Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
Clin Cancer Res ; 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39078402

ABSTRACT

Children with certain germline gene variants have an increased risk of developing myelodysplastic syndrome (MDS) and other hematopoietic malignancies (HM), such as leukemias and lymphomas. Recent studies have identified an expanding number of these predisposition genes, with variants most prevalent in children with MDS but also found in other HM. For some hematopoietic malignancy predisposition (HMP) disorders, specifically those with a high risk of MDS, early intervention through hematopoietic stem cell transplantation (HSCT) can favorably impact overall survival, providing a rationale for rigorous surveillance. A multidisciplinary panel of experts at the 2023 AACR Childhood Cancer Predisposition Workshop reviewed the latest advances in the field and updated prior 2017 surveillance recommendations for children with HMP. In addition to general guidance for all children with HMP, which includes annual physical examination, education about the signs and symptoms of HM, consultation with experienced providers, and early assessment by an HSCT specialist, the panel provided specific recommendations for individuals with a higher risk of MDS based on the affected gene. These recommendations include periodic and comprehensive surveillance for individuals with those syndromes associated with higher risk of MDS, including serial bone marrow examinations to monitor for morphologic changes and deep sequencing for somatic changes in genes associated with HM progression. This approach enables close monitoring of disease evolution based on the individual's genetic profile. As more HMP-related genes are discovered and the disorders' natural histories are better defined, these personalized recommendations will serve as a foundation for future guidelines in managing these conditions.

2.
Hepatol Commun ; 8(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38896081

ABSTRACT

BACKGROUND: Patients with telomere biology disorders (TBD) develop hepatic disease, including hepatitis, cirrhosis, and hepatopulmonary syndrome. No specific treatment exists for TBD-related liver disease, and the role of liver transplantation (LT) remains controversial. Our study objectives were to describe the clinical characteristics, management, and outcomes in patients with TBD-related liver disease, and their LT outcomes. METHODS: Data from 83 patients with TBD-associated liver disease were obtained from 17 participating centers in the Clinical Care Consortium of Telomere-Associated Ailments and by self-report for our retrospective, multicenter, international cohort study. RESULTS: Group A ("Advanced") included 40 patients with advanced liver disease. Of these, 20 underwent LT (Group AT). Group M ("Mild") included 43 patients not warranting LT evaluation, none of whom were felt to be medically unfit for liver transplantation. Supplemental oxygen requirement, pulmonary arteriovenous malformation, hepatopulmonary syndrome, and higher bilirubin and international normalized ratio values were associated with Group A. Other demographics, clinical manifestations, and laboratory findings were similar between groups. Six group A patients were declined for LT; 3 died on the waitlist. Median follow-up post-LT was 2.9 years (range 0.6-13.2 y). One-year survival post-LT was 73%. Median survival post-LT has not been reached. Group AT patients had improved survival by age compared to all nontransplant patients (log-rank test p = 0.02). Of 14 patients with pretransplant hypoxemia, 8 (57%) had improved oxygenation after transplant. CONCLUSIONS: LT recipients with TBD do not exhibit excessive posttransplant mortality, and LT improved respiratory status in 57%. A TBD diagnosis should not exclude LT consideration.


Subject(s)
Liver Transplantation , Humans , Female , Male , Retrospective Studies , Adult , Middle Aged , Telomere , Adolescent , Liver Diseases/surgery , Liver Diseases/genetics , Young Adult , Child , Treatment Outcome , Child, Preschool
3.
J Biol Chem ; 299(5): 104665, 2023 05.
Article in English | MEDLINE | ID: mdl-37003504

ABSTRACT

Telomere length maintenance is crucial to cancer cell immortality. Up to 15% of cancers utilize a telomerase-independent, recombination-based mechanism termed alternative lengthening of telomeres (ALT). Currently, the primary ALT biomarker is the C-circle, a type of circular DNA with extrachromosomal telomere repeats (cECTRs). How C-circles form is not well characterized. We investigated C-circle formation in the human cen3tel cell line, a long-telomere, telomerase+ (LTT+) cell line with progressively hyper-elongated telomeres (up to ∼100 kb). cECTR signal was observed in 2D gels and C-circle assays but not t-circle assays, which also detect circular DNA with extrachromosomal telomere repeats. Telomerase activity and C-circle signal were not separable in the analysis of clonal populations, consistent with C-circle production occurring within telomerase+ cells. We observed similar cECTR results in two other LTT+ cell lines, HeLa1.3 (∼23 kb telomeres) and HeLaE1 (∼50 kb telomeres). In LTT+ cells, telomerase activity did not directly impact C-circle signal; instead, C-circle signal correlated with telomere length. LTT+ cell lines were less sensitive to hydroxyurea than ALT+ cell lines, suggesting that ALT status is a stronger contributor to replication stress levels than telomere length. Additionally, the DNA repair-associated protein FANCM did not suppress C-circles in LTT+ cells as it does in ALT+ cells. Thus, C-circle formation may be driven by telomere length, independently of telomerase and replication stress, highlighting limitations of C-circles as a stand-alone ALT biomarker.


Subject(s)
DNA, Circular , Telomerase , Telomere , Humans , DNA Helicases/metabolism , Telomerase/metabolism , Telomere/genetics , Telomere/metabolism , Telomere Homeostasis , Cell Line , HeLa Cells , DNA Replication , Hydroxyurea , DNA Repair
4.
Am J Med Genet A ; 191(7): 1826-1835, 2023 07.
Article in English | MEDLINE | ID: mdl-37067177

ABSTRACT

The MECOM gene encodes multiple protein isoforms that are essential for hematopoietic stem cell self-renewal and maintenance. Germline MECOM variants have been associated with congenital thrombocytopenia, radioulnar synostosis and bone marrow failure; however, the phenotypic spectrum of MECOM-associated syndromes continues to expand and novel pathogenic variants continue to be identified. We describe eight unrelated patients who add to the previously known phenotypes and genetic defects of MECOM-associated syndromes. As each subject presented with unique MECOM variants, the series failed to demonstrate clear genotype-to-phenotype correlation but may suggest a role for additional modifiers that affect gene expression and subsequent phenotype. Recognition of the expanded hematologic and non-hematologic clinical features allows for rapid molecular diagnosis, early identification of life-threatening complications, and improved genetic counseling for families. A centralized international publicly accessible database to share annotated MECOM variants would advance their clinical interpretation and provide a foundation to perform functional MECOM studies.


Subject(s)
Bone Marrow Diseases , Hematologic Diseases , Pancytopenia , Humans , Bone Marrow Diseases/diagnosis , Bone Marrow Diseases/genetics , Syndrome , Bone Marrow Failure Disorders , Transcription Factors/genetics , Phenotype , MDS1 and EVI1 Complex Locus Protein/genetics
5.
bioRxiv ; 2023 Jan 27.
Article in English | MEDLINE | ID: mdl-36747763

ABSTRACT

Telomere length maintenance is crucial to cancer cell immortality. Up to 15% of cancers utilize a telomerase-independent, recombination-based mechanism termed alternative lengthening of telomeres (ALT). The primary ALT biomarker is the C-circle, a type of circular DNA with extrachromosomal telomere repeats (cECTRs). How C-circles form is not well characterized. To investigate C-circle formation in telomerase+ cells, we studied the human cen3tel cell line, in which telomeres progressively hyper-elongated post TERT -immortalization. cECTR signal was observed in 2D gels and C-circle assays but not t-circle assays, which also detect cECTRs. Telomerase activity and C-circle signal were not separable in the analysis of clonal populations, consistent with C-circle production occurring within telomerase+ cells. Two other long telomere, telomerase+ (LTT+) cell lines, HeLa1.3 (~23 kb telomeres) and HeLaE1 (~50 kb telomeres), had similar cECTR properties. Telomerase activity did not directly impact C-circle signal in LTT+ cells; instead, C-circle signal correlated with telomere length. LTT+ lines were less sensitive to hydroxyurea than an ALT+ cell line, suggesting that ALT status is a stronger contributor to replication stress levels than telomere length. Additionally, FANCM did not suppress C-circles in LTT+ cells as it does in ALT+ cells. Thus, C-circle formation may be driven by telomere length, independently of telomerase and replication stress, highlighting limitations of C-circles as a stand-alone ALT biomarker.

6.
Nat Rev Genet ; 24(2): 86-108, 2023 02.
Article in English | MEDLINE | ID: mdl-36151328

ABSTRACT

Telomeres are specialized nucleoprotein structures at the ends of linear chromosomes that prevent the activation of DNA damage response and repair pathways. Numerous factors localize at telomeres to regulate their length, structure and function, to avert replicative senescence or genome instability and cell death. In humans, Mendelian defects in several of these factors can result in abnormally short or dysfunctional telomeres, causing a group of rare heterogeneous premature-ageing diseases, termed telomeropathies, short-telomere syndromes or telomere biology disorders (TBDs). Here, we review the TBD-causing genes identified so far and describe their main functions associated with telomere biology. We present molecular aspects of TBDs, including genetic anticipation, phenocopy, incomplete penetrance and somatic genetic rescue, which underlie the complexity of these diseases. We also discuss the implications of phenotypic and genetic features of TBDs on fundamental aspects related to human telomere biology, ageing and cancer, as well as on diagnostic, therapeutic and clinical approaches.


Subject(s)
Telomerase , Telomere , Humans , Telomere/genetics , Telomere/metabolism , Aging/genetics , Telomere Homeostasis , Genomic Instability , Biology , Telomerase/genetics
7.
JCI Insight ; 7(22)2022 11 22.
Article in English | MEDLINE | ID: mdl-36219480

ABSTRACT

Acquired aplastic anemia (AA) is caused by autoreactive T cell-mediated destruction of early hematopoietic cells. Somatic loss of human leukocyte antigen (HLA) class I alleles was identified as a mechanism of immune escape in surviving hematopoietic cells of some patients with AA. However, pathogenicity, structural characteristics, and clinical impact of specific HLA alleles in AA remain poorly understood. Here, we evaluated somatic HLA loss in 505 patients with AA from 2 multi-institutional cohorts. Using a combination of HLA mutation frequencies, peptide-binding structures, and association with AA in an independent cohort of 6,323 patients from the National Marrow Donor Program, we identified 19 AA risk alleles and 12 non-risk alleles and established a potentially novel AA HLA pathogenicity stratification. Our results define pathogenicity for the majority of common HLA-A/B alleles across diverse populations. Our study demonstrates that HLA alleles confer different risks of developing AA, but once AA develops, specific alleles are not associated with response to immunosuppression or transplant outcomes. However, higher pathogenicity alleles, particularly HLA-B*14:02, are associated with higher rates of clonal evolution in adult patients with AA. Our study provides insights into the immune pathogenesis of AA, opening the door to future autoantigen identification and improved understanding of clonal evolution in AA.


Subject(s)
Anemia, Aplastic , Adult , Humans , Anemia, Aplastic/genetics , Anemia, Aplastic/pathology , Alleles , Histocompatibility Antigens Class I/genetics , HLA-B Antigens/genetics , HLA Antigens/genetics
8.
Am J Med Genet A ; 188(7): 2204-2208, 2022 07.
Article in English | MEDLINE | ID: mdl-35362179

ABSTRACT

The tumor suppressor p53 has well known roles in cancer development and germline cancer predisposition disorders, but increasing evidence supports the role of activation of this transcription factor in the pathogenesis of inherited bone marrow failure and chromosomal instability disorders. Here we report a patient with red cell aplasia, which was steroid responsive, as well as intellectual disability, seizures, microcephaly, short stature, cellular radiosensitivity, and normal telomere lengths, who had a germline heterozygous C-terminal frameshift variant in TP53 similar to others that activate the transcription factor. This is the third reported individual with a germline p53 activation syndrome, with several unique features that refine the clinical disease associated with these variants.


Subject(s)
Intellectual Disability , Tumor Suppressor Protein p53 , Genetic Predisposition to Disease , Germ Cells , Germ-Line Mutation/genetics , Humans , Intellectual Disability/genetics , Phenotype , Syndrome , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics
9.
Blood ; 140(6): 608-618, 2022 08 11.
Article in English | MEDLINE | ID: mdl-35421215

ABSTRACT

Mutations in the TINF2 gene, encoding the shelterin protein TIN2, cause telomere shortening and the inherited bone marrow (BM) failure syndrome dyskeratosis congenita (DC). A lack of suitable model systems limits the mechanistic understanding of telomere shortening in the stem cells and thus hinders the development of treatment options for BM failure. Here, we endogenously introduced TIN2-DC mutations in human embryonic stem cells (hESCs) and human hematopoietic stem and progenitor cells (HSPCs) to dissect the disease mechanism and identify a gene-editing strategy that rescued the disease phenotypes. The hESCs with the T284R disease mutation exhibited the short telomere phenotype observed in DC patients. Yet, telomeres in mutant hESCs did not trigger DNA damage responses at telomeres or show exacerbated telomere shortening when differentiated into telomerase-negative cells. Disruption of the mutant TINF2 allele by introducing a frameshift mutation in exon 2 restored telomere length in stem cells and the replicative potential of differentiated cells. Similarly, we introduced TIN2-DC disease variants in human HSPCs to assess the changes in telomere length and proliferative capacity. Lastly, we showed that editing at exon 2 of TINF2 that restored telomere length in hESCs could be generated in TINF2-DC patient HSPCs. Our study demonstrates a simple genetic intervention that rescues the TIN2-DC disease phenotype in stem cells and provides a versatile platform to assess the efficacy of potential therapeutic approaches in vivo.


Subject(s)
Dyskeratosis Congenita , Telomerase , Dyskeratosis Congenita/genetics , Dyskeratosis Congenita/therapy , Humans , Mutation , Telomerase/genetics , Telomerase/metabolism , Telomere/genetics , Telomere/metabolism , Telomere Shortening/genetics , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism
10.
Blood Adv ; 6(1): 297-306, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34758064

ABSTRACT

Shwachman-Diamond syndrome (SDS) is an inherited bone marrow failure syndrome with leukemia predisposition. An understanding of the hematologic complications of SDS with age could guide clinical management, but data are limited for this rare disease. We conducted a cohort study of 153 subjects from 143 families with confirmed biallelic SBDS mutations enrolled on the North American Shwachman Diamond Registry or Bone Marrow Failure Registry. The SBDS c.258 + 2T>C variant was present in all but 1 patient. To evaluate the association between blood counts and age, 2146 blood counts were analyzed for 119 subjects. Absolute neutrophil counts were positively associated with age (P < .0001). Hemoglobin was also positively associated with age up to 18 years (P < .0001), but the association was negative thereafter (P = .0079). Platelet counts and marrow cellularity were negatively associated with age (P < .0001). Marrow cellularity did not correlate with blood counts. Severe marrow failure necessitating transplant developed in 8 subjects at a median age of 1.7 years (range, 0.4-39.5), with 7 of 8 requiring transplant prior to age 8 years. Twenty-six subjects (17%) developed a myeloid malignancy (16 myelodysplasia and 10 acute myeloid leukemia) at a median age of 12.3 years (range, 0.5-45.0) and 28.4 years (range, 14.4-47.3), respectively. A lymphoid malignancy developed in 1 patient at the age of 16.9 years. Hematologic complications were the major cause of mortality (17/20 deaths; 85%). These data inform surveillance of hematologic complications in SDS.


Subject(s)
Bone Marrow Diseases , Exocrine Pancreatic Insufficiency , Hematologic Diseases , Adolescent , Adult , Bone Marrow Diseases/complications , Bone Marrow Diseases/genetics , Bone Marrow Diseases/pathology , Child , Child, Preschool , Cohort Studies , Exocrine Pancreatic Insufficiency/complications , Exocrine Pancreatic Insufficiency/genetics , Hematologic Diseases/complications , Humans , Infant , Middle Aged , Shwachman-Diamond Syndrome , Young Adult
11.
Nat Commun ; 12(1): 6850, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824242

ABSTRACT

The molecular mechanisms that drive hematopoietic stem cell functional decline under conditions of telomere shortening are not completely understood. In light of recent advances in single-cell technologies, we sought to redefine the transcriptional and epigenetic landscape of mouse and human hematopoietic stem cells under telomere attrition, as induced by pathogenic germline variants in telomerase complex genes. Here, we show that telomere attrition maintains hematopoietic stem cells under persistent metabolic activation and differentiation towards the megakaryocytic lineage through the cell-intrinsic upregulation of the innate immune signaling response, which directly compromises hematopoietic stem cells' self-renewal capabilities and eventually leads to their exhaustion. Mechanistically, we demonstrate that targeting members of the Ifi20x/IFI16 family of cytosolic DNA sensors using the oligodeoxynucleotide A151, which comprises four repeats of the TTAGGG motif of the telomeric DNA, overcomes interferon signaling activation in telomere-dysfunctional hematopoietic stem cells and these cells' skewed differentiation towards the megakaryocytic lineage. This study challenges the historical hypothesis that telomere attrition limits the proliferative potential of hematopoietic stem cells by inducing apoptosis, autophagy, or senescence, and suggests that targeting IFI16 signaling axis might prevent hematopoietic stem cell functional decline in conditions affecting telomere maintenance.


Subject(s)
Hematopoiesis/physiology , Telomere Shortening/physiology , Animals , Bone Marrow Failure Disorders/genetics , Bone Marrow Failure Disorders/metabolism , Bone Marrow Failure Disorders/pathology , Cell Self Renewal , Cellular Reprogramming , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Interferons/metabolism , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Nuclear Proteins/metabolism , Oligodeoxyribonucleotides/metabolism , Phosphoproteins/metabolism , Signal Transduction , Single-Cell Analysis , Telomere/chemistry , Telomere/physiology , Telomere Shortening/genetics
13.
Am J Hematol ; 96(11): 1491-1504, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34342889

ABSTRACT

The North American Pediatric Aplastic Anemia Consortium (NAPAAC) is a group of pediatric hematologist-oncologists, hematopathologists, and bone marrow transplant physicians from 46 institutions in North America with interest and expertise in aplastic anemia, inherited bone marrow failure syndromes, and myelodysplastic syndromes. The NAPAAC Bone Marrow Failure Diagnosis and Care Guidelines Working Group was established with the charge of harmonizing the approach to the diagnostic workup of aplastic anemia in an effort to standardize best practices in the field. This document outlines the rationale for initial evaluations in pediatric patients presenting with signs and symptoms concerning for severe aplastic anemia.


Subject(s)
Anemia, Aplastic/diagnosis , Anemia, Aplastic/pathology , Bone Marrow/pathology , Child , Diagnosis, Differential , Fetal Hemoglobin/analysis , HLA Antigens/analysis , Humans , North America , Severity of Illness Index
14.
Am J Med Genet A ; 185(10): 3118-3121, 2021 10.
Article in English | MEDLINE | ID: mdl-34159722

ABSTRACT

Congenital disorders of glycosylation are a group of rare monogenic inborn errors of metabolism caused by defective glycoprotein and glycolipid glycan synthesis and attachment. Here, we present a patient with galactose epimerase deficiency, also known as GALE deficiency, accompanied by pancytopenia and immune dysregulation. She was first identified by an abnormal newborn screen for galactosemia with subsequent genetic evaluation due to pancytopenia and immune dysregulation. The evaluation ultimately revealed that her known diagnosis of GALE deficiency was the cause of her hematologic and immune abnormalities. These findings further expand the clinical spectrum of disease of congenital disorders of glycosylation.


Subject(s)
Congenital Disorders of Glycosylation/genetics , Galactosemias/genetics , UDPglucose 4-Epimerase/genetics , Adult , Congenital Disorders of Glycosylation/diagnosis , Congenital Disorders of Glycosylation/pathology , Female , Galactosemias/diagnosis , Galactosemias/pathology , Glycolipids/biosynthesis , Glycolipids/genetics , Humans , Mutation/genetics , Phenotype , Polysaccharides/biosynthesis , Polysaccharides/genetics , UDPglucose 4-Epimerase/deficiency
15.
Nat Commun ; 12(1): 1334, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33637765

ABSTRACT

To understand the mechanisms that mediate germline genetic leukemia predisposition, we studied the inherited ribosomopathy Shwachman-Diamond syndrome (SDS), a bone marrow failure disorder with high risk of myeloid malignancies at an early age. To define the mechanistic basis of clonal hematopoiesis in SDS, we investigate somatic mutations acquired by patients with SDS followed longitudinally. Here we report that multiple independent somatic hematopoietic clones arise early in life, most commonly harboring heterozygous mutations in EIF6 or TP53. We show that germline SBDS deficiency establishes a fitness constraint that drives selection of somatic clones via two distinct mechanisms with different clinical consequences. EIF6 inactivation mediates a compensatory pathway with limited leukemic potential by ameliorating the underlying SDS ribosome defect and enhancing clone fitness. TP53 mutations define a maladaptive pathway with enhanced leukemic potential by inactivating tumor suppressor checkpoints without correcting the ribosome defect. Subsequent development of leukemia was associated with acquisition of biallelic TP53 alterations. These results mechanistically link leukemia predisposition to germline genetic constraints on cellular fitness, and provide a rational framework for clinical surveillance strategies.


Subject(s)
Clonal Hematopoiesis/genetics , Clonal Hematopoiesis/physiology , Shwachman-Diamond Syndrome/genetics , Shwachman-Diamond Syndrome/metabolism , Adolescent , Adult , Bone Marrow Diseases/genetics , Bone Marrow Diseases/metabolism , Child , Child, Preschool , Eukaryotic Initiation Factors/genetics , Female , Humans , Infant , Male , Middle Aged , Mutation , Ribosomes/genetics , Tumor Suppressor Protein p53/genetics , Young Adult
16.
Article in English | MEDLINE | ID: mdl-33446513

ABSTRACT

Telomere biology disorders, largely characterized by telomere lengths below the first centile for age, are caused by variants in genes associated with telomere replication, structure, or function. One of these genes, ACD, which encodes the shelterin protein TPP1, is associated with both autosomal dominantly and autosomal recessively inherited telomere biology disorders. TPP1 recruits telomerase to telomeres and stimulates telomerase processivity. Several studies probing the effect of various synthetic or patient-derived variants have mapped specific residues and regions of TPP1 that are important for interaction with TERT, the catalytic component of telomerase. However, these studies have come to differing conclusions regarding ACD haploinsufficiency. Here, we report a proband with compound heterozygous novel variants in ACD (NM_001082486.1)-c.505_507delGAG, p.(Glu169del); and c.619delG, p.(Asp207Thrfs*22)-and a second proband with a heterozygous chromosomal deletion encompassing ACD: arr[hg19] 16q22.1(67,628,846-67,813,408)x1. Clinical data, including symptoms and telomere length within the pedigrees, suggested that loss of one ACD allele was insufficient to induce telomere shortening or confer clinical features. Further analyses of lymphoblastoid cell lines showed decreased nascent ACD RNA and steady-state mRNA, but normal TPP1 protein levels, in cells containing heterozygous ACD c.619delG, p.(Asp207Thrfs*22), or the ACD-encompassing chromosomal deletion compared to controls. Based on our results, we conclude that cells are able to compensate for loss of one ACD allele by activating a mechanism to maintain TPP1 protein levels, thus maintaining normal telomere length.


Subject(s)
Germ Cells/metabolism , Serine Proteases/genetics , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/isolation & purification , Telomere/metabolism , B-Lymphocytes , Cell Line , Child, Preschool , Female , Gene Deletion , Humans , Leukoplakia, Oral/genetics , Microcephaly/genetics , Nails , Pedigree , Receptor, EphB2 , Sequence Analysis, DNA , Shelterin Complex , Skin Pigmentation , Telomerase/genetics , Telomerase/metabolism
18.
J Pediatr ; 230: 55-61.e4, 2021 03.
Article in English | MEDLINE | ID: mdl-32971146

ABSTRACT

OBJECTIVE: To describe the clinical features, therapeutic interventions, and patient outcomes of gastrointestinal (GI) hemorrhage in individuals with a telomere biology disorder, including dyskeratosis congenita, Hoyeraal-Hreidarsson syndrome, Revesz syndrome, and Coats plus. STUDY DESIGN: Clinical Care Consortium for Telomere Associated Ailments members were invited to contribute data on individuals with telomere biology disorders at their institutions who experienced GI bleeding. Patient demographic, laboratory, imaging, procedural, and treatment information and outcomes were extracted from the medical record. RESULTS: Sixteen patients who experienced GI hemorrhage were identified at 11 centers. Among 14 patients who underwent genetic testing, 8 had mutations in TINF2, 4 had mutations in CTC1 or STN1, and 1 patient each had a mutation in TERC and RTEL1. Ten patients had a history of hematopoietic cell transplantation. The patients with Coats plus and those without Coats plus had similar clinical features and courses. Angiodysplasia of the stomach and/or small bowel was described in 8 of the 12 patients who underwent endoscopy; only 4 had esophageal varices. Various medical interventions were trialed. No single intervention was uniformly associated with cessation of bleeding, although 1 patient had a sustained response to treatment with bevacizumab. Recurrence was common, and the overall long-term outcome for affected patients was poor. CONCLUSIONS: GI bleeding in patients with telomere biology disorders is associated with significant morbidity and with vascular ectasias rather than varices.


Subject(s)
Gastrointestinal Hemorrhage/etiology , Telomere/genetics , Adolescent , Adult , Ataxia/complications , Ataxia/genetics , Bone Diseases, Metabolic/complications , Bone Diseases, Metabolic/genetics , Bone Marrow/abnormalities , Brain Neoplasms/complications , Brain Neoplasms/genetics , Calcinosis/complications , Calcinosis/genetics , Central Nervous System Cysts/complications , Central Nervous System Cysts/genetics , Child , Child, Preschool , Dyskeratosis Congenita/complications , Dyskeratosis Congenita/genetics , Female , Fetal Growth Retardation/genetics , Gastrointestinal Hemorrhage/genetics , Humans , Intellectual Disability/complications , Intellectual Disability/genetics , Leukoencephalopathies/complications , Leukoencephalopathies/genetics , Male , Microcephaly/complications , Microcephaly/genetics , Muscle Spasticity/complications , Muscle Spasticity/genetics , Mutation , Retina , Retinal Diseases/complications , Retinal Diseases/genetics , Seizures/complications , Seizures/genetics , Telomere/metabolism , Telomere/pathology , Young Adult
19.
Am J Med Genet A ; 182(11): 2781-2787, 2020 11.
Article in English | MEDLINE | ID: mdl-32909658

ABSTRACT

Riboflavin transporter deficiency (RTD) (MIM #614707) is a neurogenetic disorder with its most common manifestations including sensorineural hearing loss, peripheral neuropathy, respiratory insufficiency, and bulbar palsy. Here, we present a 2-year-old boy whose initial presentation was severe macrocytic anemia necessitating multiple blood transfusions and intermittent neutropenia; he subsequently developed ataxia and dysarthria. Trio-exome sequencing detected compound heterozygous variants in SLC52A2 that were classified as pathogenic and a variant of uncertain significance. Bone marrow evaluation demonstrated megaloblastic changes. Notably, his anemia and neutropenia resolved after treatment with oral riboflavin, thus expanding the clinical phenotype of this disorder. We reiterate the importance of starting riboflavin supplementation in a young child who presents with macrocytic anemia and neurological features while awaiting biochemical and genetic work up. We detected multiple biochemical abnormalities with the help of untargeted metabolomics analysis associated with abnormal flavin adenine nucleotide function which normalized after treatment, emphasizing the reversible pathomechanisms involved in this disorder. The utility of untargeted metabolomics analysis to monitor the effects of riboflavin supplementation in RTD has not been previously reported.


Subject(s)
Anemia, Macrocytic/pathology , Bulbar Palsy, Progressive/pathology , Hearing Loss, Sensorineural/pathology , Metabolome , Riboflavin Deficiency/pathology , Riboflavin/metabolism , Adult , Anemia, Macrocytic/genetics , Anemia, Macrocytic/metabolism , Bulbar Palsy, Progressive/genetics , Bulbar Palsy, Progressive/metabolism , Female , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/metabolism , Humans , Infant , Male , Mutation , Receptors, G-Protein-Coupled/genetics , Riboflavin Deficiency/genetics , Riboflavin Deficiency/metabolism
20.
Nat Commun ; 11(1): 4766, 2020 09 21.
Article in English | MEDLINE | ID: mdl-32958778

ABSTRACT

Germline telomere maintenance defects are associated with an increased incidence of inflammatory diseases in humans, yet whether and how telomere dysfunction causes inflammation are not known. Here, we show that telomere dysfunction drives pATM/c-ABL-mediated activation of the YAP1 transcription factor, up-regulating the major pro-inflammatory factor, pro-IL-18. The colonic microbiome stimulates cytosolic receptors activating caspase-1 which cleaves pro-IL-18 into mature IL-18, leading to recruitment of interferon (IFN)-γ-secreting T cells and intestinal inflammation. Correspondingly, patients with germline telomere maintenance defects exhibit DNA damage (γH2AX) signaling together with elevated YAP1 and IL-18 expression. In mice with telomere dysfunction, telomerase reactivation in the intestinal epithelium or pharmacological inhibition of ATM, YAP1, or caspase-1 as well as antibiotic treatment, dramatically reduces IL-18 and intestinal inflammation. Thus, telomere dysfunction-induced activation of the ATM-YAP1-pro-IL-18 pathway in epithelium is a key instigator of tissue inflammation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Inflammation/pathology , Telomere/pathology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Anti-Bacterial Agents/therapeutic use , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/metabolism , Caspase 1/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Child , Colon/metabolism , Colon/microbiology , Colon/pathology , Gastrointestinal Diseases/pathology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/microbiology , Interleukin-18/genetics , Interleukin-18/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Mutant Strains , Phosphorylation , Protein Precursors/genetics , Protein Precursors/metabolism , Signal Transduction , Telomerase/genetics , Telomerase/metabolism , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL