in exon 30, were found in four mosaic JEB-nH patients. The correcting DNA changes involved a wide variety of somatic mutations, from which an indel mutation (c.2228-101_2263 + 70delins15) and a large deletion of 2,165 base pairs (c.2227 + 153_2336-318del) have not been previously observed in patients with revertant mosaicism. Our results show that there is no preference for a repair mechanism. Moreover, revertant mosaicism was confirmed on a DNA level in 6 out of 10 generalized JEB-nH patients. Further, photo-material and clinical history of the other four generalized JEB-nH patients demonstrated that each patient has revertant skin patches. In contrast, revertant mosaicism was not detected in the four localized JEB-nH patients. The fact that so many, if not all, generalized JEB-nH COL17A1 patients have revertant patches offers opportunities for cell therapies in which the patient's own naturally corrected cells are used as a source.
- Published
- 2012
43. Natural gene therapy in dystrophic epidermolysis bullosa
- Author
-
Gonnie Meijer, Peter C. van den Akker, Marcel F. Jonkman, Anna M.G. Pasmooij, Robert M.W. Hofstra, Miranda Nijenhuis, and Translational Immunology Groningen (TRIGR)
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,VII COLLAGEN ,Collagen Type VII ,Nonsense mutation ,DNA Mutational Analysis ,Dermatology ,medicine.disease_cause ,PATIENT ,Young Adult ,Germline mutation ,SIMPLEX ,medicine ,Humans ,EPIDERMIS ,Mutation ,medicine.diagnostic_test ,integumentary system ,business.industry ,MUTATIONS ,TRANSPLANTATION ,Mosaicism ,Homozygote ,Epidermolysis bullosa dystrophica ,General Medicine ,REVERTANT MOSAICISM ,medicine.disease ,REVERSION ,Skin patch ,Epidermolysis Bullosa Dystrophica ,Transplantation ,DOMINANT ,Codon, Nonsense ,Skin biopsy ,Epidermolysis bullosa ,business ,STEM-CELLS - Abstract
Background: Dystrophic epidermolysis bullosa is a genetic blistering disorder caused by mutations in the type VII collagen gene, COL7A1. In revertant mosaicism, germline mutations are corrected by somatic events resulting in a mosaic disease distribution. This "natural gene therapy" phenomenon long has been recognized in other forms of epidermolysis bullosa but only recently in dystrophic epidermolysis bullosa.Observations: We describe a 21-year-old man with recessive dystrophic epidermolysis bullosa carrying the homozygous c.6508C>T (p.Gln2170X) nonsense mutation who reported an unaffected skin patch on his neck where blisters never had occurred. Immunofluorescent type VII collagen staining was normal in 80% of the unaffected skin biopsy; however, it was strongly reduced in the affected skin. In the unaffected skin, the somatic nucleotide substitution c.6510G>T reverted line nonsense codon to tyrosine (p.Gln21.70Tyr), thereby restoring functional protein production.Conclusions: Revertant mosaicism is considered rare in recessive dystrophic epidermolysis bullosa. However, it might be more common than previously anticipated because our patient is the third in whom revertant mosaicism was identified in a short period of time. The correction mechanism is different than that previously reported. Systematic examination of patients with recessive dystrophic epidermolysis bullosa, therefore, will likely reveal more patients with revertant patches. This is important because the natural gene therapy phenomenon may provide opportunities for revertant cell therapy.
- Published
- 2011
44. Dystrophic epidermolysis bullosa: a review
- Author
-
Satoru Shinkuma
- Subjects
revertant mosaicism ,Pathology ,medicine.medical_specialty ,treatment ,medicine.diagnostic_test ,business.industry ,subtypes ,Genetic enhancement ,Mucocutaneous zone ,Review ,Dermatology ,Disease ,Immunofluorescence ,Malignancy ,medicine.disease ,gene therapy ,Dystrophic epidermolysis bullosa ,type VII collagen ,Type VII collagen ,medicine ,Lamina densa ,business ,anchoring fibril - Abstract
Dystrophic epidermolysis bullosa is a rare inherited blistering disorder caused by mutations in the COL7A1 gene encoding type VII collagen. The deficiency and/or dysfunction of type VII collagen leads to subepidermal blistering immediately below the lamina densa, resulting in mucocutaneous fragility and disease complications such as intractable ulcers, extensive scarring, malnutrition, and malignancy. The disease is usually diagnosed by immunofluorescence mapping and/or transmission electron microscopy and subsequently subclassified into one of 14 subtypes. This review provides practical knowledge on the disease, including new therapeutic strategies.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.