Next Article in Journal
Identification of Alternative Splicing Events Associated with Paratuberculosis in Dairy Cattle Using Multi-Tissue RNA Sequencing Data
Next Article in Special Issue
Identification and Somatic Characterization of the Germline PTEN Promoter Variant rs34149102 in a Family with Gastrointestinal and Breast Tumors
Previous Article in Journal
A Survey of Multigenic Protein-Altering Variant Frequency in Familial Exudative Vitreo-Retinopathy (FEVR) Patients by Targeted Sequencing of Seven FEVR-Linked Genes
Previous Article in Special Issue
Expression of FMRpolyG in Peripheral Blood Mononuclear Cells of Women with Fragile X Mental Retardation 1 Gene Premutation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Multisystemic Manifestations in Rare Diseases: The Experience of Dyskeratosis Congenita

by
Michele Callea
1,*,†,
Diego Martinelli
2,†,
Francisco Cammarata-Scalisi
3,
Chiara Grimaldi
4,
Houweyda Jilani
5,6,
Piercesare Grimaldi
7,
Colin Eric Willoughby
8 and
Antonino Morabito
4,9,*
1
Pediatric Dentistry and Special Dental Care Unit, Meyer Children’s University Hospital, 50139 Florence, Italy
2
Unit of Metabolism, Bambino Gesù Children’s Research Hospital, Piazza Sant’Onofrio, 4, 00165 Rome, Italy
3
Servicio de Pediatría, Hospital Regional de Antofagasta, Antofagasta 1240835, Chile
4
Department of Pediatric Surgery, Meyer Children’s Hospital, Viale Gaetano Pieraccini 24, 50139 Florence, Italy
5
Genetic Department, Mongi Slim Hospital, Marsa 2046, Tunisia
6
Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis 1068, Tunisia
7
Department of Public Health and Pediatric Sciences, University of Torino, 10125 Torino, Italy
8
Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine Campus, Coleraine BT52 1SA, UK
9
Department of Neurofarba, University of Florence, Viale Pieraccini 6, 50121 Florence, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Genes 2022, 13(3), 496; https://doi.org/10.3390/genes13030496
Submission received: 29 November 2021 / Revised: 7 March 2022 / Accepted: 8 March 2022 / Published: 11 March 2022
(This article belongs to the Special Issue Feature Papers: Molecular Genetics and Genomics)

Abstract

:
Dyskeratosis congenital (DC) is the first genetic syndrome described among telomeropathies. Its classical phenotype is characterized by the mucocutaneous triad of reticulated pigmentation of skin lace, nail dystrophy and oral leukoplakia. The clinical presentation, however, is heterogeneous and serious clinical complications include bone marrow failure, hematological and solid tumors. It may also involve immunodeficiencies, dental, pulmonary and liver disorders, and other minor complication. Dyskeratosis congenita shows marked genetic heterogeneity, as at least 14 genes are responsible for the shortening of telomeres characteristic of this disease. This review discusses clinical characteristics, molecular genetics, disease evolution, available therapeutic options and differential diagnosis of dyskeratosis congenita to provide an interdisciplinary and personalized medical assessment that includes family genetic counseling.

1. Clinical Aspects

Disorders in the biology of telomeres or telomeropathies comprise a number of genetic defects, of which dyskeratosis congenita (DC) was the first reported entity [1]. DC presents with a mucocutaneous triad of skin reticulated lace pigmentation [2], principally involving the neck area and the upper anterior thorax, nail dystrophy and oral leukoplakia. The clinical phenotype has expanded considerably since its initial description.
Initial dermatological signs appear in the first years; the clinical picture, however, can be heterogeneous [3]. Nail dystrophy involves at the beginning of the fingernails, then it starts with grooves and longitudinal divisions, evolving in rudimentary, small or absent nails. Leukoplakia impacts the oral mucosa, the tongue and the oropharynx [4]. Palmoplantar hyperkeratosis can lead to painful fissures and ulcers [5,6]. Approximately 30% of patients present malignant transformation to squamous cell carcinoma, thus necessitating carcinogenic surveillance, even with the execution of frequent biopsies in the involved areas [4].
Later on, it was described that DC could affect several organs prematurely, such as bone marrow failure [7], representing the most serious clinical complication. Bone marrow failure can be present in up to 80–90% of subjects when they reach their thirties and may cause more than 70% of deaths in DC patients [8]. The typical mucocutaneous triad guides the differential diagnosis between DC and other causes of bone marrow failure. DC also results in immunodeficiencies, predisposition to dental caries, hypodontia, recession and bone loss, requiring it to be differentiated from juvenile periodontitis, taurodontism, gingival inflammation, and brown intraoral pigmentation [9,10]. Presence of permanent teeth with a decreased root/crown ratio may indicate a diagnosis of DC [11]. Other disease manifestations are pulmonary fibrosis [12] and liver failure or fibrosis [13]. The risk of developing hematological and solid tumors is 50 times higher in patients with DC compared to general population; these include myelodysplastic syndrome, acute myeloid leukemia, non-Hodgkins lymphoma, cell carcinoma squamous cells of the head and neck, esophagus, anogenital cancer and basal cell carcinoma [14,15]. The most common solid tumor in DC is head and neck squamous cell carcinoma, which may present at a younger age compared to the general population (mean age of onset 32 years compared to 67 years); 50% of the patients who developed solid tumors were shown to carry a mutation in the TERC gene (Table 1).
DC can also present other “minor” features, i.e., intrauterine growth retardation [16], psychomotor delay, microcephaly [17], premature aging, early graying of hair [3] and short stature [18]. Some patients with DC also present with neurocognitive and neuropsychiatric manifestations, like adjustment disorders, anxiety disorders, ADHD, intellectual disability, mood disorders and schizophrenia [19]. Shorter telomeres seem to be associated with certain psychiatric disorders [20,21,22], but a direct biological link is still lacking. It is still not clear whether short telomeres predispose patients to develop certain neuropsychiatric conditions, and this hypothesis requires further studies.
Ocular anomalies include nasolacrimal duct stenosis, epiphora, blepharitis, sparse eyelashes, ectropion, entropion and trichiasis [23]. Retinal changes are rarely described, mainly hemorrhages, infarction of the nerve fiber layer, macular edema, preretinal fibrosis and optic atrophy. In addition, cardiomyopathy, malabsorption enteropathy, esophageal and urethral stenosis [24,25], hypothyroidism, hypogonadism, testicular atrophy [26], osteoporosis [27] and avascular necrosis of the shoulder and hip joints [28,29] may also be detected. These clinical manifestations affect patients in different ways in a variable number of cases. DC is often diagnosed late due to of the fact that mucocutaneous findings are not present in all cases. The wide spectrum of clinical presentation and the lack of conclusive laboratory tests can sometimes make clinical diagnosis challenging [30].
As in other telomere diseases, the severity of symptoms in DC correlates with the degree of telomere shortening. Most severe cases (i.e., those with greater telomere shortening 1% of the population), DC manifests in the first 10 years of life or even during pregnancy. In the less severe cases of telomeric shortening (<10% percentile), the age of onset is between 15 and 25 years; in these patients, DC can present as medullary aplasia or pulmonary fibrosis, with different probability according to the mutated gene [31]. Considering the low efficacy of therapeutic options, it is crucial to reach an early genetic diagnosis; the common genetic anticipation in DC requires a timely diagnosis also for family genetic counseling [30].

2. Related Disorders

The Hoyeraal-Hreidarsson syndrome (OMIM #305000) is the most severe form of DC, showing progressive marrow failure, intrauterine growth retardation, developmental delay, microcephaly, cerebellar hypoplasia, delayed myelination, hydrocephalus, brain atrophy, calcification [32], mental retardation, progressive immunodeficiency and the mucocutaneous triad [33]. The diagnosis is made if the subject presents four or more of by the association of cerebellar hypoplasia with additional signs of DC. Mucocutaneous symptoms and cerebral calcifications may be present [34]. Hoyeraal-Hreidarsson syndrome usually leads to death in early childhood and results from mutations in the RTEL1 and DKC1 genes, causing a decrease in telomerase activity [35,36,37]. It can also be due to pathogenic variants in the TERT, TINF2, TPP1 PARN genes [38,39,40,41].
Revesz syndrome (OMIM #268130) was described for the first time in 1992 [42]. It is another infrequent variant of DC due to pathogenic variants in the TINF2 gene [39]. The representing symptoms are the presence of bilateral exudative retinopathy, which is associated in most cases with intracranial calcification, and the classic alterations of DC, such as early bone marrow failure [43] and mucocutaneous disease. Intrauterine growth retardation, cerebellar hypoplasia and developmental delay may also be present [42].
Coats plus syndrome (OMIM #612199) is a rare AR disease due to pathogenic variants in the CTC1 gene [35], with cerebroretinal microangiopathy, intracranial calcifications, brain cysts, leukodystrophy, osteopenia, bone fractures and poor bone healing [44] and gastrointestinal bleeding [45,46].

3. Genotype–Phenotype Correlations

The genotype–phenotype correlation is made highly difficult by several factors, such as the possibility of hypomorphic gene mutations, disease anticipation and genetic and environmental modifying factors [13].
Heterozygous AD pathogenic variants in the TERT gene may be associated with adult-onset isolated bone marrow failure or pulmonary fibrosis. Compound heterozygous mutations in WRAP53 present the classic DC with the mucocutaneous triad plus or minus tongue squamous cell cancer [47,48,49].
The clinical symptoms associated with CTC1 pathogenic variants may not show the mucocutaneous symptoms; on the contrary, cytopenias, retinal exudates, intracranial calcifications or cysts, ataxia, intrauterine growth retardation, osteopenia and/or poor bone healing are frequent
DC patients without the pathogenic variant in one of the eleven known genes may present the most clinically severe phenotypes, including multiple clinical symptoms of DC, Hoyeraal-Hreisdarsson syndrome or Revesz syndrome [31,36,37,40].

4. Molecular Bases and Diagnosis

Telomeres are made up of six nucleotide repeats at the ends of chromosomes and a group of nucleoproteins located in these sequences, essential for chromosomal stability [50]. Each time a cell divides, they shorten [51]. The control of telomere length is involved in cell aging, tumorigenesis, so germline mutations in genes involved in telomere maintenance machinery may cause clinical entities like DC [52].
After the discoveries of these disorders, diagnostics test such as flow cytometry and fluorescent in situ hybridization in leukocytes have been developed [47,48,49]. A reduction in the size of telomeres in leukocytes under the first percentile for age is more than 95% sensitive and specific for DC patients [31] compared to unaffected family members or patients with other genetic causes of marrow failure. Other than being involved in the diagnosis of DC, telomere length has directed the discovery of genes associated with the development of DC [47,48,49].
Other methods employed in the diagnosis of DC s to measure the size of telomeres are quantitative polymerase chain reaction and Southern blot. Nevertheless, careful consideration is requested when selecting the method for measurement of telomeres size in a research and clinical setting [53].
A combination of candidate gene sequencing, linkage studies and, more recently, whole exome sequencing have identified at the moment 14 genes involved in telomere shortening involved with DC or similar phenotypes (Table 1) [54]. These genetic defects represent between 70–80% of patients with DC [55]. However, the genetic basis is unknown in 20–40% of cases [56].
DC exhibits diverse inheritance pattern including X-linked recessive (OMIM #305000) [16], as well as autosomal dominant (OMIM #127550) [57] and/or recessive (OMIM #224230) [58] inheritance patterns. Recessive X-linked DC (X-DC) is caused by pathogenic variants in the dyskerin 1 gene (DKC1), located in Xq28, which encodes a pseudouridine synthase [1]. X-DC occurs in males with an onset age between 5 and 12 years. Nevertheless, a variety of ages at onset, symptoms and severity may occur, even in subjects with the same mutation, making diagnosis highly complex. In X-DC, women may exhibit less severe clinical features due to lyonization, but always present at an older age [59].
The DKC1 gene has a highly conserved sequence required for the binding to small nucleolar RNAs, involved in ribosome biogenesis, and participates to telomerase complex at the moment of binding telomerase RNA or TERC [57]. In 2002, Mitchell and Collins first connected telomeres and human disease showing aberrant function of dyskerin and shortening of telomeres [60]. Therefore, germline mutations in genes involved in telomere maintenance result in abnormal shortening of these structures compared to age-matched controls, leading to chromosomal instability and progressive cell death [56].
As a result, DC represents a disease of defective telomere maintenance, leading to premature shortening, replicative senescence, premature depletion of stem cells and multisystem involvement. DC involves more prominently highly proliferating mucocutaneous tissues [61]. However, defects in telomerase and telomere components have also been shown in subjects with aplastic anemia, pulmonary fibrosis and liver disease [62].
Despite the strong link between DC and shorter telomere lengths, it is uncertain whether shortened telomeres are the exclusive cause of the phenotype. DC patients with DKC1 and TINF2 mutations usually present at a younger age than those with TERC or TERT mutations, and although they have more clinical abnormalities, there is no difference in telomere length between these patient subcategories [17]. Therefore, it is likely that pathways other than telomere maintenance are responsible for the disease phenotype.
For example, DKC1 and its associated NHP2, GAR1, NOP10 and CBF5 proteins interact with the H/ACA box of snoRNAs, which serves as a guide for pseudouridylation of ribosomal RNAs. Pseudouridylation is one of the hundred post-transcriptional modifications of RNAs (transfer, messenger, ribosomal and spliceosomal). As a result, defective dyskerin causes premature aging and acffects cell proliferation and haematopoietic potential and cancer [63]. In addition, other genes responsible for DC direct important functions other than those conected with telomere function. DKC1, TERT and TIN2 proteins are known to translocate to mitochondria, a crucial mechanism in modulating energy metabolism and ROS production under oxidative stress [64,65,66].
Due to the rarity of DC and the lack of living cells obtained from patients with specific mutations in diverse genes, researchers had to rely on animal models (zebrafish, Dictyostelium discoideum, mouse) to be able to study these mutations and dissect biochemical pathways and mechanisms involved in the disease. As increasingly demonstrated on these models, DKC1 depletion accelerates oxidative stress, which occurs prior to telomere shortening and affects ribosomal biogenesis which, in sequence, stimulates the p53 pathway [67].

5. Disease Progression

During follow-up, when the mucocutaneous triad is manifest, bone marrow failure is usually present. Sometimes, however, signs of the disease are vague and bone marrow failure or other abnormalities in another systems may present also before or in absence of the classical mucocutaneous traid [47,48,63]. Aplastic anemia, usually macrocytic with increased level of fetal hemoglobin, develops at an average age of onset of 11 years. It is associated with thrombocytopenia and then evolves to severe bone marrow failure [47,48,63]. Bone marrow failure can progress with the appearance of myelodysplasia in one or more clones [4]. Early death of can occur in 80% of cases of DC due to opportunistic infections. In DC, excessive shortening of telomeres is observed, which can lead to genome instability. Studies on electron microscopy have shown that certain DC cells have an immature embryonic core and that they can trigger a malignant transformation. In addition, the epithelial barrier is less effective than normal epithelium, so the permeability to harmful carcinogenic materials in the germ layer is higher. Leukoplastic areas can tipically undergo malignant transformation, and periodic monitoring is required. In addition, patients show a 40–50% cumulative risk incidence of malignancy at 50 years. DCS patients can develop Hodgkin’s lymphoma, larynx and bronchial cancer and GI tract adenocarcinoma, among others, of the genitourinary and skeletal system.

6. Differential Diagnosis

6.1. Disorders with Nail Dysplasia

  • Nail-patella syndrome (OMIM 161200)
  • Twenty-nail dystrophy (OMIM 161050)
  • Keratoderma with nail dystrophy and motor-sensory neuropathy (OMIM 148360)
  • Poikiloderma with neutropenia (OMIM 604173)

6.2. Disorders with Reticulated Hyperpigmentation

The Naegeli-Franceschetti-Jadassohn syndrome (OMIM #161000) due to heterozygous pathogenic variants in the keratin-14 gene (KRT14) is distinguished, as it does not present leukoplakia, bone marrow disease, and an increased risk of malignancy. However, this disease, as well as reticular pigmentosum dermatopathy (OMIM #125595) [68], an allelic disorder, may show a similar reticulated hyperpigmentation.
Fanconi anemia (OMIM #227650) [69] usually shows diffuse or uniform pigment abnormalities, and pancytopenia appears earlier compared to DC [70]. Other common findings are eye and kidney disease and limb anomalies.

6.3. Disorders with Poikiloderma and Increased Photosensitivity

Other entities include Bloom syndrome (OMIM #210900), caused by mutations in BLM gene [71]; Rothmund-Thomson syndrome (OMIM #268400), most often due to variants in RECQL4 gene [72]; and Epidermolysis Bullosa Simplex (OMIM #131900), due to mutations in 18 different genes [73], which present mottled pigmentation with similar poikiloderma. In addition, Kindler syndrome (OMIM #173650) due to FERMT1 gene mutations [74], and poikiloderma with neutropenia type Clericuzio (OMIM #604173), associated with biallelic mutations in USB1 gene [75]. In Bloom, Kindler, and Rothmund-Thomson syndromes, skin lesions may resemble those observed in DC, but they are more sensitive to the sun and have different related characteristics. Lastly, patients with graft-versus-host disease have poikiloderma, mucosal changes similar to lichen planus, and evident nail dystrophy after bone marrow transplantation [76].

6.4. Disorders with Bone Marrow Failure

Diamond-Blackfan anemia (DBA) is a disorder presenting with a profound isolated normochromic and usually macrocytic anemia with normal leukocytes and platelets; congenital malformations are observed in approximately 50% of affected individuals and growth retardation in 30%. Ninety percent of subjects with DBA present hematologic complications in the first year of life. DBA is associated with a higher risk of acute myelogenous leukemia, myelodysplastic syndrome and solid tumors. DBA is due to pathogenic variants in 16 genes encoding ribosomal proteins or in GATA1 and TSR2 genes. DBA generally shows an autosomal dominant inheritance; GATA1-related and TSR2-related DBA are inherited in an X-linked manner.
Shwachman-Diamond syndrome (SDS) is an AR disorder due to SBDS pathogenic variants and is characterized by exocrine pancreatic dysfunction with malabsorption, malnutrition, and growth failure, hematologic abnormalities with single- or multilineage cytopenia and susceptibility to myelodysplasia syndrome and acute myelogeneous leukemia, and bone abnormalities. Persistent or intermittent neutropenia is a common presenting finding, as well as short-stature and recurrent infections. Like DC, SDS may first present as bone marrow failure or gastro-intestinal malabsorption.
Differential diagnosis also includes other chromosomal breakage and reorganization syndromes such as Nijmegen break syndrome (OMIM #251260) [77], Seckel syndrome (OMIM #210600) [78], and finally the pseudo-TORCH syndrome (OMIM #251290) due to cerebral calcification [79].

7. Treatment

The integration of an interdisciplinary team is central to the management of DC and includes experts in dermatology, otorhinolaryngology, dentistry, maxillofacial surgery, oncology, gynecology and medical genetics, and including early genetic diagnostic facilities are crucial for timely family genetic counseling [4,59]. No definitive treatments are available for DC [13], and patients generally die prematurely of bone marrow failure [80]; allogeneic bone marrow transplantation is a treatment option, but it can be burdened by complications and risk of poor long-term survival. Up to 60% of DC patients with severe bone marrow failure may benefit temporarily from androgens or androgen-derivative therapy [81]. The biological mechanisms by which these compounds effectively treat bone marrow failure are not known. However, androgens can directly increase erythropoietin production or act on the erythropoietin receptor to elicit a hematologic response. Few studies on human cell lines and mouse models with aplastic anemia suggest that androgens can increase telomerase expression and, in turn, increase telomere length [82]. Furthermore, follow-up is crucial to detect the presence of tumors or severe infections due toopportunistic agents, which are among the main causes of death between the second and third decade of life [70].
The treatment of squamous cell carcinoma (SCC) of the head and neck is administered according to the anatomical region and stage of malignancy. Management of SCC can involve surgery, radiation and chemotherapy. In turn, exposure to potential carcinogens should be avoided, including ultraviolet radiation, alcohol and tobacco [83].
Systemic retinoids administered at low doses have determined some improvement in skin and nails in DC, but the side and long-term effects are uncertain [70,84].
Punch grafting is a low-cost and minimally invasive technique to enhance wound healing and has been associated with significant and quick pain reduction in DC ulcers [6,85].
Zoledronic acid treatment by intravenous injection was reported to prevent fractures in a young adult with DC [86]. According to an experimental study, zoledronic acid significantly increased bone volume and the number of hematopoietic stem cells in both young and adult mice [87].
Other exogenous therapies that can correct the telomerase defect and improve cell growth, as well as the use of modulators involved in telomere maintenance, have been suggested as new therapeutic methods for DC. Among them, the expression of a peptide derived from the dyskerin, a genetic suppressor element 24.2 (GSE24.2), which increases telomerase activity, regulates gene expression, and decreases DNA damage and oxidative stress in the cells of DC patients. Short peptides derived from GSE24.2 and GSE4, an eleven amino acid peptide have been shown to increase telomerase activity, reducing DNA damage as well as oxidative stress and cellular senescence in dyskerin-mutated cells. GSE4 expression also activated the c-myc and TERT promoters, as well as increased c-myc, TERT and TERC expression. Delivering GSE24.2, from a cDNA vector or as a peptide, reduces the pathogenic effects of Dkc1 mutations in mice, thus suggesting a new therapeutic approach [88,89,90]. On the other hand, the therapeutic efficacy of telomerase activation using adeno-associated virus (AAV9) gene therapy vectors carrying the Tert gene for telomerase was tested in two independent mouse models of aplastic anemia due to short telomeres (Trf1 and Tert deficient mice). A high dose of AAV9-Tert was found to target the bone marrow compartment, including hematopoietic stem cells. AAV9-Tert treatment after telomere attrition in bone marrow cells rescues aplastic anemia and mouse survival compared to mice treated with the empty vector. Improved survival is associated with a significant increase in telomere length in peripheral blood and bone marrow cells as well as better blood counts. These findings indicate that telomerase gene therapy represents a novel therapeutic strategy to treat aplastic anemia caused or associated with short telomeres [91].

8. Conclusions

DC is a clinically and genetically heterogeneous syndrome of bone marrow failure and a model of telomere disorders or telomeropathies. Different genetic alterations give rise to a wide spectrum of clinical symptoms with variable age of onset, so the diagnosis of DC can be a challenge in this disorder of telomere biology. Therefore, an interdisciplinary team is required. Available therapeutic options and timely family genetic counseling must be provided based on genetic diagnosis and telomere length laboratory studies.

Author Contributions

Conceptualization, M.C., D.M., F.C.-S. and A.M.; methodology, M.C., D.M., F.C.-S., A.M., C.E.W., H.J., C.G. and P.G.; writing—original draft preparation, M.C., D.M., F.C.-S. and A.M.; writing—review and editing, M.C., D.M., F.C.-S., A.M., C.E.W., H.J., C.G. and P.G.; supervision, M.C., D.M., F.C.-S., A.M. and C.E.W. 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

Refer to Michele Callea for any query, mcallea@gmail.com.

Acknowledgments

To the graduate Rosalía Gumina F., director of the Library of the Autonomous Institute Hospital Universitario de Los Andes, Universidad de Los Andes.

Conflicts of Interest

The authors declare that they have no conflict of interest.

References

  1. Heiss, N.S.; Knight, S.W.; Vulliamy, T.J.; Klauck, S.M.; Wiemann, S.; Mason, P.J.; Poustka, A.; Dokal, I. X-Linked Dyskeratosis Congenita Is Caused by Mutations in a Highly Conserved Gene with Putative Nucleolar Functions. Nat. Genet. 1998, 19, 32–38. [Google Scholar] [CrossRef] [PubMed]
  2. Zinsser, F. Atrophia Cutis Reticularis Cum Pigmentatione Dystrophia Unguium et Leukoplakia Oris. Inkonographia Dermatol. 1906, 5, 219–223. [Google Scholar]
  3. Dokal, I. Dyskeratosis Congenita. Hematol. Am. Soc. Hematol. Educ. Program 2011, 2011, 480–486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Fernández García, M.S.; Teruya-Feldstein, J. The Diagnosis and Treatment of Dyskeratosis Congenita: A Review. J. Blood Med. 2014, 5, 157–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Reimann, C.; Kloeckener-Gruissem, B.; Niemeyer, C.; Vanscheidt, W. Late Manifestation of Dyskeratosis Congenita Presenting as Chronic Dermal Ulcer in a 37-Year-Old Man. J. Eur. Acad. Dermatol. Venerol. 2008, 22, 897–898. [Google Scholar] [CrossRef] [PubMed]
  6. Quintana-Castanedo, L.; Feito-Rodríguez, M.; Pérez-Conde, I.; Maseda-Pedrero, R.; Chiloeches-Fernández, C.; Lucas Laguna, R. Chronic Sole Ulcer in a Child with Dyskeratosis Congenita: An Atypical Wound Successfully Treated with Punch Grafting. Pediatr. Dermatol. 2020, 37, 1131–1134. [Google Scholar] [CrossRef] [PubMed]
  7. Hanada, T.; Abe, T.; Nakazawa, M.; Aoki, Y.; Uyeno, K. Bone Marrow Failure in Dyskeratosis Congenita. Scand. J. Haematol. 2009, 32, 496–500. [Google Scholar] [CrossRef]
  8. Perona, R.; Sastre, L.; Callea, M.; Cammarata-Scalisi, F. Aspectos Clínicos, Etiológicos y Terapéuticos de La Disqueratosis Congénita. Revista Peruana de Investigación en Salud 2020, 4, 77–82. [Google Scholar] [CrossRef] [Green Version]
  9. Wald, C.; Diner, H. Dyskeratosis Congenita with Associated Periodontal Disease. Oral Surg. Oral Med. Oral Pathol. 1974, 37, 736–744. [Google Scholar] [CrossRef]
  10. Yavuzyilmaz, E.; Yamalik, N.; Yetgirr, S.; Kansu, O. Oral-Dental Findings in Dyskeratosis Congenita. J. Oral Pathol. Med. 1992, 21, 280–284. [Google Scholar] [CrossRef]
  11. Atkinson, J.C.; Harvey, K.E.; Domingo, D.L.; Trujillo, M.I.; Guadagnini, J.P.; Gollins, S.; Giri, N.; Hart, T.C.; Alter, B.P. Oral and Dental Phenotype of Dyskeratosis Congenita. Oral Dis. 2008, 14, 419–427. [Google Scholar] [CrossRef] [PubMed]
  12. Tsakiri, K.D.; Cronkhite, J.T.; Kuan, P.J.; Xing, C.; Raghu, G.; Weissler, J.C.; Rosenblatt, R.L.; Shay, J.W.; Garcia, C.K. Adult-Onset Pulmonary Fibrosis Caused by Mutations in Telomerase. Proc. Natl. Acad. Sci. USA 2007, 104, 7552–7557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Calado, R.T.; Regal, J.A.; Kleiner, D.E.; Schrump, D.S.; Peterson, N.R.; Pons, V.; Chanock, S.J.; Lansdorp, P.M.; Young, N.S. A Spectrum of Severe Familial Liver Disorders Associate with Telomerase Mutations. PLoS ONE 2009, 4, e7926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Alter, B.P. Diagnosis, Genetics, and Management of Inherited Bone Marrow Failure Syndromes. Hematol. Am. Soc. Hematol. Educ. Program 2007, 2007, 29–39. [Google Scholar] [CrossRef] [Green Version]
  15. Manfuso, A.; Risitano, A.M.; Copelli, C. Dyskeratosis Congenita and Squamous Cell Carcinoma of the Mandibular Alveolar Ridge. BMJ Case Rep. 2021, 14, e242459. [Google Scholar] [CrossRef]
  16. Knight, S.W.; Heiss, N.S.; Vulliamy, T.J.; Aalfs, C.M.; McMahon, C.; Richmond, P.; Jones, A.; Hennekam, R.C.M.; Poustka, A.; Mason, P.J.; et al. Unexplained Aplastic Anaemia, Immunodeficiency, and Cerebellar Hypoplasia (Hoyeraal-Hreidarsson Syndrome) Due to Mutations in the Dyskeratosis Congenita Gene, DKC1: Hoyeraal-Hreidarsson Syndrome Is Due to Mutations in DKC1. Br. J. Haematol. 1999, 107, 335–339. [Google Scholar] [CrossRef]
  17. Vulliamy, T.J.; Marrone, A.; Knight, S.W.; Walne, A.; Mason, P.J.; Dokal, I. Mutations in Dyskeratosis Congenita: Their Impact on Telomere Length and the Diversity of Clinical Presentation. Blood 2006, 107, 2680–2685. [Google Scholar] [CrossRef]
  18. Walne, A.J.; Vulliamy, T.; Beswick, R.; Kirwan, M.; Dokal, I. TINF2 Mutations Result in Very Short Telomeres: Analysis of a Large Cohort of Patients with Dyskeratosis Congenita and Related Bone Marrow Failure Syndromes. Blood 2008, 112, 3594–3600. [Google Scholar] [CrossRef] [Green Version]
  19. Rackley, S.; Pao, M.; Seratti, G.F.; Giri, N.; Rasimas, J.J.; Alter, B.P.; Savage, S.A. Neuropsychiatric Conditions Among Patients with Dyskeratosis Congenita: A Link with Telomere Biology? Psychosomatics 2012, 53, 230–235. [Google Scholar] [CrossRef] [Green Version]
  20. Hartmann, N.; Boehner, M.; Groenen, F.; Kalb, R. Telomere Length of Patients with Major Depression Is Shortened but Independent from Therapy and Severity of the Disease. Depress. Anxiety 2010, 27, 1111–1116. [Google Scholar] [CrossRef]
  21. Simon, N.M.; Smoller, J.W.; McNamara, K.L.; Maser, R.S.; Zalta, A.K.; Pollack, M.H.; Nierenberg, A.A.; Fava, M.; Wong, K.-K. Telomere Shortening and Mood Disorders: Preliminary Support for a Chronic Stress Model of Accelerated Aging. Biol. Psychiatry 2006, 60, 432–435. [Google Scholar] [CrossRef] [PubMed]
  22. Yu, W.-Y.; Chang, H.-W.; Lin, C.-H.; Cho, C.-L. Short Telomeres in Patients with Chronic Schizophrenia Who Show a Poor Response to Treatment. J. Psychiatry Neurosci. 2008, 33, 244–247. [Google Scholar] [PubMed]
  23. Tsilou, E.T.; Giri, N.; Weinstein, S.; Mueller, C.; Savage, S.A.; Alter, B.P. Ocular and Orbital Manifestations of the Inherited Bone Marrow Failure Syndromes: Fanconi Anemia and Dyskeratosis Congenita. Ophthalmology 2010, 117, 615–622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Savage, S.A.; Bertuch, A.A. The Genetics and Clinical Manifestations of Telomere Biology Disorders. Genet. Med. 2010, 12, 753–764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Arca, E.; Tuzun, A.; Tastan, H.B.; Akar, A.; Kose, O. Dyskeratosis Congenita with Esophageal and Anal Stricture. Int. J. Dermatol. 2003, 42, 555–557. [Google Scholar] [CrossRef] [PubMed]
  26. Kutbay, N.O.; Yurekli, B.S.; Erdemir, Z.; Karaca, E.; Unal, I.; Yaman, B.; Ozkinay, F.; Saygili, F. A Case of Dyskeratosis Congenita Associated with Hypothyroidism and Hypogonadism. Hormones 2016, 15, 297–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Inoue, S. Dyskeratosis Congenita With Pancytopenia: Another Constitutional Anemia. Am. J. Dis. Child. 1973, 126, 389. [Google Scholar] [CrossRef]
  28. Du, H.-Y.; Pumbo, E.; Manley, P.; Field, J.J.; Bayliss, S.J.; Wilson, D.B.; Mason, P.J.; Bessler, M. Complex Inheritance Pattern of Dyskeratosis Congenita in Two Families with 2 Different Mutations in the Telomerase Reverse Transcriptase Gene. Blood 2008, 111, 1128–1130. [Google Scholar] [CrossRef]
  29. Kalb, R.E.; Grossman, M.E.; Hutt, C. Avascular Necrosis of Bone in Dyskeratosis Congenita. Am. J. Med. 1986, 80, 511–513. [Google Scholar] [CrossRef]
  30. Trotta, L.; Norberg, A.; Taskinen, M.; Béziat, V.; Degerman, S.; Wartiovaara-Kautto, U.; Välimaa, H.; Jahnukainen, K.; Casanova, J.-L.; Seppänen, M.; et al. Diagnostics of Rare Disorders: Whole-Exome Sequencing Deciphering Locus Heterogeneity in Telomere Biology Disorders. Orphanet J. Rare Dis. 2018, 13, 139. [Google Scholar] [CrossRef] [Green Version]
  31. Alter, B.P.; Rosenberg, P.S.; Giri, N.; Baerlocher, G.M.; Lansdorp, P.M.; Savage, S.A. Telomere Length Is Associated with Disease Severity and Declines with Age in Dyskeratosis Congenita. Haematologica 2012, 97, 353–359. [Google Scholar] [CrossRef] [Green Version]
  32. Zhang, M.; Cao, Y.; Wu, H.; Li, H. Brain Imaging Features of Children with Hoyeraal-Hreidarsson Syndrome. Brain Behav. 2021, 11, e02079. [Google Scholar] [CrossRef]
  33. Khincha, P.P.; Savage, S.A. Genomic Characterization of the Inherited Bone Marrow Failure Syndromes. Semin. Hematol. 2013, 50, 333–347. [Google Scholar] [CrossRef] [Green Version]
  34. Nelson, N.D.; Bertuch, A.A. Dyskeratosis Congenita as a Disorder of Telomere Maintenance. Mutat. Res. Fundam. Mol. Mech. Mutagenesis 2012, 730, 43–51. [Google Scholar] [CrossRef] [Green Version]
  35. Anderson, B.H.; Kasher, P.R.; Mayer, J.; Szynkiewicz, M.; Jenkinson, E.M.; Bhaskar, S.S.; Urquhart, J.E.; Daly, S.B.; Dickerson, J.E.; O’Sullivan, J.; et al. Mutations in CTC1, Encoding Conserved Telomere Maintenance Component 1, Cause Coats Plus. Nat. Genet. 2012, 44, 338–342. [Google Scholar] [CrossRef]
  36. Ballew, B.J.; Yeager, M.; Jacobs, K.; Giri, N.; Boland, J.; Burdett, L.; Alter, B.P.; Savage, S.A. Germline Mutations of Regulator of Telomere Elongation Helicase 1, RTEL1, in Dyskeratosis Congenita. Hum. Genet. 2013, 132, 473–480. [Google Scholar] [CrossRef] [Green Version]
  37. Walne, A.J.; Vulliamy, T.; Kirwan, M.; Plagnol, V.; Dokal, I. Constitutional Mutations in RTEL1 Cause Severe Dyskeratosis Congenita. Am. J. Hum. Genet. 2013, 92, 448–453. [Google Scholar] [CrossRef] [Green Version]
  38. Marrone, A.; Walne, A.; Tamary, H.; Masunari, Y.; Kirwan, M.; Beswick, R.; Vulliamy, T.; Dokal, I. Telomerase Reverse-Transcriptase Homozygous Mutations in Autosomal Recessive Dyskeratosis Congenita and Hoyeraal-Hreidarsson Syndrome. Blood 2007, 110, 4198–4205. [Google Scholar] [CrossRef] [Green Version]
  39. Savage, S.A.; Giri, N.; Baerlocher, G.M.; Orr, N.; Lansdorp, P.M.; Alter, B.P. TINF2, a Component of the Shelterin Telomere Protection Complex, Is Mutated in Dyskeratosis Congenita. Am. J. Hum. Genet. 2008, 82, 501–509. [Google Scholar] [CrossRef] [Green Version]
  40. Kocak, H.; Ballew, B.J.; Bisht, K.; Eggebeen, R.; Hicks, B.D.; Suman, S.; O’Neil, A.; Giri, N.; NCI DCEG Cancer Genomics Research Laboratory; NCI DCEG Cancer Sequencing Working Group; et al. Hoyeraal-Hreidarsson Syndrome Caused by a Germline Mutation in the TEL Patch of the Telomere Protein TPP1. Genes Dev. 2014, 28, 2090–2102. [Google Scholar] [CrossRef] [Green Version]
  41. Dhanraj, S.; Gunja, S.M.R.; Deveau, A.P.; Nissbeck, M.; Boonyawat, B.; Coombs, A.J.; Renieri, A.; Mucciolo, M.; Marozza, A.; Buoni, S.; et al. Bone Marrow Failure and Developmental Delay Caused by Mutations in Poly(A)-Specific Ribonuclease (PARN). J. Med. Genet. 2015, 52, 738–748. [Google Scholar] [CrossRef] [PubMed]
  42. Revesz, T.; Fletcher, S.; Al-Gazali, L.I.; DeBuse, P. Bilateral Retinopathy, Aplastic Anaemia, and Central Nervous System Abnormalities: A New Syndrome? J. Med. Genet. 1992, 29, 673–675. [Google Scholar] [CrossRef] [PubMed]
  43. Karremann, M.; Neumaier-Probst, E.; Schlichtenbrede, F.; Beier, F.; Brümmendorf, T.H.; Cremer, F.W.; Bader, P.; Dürken, M. Revesz Syndrome Revisited. Orphanet J. Rare Dis. 2020, 15, 299. [Google Scholar] [CrossRef] [PubMed]
  44. Morgado, F.; Batista, M.; Moreno, A.; Coutinho, I. Coats plus Syndrome (Cerebroretinal Microangiopathy with Calcifications and Cysts-1): A Case Report. Pediatr. Dermatol. 2021, 38, 191–193. [Google Scholar] [CrossRef]
  45. Tolmie, J.L.; Browne, B.H.; McGettrick, P.M.; Stephenson, J.B.P. A Familial Syndrome with Coats’ Reaction Retinal Angiomas, Hair and Nail Defects and Intracranial Calcification. Eye 1988, 2, 297–303. [Google Scholar] [CrossRef]
  46. Himes, R.W.; Chiou, E.H.; Queliza, K.; Shouval, D.S.; Somech, R.; Agarwal, S.; Jajoo, K.; Ziegler, D.S.; Kratz, C.P.; Huang, J.; et al. Gastrointestinal Hemorrhage: A Manifestation of the Telomere Biology Disorders. J. Pediatr. 2021, 230, 55–61.e4. [Google Scholar] [CrossRef]
  47. Rodriguez-Centeno, J.; Perona, R.; Sastre, L. Dyskerin Mutations Present in Dyskeratosis Congenita Patients Increase Oxidative Stress and DNA Damage Signalling in Dictyostelium Discoideum. Cells 2019, 8, 1406. [Google Scholar] [CrossRef] [Green Version]
  48. Pereboom, T.C.; van Weele, L.J.; Bondt, A.; MacInnes, A.W. A Zebrafish Model of Dyskeratosis Congenita Reveals Hematopoietic Stem Cell Formation Failure Resulting from Ribosomal Protein-Mediated P53 Stabilization. Blood 2011, 118, 5458–5465. [Google Scholar] [CrossRef]
  49. Autexier, C. POT of Gold: Modeling Dyskeratosis Congenita in the Mouse: Figure 1. Genes Dev. 2008, 22, 1731–1736. [Google Scholar] [CrossRef] [Green Version]
  50. Meyne, J.; Ratliff, R.L.; Moyzis, R.K. Conservation of the Human Telomere Sequence (TTAGGG)n among Vertebrates. Proc. Natl. Acad. Sci. USA 1989, 86, 7049–7053. [Google Scholar] [CrossRef] [Green Version]
  51. Hayflick, L.; Moorhead, P.S. The Serial Cultivation of Human Diploid Cell Strains. Exp. Cell Res. 1961, 25, 585–621. [Google Scholar] [CrossRef]
  52. Shay, J.W.; Bacchetti, S. A Survey of Telomerase Activity in Human Cancer. Eur. J. Cancer 1997, 33, 787–791. [Google Scholar] [CrossRef]
  53. Khincha, P.; Dagnall, C.; Hicks, B.; Jones, K.; Aviv, A.; Kimura, M.; Katki, H.; Aubert, G.; Giri, N.; Alter, B.; et al. Correlation of Leukocyte Telomere Length Measurement Methods in Patients with Dyskeratosis Congenita and in Their Unaffected Relatives. Int. J. Mol. Sci. 2017, 18, 1765. [Google Scholar] [CrossRef] [PubMed]
  54. AlSabbagh, M.M. Dyskeratosis congenita: A literature review. J. Dtsch. Dermatol. Ges. 2020, 18, 943–967. [Google Scholar] [CrossRef]
  55. Ungar, R.A.; Giri, N.; Pao, M.; Khincha, P.P.; Zhou, W.; Alter, B.P.; Savage, S.A. Complex Phenotype of Dyskeratosis Congenita and Mood Dysregulation with Novel Homozygous RTEL1 and TPH1 Variants. Am. J. Med. Genet. 2018, 176, 1432–1437. [Google Scholar] [CrossRef] [PubMed]
  56. Savage, S.A. Dyskeratosis Congenita. In GeneReviews®; Adam, M.P., Ardinger, H.H., Pagon, R.A., Wallace, S.E., Bean, L.J., Mirzaa, G., Amemiya, A., Eds.; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  57. Vulliamy, T.; Marrone, A.; Goldman, F.; Dearlove, A.; Bessler, M.; Mason, P.J.; Dokal, I. The RNA Component of Telomerase Is Mutated in Autosomal Dominant Dyskeratosis Congenita. Nature 2001, 413, 432–435. [Google Scholar] [CrossRef]
  58. Walne, A.J.; Vulliamy, T.; Marrone, A.; Beswick, R.; Kirwan, M.; Masunari, Y.; Al-Qurashi, F.; Aljurf, M.; Dokal, I. Genetic Heterogeneity in Autosomal Recessive Dyskeratosis Congenita with One Subtype Due to Mutations in the Telomerase-Associated Protein NOP10. Hum. Mol. Genet. 2007, 16, 1619–1629. [Google Scholar] [CrossRef] [Green Version]
  59. Savage, S.A.; Dokal, I.; Armanios, M.; Aubert, G.; Cowen, E.W.; Domingo, D.L.; Giri, N.; Greene, M.H.; Orchard, P.J.; Tolar, J.; et al. Dyskeratosis Congenita: The First NIH Clinical Research Workshop: Dyskeratosis Congenita Workshop Report. Pediatr. Blood Cancer 2009, 53, 520–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Collins, K.; Mitchell, J.R. Telomerase in the Human Organism. Oncogene 2002, 21, 564–579. [Google Scholar] [CrossRef] [Green Version]
  61. Bertuch, A.A. The Molecular Genetics of the Telomere Biology Disorders. RNA Biol. 2016, 13, 696–706. [Google Scholar] [CrossRef] [Green Version]
  62. Nishio, N.; Kojima, S. Recent Progress in Dyskeratosis Congenita. Int. J. Hematol. 2010, 92, 419–424. [Google Scholar] [CrossRef]
  63. Garus, A.; Autexier, C. Dyskerin: An Essential Pseudouridine Synthase with Multifaceted Roles in Ribosome Biogenesis, Splicing, and Telomere Maintenance. RNA 2021, 27, 1441–1458. [Google Scholar] [CrossRef]
  64. Angrisani, A.; Matrone, N.; Belli, V.; Vicidomini, R.; Di Maio, N.; Turano, M.; Scialò, F.; Netti, P.A.; Porcellini, A.; Furia, M. A Functional Connection between Dyskerin and Energy Metabolism. Redox Biol. 2018, 14, 557–565. [Google Scholar] [CrossRef]
  65. Sullivan, L.B.; Santos, J.H.; Chandel, N.S. Mitochondria and Telomeres: The Promiscuous Roles of TIN2. Mol. Cell 2012, 47, 823–824. [Google Scholar] [CrossRef] [Green Version]
  66. Chen, L.-Y.; Zhang, Y.; Zhang, Q.; Li, H.; Luo, Z.; Fang, H.; Kim, S.H.; Qin, L.; Yotnda, P.; Xu, J.; et al. Mitochondrial Localization of Telomeric Protein TIN2 Links Telomere Regulation to Metabolic Control. Mol. Cell 2012, 47, 839–850. [Google Scholar] [CrossRef] [Green Version]
  67. Ibáñez-Cabellos, J.S.; Seco-Cervera, M.; Picher-Latorre, C.; Pérez-Machado, G.; García-Giménez, J.L.; Pallardó, F.V. Acute Depletion of Telomerase Components DKC1 and NOP10 Induces Oxidative Stress and Disrupts Ribosomal Biogenesis via NPM1 and Activation of the P53 Pathway. Biochim. Biophys. Acta Mol. Cell Res. 2020, 1867, 118845. [Google Scholar] [CrossRef]
  68. Heimer, W.L.; Brauner, G.; James, W.D. Dermatopathia Pigmentosa Reticularis: A Report of a Family Demonstrating Autosomal Dominant Inheritance. J. Am. Acad. Dermatol. 1992, 26, 298–301. [Google Scholar] [CrossRef]
  69. Fanconi, G. Familiäre, Infantile Perniciosähnliche Anämie (Perniziöses Blutbild Und Konstitution). Jahrbuch Kinderheilk 1927, 117, 257–280. [Google Scholar]
  70. Zhang, J.; Li, M.; Yao, Z. Updated Review of Genetic Reticulate Pigmentary Disorders. Br. J. Dermatol. 2017, 177, 945–959. [Google Scholar] [CrossRef]
  71. Bloom, D. Congenital telangiectatic erythema resembling lupus erythematosus in dwarfs: Probably a Syndrome Entity. Am. J. Dis. Child. 1954, 88, 754. [Google Scholar] [CrossRef]
  72. Rothmund, A. Über Cataracten in Verbindung Mit Einer Eigenthümlichen Hautdegeneration. Archiv für Ophthalmologie 1868, 14, 159–182. [Google Scholar]
  73. Passarge, E. Epidermolysis Bullosa Hereditaria Simplex. J. Pediatr. 1965, 67, 819–825. [Google Scholar] [CrossRef]
  74. Kindler, T. Congenital poikiloderma with traumatic bulla fokmation and progressive cutaneous atrophy. Br. J. Dermatol. 1954, 66, 104–111. [Google Scholar] [CrossRef]
  75. Clericuzio, C.; Hoyme, H.E.; Aase, J.M. Immune Deficient Poikiloderma: A New Genodermatosis. Am. J. Hum. Genet. 1991, 49, 131. [Google Scholar]
  76. Davidson, H.R.; Connor, J.M. Dyskeratosis Congenita. J. Med. Genet. 1988, 25, 843–846. [Google Scholar] [CrossRef] [Green Version]
  77. Weemaes, C.M.R.; Hustinx, T.W.J.; Scheres, J.M.J.C.; Munster, P.J.J.V.; Bakkeren, J.A.J.M.; Taalman, R.D.F.M. A new chromosomal instability disorder: The Nijmegen breakage syndrome. Acta Paediatr. 1981, 70, 557–564. [Google Scholar] [CrossRef]
  78. Seckel, H. Bird-Headed Dwarfs; Studies in Developmental Anthropology Including Human Proportions; S. Karger AG: Basel, Switzerland, 1959; ISBN 978-3-8055-1012-7. [Google Scholar]
  79. Dehmel, M.; Brenner, S.; Suttorp, M.; Hahn, G.; Schützle, H.; Dinger, J.; Di Donato, N.; Mackenroth, L.; von der Hagen, M. Novel Mutation in the DKC1 Gene: Neonatal Hoyeraal-Hreidarsson Syndrome As a Rare Differential Diagnosis in Pontocerebellar Hypoplasia, Primary Microcephaly, and Progressive Bone Marrow Failure. Neuropediatrics 2016, 47, 182–186. [Google Scholar] [CrossRef]
  80. Alcaraz-Pérez, F.; García-Castillo, J.; García-Moreno, D.; López-Muñoz, A.; Anchelin, M.; Angosto, D.; Zon, L.I.; Mulero, V.; Cayuela, M.L. A Non-Canonical Function of Telomerase RNA in the Regulation of Developmental Myelopoiesis in Zebrafish. Nat. Commun. 2014, 5, 3228. [Google Scholar] [CrossRef] [Green Version]
  81. Dokal, I. Dyskeratosis Congenita in All Its Forms: Review. Br. J. Haematol. 2000, 110, 768–779. [Google Scholar] [CrossRef]
  82. Vieri, M.; Kirschner, M.; Tometten, M.; Abels, A.; Rolles, B.; Isfort, S.; Panse, J.; Brümmendorf, T.H.; Beier, F. Comparable Effects of the Androgen Derivatives Danazol, Oxymetholone and Nandrolone on Telomerase Activity in Human Primary Hematopoietic Cells from Patients with Dyskeratosis Congenita. Int. J. Mol. Sci. 2020, 21, 7196. [Google Scholar] [CrossRef]
  83. Bongiorno, M.; Rivard, S.; Hammer, D.; Kentosh, J. Malignant Transformation of Oral Leukoplakia in a Patient with Dyskeratosis Congenita. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2017, 124, e239–e242. [Google Scholar] [CrossRef] [PubMed]
  84. DiGiovanna, J.J.; Mauro, T.; Milstone, L.M.; Schmuth, M.; Toro, J.R. Systemic Retinoids in the Management of Ichthyoses and Related Skin Types: Management of Ichthyoses and Related Skin Types. Dermatol. Ther. 2013, 26, 26–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Atkin, L.; Bućko, Z.; Montero, E.C.; Cutting, K.; Moffatt, C.; Probst, A.; Romanelli, M.; Schultz, G.S.; Tettelbach, W. Implementing TIMERS: The Race against Hard-to-Heal Wounds. J. Wound Care 2019, 28, S1–S50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Belaya, Z.; Golounina, O.; Nikitin, A.; Tarbaeva, N.; Pigarova, E.; Mamedova, E.; Vorontsova, M.; Shafieva, I.; Demina, I.; Van Hul, W. Multiple Bilateral Hip Fractures in a Patient with Dyskeratosis Congenita Caused by a Novel Mutation in the PARN Gene. Osteoporos. Int. 2021, 32, 1227–1231. [Google Scholar] [CrossRef]
  87. Soki, F.N.; Li, X.; Berry, J.; Koh, A.; Sinder, B.P.; Qian, X.; Kozloff, K.M.; Taichman, R.S.; McCauley, L.K. The Effects of Zoledronic Acid in the Bone and Vasculature Support of Hematopoietic Stem Cell Niches. J. Cell. Biochem. 2013, 114, 67–78. [Google Scholar] [CrossRef] [Green Version]
  88. Iarriccio, L.; Manguán-García, C.; Pintado-Berninches, L.; Mancheño, J.M.; Molina, A.; Perona, R.; Sastre, L. GSE4, a Small Dyskerin- and GSE24.2-Related Peptide, Induces Telomerase Activity, Cell Proliferation and Reduces DNA Damage, Oxidative Stress and Cell Senescence in Dyskerin Mutant Cells. PLoS ONE 2015, 10, e0142980. [Google Scholar] [CrossRef]
  89. Manguan-Garcia, C.; Pintado-Berninches, L.; Carrillo, J.; Machado-Pinilla, R.; Sastre, L.; Pérez-Quilis, C.; Esmoris, I.; Gimeno, A.; García-Giménez, J.L.; Pallardó, F.V.; et al. Expression of the Genetic Suppressor Element 24.2 (GSE24.2) Decreases DNA Damage and Oxidative Stress in X-Linked Dyskeratosis Congenita Cells. PLoS ONE 2014, 9, e101424. [Google Scholar] [CrossRef] [Green Version]
  90. Machado-Pinilla, R.; Sánchez-Pérez, I.; Ramón Murguía, J.; Sastre, L.; Perona, R. A Dyskerin Motif Reactivates Telomerase Activity in X-Linked Dyskeratosis Congenita and in Telomerase-Deficient Human Cells. Blood 2008, 111, 2606–2614. [Google Scholar] [CrossRef]
  91. Bär, C.; Povedano, J.M.; Serrano, R.; Benitez-Buelga, C.; Popkes, M.; Formentini, I.; Bobadilla, M.; Bosch, F.; Blasco, M.A. Telomerase Gene Therapy Rescues Telomere Length, Bone Marrow Aplasia, and Survival in Mice with Aplastic Anemia. Blood 2016, 127, 1770–1779. [Google Scholar] [CrossRef] [Green Version]
Table 1. Genes responsible of abnormalities in telomere biology responsible of dyskeratosis congenita.
Table 1. Genes responsible of abnormalities in telomere biology responsible of dyskeratosis congenita.
Mechanism of ActionGeneMost Important MutationsInheritance Pattern
Telomerase holoenzyme complexDKC1 *p.Ile38Thr, p.Thr49Met, p.Ser121GlyXL
TERC *-AD
TERT *p.Ala202Thr, p.His412TyrAD or AR
NOP10 *p.Arg34TrpAR
NHP2 *p.Tyr139His, p.Val126Met, p.Ter154ArgAR
Shelterin complexTPP1p.Lys170del, p.Pro491ThrAD or AR
TINF2 *p.Lys280Glu, p.Arg282His, p.Arg282SerAD
POT1p.Ser322Leu, p.Lys242LeuAD
Telomere-limiting proteinsCTC1p.Lys242Leufs * 41, p.Arg987TrpAR
STN1p.Arg136Thr, p.Asp157TyrAR
Other proteins that interact directly or indirectly with key cellular processesRTEL1p.Arg981TrpAD or AR
NAF1--
WRAP53p.Phe164Leu, p.Arg398TrpAR
PARNp.Ala383Val, p.Asn288Lysfs * 23AR
* Responsible for regulation of telomere length. AD: autosomal dominant. AR: autosomal recessive. XL: X-linked recessive.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Callea, M.; Martinelli, D.; Cammarata-Scalisi, F.; Grimaldi, C.; Jilani, H.; Grimaldi, P.; Willoughby, C.E.; Morabito, A. Multisystemic Manifestations in Rare Diseases: The Experience of Dyskeratosis Congenita. Genes 2022, 13, 496. https://doi.org/10.3390/genes13030496

AMA Style

Callea M, Martinelli D, Cammarata-Scalisi F, Grimaldi C, Jilani H, Grimaldi P, Willoughby CE, Morabito A. Multisystemic Manifestations in Rare Diseases: The Experience of Dyskeratosis Congenita. Genes. 2022; 13(3):496. https://doi.org/10.3390/genes13030496

Chicago/Turabian Style

Callea, Michele, Diego Martinelli, Francisco Cammarata-Scalisi, Chiara Grimaldi, Houweyda Jilani, Piercesare Grimaldi, Colin Eric Willoughby, and Antonino Morabito. 2022. "Multisystemic Manifestations in Rare Diseases: The Experience of Dyskeratosis Congenita" Genes 13, no. 3: 496. https://doi.org/10.3390/genes13030496

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