scholarly journals Conditional TRF1 knockout in the hematopoietic compartment leads to bone marrow failure and recapitulates clinical features of dyskeratosis congenita

Blood ◽  
2012 ◽  
Vol 120 (15) ◽  
pp. 2990-3000 ◽  
Author(s):  
Fabian Beier ◽  
Miguel Foronda ◽  
Paula Martinez ◽  
Maria A. Blasco

Abstract TRF1 is part of the shelterin complex, which binds telomeres and it is essential for their protection. Ablation of TRF1 induces sister telomere fusions and aberrant numbers of telomeric signals associated with telomere fragility. Dyskeratosis congenita is characterized by a mucocutaneous triad, bone marrow failure (BMF), and presence of short telomeres because of mutations in telomerase. A subset of patients, however, show mutations in the shelterin component TIN2, a TRF1-interacting protein, presenting a more severe phenotype and presence of very short telomeres despite normal telomerase activity. Allelic variations in TRF1 have been found associated with BMF. To address a possible role for TRF1 dysfunction in BMF, here we generated a mouse model with conditional TRF1 deletion in the hematopoietic system. Chronic TRF1 deletion results in increased DNA damage and cellular senescence, but not increased apoptosis, in BM progenitor cells, leading to severe aplasia. Importantly, increased compensatory proliferation of BM stem cells is associated with rapid telomere shortening and further increase in senescent cells in vivo, providing a mechanism for the very short telomeres of human patients with mutations in the shelterin TIN2. Together, these results represent proof of principle that mutations in TRF1 lead to the main clinical features of BMF.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4052-4052
Author(s):  
Travis Witt ◽  
Aloysius Klingelhutz ◽  
Erik Westin ◽  
Preeti Satyanarayana ◽  
Peter M. Lansdorp ◽  
...  

Abstract Dyskeratosis congenita (DC) is an inherited multisystem disorder of premature aging, typically characterized by bone marrow failure, mucosal leukoplakia, abnormal skin pigmentation, and nail dystrophy. The X-linked and autosomal dominant forms of DC are associated with mutations in genes that affect telomerase activity resulting in a decrease in telomere length. DC, like other bone marrow failure disorders, is associated with ineffective hematopoiesis and a cancer predisposition. Standard treatment of bone marrow failure or cancer requires cytotoxic therapy, and clinical observations suggest DC patients have an increased sensitivity to cytotoxic therapy. To explain this, we hypothesized that the short telomeres in somatic cells from DC patients could alter the activity and/or expression of several proteins involved in DNA repair or the response to cellular stress including p16, p53 and p21. Lymphocytes from five DC subjects and age-matched controls were stimulated to grow in vitro in the presence of various cytotoxic agents with different modes of action, including Taxol (antimitotic agent and microtubule inhibitor) and Etoposide (topoisomerase inhibitor and DNA damaging agent). In addition, we tested fibroblasts and keratinocyte extracted from skin biopsies from DC and control subjects that were serially passaged. Cellular proliferation and cell death were monitored by cell counts and flow cytometry. Western blotting was used to measure steady state and DNA damage- induced expression of tumor suppressor protein p53 and other proteins involved in DNA damage response signaling pathway, including p16 and p21 in relation to telomere length. Results of flow cytometry accompanied by direct visualization showed a decreased proliferation of DC lymphocytes compared to normal cells, and this growth disadvantage was further accentuated following cell exposure to cytotoxic agents. DC lymphocytes exposed to 10−6 M Taxol showed a decrease in cellular proliferation between 3 and 8 fold while normal control cells exposed to the same agents exhibited only a 3 to 4 fold decrease in cell growth. Similarly DC lymphocytes exposed to Etoposide were inhibited to a greater extent than control cells. Western blot analysis of whole cell lysates indicated a difference in DNA damage response proteins. Of note, lymphocytes from several DC subjects exposed to Taxol did not upregulate p53 expression, while inducible levels were noted in Taxol-treated control cells. In contrast, DC and control lymphocytes exposed to Etoposide upregulated p53 in a similar dose dependent manner. No differences were noted in DC versus control lymphocytes with regards to basal or chemotherapy induced p16 expression. Interestingly, late passage DC fibroblasts displayed enhanced basal expression of p16. These results support the clinical observation of increased “chemosensitivity” in DC subjects and suggest that diminished telomerase activity and premature telomere shortening may interfere with normal DNA damage and stress response pathways. These data are also consistent with our finding that DC fibroblasts, keratinocytes, and lymphocytes have a reduced cell proliferative lifespan. Further studies are needed to dissect the role of telomeres in the cellular response to various types of DNA damage.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2230-2230
Author(s):  
Blanche P Alter ◽  
Neelam Giri ◽  
Peter M. Lansdorp ◽  
Gabriela M. Baerlocher ◽  
Philip S Rosenberg ◽  
...  

Abstract Abstract 2230 Dyskeratosis congenita (DC) is an inherited bone marrow failure syndrome with a complex clinical phenotype, including dysplastic nails, lacy reticular pigmentation, and oral leukoplakia (the diagnostic triad). Numerous other physical abnormalities may be present, in addition to cytopenias due to bone marrow failure, and a high risk of leukemia or solid tumors. However, many patients have no physical findings at diagnosis. Patients with DC have very short telomeres, and approximately one-half have a mutation in one of six genes important in telomere biology. Telomere length in leukocyte subsets, measured by automated flow fluorescence in situ hybridization (flow-FISH), is both sensitive and specific for identifying individuals with DC. Telomeres consist of nucleotide repeats and a protein complex at chromosome ends that are critical in chromosomal stability which shorten during normal cell division. Cross-sectional studies of normal individuals suggest that telomere length decreases with age in a sigmoid pattern from birth to old age. In a cross-sectional analysis of 26 patients with DC, we previously observed that telomere length appeared to be stable or even to slightly increase with age (BP Alter et al, Blood 110:149, 2007). Similar results were shown in 23 different DC patients by others (M Bessler et al, FEBS Lett 2010 in press). We speculated that these data were influenced by early presentation (or recognition) of clinically more severe patients, while patients with similar telomere length who were clinically milder were identified at older ages. In this pilot study, we examined, for the first time, the longitudinal age-association of telomere attrition in nine patients with DC who were followed for five to seven years (currently 8 – 50 years of age). These include three patients with mutations in TERC, and two each with TINF2, TERT, and DKC1 mutations. When first studied, four had normal hematopoiesis, three moderate cytopenias, one was receiving androgens, and one was on transfusions. Subsequently, one with normal hematopoiesis developed mild thrombocytopenia, one who was on transfusions responded to androgens, and one with moderate aplastic anemia became severe. In all cases, telomere length decreased with age. In a linear regression model, the average annual decrease in telomere length in lymphocytes was 167 base pairs/year (bp/yr) + 104, similar to the rate in granulocytes, 159 + 92 bp/yr. According to the literature, the rate of telomere attrition in longitudinal studies in normal blood is ∼45-50 bp/yr, albeit by methods other than flow-FISH; the rate of telomere shortening appears to decrease with increasing age. The average patient Z-scores at the beginning of the study were -3.9 standard deviations below the median for age in healthy normal controls, and were -4.3 at the end, consistent with the impression that DC patient telomeres shorten somewhat more than expected from normal aging. These data support the hypothesis that the earlier cross-sectional results for patients with DC indeed were influenced by the cross-sectional rather than longitudinal nature of the data. The current longitudinal data suggest that telomere shortening could possibly be accelerated in patients with DC, but larger studies are required. Our results indicate that patients with DC have telomeres that are much shorter than normal for their age, and that over time they continue to shorten, consistent with DC being classified as a disorder of premature aging. Disclosures: Lansdorp: Repeat Diagnostics: Equity Ownership.


Blood ◽  
2008 ◽  
Vol 111 (9) ◽  
pp. 4446-4455 ◽  
Author(s):  
Rodrigo T. Calado ◽  
Neal S. Young

AbstractAcquired and congenital aplastic anemias recently have been linked molecularly and pathophysiologically by abnormal telomere maintenance. Telomeres are repeated nucleotide sequences that cap the ends of chromosomes and protect them from damage. Telomeres are eroded with cell division, but in hematopoietic stem cells, maintenance of their length is mediated by telomerase. Accelerated telomere shortening is virtually universal in dyskeratosis congenita, caused by mutations in genes encoding components of telomerase or telomere-binding protein (TERT, TERC, DKC1, NOP10, or TINF2). About one-third of patients with acquired aplastic anemia also have short telomeres, which in some cases associate with TERT or TERC mutations. These mutations cause low telomerase activity, accelerated telomere shortening, and diminished proliferative capacity of hematopoietic progenitors. As in other genetic diseases, additional environmental, genetic, and epigenetic modifiers must contribute to telomere erosion and ultimately to disease phenotype. Short telomeres also may cause genomic instability and malignant progression in these marrow failure syndromes. Identification of short telomeres has potential clinical implications: it may be useful in dyskeratosis congenita diagnosis, in suggesting mutations in patients with acquired aplastic anemia, and for selection of suitable hematopoietic stem cell family donors for transplantation in telomerase-deficient patients.


2020 ◽  
Vol 78 (4) ◽  
pp. 369-372
Author(s):  
Virgínia Vinha Zanuncio ◽  
Kelvin Oliveira Rocha

Dyskeratosis Congenita (DC) is a hereditary and multisystemic syndrome, with heterogeneous clinical and genetic manifestations, characterized as a disease associated with maintenance of defects and early telomere shortening. It is a rare condition, with an estimated annual incidence of 1 in 1 million individuals, and is more common in males than females, with an approximate ratio of 10:1. The main initial clinical manifestations are disorders of cutaneous pigmentation, nail dystrophy and abnormalities in the oropharynx, usually occurring between five and twelve years of age. Bone marrow failure (BMF) is the leading cause of death related to this disease. We present a case report of a child, female, two years old, who presented the cardinal signs and symptoms of DC at an early age, and we emphasize the importance of multidisciplinary monitoring of the patient.


2008 ◽  
Vol 28 (7) ◽  
pp. 2332-2341 ◽  
Author(s):  
Kalpana Kannan ◽  
Andrew D. L. Nelson ◽  
Dorothy E. Shippen

ABSTRACT Dyskerin binds the H/ACA box of human telomerase RNA and is a core telomerase subunit required for RNP biogenesis and enzyme function in vivo. Missense mutations in dyskerin result in dyskeratosis congenita, a complex syndrome characterized by bone marrow failure, telomerase enzyme deficiency, and progressive telomere shortening. Here we demonstrate that dyskerin also contributes to telomere maintenance in Arabidopsis thaliana. We report that both AtNAP57, the Arabidopsis dyskerin homolog, and AtTERT, the telomerase catalytic subunit, accumulate in the plant nucleolus, and AtNAP57 associates with active telomerase RNP particles in an RNA-dependent manner. Furthermore, AtNAP57 interacts in vitro with AtPOT1a, a novel component of Arabidopsis telomerase. Although a null mutation in AtNAP57 is lethal, AtNAP57, like AtTERT, is not haploinsufficient for telomere maintenance in Arabidopsis. However, introduction of an AtNAP57 allele containing a T66A mutation decreased telomerase activity in vitro, disrupted telomere length regulation on individual chromosome ends in vivo, and established a new, shorter telomere length set point. These results imply that T66A NAP57 behaves as a dominant-negative inhibitor of telomerase. We conclude that dyskerin is a conserved component of the telomerase RNP complex in higher eukaryotes that is required for maximal enzyme activity in vivo.


Blood ◽  
2007 ◽  
Vol 110 (13) ◽  
pp. 4198-4205 ◽  
Author(s):  
Anna Marrone ◽  
Amanda Walne ◽  
Hannah Tamary ◽  
Yuka Masunari ◽  
Michael Kirwan ◽  
...  

Dyskeratosis congenita (DC) is a multisystem bone marrow failure syndrome characterized by a triad of mucocutaneous abnormalities and an increased predisposition to malignancy. X-linked DC is due to mutations in DKC1, while heterozygous mutations in TERC (telomerase RNA component) and TERT (telomerase reverse transcriptase) have been found in autosomal dominant DC. Many patients with DC remain uncharacterized, particularly families displaying autosomal recessive (AR) inheritance. We have now identified novel homozygous TERT mutations in 2 unrelated consanguineous families, where the index cases presented with classical DC or the more severe variant, Hoyeraal-Hreidarsson (HH) syndrome. These TERT mutations resulted in reduced telomerase activity and extremely short telomeres. As these mutations are homozygous, these patients are predicted to have significantly reduced telomerase activity in vivo. Interestingly, in contrast to patients with heterozygous TERT mutations or hemizygous DKC1 mutations, these 2 homozygous TERT patients were observed to have higher-than-expected TERC levels compared with controls. Collectively, the findings from this study demonstrate that homozygous TERT mutations, resulting in a pure but severe telomerase deficiency, produce a phenotype of classical AR-DC and its severe variant, the HH syndrome.


Blood ◽  
2005 ◽  
Vol 105 (2) ◽  
pp. 682-688 ◽  
Author(s):  
Matt Knudson ◽  
Shashikant Kulkarni ◽  
Zuhair K. Ballas ◽  
Monica Bessler ◽  
Frederick Goldman

Abstract Dyskeratosis congenita (DC) is an inherited bone marrow failure disorder characterized by abnormal skin pigmentation and nail dystrophy. We have recently described, in 10 members of a large 3-generation family, an autosomal-dominant form of DC (AD DC) that is due to a mutation in the gene-encoding human telomerase RNA (TERC), resulting in telomere shortening. In studying the immunologic consequences of TERC mutations, severe B lymphopenia and decreased immunoglobulin M (IgM) levels were noted. T cells were found to overexpress senescent markers, including CD57 and Fas receptor, and were moderately reduced in cell number. To determine whether these in vivo findings were related to cellular replicative defects, short-term cultures of AD DC lymphocytes were established to measure proliferation, mitoses, and apoptosis. AD DC lymphocytes displayed a markedly reduced proliferative capacity and increased basal apoptotic rate. Finally, telomere shortening was most prominent in third-generation subjects, and there appeared to be a correlation between telomere length and in vivo and in vitro immune findings. In summary, the observed lymphopenia and hypogammaglobulinemia in AD DC is likely a consequence of replicative failure and premature senescence of lymphocytes, supporting a role of telomerase activity in immune homeostasis.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 992-992 ◽  
Author(s):  
Joshua A. Regal ◽  
Rodrigo T. Calado ◽  
Aarthi Shenoy ◽  
Peter M. Lansdorp ◽  
Neal S. Young

Abstract Mutations in telomere repair complex genes TERT (encoding telomerase reverse transcriptase) and TERC (telomerase RNA component) are associated with bone marrow failure, especially acquired aplastic anemia and dyskeratosis congenita. Low telomerase activity leads to short telomeres of leukocytes, predisposing highly proliferative tissues such as the bone marrow to early senescence and exhaustion of the stem cell compartment. Telomere repair gene mutations have been suggested to result in disease anticipation, defined as earlier and/or worsening clinical manifestations in successive generations. We have identified a six-generation pedigree in a large Mennonite family carrying a novel TERT mutation (K570N), which localizes in the catalytic domain with reverse transcriptase activity (RT domain). The index patient is a 26 year old male dairy farmer with a ten-year history of severe aplastic anemia (5% bone marrow cellularity) unresponsive to immunosuppression. A long history of hematologic diseases was well known and named in the family--the patient’s paternal great-great-grandmother had died of a severe blood disorder at age 65 years. However, the great-grandmother and the grandfather had never presented any hematological disease. The patient’s father had myelodysplastic syndrome at age 33 years, evolving to acute myeloid leukemia and death due to failure to recover blood counts after chemotherapy. One of the proband’s paternal aunts had aplastic anemia develop when she was a young woman and has been transfusion-independent for decades in response to chronic androgen therapy. A second proband’s paternal aunt underwent a liver transplant at age 20 for submassive hepatic necrosis with fibrosis. A third proband’s paternal aunt has macrocytosis only at age 47. Two sisters (ages 21 and 23) also have macrocytosis in the absence of other hematological abnormality, and two other sisters are healthy. Genetic analysis showed that TERT K570N mutation is present in the patient’s paternal (including grandfather, three aunts and two affected sisters) but not maternal relatives (making his father an obligatory carrier). The patient’s oldest of three sons, now age four years, has the mutation but is asymptomatic. There was no nail dystrophy, leukoplakia or skin hyperpigmentation in any of the TERT K570N carriers; although the index patient and some of his relatives showed early graying of hair, this characteristic did not track with the mutation. Telomere shortening of leukocytes, as measured by Flow-FISH, tracked to the mutation in three generations analyzed, being shortest in the proband and in his aunt with marrow failure. Mutagenized TERT vectors transfected into telomerase-deficient VA13 cell lines yielded no telomerase activity using the telomeric repeat amplification protocol (TRAP) assay, whereas when wild-type TERT vectors were co-transfected, telomerase activity was approximately half of wild-type transfected only, indicating haploinsufficiency as a mechanism of telomere shortening. Our results confirm the association between aplastic anemia and TERT mutations. The pattern of hematologic disease in this kindred does not support disease anticipation in TERT mutations. Most remarkably, there is a likely relationship between a telomerase gene mutation and hematological malignancy and severe liver disease.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1079-1079
Author(s):  
Rodrigo T. Calado ◽  
Joshua A. Regal ◽  
Sachiko Kajigaya ◽  
Neal S Young

Abstract Abstract 1079 Poster Board I-101 Loss-of-function mutations in telomerase complex genes reduce telomerase activity and shorten overall telomere length in leukocytes, and they can clinically manifest as bone marrow failure (aplastic anemia and dyskeratosis congenita), familial pulmonary fibrosis, and hepatic cirrhosis. The double-stranded tandem telomeric TTAGGG repeats are followed by a 3' G-rich single-stranded overhang, a crucial structural component responsible for protective t-loop formation. We investigated the length of telomeric overhangs in 25 healthy individuals from 0 to 76 years, 16 patients with aplastic anemia, and 13 of their immediate relatives using a non-denaturing in-gel method and the telomere-oligonucleotide ligation assay (T-OLA). Among healthy controls, overall telomeric length of leukocytes shortened as a function of age. It was longest in umbilical cord blood samples and eroded as a function of a third-order polynomial until 76 years (R2=0.9517; Spearman=0.90; P<0.0001). However, the lengths of single-stranded overhangs were constant with age, as determined by both non-denaturing and the T-OLA methods. In contrast, most patients with marrow failure carrying a telomerase gene mutation showed marked erosion of telomeric overhangs associated with critically short telomeres (P<0.05). Seven telomerase complex-mutant aplastic patients had overhang lengths below the 5% percentile established for healthy volunteers. In aplastic patients with normal genotypes, normal overall telomere lengths, and who effectively responded to immunosuppressive therapy, telomeric overhangs were maintained and were all within the range established for control individuals. In conclusion, telomeric overhang erosion does not participate in physiological aging but eroded telomeric overhangs and abnormal telomere structure appear in pathologic shortening of telomeres, especially caused by loss-of-function telomerase mutations. Disrupted telomere structure caused by short telomeric overhangs may contribute to the mechanisms of abnormal hematopoietic compartment senescence and chromosomal instability in human bone marrow failure. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2468-2468 ◽  
Author(s):  
Bai-wei Gu ◽  
Jason A. Mills ◽  
Marisa Apicella ◽  
Jian-meng Fan ◽  
Deborah L. French ◽  
...  

Abstract Telomerase is a ribonucleoprotein that adds telomeric repeats onto the chromosome ends, preventing the replication-dependent loss of telomere repeats and cellular senescence in highly proliferative germ-line cells and in stem cells and cancer cells. Dyskeratosis Congenita (DC) is a rare bone marrow failure syndrome, which affects tissues that need constant renewal by stem cell activity. So far 8 genes have been found whose mutation causes DC and they all encode products that play a role in telomere maintenance. About 35% of DC patients show X-linked-recessive inheritance due to mutations in the DKC1gene encoding dyskerin, a protein important in telomere maintenance and ribosomal RNA biogenesis. Mutant dyskerin can destabilize telomerase RNA leading to rapidly shortening telomeres, accelerated stem cell aging and bone marrow failure. However the precise mechanism by which this occurs is not known and some results suggest dyskerin may play a more direct role in telomerase action. So far studies of the cell biology of DC stem cells have been hampered by their scarcity in patients and their short life span and attempts to create mouse models have suffered from differences in both telomere biology and hematopoiesis between mouse and human. In this study, to investigate disease pathogenesis of X-linked DC, we generated induced pluripotent stem cells (iPSC) from patients’ skin fibroblast cell carrying DKC1 mutations Q31E, Δ37 and A353V. The recurrent A353V mutation accounts for about 40% of DKC1 mutations and usually causes a severe clinical phenotype while patients carrying Q31E and Δ37 mutations show a milder phenotype. We found that dyskerin protein expression in all of these dyskerin mutant iPS cells is decreased in agreement with our mouse studies that show mutant proteins are relatively unstable. These iPS cells show dramatically decreased TERC RNA levels and telomerase activity. Telomere length measurement revealed that mutant iPS cells lose the ability to elongate telomeres during the reprogramming processing; telomere erosion was particularly rapid in A353V cells. To further investigate whether dyskerin protein could play direct role in regulating telomerase activity other than stabilization of TERC RNA during the processing of assembly, we tested if the defect in telomerase function in these iPS cells could be rescued by expressing wild type dyskerin or TERC RNA. We expressed the rescuing genes by using the zinc-finger nuclease-mediated method to insert them into the safe harbor AAVs1 site, initially of DKC1Δ37 cells. After testing the telomerase function, we found that expressing WT-dyskerin protein in Δ37 iPS cells fully restores the mature Terc RNA expression level and the telomerase activity to normal levels. However, although Δ37 iPS with over-expressed WT TERC RNA can accumulate normal level of TERC RNA, these cells fail to increase telomerase activity. These results suggest that defective telomerase activity cause by DKC1 mutations can only reversed by expressing WT dyskerin but not by TERC RNA. These data suggest that, as one of three core components of the telomerase complex, dyskerin may play a direct role in telomerase activity. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document