Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 50(21): 12400-12424, 2022 11 28.
Article in English | MEDLINE | ID: mdl-35947650

ABSTRACT

Trimethylguanosine synthase 1 (TGS1) is a highly conserved enzyme that converts the 5'-monomethylguanosine cap of small nuclear RNAs (snRNAs) to a trimethylguanosine cap. Here, we show that loss of TGS1 in Caenorhabditis elegans, Drosophila melanogaster and Danio rerio results in neurological phenotypes similar to those caused by survival motor neuron (SMN) deficiency. Importantly, expression of human TGS1 ameliorates the SMN-dependent neurological phenotypes in both flies and worms, revealing that TGS1 can partly counteract the effects of SMN deficiency. TGS1 loss in HeLa cells leads to the accumulation of immature U2 and U4atac snRNAs with long 3' tails that are often uridylated. snRNAs with defective 3' terminations also accumulate in Drosophila Tgs1 mutants. Consistent with defective snRNA maturation, TGS1 and SMN mutant cells also exhibit partially overlapping transcriptome alterations that include aberrantly spliced and readthrough transcripts. Together, these results identify a neuroprotective function for TGS1 and reinforce the view that defective snRNA maturation affects neuronal viability and function.


Subject(s)
Methyltransferases , Motor Neurons , RNA, Small Nuclear , Animals , Humans , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Drosophila/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , HeLa Cells , Motor Neurons/metabolism , Motor Neurons/pathology , Phenotype , RNA, Small Nuclear/metabolism , Methyltransferases/metabolism
2.
FEBS Lett ; 596(1): 42-52, 2022 01.
Article in English | MEDLINE | ID: mdl-34817067

ABSTRACT

Mutations in many genes that control the expression, the function, or the stability of telomerase cause telomere biology disorders (TBDs), such as dyskeratosis congenita, pulmonary fibrosis, and aplastic anemia. Mutations in a subset of the genes associated with TBDs cause reductions of the telomerase RNA moiety hTR, thus limiting telomerase activity. We have recently found that loss of the trimethylguanosine synthase TGS1 increases both hTR abundance and telomerase activity and leads to telomere elongation. Here, we show that treatment with the S-adenosylmethionine analog sinefungin inhibits TGS1 activity, increases the hTR levels, and promotes telomere lengthening in different cell types. Our results hold promise for restoring telomere length in stem and progenitor cells from TBD patients with reduced hTR levels.


Subject(s)
Methyltransferases
3.
Nature ; 597(7878): 715-719, 2021 09.
Article in English | MEDLINE | ID: mdl-34526722

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer deaths worldwide1. Studies in human tissues and in mouse models have suggested that for many cancers, stem cells sustain early mutations driving tumour development2,3. For the pancreas, however, mechanisms underlying cellular renewal and initiation of PDAC remain unresolved. Here, using lineage tracing from the endogenous telomerase reverse transcriptase (Tert) locus, we identify a rare TERT-positive subpopulation of pancreatic acinar cells dispersed throughout the exocrine compartment. During homeostasis, these TERThigh acinar cells renew the pancreas by forming expanding clones of acinar cells, whereas randomly marked acinar cells do not form these clones. Specific expression of mutant Kras in TERThigh acinar cells accelerates acinar clone formation and causes transdifferentiation to ductal pre-invasive pancreatic intraepithelial neoplasms by upregulating Ras-MAPK signalling and activating the downstream kinase ERK (phospho-ERK). In resected human pancreatic neoplasms, we find that foci of phospho-ERK-positive acinar cells are common and frequently contain activating KRAS mutations, suggesting that these acinar regions represent an early cancer precursor lesion. These data support a model in which rare TERThigh acinar cells may sustain KRAS mutations, driving acinar cell expansion and creating a field of aberrant cells initiating pancreatic tumorigenesis.


Subject(s)
Acinar Cells/cytology , Carcinogenesis , Pancreas/cytology , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Transdifferentiation , Cell Transformation, Neoplastic/genetics , Homeostasis , Humans , MAP Kinase Signaling System , Mice , Mutation , Pancreas/pathology , Pancreas/physiology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Telomerase/genetics
4.
Hum Pathol (N Y) ; 252021 Sep.
Article in English | MEDLINE | ID: mdl-34522616

ABSTRACT

Dyskeratosis congenita is a disease of impaired tissue maintenance downstream of telomere dysfunction. Characteristically, patients present with the clinical triad of nail dystrophy, oral leukoplakia, and skin pigmentation defects, but the disease involves degenerative changes in multiple organs. Mutations in telomere-binding proteins such as TINF2 (TRF1-interacting nuclear factor 2) or in telomerase, the enzyme that counteracts age related telomere shortening, are causative in dyskeratosis congenita. We present a patient who presented with severe hypoxemia at age 13. The patient had a history of myelodysplastic syndrome treated with bone marrow transplant at the age of 5. At age 18 she was hospitalized for an acute pneumonia progressing to respiratory failure, developed renal failure and ultimately, she and her family opted to withdraw support as she was not a candidate for a lung transplant. Sequencing of the patient's TINF2 locus revealed a heterozygous mutation (c.844C > T, Arg282Cys) which has previously been reported in a subset of dyskeratosis congenita patients. Tissue sections from multiple organs showed degenerative changes including disorganized bone remodeling, diffuse alveolar damage and small vessel proliferation in the lung, and hyperkeratosis with hyperpigmentation of the skin. Autopsy samples revealed a bimodal distribution of telomere length, with telomeres from donor hematopoietic tissues being an age-appropriate length and those from patient tissues showing pathogenic shortening, with the shortest telomeres in lung, liver, and kidney. We report for the first time a survey of degenerative changes and telomere lengths in multiple organs in a patient with dyskeratosis congenita.

5.
Nat Rev Mol Cell Biol ; 21(7): 384-397, 2020 07.
Article in English | MEDLINE | ID: mdl-32242127

ABSTRACT

Telomerase is a ribonucleoprotein complex, the catalytic core of which includes the telomerase reverse transcriptase (TERT) and the non-coding human telomerase RNA (hTR), which serves as a template for the addition of telomeric repeats to chromosome ends. Telomerase expression is restricted in humans to certain cell types, and telomerase levels are tightly controlled in normal conditions. Increased levels of telomerase are found in the vast majority of human cancers, and we have recently begun to understand the mechanisms by which cancer cells increase telomerase activity. Conversely, germline mutations in telomerase-relevant genes that decrease telomerase function cause a range of genetic disorders, including dyskeratosis congenita, idiopathic pulmonary fibrosis and bone marrow failure. In this Review, we discuss the transcriptional regulation of human TERT, hTR processing, assembly of the telomerase complex, the cellular localization of telomerase and its recruitment to telomeres, and the regulation of telomerase activity. We also discuss the disease relevance of each of these steps of telomerase biogenesis.


Subject(s)
Gene Expression Regulation , Homeostasis , Mutation , Neoplasms/genetics , Neoplasms/pathology , Telomerase/metabolism , Telomere/physiology , Humans , Neoplasms/metabolism
6.
Cell Rep ; 30(5): 1358-1372.e5, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32023455

ABSTRACT

Biogenesis of the human telomerase RNA (hTR) involves a complex series of posttranscriptional modifications, including hypermethylation of the 5' mono-methylguanosine cap to a tri-methylguanosine cap (TMG). How the TMG cap affects hTR maturation is unknown. Here, we show that depletion of trimethylguanosine synthase 1 (TGS1), the enzyme responsible for cap hypermethylation, increases levels of hTR and telomerase. Diminished trimethylation increases hTR association with the cap-binding complex (CBC) and with Sm chaperone proteins. Loss of TGS1 causes an increase in accumulation of mature hTR in both the nucleus and the cytoplasm compared with controls. In TGS1 mutant cells, increased hTR assembles with telomerase reverse transcriptase (TERT) protein to yield elevated active telomerase complexes and increased telomerase activity, resulting in telomere elongation in cultured human cells. Our results show that TGS1-mediated hypermethylation of the hTR cap inhibits hTR accumulation, restrains levels of assembled telomerase, and limits telomere elongation.


Subject(s)
Methyltransferases/deficiency , RNA/metabolism , Telomerase/metabolism , Telomere/metabolism , Biocatalysis , Coiled Bodies/metabolism , Guanosine/metabolism , HEK293 Cells , HeLa Cells , Humans , Methylation , Methyltransferases/genetics , Models, Biological , Mutation/genetics , Polyadenylation , RNA Caps/metabolism , Subcellular Fractions/metabolism
7.
Nat Methods ; 16(6): 489-492, 2019 06.
Article in English | MEDLINE | ID: mdl-31133759

ABSTRACT

Modular domains of long non-coding RNAs can serve as scaffolds to bring distant regions of the linear genome into spatial proximity. Here, we present HiChIRP, a method leveraging bio-orthogonal chemistry and optimized chromosome conformation capture conditions, which enables interrogation of chromatin architecture focused around a specific RNA of interest down to approximately ten copies per cell. HiChIRP of three nuclear RNAs reveals insights into promoter interactions (7SK), telomere biology (telomerase RNA component) and inflammatory gene regulation (lincRNA-EPS).


Subject(s)
Chromatin/chemistry , Chromatin/genetics , Embryonic Stem Cells/metabolism , Gene Expression Regulation , RNA, Long Noncoding/genetics , RNA/chemistry , Telomerase/chemistry , Animals , Cells, Cultured , Chromosomes , Embryonic Stem Cells/cytology , Genome , Mice , Promoter Regions, Genetic , RNA/genetics , Telomerase/genetics
8.
Mol Cell ; 74(4): 688-700.e3, 2019 05 16.
Article in English | MEDLINE | ID: mdl-30930056

ABSTRACT

Mutations in RNA-processing enzymes are increasingly linked to human disease. Telomerase RNA and related noncoding RNAs require 3' end-processing steps, including oligoadenylation. Germline mutations in poly(A)ribonuclease (PARN) cause accumulation of extended human telomerase RNA (hTR) species and precipitate dyskeratosis congenita and pulmonary fibrosis. Here, we develop nascent RNAend-seq to measure processing rates of RNA precursors. We find that mature hTR derives from extended precursors but that in PARN-mutant cells hTR maturation kinetically stalls and unprocessed precursors are degraded. Loss of poly(A)polymerase PAPD5 in PARN-mutant cells accelerates hTR maturation and restores hTR processing, indicating that oligoadenylation and deadenylation set rates of hTR maturation. The H/ACA domain mediates hTR maturation by precisely defining the 3' end, recruiting poly(A)polymerase activity, and conferring sensitivity to PARN regulation. These data reveal a feedforward circuit in which post-transcriptional oligoadenylation controls RNA maturation kinetics. Similar alterations in RNA processing rates may contribute to mechanisms of RNA-based human disease.


Subject(s)
Dyskeratosis Congenita/genetics , Exoribonucleases/genetics , RNA Nucleotidyltransferases/genetics , RNA/genetics , Telomerase/genetics , Dyskeratosis Congenita/pathology , Germ-Line Mutation/genetics , HeLa Cells , Humans , Kinetics , RNA Processing, Post-Transcriptional/genetics
9.
Cell ; 174(1): 218-230.e13, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29804836

ABSTRACT

Ribonucleoprotein enzymes require dynamic conformations of their RNA constituents for regulated catalysis. Human telomerase employs a non-coding RNA (hTR) with a bipartite arrangement of domains-a template-containing core and a distal three-way junction (CR4/5) that stimulates catalysis through unknown means. Here, we show that telomerase activity unexpectedly depends upon the holoenzyme protein TCAB1, which in turn controls conformation of CR4/5. Cells lacking TCAB1 exhibit a marked reduction in telomerase catalysis without affecting enzyme assembly. Instead, TCAB1 inactivation causes unfolding of CR4/5 helices that are required for catalysis and for association with the telomerase reverse-transcriptase (TERT). CR4/5 mutations derived from patients with telomere biology disorders provoke defects in catalysis and TERT binding similar to TCAB1 inactivation. These findings reveal a conformational "activity switch" in human telomerase RNA controlling catalysis and TERT engagement. The identification of two discrete catalytic states for telomerase suggests an intramolecular means for controlling telomerase in cancers and progenitor cells.


Subject(s)
RNA, Untranslated/chemistry , Telomerase/metabolism , Biocatalysis , Cell Line , HeLa Cells , Humans , Molecular Chaperones , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleic Acid Conformation , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , RNA, Untranslated/metabolism , Telomerase/antagonists & inhibitors , Telomerase/chemistry , Telomerase/genetics , Telomere/metabolism
10.
Cell ; 170(1): 8-9, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28666127

ABSTRACT

Functions of the telomeric repeat-containing RNA (TERRA), the long noncoding RNA (lncRNA) transcribed from telomeres, have eluded researchers. In this issue of Cell, Graf el al. and Chu et al. uncover new regulatory roles for TERRA at the telomere and at distant genomic sites.


Subject(s)
RNA, Untranslated/genetics , Telomere , Genomics , Humans , RNA , RNA, Long Noncoding
11.
Article in English | MEDLINE | ID: mdl-28289249

ABSTRACT

Telomeres, the nucleoprotein complex at the ends of eukaryotic chromosomes, perform an essential cellular role in part by preventing the chromosomal end from initiating a DNA-damage response. This function of telomeres can be compromised as telomeres erode either as a consequence of cell division in culture or as a normal part of cellular ageing in proliferative tissues. Telomere dysfunction in this context leads to DNA-damage signaling and activation of the tumor-suppressor protein p53, which then can prompt either cellular senescence or apoptosis. By culling cells with dysfunctional telomeres, p53 plays a critical role in protecting tissues against the effects of critically short telomeres. However, as telomere dysfunction worsens, p53 likely exacerbates short telomere-driven tissue failure diseases, including pulmonary fibrosis, aplastic anemia, and liver cirrhosis. In cells lacking p53, unchecked telomere shortening drives chromosomal end-to-end fusions and cycles of chromosome fusion-bridge-breakage. Incipient cancer cells confronting these telomere barriers must disable p53 signaling to avoid senescence and eventually up-regulate telomerase to achieve cellular immortality. The recent findings of highly recurrent activating mutations in the promoter for the telomerase reverse transcriptase (TERT) gene in diverse human cancers, together with the widespread mutations in p53 in cancer, provide support for the idea that circumvention of a telomere-p53 checkpoint is essential for malignant progression in human cancer.


Subject(s)
Cellular Senescence/genetics , Neoplasms/genetics , Signal Transduction , Telomerase/physiology , Telomere/genetics , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis , Cell Cycle Checkpoints , DNA Damage/physiology , Humans , Mice , Mutation , Neoplasms/metabolism , Telomerase/metabolism , Telomere/physiology , Telomere Shortening , Tumor Suppressor Protein p53/metabolism , Up-Regulation
13.
Cancer Cell ; 28(3): 277-9, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26373274

ABSTRACT

In this issue of Cancer Cell, Ramamoorthy and Smith report that cancer cells that maintain their chromosome ends through alternative lengthening of telomeres (ALT) display persistent sister telomere cohesion. This delayed resolution of sister telomere cohesion depends upon the loss of ATRX and its histone-sequestering function and is associated with increased recombination between sister telomeres.


Subject(s)
DNA Helicases/genetics , Nuclear Proteins/genetics , Recombination, Genetic/genetics , Telomere/genetics , Humans
14.
Cell Cycle ; 10(16): 2806-15, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21822053

ABSTRACT

Small cell lung carcinoma (SCLC) is a neuroendocrine subtype of lung cancer that affects more than 200,000 people worldwide every year with a very high mortality rate. Here, we used a mouse genetics approach to characterize the cell of origin for SCLC; in this mouse model, tumors are initiated by the deletion of the Rb and p53 tumor suppressor genes in the lung epithelium of adult mice. We found that mouse SCLCs often arise in the lung epithelium, where neuroendocrine cells are located, and that the majority of early lesions were composed of proliferating neuroendocrine cells. In addition, mice in which Rb and p53 are deleted in a variety of non-neuroendocrine lung epithelial cells did not develop SCLC. These data indicate that SCLC likely arises from neuroendocrine cells in the lung.


Subject(s)
Genes, Retinoblastoma/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neuroendocrine Cells/pathology , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Tumor Suppressor Protein p53/genetics , Animals , Epithelium , Mice , Mice, Transgenic , Sequence Deletion
15.
Mol Cell Biol ; 30(7): 1729-45, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20100864

ABSTRACT

In cancer cells, the retinoblastoma tumor suppressor RB is directly inactivated by mutation in the RB gene or functionally inhibited by abnormal activation of cyclin-dependent kinase activity. While variations in RB levels may also provide an important means of controlling RB function in both normal and cancer cells, little is known about the mechanisms regulating RB transcription. Here we show that members of the RB and E2F families bind directly to the RB promoter. To investigate how the RB/E2F pathway may regulate Rb transcription, we generated reporter mice carrying an eGFP transgene inserted into a bacterial artificial chromosome containing most of the Rb gene. Expression of eGFP largely parallels that of Rb in transgenic embryos and adult mice. Using these reporter mice and mutant alleles for Rb, p107, and p130, we found that RB family members modulate Rb transcription in specific cell populations in vivo and in culture. Interestingly, while Rb is a target of the RB/E2F pathway in mouse and human cells, Rb expression does not strictly correlate with the cell cycle status of these cells. These experiments identify novel regulatory feedback mechanisms within the RB pathway in mammalian cells.


Subject(s)
E2F Transcription Factors/metabolism , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p130/metabolism , Transcription, Genetic , Animals , Cell Cycle/physiology , E2F Transcription Factors/genetics , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Genes, Reporter , Humans , Mice , Mice, Transgenic , NIH 3T3 Cells , Promoter Regions, Genetic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p130/genetics , Tissue Distribution , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...