Background: Telomeres are the end-capping structures of all eukaryotic chromosomes thereby protecting the genome from damage and degradation. During the aging process, telomeres shorten continuously with each cell division until critically short telomeres prevent further proliferation whereby cells undergo terminal differentiation, senescence, or apoptosis. Premature aging due to critically short telomere length (TL) can also result from pathogenic germline variants in the telomerase complex or related genes that typically counteract replicative telomere shortening in germline and certain somatic cell populations, e.g., hematopoetic stem cells. Inherited diseases that result in altered telomere maintenance are summarized under the term telomere biology disorder (TBD). Summary: Since TL both reflects but more importantly restricts the replicative capacity of various human tissues, a sufficient telomere reserve is particularly important in cells with high proliferative activity (e.g., hematopoiesis, immune cells, intestinal cells, liver, lung, and skin). Consequently, altered telomere maintenance as observed in TBDs typically results in premature replicative cellular exhaustion in the respective organ systems eventually leading to life-threatening complications such as bone marrow failure (BMF), pulmonary fibrosis, and liver cirrhosis. Key Messages: The recognition of a potential congenital origin in approximately 10% of adult patients with clinical BMF is of utmost importance for the proper diagnosis, appropriate patient and family counseling, to prevent the use of inefficient treatment and to avoid therapy-related toxicities including appropriate donor selection when patients have to undergo stem cell transplantation from related donors. This review summarizes the current state of knowledge about TBDs with particular focus on the clinical manifestation patterns in children (termed early onset TBD) compared to adults (late-onset TBD) including typical treatment- and disease course-related complications as well as their prognosis and adequate therapy. Thereby, it aims to raise awareness for a disease group that is currently still highly underdiagnosed particularly when it first manifests itself in adulthood.

The end structure of all chromosomes was identified in 1938 by Hermann Mueller as “telomeres” in flies and a little later by Barbara McClintock in corn [1, 2]. McClintock also recognized the importance of proper telomere maintenance for chromosomal stability by demonstrating that telomeres are required to prevent (otherwise free) chromosome ends from fusion with each other [1]. It took almost four decades until the underlying tandem repeat sequence of telomeres was first described by Elizabeth Blackburn in 1978 [3]. This noncoding, three-dimensional telomeric loop structure organizes and protects genetic information encoded in subtelomeric DNA [1, 4]. In the course of this review, we will (1) introduce the structure of the telomere complex including telomere-associated proteins as well as telomerase, (2) describe diseases that are thought to result from impaired telomere maintenance, (3) focus on the differential symptomology associated with early onset (classical “dyskeratosis congenita,” DKC) as well as late-onset (so-called cryptic) telomere biology disorders (TBDs), and finally, (4) explain existing diagnostic and therapeutic options for patients with TBD.

The typical (TTAGGG)n telomere repeat sequences at the 192 ends of the 48 chromosomes of a human cell are organized by a sophisticated interplay between different proteins and nucleic acids that build various complexes essential for proper telomere maintenance and protection [4]. The most important complexes are the shelterin complex, telomerase and associated modifiers, the CST, cytoplasmic iron-sulfur assembly, and the Apollo complexes [4‒8].

The shelterin complex contributes essentially to the formation of the T-loop end structure of the chromosomes and is comprised of proteins such as telomeric repeat binding factor 1 (TERF1), TERF2, TERF1 interacting nuclear factor 2 (TINF2) (=TIN2), repressor/activator protein 1 (RAP1), adrenocortical dysplasia protein homolog (TPP1), and protection of telomeres 1 (POT1) [6, 9]. The complex functionally constitutes a protective cap around the telomeres thereby protecting them from degradation and from being (mis)recognized as a double-strand break by the DNA repair machinery (shown in Fig. 1) [6, 9].

Fig. 1.

Telomeres, telomerase, and TBDs. The figure shows parts of the telomerase and shelterin complex and their interaction with the telomere repeat region. The components are shown in different colors depending on the described disease-causing heritability. Red indicates components that are inherited in an autosomal dominant (and/or autosomal recessive) manner. Blue indicates components that are mostly inherited in an autosomal recessive manner. Green indicates the x-linked inheritance of DKC1, orange shows TINF2 (often de novo), and gray indicates components for which the current literature is sparse or not present. Potential symptoms of telomere biology disorders (TBDs) are indicated in the bottom part of the figure. The classical triad of dyskeratosis congenita (DKC) is marked with a yellow star. Adapted from “Telomeres and Telomerase,” by BioRender.com (2024). Retrieved from “https://app.BioRender.com/biorender-templates”.

Fig. 1.

Telomeres, telomerase, and TBDs. The figure shows parts of the telomerase and shelterin complex and their interaction with the telomere repeat region. The components are shown in different colors depending on the described disease-causing heritability. Red indicates components that are inherited in an autosomal dominant (and/or autosomal recessive) manner. Blue indicates components that are mostly inherited in an autosomal recessive manner. Green indicates the x-linked inheritance of DKC1, orange shows TINF2 (often de novo), and gray indicates components for which the current literature is sparse or not present. Potential symptoms of telomere biology disorders (TBDs) are indicated in the bottom part of the figure. The classical triad of dyskeratosis congenita (DKC) is marked with a yellow star. Adapted from “Telomeres and Telomerase,” by BioRender.com (2024). Retrieved from “https://app.BioRender.com/biorender-templates”.

Close modal

The telomerase complex consists of the proteins telomerase reverse transcriptase (TERT) with the corresponding telomerase RNA component (TERC), the protein dyskerin (DKC1), the H/ACA ribonucleoprotein complex subunit 2 (NHP2), the nucleolar protein 10 (NOP10), GAR1 ribonucleoprotein (GAR1), and the WD repeat containing antisense to TP53 (TCAB1) [4, 5]. The complex is of particular importance in germline and certain somatic (stem) cell populations and promotes the active lengthening or net stabilization of telomeres, e.g., in highly proliferative organs (shown in Fig. 1) [4, 5].

The CST complex includes the CST telomere replication complex component 1 (CTC1), the TEN1 subunit of CST complex (TEN1) and the STN1 subunit of CST complex (STN1) and is essential for telomere maintenance especially under conditions of replicative stress [6, 10]. There are also other complexes described as the cytoplasmic iron-sulfur assembly complex and the Apollo complex (contains the protein DNA cross-link repair 1B, DCLRE1B, = Apollo) [7, 11, 12]. Both complexes play a fundamental role in DNA repair and protection of telomeric DNA [7, 11, 12].

In general, many of the proteins involved in telomere regulation are important for DNA integrity, intracellular signaling cascades, and ribosomal function [8, 13, 14]. These proteins protect the telomeric region and regulate its tertiary and quaternary structure thereby avoiding genetic instability leading to end-to-end fusions, unbalanced translocations, and eventually loss of chromosomal DNA [8].

In most somatic cells, telomeres shorten with each cell division due to the so-called “end replication problem” [1]. This physiological mechanism leads to age-related telomere length (TL) shortening thereby limiting the lifespan of cellular organisms [1, 15, 16]. Cells that reach critically short TL undergo replicative senescence and/or apoptosis [17]. Non-replication dependent reduction of TL can also occur due to oxidative damage [18, 19] or increased activity of the telomeric zinc finger-associated protein (TZAP) that can actively trim telomeres [20]. TL shortening can both act as an efficient regulator of cell proliferation as well as a tumor preventive mechanism by limiting the number of cell divisions a somatic cell can undergo under normal circumstances, a condition first described in 1961 as the so-called Hayflick limit. For most somatic cell types, the Hayflick limit is assumed to be reached after around 50–80 mitoses in somatic cells [21]. With increasingly critical shortness, telomeres can destabilize DNA leading to increased DNA degradation, increased fusion between different chromosomes, eventually resulting in dicentric chromosomes, chromosomal breakage, or aneuploidy [8].

The inverse correlation between an individual cell’s TL and its lifespan was suggested to act in the sense of a biological clock [1, 17]. Nevertheless, some cellular compartments are capable to counteract replicative TL shortening via various mechanisms. In 1985, the telomerase complex that enables the elongation of the telomere repeat sequence was first discovered [22]. Telomerase is primarily recruited to the telomeric region by interacting with proteins of the shelterin complex [4, 8, 9]. Embryonic stem cells, sperm cells, but also some adult cell populations (e.g., hematopoietic stem cells [HSCs], epidermal cells, and activated (B-)lymphocytes) are able to (re)express and actively use the function of this protein [8]. Nevertheless, in steady-state, most somatic cells do not express telomerase which means that telomeres require special protective mechanisms [4]. Apart from telomerase activity, other mechanisms known to be able to sustain and increase TL are summarized under the term “alternative lengthening of telomeres” (ALT) [23].

Historically, DKC was first described on the basis of skin manifestations such as leukoplakia and nail dystrophy by the dermatologist Zinsser [24], as well as Engman [25] and Cole [26]. One of the first systematic reviews of dyskeratosis congenita was published by Costello and Buncke [27] in 1956 and later reports expanded the spectrum of clinical symptoms related to dysfunction of other organ systems such as bone marrow failure (BMF) [28, 29]. Only in 1998, the connection between a defective telomerase component leading to prematurely shortened telomeres was discovered, thereby proving the causal link between a pathogenic variant in a single gene and the multisystem pathophysiology of DKC [30, 31]. In the following years, techniques to routinely measure TL in the peripheral blood of normal individuals and patients with HSC-associated disorders were developed [32‒35] enabling the functional screening of patients for an underlying TBD as practiced today [36, 37].

Whereas accelerated telomere shortening had first been described in blood cells from patients with acquired aplastic anemia in 1998 [38], this was initially suspected to be a secondary phenomenon, i.e., to result from increased stem cell turnover of residual HSCs as a response to continuous autoimmune-mediated damage to the stem cell compartment [39]. Subsequently, in 2005 it was shown that pathogenic germline variants in the telomerase reverse transcriptase (TERT) gene can be detected in adult patients with aplastic anemia thereby defining the so-called late-onset, adult TBDs [40, 41]. Due to the rapid development of sequencing techniques, a larger number of other genes with pathogenic variants were detected in juvenile and adult TBD patients in the following years, such as variants in the regulator of telomere elongation helicase 1 (RTEL1) or the poly(A)-specific ribonuclease (PARN) in patients with interstitial lung disease [42].

At the moment, there is sufficiently strong evidence that certain variants of the following genes might cause a clinically relevant TBD phenotype (see Table 1): adrenocortical dysplasia protein homolog (ACD) (=TPP1), CST telomere replication complex component 1 (CTC1), DNA cross-link repair 1B (DCLRE1B; =SNM1B; = Apollo), dyskerin pseudouridine synthase 1 (DKC1), MDM4 regulator of p53 (MDM4), nuclear assembly factor 1 ribonucleoprotein (NAF1), H/ACA ribonucleoprotein complex subunit 2 (NHP2) (=NOLA2), nucleolar protein 10 (NOP10) (=NOLA3), nucleophosmin 1 (NPM1), PARN, protection of telomeres 1 (POT1), replication protein A1 (RPA1), RTEL1, STN1 subunit of CST complex (STN1), telomerase RNA component (TERC), TERT, TERF1 interacting nuclear factor 2 (TINF2) (=TIN2), WD repeat containing antisense to TP53 (WRAP53) (=TCAB1), and zinc finger CCHC-type containing 8 (ZCCHC8) [17, 43‒45].

Table 1.

Genes that have been proven to cause telomere biology disorders (TBDs)

GeneProteinFunctionPotential consequence of pathogenic variantsEstimated proportion of all TBDs, n (%)Year reported (clinical condition)Nature of inheritance
Adrenocortical dysplasia protein homolog (ACD) (=TPP1) [46, 47TPP1 (not (!) tripeptidyl peptidase 1) 
  • Part of the shelterin complex (Capping/Bridging shelterin)

  • Bridging component

  • Recruits POT1 and telomerase

  • Binds to the POT1 protein, TINF2, and the ataxia telangiectasia and Rad3-related protein (ATR)

  • Regulates TL and supports stability

  • Regulates DNA repair

 
  • Decreased recruitment of the telomerase

 
1.5 2014 Autosomal dominant or autosomal recessive 
CST telomere replication complex component 1 (CTC1) [48, 49CTC1 
  • Part of the CST complex

  • Replication of the telomere repeats

  • Stimulation of DNA polymerase α-primase

 
  • Instable telomeres

  • Impaired replication of the telomere repeats

  • Sometimes less prominent/no change in TL

 
2012 Autosomal recessive 
DNA cross-link repair 1B (DCLRE1B) (=SNM1B) (=Apollo) [11, 50Apollo 
  • Interacts with TERF2

  • Protects telomeres during and after replication

  • Function in DNA repair

  • Overhang processing

 
  • Genetic instability

  • Often with normal TL (in some cases very short telomeres were only detected by using more sensitive methods)

 
<1 2010 Autosomal recessive 
Dyskerin pseudouridine synthase 1 (DKC1) [30Dyskerin 
  • Part of the telomerase complex

  • Stabilization of TERC

 
  • Reduced telomerase activity

  • Instability of TERC

 
25 1998 X-linked 
MDM4 regulator of p53 (MDM4) [51MDM4 
  • Regulates and degrades TP53

 
  • Increased TP53 activity

  • Short TL

 
<1 2020 Autosomal dominant 
Nuclear assembly factor 1 ribonucleoprotein (NAF1) [52NAF1 
  • Regulation of telomerase

  • Enables protein binding and binding of TERC

  • Stabilization of TERC and H/ACA small ribonucleoproteins (chaperon function)

  • Important factor for the biosynthesis of ribosomes

 
  • Reduced telomerase activity

  • Instability of TERC

 
<1 2016 Autosomal dominant 
H/ACA ribonucleoprotein complex subunit 2 (NHP2) (=NOLA2NHP2 
  • Part of the telomerase/dyskerin complex

  • (Non-catalytic) component of H/ACA small nucleolar ribonucleoproteins

  • Biogenesis of telomerase

  • Stabilization of TERC

  • Essential for the biogenesis of ribosomes

 
  • Reduced telomerase activity

  • Instability of TERC

 
<1 2008 Autosomal recessive 
Nucleolar protein 10 (NOP10) (=NOLA3) [53, 54NOP10 
  • Part of the telomerase/dyskerin complex

  • Biogenesis of telomerase

  • Stabilization of TERC

 
  • Reduced telomerase activity

  • Instability of TERC

 
<1 2007 Autosomal recessive 
Nucleophosmin 1 (NPM1) [55NPM1 
  • Modifying rRNA (for example regulating ribosomal RNA 2′-O-methylations (2′-O-Me))

  • Interacts with NHP2 and NOP10

 
  • Impaired ribosome function by aberrant 2′-O-Me on rRNA residues

 
<1 2019 Autosomal dominant 
Poly(A)-specific ribonuclease (PARN) [56‒60PARN 
  • Processing of TERC

  • Stabilization of TERC

 
  • Reduced telomerase activity

  • Instability of TERC

  • Sometimes only with subtle change in TL (close to the 10% percentile)

 
>1% (especially in patients with idiopathic PF) 2015 Autosomal dominant or autosomal recessive 
Protection of telomeres 1 (POT1) [61, 62POT1 
  • Part of the shelterin complex

  • Binds to telomeres (3’ overhang) and interacts with the CST complex

  • Binds to TPP1

  • Binds to ATR and inhibits ATR-mediated DNA damage response

  • Inhibits end-to-end fusion/protects the G-strand

 
  • Impaired replication of the telomere repeats

  • Dysfunctional regulation of the telomerase

  • Defective binding to the telomere overhang

 
<1 2016 Autosomal recessive 
Replication protein A1 (RPA1) [63RPA1 
  • Important for DNA replication and telomere maintenance

  • Functions in DNA repair

  • Binds single-stranded DNA and protect its structure

 
  • Increased amount of unfolded telomeres

  • Increased binding affinity to single-stranded and telomeric DNA (gain of function)

 
<1 2022 Autosomal dominant 
Regulator of telomere elongation helicase 1 (RTEL1) [64‒68RTEL1 
  • Replication of the telomeres

  • Dissociation and stability of T-loops

  • Target of the cytosolic iron-sulfur protein assembly (CIA) complex

  • Prevents loss of telomeres

 
  • Decreased/impaired telomere replication

  • Decreased telomere stability

 
10 2013 Autosomal dominant or autosomal recessive 
STN1 subunit of CST complex (STN1) [69STN1 
  • Part of the CST complex

  • Important for telomere replication

 
  • Decreased/impaired telomere replication

 
<1 2016 Autosomal recessive 
Telomerase RNA component (TERC) [70TERC 
  • Part of the telomerase complex

  • Elongation of telomeres

 
  • Reduced telomerase activity

 
10 2001 Autosomal dominant 
Telomerase reverse transcriptase (TERT) [40, 41TERT 
  • Part of the telomerase complex

  • Recruitment of telomerase

  • Elongation of telomeres

  • Impacts the Wnt signaling pathway

  • Might promote pluripotency and mobilization of stem cells

 
  • Reduced telomerase activity

  • Impaired telomerase recruitment

 
2005 Autosomal dominant or autosomal recessive 
TERF1 interacting nuclear factor 2 (TINF2) (=TIN2) [71, 72TIN2 
  • Part of the shelterin complex (bridging shelterin)

  • Stabilizes the shelterin complex

  • Protects telomeres and regulates TL

  • Recruits and regulates telomerase

  • Binds to TPP1, TERF1, and TERF2. Stabilizes TERF1/TERF2 interaction

  • Inhibits PARsylation of TERF1

 
  • Impaired telomere maintenance

 
20 2008 Autosomal dominant (often as a de novo mutation) 
WD repeat containing antisense to TP53 (WRAP53) (=TCAB1) [73, 74TCAB1 
  • Associated with the telomerase complex

  • Facilitates (physiological) protein-protein and protein-RNA interactions

  • Interacts with Dyskerin, TERT, and TERC

  • Recruits telomerase to the telomeres

  • Recruits proteins to the sides of Cajal bodies and sides of DNA damage

  • Transcript regulates levels of TP53 RNA

 
  • Impaired trafficking and recruitment of telomerase

 
<1 2011 Autosomal recessive 
Zinc finger CCHC-type containing 8 (ZCCHC8) [75ZCCHC8 
  • TERC processing and maturation

 
  • Decreased/impaired telomerase function

  • Accumulation of extended TERC/decreased maturation of TERC

 
<1 2019 Autosomal dominant 
Unknown [17, 45   ≈20   
GeneProteinFunctionPotential consequence of pathogenic variantsEstimated proportion of all TBDs, n (%)Year reported (clinical condition)Nature of inheritance
Adrenocortical dysplasia protein homolog (ACD) (=TPP1) [46, 47TPP1 (not (!) tripeptidyl peptidase 1) 
  • Part of the shelterin complex (Capping/Bridging shelterin)

  • Bridging component

  • Recruits POT1 and telomerase

  • Binds to the POT1 protein, TINF2, and the ataxia telangiectasia and Rad3-related protein (ATR)

  • Regulates TL and supports stability

  • Regulates DNA repair

 
  • Decreased recruitment of the telomerase

 
1.5 2014 Autosomal dominant or autosomal recessive 
CST telomere replication complex component 1 (CTC1) [48, 49CTC1 
  • Part of the CST complex

  • Replication of the telomere repeats

  • Stimulation of DNA polymerase α-primase

 
  • Instable telomeres

  • Impaired replication of the telomere repeats

  • Sometimes less prominent/no change in TL

 
2012 Autosomal recessive 
DNA cross-link repair 1B (DCLRE1B) (=SNM1B) (=Apollo) [11, 50Apollo 
  • Interacts with TERF2

  • Protects telomeres during and after replication

  • Function in DNA repair

  • Overhang processing

 
  • Genetic instability

  • Often with normal TL (in some cases very short telomeres were only detected by using more sensitive methods)

 
<1 2010 Autosomal recessive 
Dyskerin pseudouridine synthase 1 (DKC1) [30Dyskerin 
  • Part of the telomerase complex

  • Stabilization of TERC

 
  • Reduced telomerase activity

  • Instability of TERC

 
25 1998 X-linked 
MDM4 regulator of p53 (MDM4) [51MDM4 
  • Regulates and degrades TP53

 
  • Increased TP53 activity

  • Short TL

 
<1 2020 Autosomal dominant 
Nuclear assembly factor 1 ribonucleoprotein (NAF1) [52NAF1 
  • Regulation of telomerase

  • Enables protein binding and binding of TERC

  • Stabilization of TERC and H/ACA small ribonucleoproteins (chaperon function)

  • Important factor for the biosynthesis of ribosomes

 
  • Reduced telomerase activity

  • Instability of TERC

 
<1 2016 Autosomal dominant 
H/ACA ribonucleoprotein complex subunit 2 (NHP2) (=NOLA2NHP2 
  • Part of the telomerase/dyskerin complex

  • (Non-catalytic) component of H/ACA small nucleolar ribonucleoproteins

  • Biogenesis of telomerase

  • Stabilization of TERC

  • Essential for the biogenesis of ribosomes

 
  • Reduced telomerase activity

  • Instability of TERC

 
<1 2008 Autosomal recessive 
Nucleolar protein 10 (NOP10) (=NOLA3) [53, 54NOP10 
  • Part of the telomerase/dyskerin complex

  • Biogenesis of telomerase

  • Stabilization of TERC

 
  • Reduced telomerase activity

  • Instability of TERC

 
<1 2007 Autosomal recessive 
Nucleophosmin 1 (NPM1) [55NPM1 
  • Modifying rRNA (for example regulating ribosomal RNA 2′-O-methylations (2′-O-Me))

  • Interacts with NHP2 and NOP10

 
  • Impaired ribosome function by aberrant 2′-O-Me on rRNA residues

 
<1 2019 Autosomal dominant 
Poly(A)-specific ribonuclease (PARN) [56‒60PARN 
  • Processing of TERC

  • Stabilization of TERC

 
  • Reduced telomerase activity

  • Instability of TERC

  • Sometimes only with subtle change in TL (close to the 10% percentile)

 
>1% (especially in patients with idiopathic PF) 2015 Autosomal dominant or autosomal recessive 
Protection of telomeres 1 (POT1) [61, 62POT1 
  • Part of the shelterin complex

  • Binds to telomeres (3’ overhang) and interacts with the CST complex

  • Binds to TPP1

  • Binds to ATR and inhibits ATR-mediated DNA damage response

  • Inhibits end-to-end fusion/protects the G-strand

 
  • Impaired replication of the telomere repeats

  • Dysfunctional regulation of the telomerase

  • Defective binding to the telomere overhang

 
<1 2016 Autosomal recessive 
Replication protein A1 (RPA1) [63RPA1 
  • Important for DNA replication and telomere maintenance

  • Functions in DNA repair

  • Binds single-stranded DNA and protect its structure

 
  • Increased amount of unfolded telomeres

  • Increased binding affinity to single-stranded and telomeric DNA (gain of function)

 
<1 2022 Autosomal dominant 
Regulator of telomere elongation helicase 1 (RTEL1) [64‒68RTEL1 
  • Replication of the telomeres

  • Dissociation and stability of T-loops

  • Target of the cytosolic iron-sulfur protein assembly (CIA) complex

  • Prevents loss of telomeres

 
  • Decreased/impaired telomere replication

  • Decreased telomere stability

 
10 2013 Autosomal dominant or autosomal recessive 
STN1 subunit of CST complex (STN1) [69STN1 
  • Part of the CST complex

  • Important for telomere replication

 
  • Decreased/impaired telomere replication

 
<1 2016 Autosomal recessive 
Telomerase RNA component (TERC) [70TERC 
  • Part of the telomerase complex

  • Elongation of telomeres

 
  • Reduced telomerase activity

 
10 2001 Autosomal dominant 
Telomerase reverse transcriptase (TERT) [40, 41TERT 
  • Part of the telomerase complex

  • Recruitment of telomerase

  • Elongation of telomeres

  • Impacts the Wnt signaling pathway

  • Might promote pluripotency and mobilization of stem cells

 
  • Reduced telomerase activity

  • Impaired telomerase recruitment

 
2005 Autosomal dominant or autosomal recessive 
TERF1 interacting nuclear factor 2 (TINF2) (=TIN2) [71, 72TIN2 
  • Part of the shelterin complex (bridging shelterin)

  • Stabilizes the shelterin complex

  • Protects telomeres and regulates TL

  • Recruits and regulates telomerase

  • Binds to TPP1, TERF1, and TERF2. Stabilizes TERF1/TERF2 interaction

  • Inhibits PARsylation of TERF1

 
  • Impaired telomere maintenance

 
20 2008 Autosomal dominant (often as a de novo mutation) 
WD repeat containing antisense to TP53 (WRAP53) (=TCAB1) [73, 74TCAB1 
  • Associated with the telomerase complex

  • Facilitates (physiological) protein-protein and protein-RNA interactions

  • Interacts with Dyskerin, TERT, and TERC

  • Recruits telomerase to the telomeres

  • Recruits proteins to the sides of Cajal bodies and sides of DNA damage

  • Transcript regulates levels of TP53 RNA

 
  • Impaired trafficking and recruitment of telomerase

 
<1 2011 Autosomal recessive 
Zinc finger CCHC-type containing 8 (ZCCHC8) [75ZCCHC8 
  • TERC processing and maturation

 
  • Decreased/impaired telomerase function

  • Accumulation of extended TERC/decreased maturation of TERC

 
<1 2019 Autosomal dominant 
Unknown [17, 45   ≈20   

The table summarizes genes with the respective protein name and function that might harbor pathogenic variants that lead to impaired telomere maintenance and clinical manifestations. Potential consequences of pathogenic variants, the estimated proportion of pathogenic variants in this gene among all telomere biology disorders (TBDs), the year of the first clinical description and the suspected inheritance pattern are shown [43, 45, 76, 77].

While initially DKC was considered an appropriate term for all telomere-related diseases, it later became clear that on the one hand side, certain pediatric forms such as the Revesz syndrome, the Hoyeraal-Hreidarsson syndrome, or the Coats plus syndrome are characterized by a strikingly more severe phenotype and earlier disease onset clearly distinct from classical DKC [48, 64, 78]. On the other hand, adult-onset forms of TBDs sometimes only manifest themselves beyond 40 years of age, often lacking a skin phenotype and displaying distinct and highly variable organ system manifestations from classical DKC [36, 45].

Consequently, in parallel to the identification of a growing number of disease-causing genes, there was also a change in nomenclature [7, 44]. For instance, some patients with a Coats plus syndrome harbor a pathogenic variant in a gene important for telomere maintenance and show a TBD phenotype, but do not have shorter TL [43, 48, 61]. This led to the change in terminology away from the term telomeropathy or telomere disease to the currently used term TBD [17, 43].

The main clinical symptoms and diagnostic of TBDs are shown below. We proceed chronologically according to age of first disease manifestation. TBDs that will be discussed include early pediatric forms, and classical adolescence DKC, but the focus of this review is on different forms and manifestations of late-onset (previously called “cryptic”) TBDs.

In general, there are several techniques available to measure TL [33, 36, 37]. The current standard for clinical detection of shortened TL is the combination of a quantitatively telomere-binding FISH probe in combination with flow cytometry (flow-FISH) [32, 33]. This method was developed for the analysis of clinical specimen, i.e., to allow for high cell numbers (see telomere Q-FISH for comparison [79]) in high throughput and to be able to analyze at the same time different blood cell subpopulations involved in disease pathophysiology [34, 39, 80‒82]. Interestingly, lymphocyte TL measured via flow-FISH is sensitive and highly specific and was empirically shown to be superior for diagnosis of inherited TBD over granulocyte TL probably because the latter population is more directly involved in the disease course of AA itself (see above) while lymphocyte TL more specifically reflects the hereditary “telomere genotype” [15, 43, 83]. In analogy to percentile-based growth curves that are used to track physical development in children, TBDs can be identified on the basis of differences in TL from age-adapted percentiles [36, 83]. Thereby, TL in the peripheral blood shows a negative correlation with age and it follows an at least bi-phasic kinetic [33]. The most frequently used diagnostic threshold for TBDs is currently a TL value below the 1% percentile of normal individuals measured by flow-FISH in lymphocytes. Due to the substantially larger “diagnostic window” in children, conventional methods for TL measurement like PCR with lower sensitivity are still being used in pediatric patients [37]. In borderline cases or in scenarios with a high degree of clinical suspicion for an underlying TBD (e.g., due to family history, clinical phenotype or other aspects), detection of TL in lymphocyte subsets might also help increase sensitivity and specificity [15, 43, 83]. Although overall lymphocyte TL is the most established marker to diagnose TBDs, more recent publications indicate that the simultaneous detection of lymphocyte and granulocyte TL should be preferred [43, 84]. In case of short TL below the 1% percentile, subsequent genetic testing is recommended to test for disease causing germline variants in so-far known TBD-related genes [36, 37, 83]. However, based on current knowledge a pathogenic variant cannot be detected in all clinical TBD cases primarily suggesting that not all gene alterations involved in telomere maintenance are yet known and secondly, highlighting the diagnostic value of TL measurements as the yet only functional test available for primary screening of TBDs [17, 45].

Early severe pediatric forms of TBDs with often characteristic underlying genetic variants are the Revesz syndrome, the Hoyeraal-Hreidarsson syndrome, and the Coats plus syndrome [45, 64, 69, 78]. Revesz and Hoyeraal-Hreidarsson syndrome are characterized by a severe disease course, very short telomeres (considerably below the 1% percentile), and a disease onset in the early childhood [45, 65, 78]. In comparison, the Coats plus syndrome does not necessarily have to be accompanied by short TL (in this rare case of a TBD leading to a limited diagnostic value of TL screening) but otherwise shows classical signs of TBD [61, 69, 76]. All three forms overlap clinically with classical DKC and are associated with significantly reduced survival [45, 85].

Revesz syndrome typically features bilateral exudative retinopathy, intracranial calcification, cerebellar hypoplasia thereby causing ataxia, (intrauterine) growth restriction, general developmental delay, and fine hairs. Affected patients have a high risk for BMF. It is often caused by variants in the TINF2 gene [78, 77].

Hoyeraal-Hreidarsson syndrome, on the other hand, shares some signs of Revesz syndrome such as cerebellar hypoplasia and intrauterine growth restriction, but can also present with progressive immunodeficiency, microcephaly, and moderate to severe mental retardation. Also, progressive pancytopenia (early onset) is frequently observed. In many cases, pathogenic germline variants in the DKC1 gene are detected as the underlying cause of the syndrome [11, 64, 65, 86, 73].

Characteristic findings of Coats plus syndrome are exudative retinopathy, cerebroretinal microangiopathy with concomitant brain calcifications, brain cysts, loss of the white matter (leukodystrophy), gastrointestinal bleedings, and bone abnormalities (e.g., osteopenia). Abnormalities of the skeletal system are often accompanied by impaired bone healing. The leukodystrophy can lead to a progressive cognitive decline [48]. Also, anemia and thrombocytopenia (with or without BMF) are common symptoms. Variants in the genes CTC1, STN1 and POT1 are frequently found in patients with Coat plus syndrome [48, 61, 69].

The paradigmatic TBD is DKC or Zinsser-Engman-Cole syndrome. Classical DKC can be caused by different genetic variants in genes affecting telomerase function, telomere maintenance, and telomere interaction. Classical DKC is often diagnosed during childhood or early adolescence. In classical DKC pathogenic variants are found in genes such as DKC1, TERT, TERC, TINF2, RTEL1, NOP10, NHP2, CTC1, ACD (=TPP1), PARN and WRAP53 (=TCBA1) [30, 49, 53, 56, 66, 67, 70, 71, 74]. Most variants lead to reduced telomerase activity and increased telomere erosion [15, 83]. More recent reports indicate that variants in RPA1, STN1, DCLRE1B (Apollo), and NPM1 can also cause (classical) DKC [50, 55, 63].

Classical DKC shows severe manifestations early in life and the typical mucocutaneous triad with leukoplakia, reticulated skin pigmentations and nail dystrophy (classical DKC, shown in Fig. 1) [44, 83, 85]. Apart from the mucocutaneous triad, the characteristic clinical disease course includes an increased risk for BMF and cancer development [43, 44]. Additional symptoms reflecting the multi-systemic nature of TBDs include early hair graying and alopecia, palmoplantar hyperkeratosis and hyperhidrosis, short stature and osteoporosis, avascular bone necrosis, lung fibrosis and lung emphysema, liver disease including liver fibrosis and liver cirrhosis, arteriovenous malformations, hypogonadism, different gastrointestinal diseases, neurological symptoms, visual disorders, atresia of the lacrimal duct, severe dental caries, esophageal strictures, and stenosis of the urethra (shown in Fig. 1; Table 2) [15, 44, 72, 83, 85, 87, 88].

Table 2.

(Inherited) telomere biology disorders (TBDs) together with accompanying symptoms

DiseaseGenes that might harbor pathogenic variantsClinical manifestations
(Classical) Dyskeratosis congenita (DKC) (=Zinsser-Engman-Cole syndrome) [30, 49, 53, 55, 56, 66, 67, 70, 71, 74] (Manifestation: childhood or (early) adolescence) DKC1, TERT, TERC, TINF2, RTEL1, NOP10, NHP2, CTC1, ACD (=TPP1), PARN, RPA1, WRAP53 (=TCBA1), DCLRE1B (Apollo), NPM1 
  • Dystrophic nails*

  • Skin pigmentations* (e.g., hyper/hypopigmented areas, often with a reticular pattern)

  • Oral leukoplakia*(potential precancerous)

  • BMF

  • Gray hairs and alopecia

  • Palmoplantar hyperkeratosis and hyperhidrosis

  • Short stature and osteoporosis

  • Avascular necrosis (e.g., hip or shoulder)

  • Lung fibrosis and emphysema

  • (Pulmonary) arteriovenous malformations

  • Liver fibrosis, liver cirrhosis, and various other liver diseases

  • (Gastro)intestinal diseases

  • Neurological symptoms

  • Visual disorders

  • Hypogonadism

  • Increased watering of the eyes due to lacrimal duct atresia

  • Dental caries

  • Esophageal strictures

  • Stenosis of the urethra

  • Increased cancer and leukemia risk

 
Hoyeraal-Hreidarsson syndrome [11, 64, 65, 73, 86] (severe phenotypic variant of DKC) (Manifestation: early childhood) DKC1, TERT (autosomal recessive), TINF2, PARN, WRAP53 (=TCBA1), ACD (=TPP1), RTEL1, NHP2, DCLRE1B (Apollo
  • Possible overlap with most characteristics of DKC

  • Cerebellar hypoplasia and microcephaly

  • Immunodeficiency (progressive)

  • (Intrauterine) growth restriction

  • Mental retardation

  • BMF (early onset and progressive)

  • Mucocutaneous lesions (e.g., hyper/hypopigmentation, nail dystrophy, and premalignant leukoplakia [oral + gastrointestinal])

  • Higher risk of cancer and PF

 
Revesz syndrome [71] (severe phenotypic variant of DKC) (Manifestation: early childhood) TINF2 
  • Bilateral exudative retinopathy

  • Skin hyper/hypopigmentation

  • Nail dystrophy

  • Oral leukoplakia

  • Fine and sparse hairs

  • Intracranial calcifications

  • Balance problems/ataxia

  • (Intrauterine) growth restriction

  • High risk of BMF

  • Risk of liver and lung fibrosis

  • High risk of cancer (e.g., leukemia)

 
Coats plus syndrome [48, 61, 69] (Manifestation: (early) childhood or (early) adolescence) CTC1, STN1, POT1 
  • Dilatation of blood vessels in the retina (exudative retinopathy/cerebroretinal microangiopathy)

  • (Brain) calcifications

  • (Brain) cysts

  • Gastrointestinal bleedings

  • Bone abnormalities (e.g., osteopenia)

  • Impaired bone healing

  • Leukodystrophy/leukoencephalopathy

  • Ataxia, seizures, spasticity, and cognitive decline

  • Anemia and thrombocytopenia

  • Changed skin pigmentations (café-au-lait spots)

  • Malformations of the nails

  • Prematurely gray hairs

  • Portal hypertension

  • Often without short TL

 
CrypticDKCs/TBDs(Manifestation: adolescence or (late) adulthood
BMF/aplastic anemia [41, 46, 50, 51, 55, 58, 63, 72DKC1, TERT, TERC, NOP10, NHP2, RTEL1, ACD (=TPP1), DCLRE1B (Apollo), TINF2, NPM1, MDM4, RPA1 
  • Pancytopenia (anemia, thrombocytopenia, neutropenia)

  • Hypocellular BM

  • Increased bleeding tendency

  • Increased susceptibility to infections

 
Idiopathic PF, interstitial lung diseases, emphysema [42, 47, 52, 54, 59, 62, 63, 75, 86, 89, 90DKC1, TERT, TERC, NOP10, NHP2, ACD (=TPP1), RTEL1, PARN, RPA1, NAF1, TINF2, ZCCHC8, POT1 
  • (Progressive) shortness of breath

  • Shallow and fast breathing

  • (Dry) cough – increased fatigue

  • (Unintended) weight loss

  • Clubbing of the finger tips (and toes)

 
Idiopathic liver cirrhosis/nodular regenerative hyperplasia/liver disease [68, 91, 92TERT, TERC, RTEL1, TINF2, DKC1, NHP2, NOP10, NAF1, WRAP53 (=TCBA1
  • Increased fatigue

  • Increased bleeding tendency/bruising

  • Nausea and decreased appetite

  • (Unintended) weight loss

  • Edema

  • Jaundice of the skin and eyes

  • Itchy skin

 
Head and neck cancer (HNSCC), squamous cell carcinoma (e.g., anogenital cancer, skin, or esophagus) and other cancers [51, 93‒100TERT (but also TERT promoter**), TERC, CTC1, MDM4, NAF1, POT1*** 
  • Cancer at a young age

  • Absence of risk factors

  • Family history of cancer or symptoms typical of TBD

  • Increased therapy-related toxicity

 
Others: MDS/hematopoietic diseases (e.g., TERT, TERC, RPA1, NAF1) [52, 63, 101, 102], immunodeficiencies/(auto)inflammation (e.g., TERT, TERC, DKC1, RTEL1) [84, 103, 104], and inflammatory bowel diseases/gastrointestinal manifestations [105, 106
DiseaseGenes that might harbor pathogenic variantsClinical manifestations
(Classical) Dyskeratosis congenita (DKC) (=Zinsser-Engman-Cole syndrome) [30, 49, 53, 55, 56, 66, 67, 70, 71, 74] (Manifestation: childhood or (early) adolescence) DKC1, TERT, TERC, TINF2, RTEL1, NOP10, NHP2, CTC1, ACD (=TPP1), PARN, RPA1, WRAP53 (=TCBA1), DCLRE1B (Apollo), NPM1 
  • Dystrophic nails*

  • Skin pigmentations* (e.g., hyper/hypopigmented areas, often with a reticular pattern)

  • Oral leukoplakia*(potential precancerous)

  • BMF

  • Gray hairs and alopecia

  • Palmoplantar hyperkeratosis and hyperhidrosis

  • Short stature and osteoporosis

  • Avascular necrosis (e.g., hip or shoulder)

  • Lung fibrosis and emphysema

  • (Pulmonary) arteriovenous malformations

  • Liver fibrosis, liver cirrhosis, and various other liver diseases

  • (Gastro)intestinal diseases

  • Neurological symptoms

  • Visual disorders

  • Hypogonadism

  • Increased watering of the eyes due to lacrimal duct atresia

  • Dental caries

  • Esophageal strictures

  • Stenosis of the urethra

  • Increased cancer and leukemia risk

 
Hoyeraal-Hreidarsson syndrome [11, 64, 65, 73, 86] (severe phenotypic variant of DKC) (Manifestation: early childhood) DKC1, TERT (autosomal recessive), TINF2, PARN, WRAP53 (=TCBA1), ACD (=TPP1), RTEL1, NHP2, DCLRE1B (Apollo
  • Possible overlap with most characteristics of DKC

  • Cerebellar hypoplasia and microcephaly

  • Immunodeficiency (progressive)

  • (Intrauterine) growth restriction

  • Mental retardation

  • BMF (early onset and progressive)

  • Mucocutaneous lesions (e.g., hyper/hypopigmentation, nail dystrophy, and premalignant leukoplakia [oral + gastrointestinal])

  • Higher risk of cancer and PF

 
Revesz syndrome [71] (severe phenotypic variant of DKC) (Manifestation: early childhood) TINF2 
  • Bilateral exudative retinopathy

  • Skin hyper/hypopigmentation

  • Nail dystrophy

  • Oral leukoplakia

  • Fine and sparse hairs

  • Intracranial calcifications

  • Balance problems/ataxia

  • (Intrauterine) growth restriction

  • High risk of BMF

  • Risk of liver and lung fibrosis

  • High risk of cancer (e.g., leukemia)

 
Coats plus syndrome [48, 61, 69] (Manifestation: (early) childhood or (early) adolescence) CTC1, STN1, POT1 
  • Dilatation of blood vessels in the retina (exudative retinopathy/cerebroretinal microangiopathy)

  • (Brain) calcifications

  • (Brain) cysts

  • Gastrointestinal bleedings

  • Bone abnormalities (e.g., osteopenia)

  • Impaired bone healing

  • Leukodystrophy/leukoencephalopathy

  • Ataxia, seizures, spasticity, and cognitive decline

  • Anemia and thrombocytopenia

  • Changed skin pigmentations (café-au-lait spots)

  • Malformations of the nails

  • Prematurely gray hairs

  • Portal hypertension

  • Often without short TL

 
CrypticDKCs/TBDs(Manifestation: adolescence or (late) adulthood
BMF/aplastic anemia [41, 46, 50, 51, 55, 58, 63, 72DKC1, TERT, TERC, NOP10, NHP2, RTEL1, ACD (=TPP1), DCLRE1B (Apollo), TINF2, NPM1, MDM4, RPA1 
  • Pancytopenia (anemia, thrombocytopenia, neutropenia)

  • Hypocellular BM

  • Increased bleeding tendency

  • Increased susceptibility to infections

 
Idiopathic PF, interstitial lung diseases, emphysema [42, 47, 52, 54, 59, 62, 63, 75, 86, 89, 90DKC1, TERT, TERC, NOP10, NHP2, ACD (=TPP1), RTEL1, PARN, RPA1, NAF1, TINF2, ZCCHC8, POT1 
  • (Progressive) shortness of breath

  • Shallow and fast breathing

  • (Dry) cough – increased fatigue

  • (Unintended) weight loss

  • Clubbing of the finger tips (and toes)

 
Idiopathic liver cirrhosis/nodular regenerative hyperplasia/liver disease [68, 91, 92TERT, TERC, RTEL1, TINF2, DKC1, NHP2, NOP10, NAF1, WRAP53 (=TCBA1
  • Increased fatigue

  • Increased bleeding tendency/bruising

  • Nausea and decreased appetite

  • (Unintended) weight loss

  • Edema

  • Jaundice of the skin and eyes

  • Itchy skin

 
Head and neck cancer (HNSCC), squamous cell carcinoma (e.g., anogenital cancer, skin, or esophagus) and other cancers [51, 93‒100TERT (but also TERT promoter**), TERC, CTC1, MDM4, NAF1, POT1*** 
  • Cancer at a young age

  • Absence of risk factors

  • Family history of cancer or symptoms typical of TBD

  • Increased therapy-related toxicity

 
Others: MDS/hematopoietic diseases (e.g., TERT, TERC, RPA1, NAF1) [52, 63, 101, 102], immunodeficiencies/(auto)inflammation (e.g., TERT, TERC, DKC1, RTEL1) [84, 103, 104], and inflammatory bowel diseases/gastrointestinal manifestations [105, 106

Shown are different manifestations of telomere biology disorders (TBDs) and clinical symptoms which fall within the spectrum of a TBD. The table demonstrates potential clinical manifestations as well as genes that were predominantly described in the literature for different variants of TBD. The table shows diseases that often manifest in the early childhood (Revesz and Hoyeraal-Hreidarsson syndrome), childhood and early adolescence (DKC and Coats plus syndrome) and during late adulthood (cryptic TBDs). Important clinical features are printed in bold.

*Classical DKC triad.

**Associated with longer TL.

***Might also cause longer telomeres.

About 80% of all cases show signs of BMF leading to significantly increased mortality. Especially patients with hematopoietic insufficiency experience a substantially increased risk of developing secondary hematopoietic malignancies such as myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) [85]. The cumulative risk of developing a secondary malignancy by the age of 50 years is about 40–50% [85]. As a result, median overall survival for patients with DKC was shown to be reduced to about 42 years in two independent cohorts [85].

In addition to TBDs first manifesting themselves in childhood or adolescence, there is nowadays increasing awareness about so-called late-onset TBDs also referred to as cryptic TBDs or adult-onset TBDs [36, 91]. Despite of this increasing perception as an important clinical subgroup, e.g., of patients with aplastic anemia (and/or liver cirrhosis and/or lung fibrosis), adult-onset hereditary syndromes in general and TBDs in particular are still severely underdiagnosed presumably because non-pediatricians are simply not as sensitized for a hereditary disease manifesting itself for the first time in adulthood [45]. What also contributes to this underdiagnosis is the fact that clinical symptoms are often subtle and highly variable between individuals as well as heterogeneously distributed over the organ systems involved [36, 43, 45]. The age of first manifestation depends very much on the respective genetic alteration and its respective impact on telomere biology (with or without the influence of additional external factors) [43, 76, 77]. Furthermore, TBDs are characterized by disease anticipation, i.e., age of critical telomere shortening and as a consequence, clinical onset of symptoms tends to decrease with consecutive generations [43, 44]. First manifestations often take place in middle-aged adulthood [36, 91]. The spectrum of cryptic TBDs might also include individuals who remain oligo- to even asymptomatic for most of their lives. Symptoms are often unspecific, making it very challenging to accurately diagnose a TBD [36, 91]. Cryptic TBD patients can present with only a single manifestation typically affecting lung, bone marrow, liver, or other TBD-associated symptoms [36, 45, 57, 91]. The four quantitatively most important clinical manifestations are given below.

Due to the high cell turnover of the hematopoietic system with daily blood cell productions exceeding 1012 cells in steady-state [107], blood and bone marrow cells are particularly susceptible toward defects in telomere maintenance genes [17, 45]. It is assumed that critically short telomeres functionally impact the individual bone marrow replicative capacity until eventually, the bone marrow function exhausts due to HSCs depletion [17]. TL can therefore be used as a (bio-)marker to approximate the degree of replicative exhaustion of the HSC compartment (very short TL indicates long-lasting and intensive compensatory proliferation of the residual HSC compartment) and/or severity of the underlying genetic defect. Pathogenic variants in DKC1, TERT, TERC, NOP10, NHP2, RTEL1, ACD (=TPP1), DCLRE1B (Apollo), TINF2, RTEL1, NPM1, MDM4, and RPA1 have been described in patients showing signs of BMF or aplastic anemia [41, 46, 50, 51, 55, 58, 63, 72]. Furthermore, pathogenic variants in TERT, TERC, RPA1, and NAF1 have been recently associated with MDS-typical changes in the bone marrow [52, 63, 101, 102]. Ineffective hematopoiesis and an increased risk of malignant transformation can therefore be considered a consequence of telomere-mediated genetic instability [41, 108].

Due to the fact that many TBD-associated diseases may lead to cytopenia and BMF, exclusion of a TBD is part of the comprehensive diagnostic assessment in every newly diagnosed case of BMF [43, 109]. In particular, the clarification of the family history, examination of the patient for TBD-typical stigmata (see Fig. 1) and TL measurement are indispensable (see Fig. 2) [43]. The assessment of TL represents a relatively cheap and essential (pre)screening tool which should be followed (in case of significantly shortened TL) by a further work-up with a next-generation sequencing approach that focuses on pathogenic variants in genes that are involved in telomere maintenance (Fig. 2).

Fig. 2.

Diagnostic procedures to confirm a suspected telomere biology disorder (TBD). The step-by-step diagnostic algorithm for proper identification of a telomere biology disorders (TBD) is shown as follows: (1) clinical presentation + basic diagnostics: the clinical appearance is often the first and sometimes the only indicator for a present TBD; (2) basic diagnostics such as a routine laboratory work-up mainly serve to exclude other causes that explain the patient’s phenotype; (3) flow-FISH diagnostic: to measure the absolute telomere length (TL) in peripheral blood lymphocytes and granulocytes; (4) advanced diagnostics: variable and highly dependent on the clinical presentation of the patient. Complications such as PF or liver fibrosis/liver cirrhosis can be diagnosed and quantified; (5) genetic work-up: NGS diagnostic, whole exome or whole-genome sequencing to detect a genetic cause of TBD. A chromosome breakage analysis should be performed to rule out other differential diagnoses that might lead to chromosomal instability and (secondary) telomere shortening (especially Fanconi anemia). Graph: possible results of telomere measurements and typical results for various diseases. Created with “BioRender.com.”

Fig. 2.

Diagnostic procedures to confirm a suspected telomere biology disorder (TBD). The step-by-step diagnostic algorithm for proper identification of a telomere biology disorders (TBD) is shown as follows: (1) clinical presentation + basic diagnostics: the clinical appearance is often the first and sometimes the only indicator for a present TBD; (2) basic diagnostics such as a routine laboratory work-up mainly serve to exclude other causes that explain the patient’s phenotype; (3) flow-FISH diagnostic: to measure the absolute telomere length (TL) in peripheral blood lymphocytes and granulocytes; (4) advanced diagnostics: variable and highly dependent on the clinical presentation of the patient. Complications such as PF or liver fibrosis/liver cirrhosis can be diagnosed and quantified; (5) genetic work-up: NGS diagnostic, whole exome or whole-genome sequencing to detect a genetic cause of TBD. A chromosome breakage analysis should be performed to rule out other differential diagnoses that might lead to chromosomal instability and (secondary) telomere shortening (especially Fanconi anemia). Graph: possible results of telomere measurements and typical results for various diseases. Created with “BioRender.com.”

Close modal

Correct identification of TBD patients is of utmost importance as patients with TBDs typically show no relevant clinical response to standard treatment for aplastic anemia like antithymocyte globulin, cyclosporine A, or eltrombopag [110]. In addition, TBDs are characterized by a significantly poorer outcome after allogeneic stem cell transplantation (allo-SCT) [111, 112]. Especially, the choice of a potentially affected family donor with the same genetic aberration should be avoided due to an increased risk of complications after allo-SCT (graft-failure, accelerated telomere shortening, and graft-versus-host disease) [112, 113]. A very careful donor selection, an adapted therapy regime and (if possible) avoidance of radiotherapy due to the increased risk of secondary malignancy as well as the development of fatal interstitial lung diseases are preferable for TBD patients [111, 113, 114]. In addition, special check-ups as well as follow-ups after treatment are recommended in order to recognize common secondary diseases (e.g., secondary malignancies) [44]. Moreover, the introduction of affected patients to a specialized center for a second opinion, study inclusion, and inclusion in central registries should be standard of care. There are a few registries world-wide that systematically collect patient data, document the disease course of TBD patients and systematically archive patient material (see Acknowledgment).

Pulmonary fibrosis (PF) is a disease characterized by progressive scar formation and thickening of the tissue around the alveoli in the lung thereby impairing the vital process of gas exchange [115]. The disease can be caused by external factors like air pollution, or previous medical treatments as radiation therapy or drugs with pulmonary toxicity (e.g., bleomycin, busulfan, and several other medications) [111, 116]. Some cases remain etiologically unclear and are therefore described as idiopathic [59, 115]. Usually, PF occurs in middle-aged and older individuals so that young patients with a family history raise a strong suspicion that the disease might be caused by pathogenic germline variants [115]. However, in analogy to inherited BMF, there are also reports indicating that even older patients suffering from PF might harbor pathogenic variants in TBD-associated genes that are inherited in an autosomal dominant manner [89]. Several studies showed that idiopathic PF can be caused by pathogenic variants in TBD-associated genes such as DKC1, TERT, TERC, NOP10, NHP2, ACD (=TPP1), RTEL1, PARN, RPA1, NAF1, TINF2, ZCCHC8, POT1, RPA1 [42, 47, 52, 54, 59, 62, 63, 75, 86]. It has also been shown that genetically impaired telomere maintenance is associated with several other interstitial lung diseases [59]. Symptom severity and type of manifestation differed greatly between patients with TBD-associated PF, but for all patients a continuously progressive disease course was observed [59].

Liver diseases are an increasing problem in the Western world with a significantly rising number of cases with non-alcoholic fatty liver disease and nonalcoholic steatohepatitis [117, 118]. In around 5% of all cases with liver cirrhosis, the underlying etiology cannot be identified, leading to the fact that these cases being classified as cryptogenic cirrhosis [118]. In TBD patients, a hepatic involvement is common (in some cohorts 40% of all TBD patients [119]), difficult to specifically detect [91] and contributes to mortality to a highly variable degree [91, 119]. Interestingly, the presence of a TBD-causing pathogenic variant was a risk factor for the progression from liver fibrosis to liver cirrhosis [120]. Studies in patients with liver cirrhosis who undergo liver transplantation have shown that cryptic TBD can be the cause of isolated liver fibrosis/liver cirrhosis [68, 92, 120]. It was demonstrated that aberrations in telomere maintenance genes such as TERT, TERC, RTEL1, TINF2, DKC1, NHP2, NOP10, NAF1, and WRAP53 (=TCBA1) were able to cause severe hepatic manifestations [68, 91, 92, 121].

Even though the risk for many cancers including head and neck squamous cell carcinoma (HNSCC) are associated with somatic TERT promoter and POT1 variants leading to longer TL [93, 94, 122], there are also indications that pathogenic germline variants in TBD-associated genes promote cancer development [17, 45]. Many tumor types that occur in the context of TBDs also show a higher incidence in the presence of (very) short telomeres in the general population [85, 108, 123‒125]. For example, variants in TERT, TERC, CTC1, and MDM4 lead to a higher risk for HNSCC and other squamous cell carcinomas (SCCs) (unpublished data) [51, 95, 126].

Patients with DKC have hundreds-fold greater risk of developing HNSCCs, SCCs of the skin and the anogenital region [85, 108]. 30% of the macroscopic oral leukoplakia in TBD patients might transform to HNSCC [108, 127]. Also, a higher risk of stomach cancer, lung, esophagus, and colon cancer is described in DKC patients [85]. Even the risk for AML development, Hodgkin’s disease as well as non-Hodgkin lymphoma was found to be increased [108]. Future studies are needed to address the question whether the increased susceptibility toward carcinogenesis in TBDs is explained by increased genetic instability only and/or by impaired tumor surveillance by the immune system [103]. Importantly, treatment of TBD-associated malignancies is expected to result in increased treatment-related toxicities due to a higher risk of functional organ damages, secondary BMF and increased mortality [87, 111, 116, 128]. For this reason, adapted therapy regimes (and components) including potential dose reductions in TBD patients suffering from cancer should be discussed individually with specialized TBD centers.

Androgen derivatives as cyclostragenol, oxymetholone, and danazol are molecules that may increase telomerase activity [129‒131]. The first case of a patient with AA who continuously showed in vivo multi-lineage telomere elongation in peripheral blood cells associated with transfusion independence following androgen treatment was published in 2012 [132]. For various androgen derivatives such as oxymetholone and danazol, elongation of TL in the peripheral blood was shown for TBD patients [109, 132‒136]. About 69% of all patients had a hematologic response to oxymetholone treatment [135], but side effects such as liver toxicity and virilization were frequent and were primarily observed in female patients [109, 135]. For danazol, on the other hand, side-effects were less frequent and about 50–100% of all TBD patients had at least a short-term response in the sense of an increase in blood counts [109, 135, 136]. Previous data showed that androgens are intracellularly converted to estrogens acting on the estrogen sensitive promotor of the telomerase gene resulting in an increased telomerase activity [129]. In line with this mechanism, an increased telomerase activity was observed after androgen treatment [129, 135]. Another study demonstrated that 11/12 patients under danazol treatment gained TL (mean increase of 386 bp) and 83% showed a hematologic response after 24 months [134].

Even though allo-SCT can reconstitute cytopenias in the hematopoietic system in TBD-associated BMF, long-term survival remains low in TBD patients with only 23% after 10 years [112]. One frequent long-term complication for TBD patients after allo-SCT was progressive PF [111, 112]. Due to the fact that increased (therapy-related) toxicity in TBD patients has been described [112], it was shown that a non-myeloablative protocol with reduced intensity and the avoidance of radiation were able to improve overall outcome [113, 114]. There are some indications that allo-SCT should not be enforced in patients with pre-existing organ damages, whereby patients with progressive BMF more likely benefit from the procedure [111, 113].

In the future, telomerase gene therapy represents a highly promising potential therapeutic strategy for patients with TBDs especially in patients with early BMF. Even though this concept is still experimental, first studies have already been carried out (NCT04211714). In line with this, in telomere-deficient mice that recapitulated the phenotype of BMF/aplastic anemia, cytopenias were successfully treated by using a Tert gene therapy system that was based on an adeno-associated virus serotype 9 (AAV9) vector [137]. Also, as another promising therapeutic approach, it was shown that PAPD5 inhibitors are able to restore telomerase activity in stem cells thereby implying that these drugs might be useful to restore the replicative capability of stem cells in TBD patients [138].

TBDs are characterized by impaired telomere maintenance that is often caused by pathogenic variants in various telomere-associated genes. Most patients with TBDs are prone to develop premature and eventually functional telomere shortening. Even though classical TBDs are very rare, the number of unreported cases of adult-onset TBDs is expected to be higher and every physician and particularly hematologist, hepatologist and pulmonologist should be aware of the classical symptom complexes and particularly, the heterogeneity of this hereditary disease group. TL screening by flow-FISH represents a sensitive and cost-effective method to functionally identify patients with suspected TBDs. This is of high importance as TBD patients require specific and individual treatment decisions, are prone to (often immediately life-threatening) complications and careful follow-up care concepts are needed to detect secondary (particularly malignant) disorders at the earliest possible time point (e.g., where they are still locally resectable). Furthermore, adequate counseling is of utmost importance in affected families. Inclusion into innovative clinical trials and registries including proper biobanking as well as connection of the patients with specialized patient support groups (see Acknowledgment) should be offered, e.g., with the help of a specialized tertiary hematology center.

We would like to take this opportunity to thank our dedicated telomere diagnostics laboratory at the University Hospital RWTH Aachen, especially Lucia Vankann and Anne Abels, whose dedicated work ensures that we can perform clinically valid and highly accurate TL measurements for centers in Europe and all over the world (https://www.ukaachen.de/kliniken-institute/klinik-fuer-haematologie-onkologie-haemostaseologie-und-stammzelltransplantation-med-klinik-iv/forschung/zentrum-fuer-telomererkrankungen-telomeropathien/). We would also like to thank our TBDs registry at the RWTH Aachen University, “https://teamtelomere.org,” “AA/PNH e.V.” and “Stiftung Lichterzellen” for their excellent support of collaborative research and targeted patient education. Furthermore, we would like to acknowledge several well-established TBD registers which are spread all over the globe (incomplete list): Bone Marrow Failure Registry (RWTH Aachen, Germany, covering German-speaking centers in Germany, Austria, and Switzerland), French National Registry of Bone Marrow Failures (RIME) (France), Canadian Inherited Marrow Failure Registry (Canada), Australian Aplastic Anemia and other Bone Marrow Failure Syndromes Registry (AAR) (Australia), Israeli Inherited Bone Marrow Failure Registry (Israel), NCI Inherited Bone Marrow Failure Syndrome Cohort Study (IBMFS) (USA), Bone Marrow Failure Research Study and Repository (Philadelphia) (USA), Pediatric Bone Marrow Failure and MDS Registry (Boston Children’s Hospital) (USA), and the Bone Marrow Failure and MDS registry (Dana Farber Cancer Center) (USA).

FB receives scientific support from RepeatDx, Vancouver. For all other authors, there is no conflict of interest to disclose in relation to this work.

B.R. received a Mildred-Scheel scholarship from the German Cancer Aid (“Deutsche Krebshilfe”) (No. 70114570). The funder had no role in the design, data collection, data analysis, or reporting of this study.

B.R. and T.H.B. conceptualized the project, designed and wrote the manuscript. B.R. has created the figures by using “BioRender.com.” M.T., M.K., R.M., and F.B. provided scientific input, revised, and updated the manuscript, figures, and tables accordingly. All authors approved the final version of the manuscript including figures and tables.

1.
Chakravarti
D
,
LaBella
KA
,
DePinho
RA
.
Telomeres: history, health, and hallmarks of aging
.
Cell
.
2021
;
184
(
2
):
306
22
.
2.
Muller
HJ
.
The remaking of chromosomes
.
Collecting Net
.
1938
;
8
:
198
.
3.
Blackburn
EH
,
Gall
JG
.
A tandemly repeated sequence at the termini of the extrachromosomal ribosomal RNA genes in Tetrahymena
.
J Mol Biol
.
1978
;
120
(
1
):
33
53
.
4.
Shay
JW
,
Wright
WE
.
Telomeres and telomerase: three decades of progress
.
Nat Rev Genet
.
2019
;
20
(
5
):
299
309
.
5.
Ghanim
GE
,
Fountain
AJ
,
van Roon
AMM
,
Rangan
R
,
Das
R
,
Collins
K
, et al
.
Structure of human telomerase holoenzyme with bound telomeric DNA
.
Nature
.
2021
;
593
(
7859
):
449
53
.
6.
Lim
CJ
,
Cech
TR
.
Shaping human telomeres: from shelterin and CST complexes to telomeric chromatin organization
.
Nat Rev Mol Cell Biol
.
2021
;
22
(
4
):
283
98
.
7.
Wlodarski
MW
.
The rise of Apollo, protector of telomeres
.
Blood
.
2022
;
139
(
16
):
2415
6
.
8.
Roake
CM
,
Artandi
SE
.
Regulation of human telomerase in homeostasis and disease
.
Nat Rev Mol Cell Biol
.
2020
;
21
(
7
):
384
97
.
9.
Lim
CJ
,
Zaug
AJ
,
Kim
HJ
,
Cech
TR
.
Reconstitution of human shelterin complexes reveals unexpected stoichiometry and dual pathways to enhance telomerase processivity
.
Nat Commun
.
2017
;
8
(
1
):
1075
.
10.
Zaug
AJ
,
Goodrich
KJ
,
Song
JJ
,
Sullivan
AE
,
Cech
TR
.
Reconstitution of a telomeric replicon organized by CST
.
Nature
.
2022
;
608
(
7924
):
819
25
.
11.
Touzot
F
,
Callebaut
I
,
Soulier
J
,
Gaillard
L
,
Azerrad
C
,
Durandy
A
, et al
.
Function of Apollo (SNM1B) at telomere highlighted by a splice variant identified in a patient with Hoyeraal-Hreidarsson syndrome
.
Proc Natl Acad Sci U S A
.
2010
;
107
(
22
):
10097
102
.
12.
Petronek
MS
,
Allen
BG
.
Maintenance of genome integrity by the late-acting cytoplasmic iron-sulfur assembly (CIA) complex
.
Front Genet
.
2023
;
14
:
1152398
.
13.
Gonzalez
OG
,
Assfalg
R
,
Koch
S
,
Schelling
A
,
Meena
JK
,
Kraus
J
, et al
.
Telomerase stimulates ribosomal DNA transcription under hyperproliferative conditions
.
Nat Commun
.
2014
;
5
:
4599
.
14.
Nassour
J
,
Aguiar
LG
,
Correia
A
,
Schmidt
TT
,
Mainz
L
,
Przetocka
S
, et al
.
Telomere-to-mitochondria signalling by ZBP1 mediates replicative crisis
.
Nature
.
2023
;
614
(
7949
):
767
73
.
15.
Alter
BP
,
Rosenberg
PS
,
Giri
N
,
Baerlocher
GM
,
Lansdorp
PM
,
Savage
SA
.
Telomere length is associated with disease severity and declines with age in dyskeratosis congenita
.
Haematologica
.
2012
;
97
(
3
):
353
9
.
16.
Brümmendorf
TH
,
Mak
J
,
Sabo
KM
,
Baerlocher
GM
,
Dietz
K
,
Abkowitz
JL
, et al
.
Longitudinal studies of telomere length in feline blood cells: implications for hematopoietic stem cell turnover in vivo
.
Exp Hematol
.
2002
;
30
(
10
):
1147
52
.
17.
Revy
P
,
Kannengiesser
C
,
Bertuch
AA
.
Genetics of human telomere biology disorders
.
Nat Rev Genet
.
2023
;
24
(
2
):
86
108
.
18.
von Zglinicki
T
.
Oxidative stress shortens telomeres
.
Trends Biochem Sci
.
2002
;
27
(
7
):
339
44
.
19.
Rolles
B
,
Ferreira
MSV
,
Vieri
M
,
Rheinwalt
KP
,
Schmitz
SM
,
Alizai
PH
, et al
.
Telomere length dynamics measured by flow-FISH in patients with obesity undergoing bariatric surgery
.
Sci Rep
.
2023
;
13
(
1
):
304
.
20.
Li
JSZ
,
Miralles Fusté
J
,
Simavorian
T
,
Bartocci
C
,
Tsai
J
,
Karlseder
J
, et al
.
TZAP: a telomere-associated protein involved in telomere length control
.
Science
.
2017
;
355
(
6325
):
638
41
.
21.
Lee
KH
,
Kimmel
M
.
Analysis of two mechanisms of telomere maintenance based on the theory of g-Networks and stochastic automata networks
.
BMC genomics
.
2020
;
21
(
Suppl 9
):
587
.
22.
Greider
CW
,
Blackburn
EH
.
Identification of a specific telomere terminal transferase activity in Tetrahymena extracts
.
Cell
.
1985
;
43
(
2 Pt 1
):
405
13
.
23.
Cesare
AJ
,
Reddel
RR
.
Alternative lengthening of telomeres: models, mechanisms and implications
.
Nat Rev Genet
.
2010
;
11
(
5
):
319
30
.
24.
Zinsser
F
.
Atrophia cutis reticularis cum pigmentatione dystrophia unguium et leukoplakia oris
.
Ikonogr Dermat Kioto
.
1910
;
5
:
219
23
.
25.
Engman
MF
.
Congenital atrophy of the skin, with reticular pigmentation
.
JAMA
.
1935
;
105
(
16
):
1252
.
26.
Cole
HN
.
Dyskeratosis congenita with pigmentation, dystrophia unguis and leukokeratosis oris
.
Arch Dermatol
.
1930
;
21
(
1
):
71
.
27.
Costello
MJ
,
Buncke
CM
.
Dyskeratosis congenita
.
JAMA Arch Derm
.
1956
;
73
(
2
):
123
32
.
28.
Sorrow
JM
,
Hitch
JM
.
Dyskeratosis congenita. First report of its occurrence in a female and a review of the literature
.
Arch Dermatol
.
1963
;
88
:
340
7
.
29.
Steier
W
,
van Voolen
GA
,
Selmanowitz
VJ
.
Dyskeratosis congenita: relationship to fanconi’s anemia
.
Blood
.
1972
;
39
(
4
):
510
21
.
30.
Heiss
NS
,
Knight
SW
,
Vulliamy
TJ
,
Klauck
SM
,
Wiemann
S
,
Mason
PJ
, et al
.
X-linked dyskeratosis congenita is caused by mutations in a highly conserved gene with putative nucleolar functions
.
Nat Genet
.
1998
;
19
(
1
):
32
8
.
31.
Knight
SW
,
Vulliamy
TJ
,
Heiss
NS
,
Matthijs
G
,
Devriendt
K
,
Connor
JM
, et al
.
1.4 Mb candidate gene region for X linked dyskeratosis congenita defined by combined haplotype and X chromosome inactivation analysis
.
J Med Genet
.
1998
;
35
(
12
):
993
6
.
32.
Rufer
N
,
Dragowska
W
,
Thornbury
G
,
Roosnek
E
,
Lansdorp
PM
.
Telomere length dynamics in human lymphocyte subpopulations measured by flow cytometry
.
Nat Biotechnol
.
1998
;
16
(
8
):
743
7
.
33.
Rufer
N
,
Brümmendorf
TH
,
Kolvraa
S
,
Bischoff
C
,
Christensen
K
,
Wadsworth
L
, et al
.
Telomere fluorescence measurements in granulocytes and T lymphocyte subsets point to a high turnover of hematopoietic stem cells and memory T cells in early childhood
.
J Exp Med
.
1999
;
190
(
2
):
157
67
.
34.
Brümmendorf
TH
,
Holyoake
TL
,
Rufer
N
,
Barnett
MJ
,
Schulzer
M
,
Eaves
CJ
, et al
.
Prognostic implications of differences in telomere length between normal and malignant cells from patients with chronic myeloid leukemia measured by flow cytometry
.
Blood
.
2000
;
95
(
6
):
1883
90
.
35.
Brümmendorf
TH
,
Rufer
N
,
Holyoake
TL
,
Maciejewski
J
,
Barnett
MJ
,
Eaves
CJ
, et al
.
Telomere length dynamics in normal individuals and in patients with hematopoietic stem cell-associated disorders
.
Ann N Y Acad Sci
.
2001
;
938
:
293
304
; discussion 303–4.
36.
Tometten
M
,
Kirschner
M
,
Meyer
R
,
Begemann
M
,
Halfmeyer
I
,
Vieri
M
, et al
.
Identification of adult patients with classical dyskeratosis congenita or cryptic telomere biology disorder by telomere length screening using age-modified criteria
.
Hemasphere
.
2023
;
7
(
5
):
e874
.
37.
Ferreira
MSV
,
Kirschner
M
,
Halfmeyer
I
,
Estrada
N
,
Xicoy
B
,
Isfort
S
, et al
.
Comparison of flow-FISH and MM-qPCR telomere length assessment techniques for the screening of telomeropathies
.
Ann N Y Acad Sci
.
2020
;
1466
(
1
):
93
103
.
38.
Ball
SE
,
Gibson
FM
,
Rizzo
S
,
Tooze
JA
,
Marsh
JC
,
Gordon-Smith
EC
.
Progressive telomere shortening in aplastic anemia
.
Blood
.
1998
;
91
(
10
):
3582
92
.
39.
Brümmendorf
TH
,
Maciejewski
JP
,
Mak
J
,
Young
NS
,
Lansdorp
PM
.
Telomere length in leukocyte subpopulations of patients with aplastic anemia
.
Blood
.
2001
;
97
(
4
):
895
900
.
40.
Yamaguchi
H
,
Calado
RT
,
Ly
H
,
Kajigaya
S
,
Baerlocher
GM
,
Chanock
SJ
, et al
.
Mutations in TERT, the gene for telomerase reverse transcriptase, in aplastic anemia
.
N Engl J Med
.
2005
;
352
(
14
):
1413
24
.
41.
Vulliamy
TJ
,
Walne
A
,
Baskaradas
A
,
Mason
PJ
,
Marrone
A
,
Dokal
I
.
Mutations in the reverse transcriptase component of telomerase (TERT) in patients with bone marrow failure
.
Blood Cell Mol Dis
.
2005
;
34
(
3
):
257
63
.
42.
Stuart
BD
,
Choi
J
,
Zaidi
S
,
Xing
C
,
Holohan
B
,
Chen
R
, et al
.
Exome sequencing links mutations in PARN and RTEL1 with familial pulmonary fibrosis and telomere shortening
.
Nat Genet
.
2015
;
47
(
5
):
512
7
.
43.
Niewisch
MR
,
Beier
F
,
Savage
SA
.
Clinical manifestations of telomere biology disorders in adults
.
Hematol Am Soc Hematol Educ Program
.
2023
;
2023
(
1
):
563
72
.
44.
Dokal
I
.
Dyskeratosis congenita
.
Hematol Am Soc Hematol Educ Program
.
2011
;
2011
:
480
6
.
45.
Savage
SA
,
Niewisch
MR
.
GeneReviews®: dyskeratosis congenita and related telomere biology disorders
.
Seattle (WA)
;
1993
.
46.
Guo
Y
,
Kartawinata
M
,
Li
J
,
Pickett
HA
,
Teo
J
,
Kilo
T
, et al
.
Inherited bone marrow failure associated with germline mutation of ACD, the gene encoding telomere protein TPP1
.
Blood
.
2014
;
124
(
18
):
2767
74
.
47.
Hoffman
TW
,
van der Vis
JJ
,
van der Smagt
JJ
,
Massink
MPG
,
Grutters
JC
,
van Moorsel
CHM
.
Pulmonary fibrosis linked to variants in the ACD gene, encoding the telomere protein TPP1
.
Eur Respir J
.
2019
;
54
(
6
):
1900809
.
48.
Anderson
BH
,
Kasher
PR
,
Mayer
J
,
Szynkiewicz
M
,
Jenkinson
EM
,
Bhaskar
SS
, et al
.
Mutations in CTC1, encoding conserved telomere maintenance component 1, cause Coats plus
.
Nat Genet
.
2012
;
44
(
3
):
338
42
.
49.
Keller
RB
,
Gagne
KE
,
Usmani
GN
,
Asdourian
GK
,
Williams
DA
,
Hofmann
I
, et al
.
CTC1 Mutations in a patient with dyskeratosis congenita
.
Pediatr Blood Cancer
.
2012
;
59
(
2
):
311
4
.
50.
Kermasson
L
,
Churikov
D
,
Awad
A
,
Smoom
R
,
Lainey
E
,
Touzot
F
, et al
.
Inherited human Apollo deficiency causes severe bone marrow failure and developmental defects
.
Blood
.
2022
;
139
(
16
):
2427
40
.
51.
Toufektchan
E
,
Lejour
V
,
Durand
R
,
Giri
N
,
Draskovic
I
,
Bardot
B
, et al
.
Germline mutation of MDM4, a major p53 regulator, in a familial syndrome of defective telomere maintenance
.
Sci Adv
.
2020
;
6
(
15
):
eaay3511
.
52.
Stanley
SE
,
Gable
DL
,
Wagner
CL
,
Carlile
TM
,
Hanumanthu
VS
,
Podlevsky
JD
, et al
.
Loss-of-function mutations in the RNA biogenesis factor NAF1 predispose to pulmonary fibrosis-emphysema
.
Sci Transl Med
.
2016
;
8
(
351
):
351ra107
.
53.
Walne
AJ
,
Vulliamy
T
,
Marrone
A
,
Beswick
R
,
Kirwan
M
,
Masunari
Y
, et al
.
Genetic heterogeneity in autosomal recessive dyskeratosis congenita with one subtype due to mutations in the telomerase-associated protein NOP10
.
Hum Mol Genet
.
2007
;
16
(
13
):
1619
29
.
54.
Kannengiesser
C
,
Manali
ED
,
Revy
P
,
Callebaut
I
,
Ba
I
,
Borgel
A
, et al
.
First heterozygous NOP10 mutation in familial pulmonary fibrosis
.
Eur Respir J
.
2020
;
55
(
6
):
1902465
.
55.
Nachmani
D
,
Bothmer
AH
,
Grisendi
S
,
Mele
A
,
Bothmer
D
,
Lee
JD
, et al
.
Germline NPM1 mutations lead to altered rRNA 2'-O-methylation and cause dyskeratosis congenita
.
Nat Genet
.
2019
;
51
(
10
):
1518
29
.
56.
Tummala
H
,
Walne
A
,
Collopy
L
,
Cardoso
S
,
La Fuente
J
,
Lawson
S
, et al
.
Poly(A)-specific ribonuclease deficiency impacts telomere biology and causes dyskeratosis congenita
.
J Clin Invest
.
2015
;
125
(
5
):
2151
60
.
57.
Feurstein
S
,
Adegunsoye
A
,
Mojsilovic
D
,
Vij
R
,
West DePersia
AH
,
Rajagopal
PS
, et al
.
Telomere biology disorder prevalence and phenotypes in adults with familial hematologic and/or pulmonary presentations
.
Blood Adv
.
2020
;
4
(
19
):
4873
86
.
58.
Dhanraj
S
,
Gunja
SMR
,
Deveau
AP
,
Nissbeck
M
,
Boonyawat
B
,
Coombs
AJ
, et al
.
Bone marrow failure and developmental delay caused by mutations in poly(A)-specific ribonuclease (PARN)
.
J Med Genet
.
2015
;
52
(
11
):
738
48
.
59.
Newton
CA
,
Batra
K
,
Torrealba
J
,
Kozlitina
J
,
Glazer
CS
,
Aravena
C
, et al
.
Telomere-related lung fibrosis is diagnostically heterogeneous but uniformly progressive
.
Eur Respir J
.
2016
;
48
(
6
):
1710
20
.
60.
Philippot
Q
,
Kannengiesser
C
,
Debray
MP
,
Gauvain
C
,
Ba
I
,
Vieri
M
, et al
.
Interstitial lung diseases associated with mutations of poly(A)-specific ribonuclease: a multicentre retrospective study
.
Respirology
.
2022
;
27
(
3
):
226
35
.
61.
Takai
H
,
Jenkinson
E
,
Kabir
S
,
Babul-Hirji
R
,
Najm-Tehrani
N
,
Chitayat
DA
, et al
.
A POT1 mutation implicates defective telomere end fill-in and telomere truncations in Coats plus
.
Genes Dev
.
2016
;
30
(
7
):
812
26
.
62.
Kelich
J
,
Aramburu
T
,
van der Vis
JJ
,
Showe
L
,
Kossenkov
A
,
van der Smagt
J
, et al
.
Telomere dysfunction implicates POT1 in patients with idiopathic pulmonary fibrosis
.
J Exp Med
.
2022
;
219
(
5
):
e20211681
.
63.
Sharma
R
,
Sahoo
SS
,
Honda
M
,
Granger
SL
,
Goodings
C
,
Sanchez
L
, et al
.
Gain-of-function mutations in RPA1 cause a syndrome with short telomeres and somatic genetic rescue
.
Blood
.
2022
;
139
(
7
):
1039
51
.
64.
Deng
Z
,
Glousker
G
,
Molczan
A
,
Fox
AJ
,
Lamm
N
,
Dheekollu
J
, et al
.
Inherited mutations in the helicase RTEL1 cause telomere dysfunction and Hoyeraal-Hreidarsson syndrome
.
Proc Natl Acad Sci U S A
.
2013
;
110
(
36
):
E3408
16
.
65.
Le Guen
T
,
Jullien
L
,
Touzot
F
,
Schertzer
M
,
Gaillard
L
,
Perderiset
M
, et al
.
Human RTEL1 deficiency causes Hoyeraal-Hreidarsson syndrome with short telomeres and genome instability
.
Hum Mol Genet
.
2013
;
22
(
16
):
3239
49
.
66.
Ballew
BJ
,
Yeager
M
,
Jacobs
K
,
Giri
N
,
Boland
J
,
Burdett
L
, et al
.
Germline mutations of regulator of telomere elongation helicase 1, RTEL1, in Dyskeratosis congenita
.
Hum Genet
.
2013
;
132
(
4
):
473
80
.
67.
Walne
AJ
,
Vulliamy
T
,
Kirwan
M
,
Plagnol
V
,
Dokal
I
.
Constitutional mutations in RTEL1 cause severe dyskeratosis congenita
.
Am J Hum Genet
.
2013
;
92
(
3
):
448
53
.
68.
Chiu
V
,
Hogen
R
,
Sher
L
,
Wadé
N
,
Conti
D
,
Martynova
A
, et al
.
Telomerase variants in patients with cirrhosis awaiting liver transplantation
.
Hepatology
.
2019
;
69
(
6
):
2652
63
.
69.
Simon
AJ
,
Lev
A
,
Zhang
Y
,
Weiss
B
,
Rylova
A
,
Eyal
E
, et al
.
Mutations in STN1 cause Coats plus syndrome and are associated with genomic and telomere defects
.
J Exp Med
.
2016
;
213
(
8
):
1429
40
.
70.
Vulliamy
T
,
Marrone
A
,
Goldman
F
,
Dearlove
A
,
Bessler
M
,
Mason
PJ
, et al
.
The RNA component of telomerase is mutated in autosomal dominant dyskeratosis congenita
.
Nature
.
2001
;
413
(
6854
):
432
5
.
71.
Savage
SA
,
Giri
N
,
Baerlocher
GM
,
Orr
N
,
Lansdorp
PM
,
Alter
BP
.
TINF2, a component of the shelterin telomere protection complex, is mutated in dyskeratosis congenita
.
Am J Hum Genet
.
2008
;
82
(
2
):
501
9
.
72.
Walne
AJ
,
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
(
9
):
3594
600
.
73.
Bergstrand
S
,
Böhm
S
,
Malmgren
H
,
Norberg
A
,
Sundin
M
,
Nordgren
A
, et al
.
Biallelic mutations in WRAP53 result in dysfunctional telomeres, Cajal bodies and DNA repair, thereby causing Hoyeraal-Hreidarsson syndrome
.
Cell Death Dis
.
2020
;
11
(
4
):
238
.
74.
Zhong
F
,
Savage
SA
,
Shkreli
M
,
Giri
N
,
Jessop
L
,
Myers
T
, et al
.
Disruption of telomerase trafficking by TCAB1 mutation causes dyskeratosis congenita
.
Genes Dev
.
2011
;
25
(
1
):
11
6
.
75.
Gable
DL
,
Gaysinskaya
V
,
Atik
CC
,
Talbot
CC
,
Kang
B
,
Stanley
SE
, et al
.
ZCCHC8, the nuclear exosome targeting component, is mutated in familial pulmonary fibrosis and is required for telomerase RNA maturation
.
Genes Dev
.
2019
;
33
(
19–20
):
1381
96
.
76.
Savage
SA
.
Dyskeratosis congenita and telomere biology disorders
.
Hematol Am Soc Hematol Educ Program
.
2022
;
2022
(
1
):
637
48
.
77.
Niewisch
MR
,
Giri
N
,
McReynolds
LJ
,
Alsaggaf
R
,
Bhala
S
,
Alter
BP
, et al
.
Disease progression and clinical outcomes in telomere biology disorders
.
Blood
.
2022
;
139
(
12
):
1807
19
.
78.
Karremann
M
,
Neumaier-Probst
E
,
Schlichtenbrede
F
,
Beier
F
,
Brümmendorf
TH
,
Cremer
FW
, et al
.
Revesz syndrome revisited
.
Orphanet J Rare Dis
.
2020
;
15
(
1
):
299
.
79.
Martens
UM
,
Zijlmans
JM
,
Poon
SS
,
Dragowska
W
,
Yui
J
,
Chavez
EA
, et al
.
Short telomeres on human chromosome 17p
.
Nat Genet
.
1998
;
18
(
1
):
76
80
.
80.
Beier
F
,
Balabanov
S
,
Buckley
T
,
Dietz
K
,
Hartmann
U
,
Rojewski
M
, et al
.
Accelerated telomere shortening in glycosylphosphatidylinositol (GPI)-negative compared with GPI-positive granulocytes from patients with paroxysmal nocturnal hemoglobinuria (PNH) detected by proaerolysin flow-FISH
.
Blood
.
2005
;
106
(
2
):
531
3
.
81.
Mathioudakis
G
,
Storb
R
,
McSweeney
PA
,
Torok-Storb
B
,
Lansdorp
PM
,
Brümmendorf
TH
, et al
.
Polyclonal hematopoiesis with variable telomere shortening in human long-term allogeneic marrow graft recipients
.
Blood
.
2000
;
96
(
12
):
3991
4
.
82.
Rufer
N
,
Brümmendorf
TH
,
Chapuis
B
,
Helg
C
,
Lansdorp
PM
,
Roosnek
E
.
Accelerated telomere shortening in hematological lineages is limited to the first year following stem cell transplantation
.
Blood
.
2001
;
97
(
2
):
575
7
.
83.
Alter
BP
,
Baerlocher
GM
,
Savage
SA
,
Chanock
SJ
,
Weksler
BB
,
Willner
JP
, et al
.
Very short telomere length by flow fluorescence in situ hybridization identifies patients with dyskeratosis congenita
.
Blood
.
2007
;
110
(
5
):
1439
47
.
84.
Rolles
B
,
Caballero-Oteyza
A
,
Proietti
M
,
Goldacker
S
,
Warnatz
K
,
Camacho-Ordonez
N
, et al
.
Telomere biology disorders may manifest as common variable immunodeficiency (CVID)
.
Clin Immunol
.
2023
;
257
:
109837
.
85.
Alter
BP
,
Giri
N
,
Savage
SA
,
Rosenberg
PS
.
Cancer in dyskeratosis congenita
.
Blood
.
2009
;
113
(
26
):
6549
57
.
86.
Benyelles
M
,
O’Donohue
MF
,
Kermasson
L
,
Lainey
E
,
Borie
R
,
Lagresle-Peyrou
C
, et al
.
NHP2 deficiency impairs rRNA biogenesis and causes pulmonary fibrosis and Høyeraal-Hreidarsson syndrome
.
Hum Mol Genet
.
2020
;
29
(
6
):
907
22
.
87.
Pereboeva
L
,
Westin
E
,
Patel
T
,
Flaniken
I
,
Lamb
L
,
Klingelhutz
A
, et al
.
DNA damage responses and oxidative stress in dyskeratosis congenita
.
PLoS One
.
2013
;
8
(
10
):
e76473
.
88.
Walne
AJ
,
Collopy
L
,
Cardoso
S
,
Ellison
A
,
Plagnol
V
,
Albayrak
C
, et al
.
Marked overlap of four genetic syndromes with dyskeratosis congenita confounds clinical diagnosis
.
Haematologica
.
2016
;
101
(
10
):
1180
9
.
89.
Armanios
MY
,
Chen
JJL
,
Cogan
JD
,
Alder
JK
,
Ingersoll
RG
,
Markin
C
, et al
.
Telomerase mutations in families with idiopathic pulmonary fibrosis
.
N Engl J Med
.
2007
;
356
(
13
):
1317
26
.
90.
Stanley
SE
,
Chen
JJL
,
Podlevsky
JD
,
Alder
JK
,
Hansel
NN
,
Mathias
RA
, et al
.
Telomerase mutations in smokers with severe emphysema
.
J Clin Invest
.
2015
;
125
(
2
):
563
70
.
91.
Tometten
M
,
Kirschner
M
,
Isfort
S
,
Berres
ML
,
Brümmendorf
TH
,
Beier
F
.
Transient elastography in adult patients with cryptic dyskeratosis congenita reveals subclinical liver fibrosis: a retrospective analysis of the Aachen telomere biology disease registry
.
Orphanet J Rare Dis
.
2021
;
16
(
1
):
395
.
92.
Gorgy
AI
,
Jonassaint
NL
,
Stanley
SE
,
Koteish
A
,
DeZern
AE
,
Walter
JE
, et al
.
Hepatopulmonary syndrome is a frequent cause of dyspnea in the short telomere disorders
.
Chest
.
2015
;
148
(
4
):
1019
26
.
93.
Annunziata
C
,
Pezzuto
F
,
Greggi
S
,
Ionna
F
,
Losito
S
,
Botti
G
, et al
.
Distinct profiles of TERT promoter mutations and telomerase expression in head and neck cancer and cervical carcinoma
.
Int J Cancer
.
2018
;
143
(
5
):
1153
61
.
94.
Yu
Y
,
Fan
D
,
Song
X
,
Zakeri
K
,
Chen
L
,
Kang
J
, et al
.
TERT promoter mutations are enriched in oral cavity cancers and associated with locoregional recurrence
.
JCO Precis Oncol
.
2021
;
5
:
1259
69
.
95.
Gu
Y
,
Yu
C
,
Miao
L
,
Wang
L
,
Xu
C
,
Xue
W
, et al
.
Telomere length, genetic variants and risk of squamous cell carcinoma of the head and neck in Southeast Chinese
.
Sci Rep
.
2016
;
6
:
20675
.
96.
Li
Z
,
Song
Y
,
Xu
Y
,
Shen
Y
,
Zhang
N
,
Yang
M
, et al
.
Identification of Leukocyte telomere length-related genetic variants contributing to predisposition of Esophageal Squamous Cell Carcinoma
.
J Cancer
.
2020
;
11
(
17
):
5025
31
.
97.
Baird
DM
.
Variation at the TERT locus and predisposition for cancer
.
Expert Rev Mol Med
.
2010
;
12
:
e16
.
98.
Campa
D
,
Matarazzi
M
,
Greenhalf
W
,
Bijlsma
M
,
Saum
KU
,
Pasquali
C
, et al
.
Genetic determinants of telomere length and risk of pancreatic cancer: a PANDoRA study
.
Int J Cancer
.
2019
;
144
(
6
):
1275
83
.
99.
Hosgood
HD
,
Cawthon
R
,
He
X
,
Chanock
S
,
Lan
Q
.
Genetic variation in telomere maintenance genes, telomere length, and lung cancer susceptibility
.
Lung Cancer
.
2009
;
66
(
2
):
157
61
.
100.
McKay
JD
,
Hung
RJ
,
Gaborieau
V
,
Boffetta
P
,
Chabrier
A
,
Byrnes
G
, et al
.
Lung cancer susceptibility locus at 5p15.33
.
Nat Genet
.
2008
;
40
(
12
):
1404
6
.
101.
Kirwan
M
,
Vulliamy
T
,
Marrone
A
,
Walne
AJ
,
Beswick
R
,
Hillmen
P
, et al
.
Defining the pathogenic role of telomerase mutations in myelodysplastic syndrome and acute myeloid leukemia
.
Hum Mutat
.
2009
;
30
(
11
):
1567
73
.
102.
Reilly
CR
,
Myllymäki
M
,
Redd
R
,
Padmanaban
S
,
Karunakaran
D
,
Tesmer
V
, et al
.
The clinical and functional effects of TERT variants in myelodysplastic syndrome
.
Blood
.
2021
;
138
(
10
):
898
911
.
103.
Schratz
KE
,
Flasch
DA
,
Atik
CC
,
Cosner
ZL
,
Blackford
AL
,
Yang
W
, et al
.
T cell immune deficiency rather than chromosome instability predisposes patients with short telomere syndromes to squamous cancers
.
Cancer Cell
.
2023
;
41
(
4
):
807
17.e6
.
104.
Wagner
CL
,
Hanumanthu
VS
,
Talbot
CC
,
Abraham
RS
,
Hamm
D
,
Gable
DL
, et al
.
Short telomere syndromes cause a primary T cell immunodeficiency
.
J Clin Invest
.
2018
;
128
(
12
):
5222
34
.
105.
Trotta
L
,
Norberg
A
,
Taskinen
M
,
Béziat
V
,
Degerman
S
,
Wartiovaara-Kautto
U
, et al
.
Diagnostics of rare disorders: whole-exome sequencing deciphering locus heterogeneity in telomere biology disorders
.
Orphanet J Rare Dis
.
2018
;
13
(
1
):
139
.
106.
Ziv
A
,
Werner
L
,
Konnikova
L
,
Awad
A
,
Jeske
T
,
Hastreiter
M
, et al
.
An RTEL1 mutation links to infantile-onset ulcerative colitis and severe immunodeficiency
.
J Clin Immunol
.
2020
;
40
(
7
):
1010
9
.
107.
Higgins
JM
.
Red blood cell population dynamics
.
Clin Lab Med
.
2015
;
35
(
1
):
43
57
.
108.
Alter
BP
,
Giri
N
,
Savage
SA
,
Rosenberg
PS
.
Cancer in the National Cancer Institute inherited bone marrow failure syndrome cohort after fifteen years of follow-up
.
Haematologica
.
2018
;
103
(
1
):
30
9
.
109.
Calado
RT
,
Clé
DV
.
Treatment of inherited bone marrow failure syndromes beyond transplantation
.
Hematol Am Soc Hematol Educ Program
.
2017
;
2017
(
1
):
96
101
.
110.
Sakaguchi
H
,
Nishio
N
,
Hama
A
,
Kawashima
N
,
Wang
X
,
Narita
A
, et al
.
Peripheral blood lymphocyte telomere length as a predictor of response to immunosuppressive therapy in childhood aplastic anemia
.
Haematologica
.
2014
;
99
(
8
):
1312
6
.
111.
Gadalla
SM
,
Sales-Bonfim
C
,
Carreras
J
,
Alter
BP
,
Antin
JH
,
Ayas
M
, et al
.
Outcomes of allogeneic hematopoietic cell transplantation in patients with dyskeratosis congenita
.
Biol Blood Marrow Transplant
.
2013
;
19
(
8
):
1238
43
.
112.
Elmahadi
S
,
Muramatsu
H
,
Kojima
S
.
Allogeneic hematopoietic stem cell transplantation for dyskeratosis congenita
.
Curr Opin Hematol
.
2016
;
23
(
6
):
501
7
.
113.
Fioredda
F
,
Iacobelli
S
,
Korthof
ET
,
Knol
C
,
van Biezen
A
,
Bresters
D
, et al
.
Outcome of haematopoietic stem cell transplantation in dyskeratosis congenita
.
Br J Haematol
.
2018
;
183
(
1
):
110
8
. Or R.
114.
Nelson
AS
,
Marsh
RA
,
Myers
KC
,
Davies
SM
,
Jodele
S
,
O'Brien
TA
, et al
.
A reduced-intensity conditioning regimen for patients with dyskeratosis congenita undergoing hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2016
;
22
(
5
):
884
8
.
115.
Courtwright
AM
,
El-Chemaly
S
.
Telomeres in interstitial lung disease: the short and the long of it
.
Ann Am Thorac Soc
.
2019
;
16
(
2
):
175
81
.
116.
Schwaiblmair
M
,
Behr
W
,
Haeckel
T
,
Märkl
B
,
Foerg
W
,
Berghaus
T
.
Drug induced interstitial lung disease
.
Open Respir Med J
.
2012
;
6
:
63
74
.
117.
Schuppan
D
,
Afdhal
NH
.
Liver cirrhosis
.
Lancet
.
2008
;
371
(
9615
):
838
51
.
118.
Ayata
G
,
Gordon
FD
,
Lewis
WD
,
Pomfret
E
,
Pomposelli
JJ
,
Jenkins
RL
, et al
.
Cryptogenic cirrhosis: clinicopathologic findings at and after liver transplantation
.
Hum Pathol
.
2002
;
33
(
11
):
1098
104
.
119.
Kapuria
D
,
Ben-Yakov
G
,
Ortolano
R
,
Cho
MH
,
Kalchiem-Dekel
O
,
Takyar
V
, et al
.
The spectrum of hepatic involvement in patients with telomere disease
.
Hepatology
.
2019
;
69
(
6
):
2579
85
.
120.
Calado
RT
,
Brudno
J
,
Mehta
P
,
Kovacs
JJ
,
Wu
C
,
Zago
MA
, et al
.
Constitutional telomerase mutations are genetic risk factors for cirrhosis
.
Hepatology
.
2011
;
53
(
5
):
1600
7
.
121.
Hartmann
D
,
Srivastava
U
,
Thaler
M
,
Kleinhans
KN
,
N’kontchou
G
,
Scheffold
A
, et al
.
Telomerase gene mutations are associated with cirrhosis formation
.
Hepatology
.
2011
;
53
(
5
):
1608
17
.
122.
DeBoy
EA
,
Tassia
MG
,
Schratz
KE
,
Yan
SM
,
Cosner
ZL
,
McNally
EJ
, et al
.
Familial clonal hematopoiesis in a long telomere syndrome
.
N Engl J Med
.
2023
;
388
(
26
):
2422
33
.
123.
Liu
X
,
Bao
G
,
Huo
T
,
Wang
Z
,
He
X
,
Dong
G
.
Constitutive telomere length and gastric cancer risk: case-control analysis in Chinese Han population
.
Cancer Sci
.
2009
;
100
(
7
):
1300
5
.
124.
Ma
H
,
Zhou
Z
,
Wei
S
,
Liu
Z
,
Pooley
KA
,
Dunning
AM
, et al
.
Shortened telomere length is associated with increased risk of cancer: a meta-analysis
.
PLoS One
.
2011
;
6
:
e20466
.
125.
Willeit
P
,
Willeit
J
,
Mayr
A
,
Weger
S
,
Oberhollenzer
F
,
Brandstätter
A
, et al
.
Telomere length and risk of incident cancer and cancer mortality
.
JAMA
.
2010
;
304
(
1
):
69
75
.
126.
Sun
P
,
Wei
P
,
Liu
H
,
Wu
J
,
Gross
ND
,
Sikora
AG
, et al
.
GWAS-identified telomere length associated genetic variants predict risk of recurrence of HPV-positive oropharyngeal cancer after definitive radiotherapy
.
EBioMedicine
.
2023
;
94
:
104722
.
127.
Liu
AQ
,
Deane
EC
,
Prisman
E
,
Durham
JS
.
Dyskeratosis congenita and squamous cell cancer of the head and neck: a case report and systematic review
.
Ann Otol Rhinol Laryngol
.
2022
;
131
(
9
):
1036
42
.
128.
Rolles
B
,
Gorgulho
J
,
Tometten
M
,
Roderburg
C
,
Vieri
M
,
Abels
A
, et al
.
Telomere shortening in peripheral leukocytes is associated with poor survival in cancer patients treated with immune checkpoint inhibitor therapy
.
Front Oncol
.
2021
;
11
:
729207
.
129.
Calado
RT
,
Yewdell
WT
,
Wilkerson
KL
,
Regal
JA
,
Kajigaya
S
,
Stratakis
CA
, et al
.
Sex hormones, acting on the TERT gene, increase telomerase activity in human primary hematopoietic cells
.
Blood
.
2009
;
114
(
11
):
2236
43
.
130.
Yilmaz
S
,
Bedir
E
,
Ballar Kirmizibayrak
P
.
The role of cycloastragenol at the intersection of NRF2/ARE, telomerase, and proteasome activity
.
Free Radic Biol Med
.
2022
;
188
:
105
16
.
131.
Vieri
M
,
Kirschner
M
,
Tometten
M
,
Abels
A
,
Rolles
B
,
Isfort
S
, et al
.
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
(
19
):
7196
.
132.
Ziegler
P
,
Schrezenmeier
H
,
Akkad
J
,
Brassat
U
,
Vankann
L
,
Panse
J
, et al
.
Telomere elongation and clinical response to androgen treatment in a patient with aplastic anemia and a heterozygous hTERT gene mutation
.
Ann Hematol
.
2012
;
91
(
7
):
1115
20
.
133.
Islam
A
,
Rafiq
S
,
Kirwan
M
,
Walne
A
,
Cavenagh
J
,
Vulliamy
T
, et al
.
Haematological recovery in dyskeratosis congenita patients treated with danazol
.
Br J Haematol
.
2013
;
162
(
6
):
854
6
.
134.
Townsley
DM
,
Dumitriu
B
,
Liu
D
,
Biancotto
A
,
Weinstein
B
,
Chen
C
, et al
.
Danazol treatment for telomere diseases
.
N Engl J Med
.
2016
;
374
(
20
):
1922
31
.
135.
Khincha
PP
,
Wentzensen
IM
,
Giri
N
,
Alter
BP
,
Savage
SA
.
Response to androgen therapy in patients with dyskeratosis congenita
.
Br J Haematol
.
2014
;
165
(
3
):
349
57
.
136.
Kirschner
M
,
Vieri
M
,
Kricheldorf
K
,
Ferreira
MSV
,
Wlodarski
MW
,
Schwarz
M
, et al
.
Androgen derivatives improve blood counts and elongate telomere length in adult cryptic dyskeratosis congenita
.
Br J Haematol
.
2021
;
193
(
3
):
669
73
.
137.
Bär
C
,
Povedano
JM
,
Serrano
R
,
Benitez-Buelga
C
,
Popkes
M
,
Formentini
I
, et al
.
Telomerase gene therapy rescues telomere length, bone marrow aplasia, and survival in mice with aplastic anemia
.
Blood
.
2016
;
127
(
14
):
1770
9
.
138.
Nagpal
N
,
Wang
J
,
Zeng
J
,
Lo
E
,
Moon
DH
,
Luk
K
, et al
.
Small-molecule PAPD5 inhibitors restore telomerase activity in patient stem cells
.
Cell Stem Cell
.
2020
;
26
(
6
):
896
909.e8
.