Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Formos Med Assoc ; 2024 Jun 23.
Article in English | MEDLINE | ID: mdl-38914514

ABSTRACT

BACKGROUND/PURPOSE: Insufficient numbers of peripheral blood stem cells (PBSC) after granulocyte colony-stimulating factor (G-CSF) mobilization occurs in a significant proportion of PBSC collections, often from older age donors. Telomere length (TL) is often used as an indicator of an individual's biological age. This study aimed to investigate the relationship between donors' leukocyte TL and the outcome of G-CSF-induced PBSC mobilization in healthy unrelated donors. METHODS: Donors' leukocyte TLs and the outcome of G-CSF-induced PBSC mobilization, as assessed by pre-harvest CD34+ cell counts, were analyzed in 39 healthy PBSC donors. TL in a non-mobilized general population (n = 90) was included as a control group. G-CSF mobilization effect was categorized into three groups according to pre-harvest CD34+ cell count: poor (≤25/µL, PMD), intermediate (between 25 and 180/µL), and good (≥180/µl, GMD). RESULTS: Leukocyte TL of PBSC donors correlated well with pre-harvest CD34+ cell counts (r = 0.645, p < 0.001). Leukocyte TLs of PMDs (n = 8) were significantly shorter than those of GMDs (n = 9) and non-mobilization controls (p < 0.05). Moreover, all PMD TLs were below the 50th percentile, and 62.5% of PMDs had TLs below the 10th percentile of age-matched control participants. In contrast, no GMD TLs were below the 10th percentile; in fact, 33.3% (3/9) of them were above the 90th percentile. CONCLUSION: Our results indicate that shorter donor leukocyte TL is associated with poor G-CSF-induced PBSC mobilization. TL, which represents a donor's biological age, could be a potential predictor for mobilization outcome.

2.
Nucleic Acids Res ; 52(11): 6472-6489, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38752489

ABSTRACT

Orphan nuclear receptors (NRs), such as COUP-TF1, COUP-TF2, EAR2, TR2 and TR4, are implicated in telomerase-negative cancers that maintain their telomeres through the alternative lengthening of telomeres (ALT) mechanism. However, how telomere association of orphan NRs is involved in ALT activation remains unclear. Here, we demonstrate that telomeric tethering of orphan NRs in human fibroblasts initiates formation of ALT-associated PML bodies (APBs) and features of ALT activity, including ALT telomere DNA synthesis, telomere sister chromatid exchange, and telomeric C-circle generation, suggesting de novo ALT induction. Overexpression of orphan NRs exacerbates ALT phenotypes in ALT cells, while their depletion limits ALT. Orphan NRs initiate ALT via the zinc finger protein 827, suggesting the involvement of chromatin structure alterations for ALT activation. Furthermore, we found that orphan NRs and deficiency of the ALT suppressor ATRX-DAXX complex operate in concert to promote ALT activation. Moreover, PML depletion by gene knockout or arsenic trioxide treatment inhibited ALT induction in fibroblasts and ALT cancer cells, suggesting that APB formation underlies the orphan NR-induced ALT activation. Importantly, arsenic trioxide administration abolished APB formation and features of ALT activity in ALT cancer cell line-derived mouse xenografts, suggesting its potential for further therapeutic development to treat ALT cancers.


Subject(s)
Fibroblasts , Promyelocytic Leukemia Protein , Telomere Homeostasis , Humans , Animals , Promyelocytic Leukemia Protein/metabolism , Promyelocytic Leukemia Protein/genetics , Mice , Fibroblasts/metabolism , Telomere/metabolism , Telomere/genetics , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Sister Chromatid Exchange , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Arsenic Trioxide/pharmacology , Molecular Chaperones
3.
Elife ; 122024 May 16.
Article in English | MEDLINE | ID: mdl-38752723

ABSTRACT

A causal relationship exists among the aging process, organ decay and disfunction, and the occurrence of various diseases including cancer. A genetically engineered mouse model, termed Klf1K74R/K74R or Klf1(K74R), carrying mutation on the well-conserved sumoylation site of the hematopoietic transcription factor KLF1/EKLF has been generated that possesses extended lifespan and healthy characteristics, including cancer resistance. We show that the healthy longevity characteristics of the Klf1(K74R) mice, as exemplified by their higher anti-cancer capability, are likely gender-, age-, and genetic background-independent. Significantly, the anti-cancer capability, in particular that against melanoma as well as hepatocellular carcinoma, and lifespan-extending property of Klf1(K74R) mice, could be transferred to wild-type mice via transplantation of their bone marrow mononuclear cells at a young age of the latter. Furthermore, NK(K74R) cells carry higher in vitro cancer cell-killing ability than wild-type NK cells. Targeted/global gene expression profiling analysis has identified changes in the expression of specific proteins, including the immune checkpoint factors PDCD and CD274, and cellular pathways in the leukocytes of the Klf1(K74R) that are in the directions of anti-cancer and/or anti-aging. This study demonstrates the feasibility of developing a transferable hematopoietic/blood system for long-term anti-cancer and, potentially, for anti-aging.


Subject(s)
Kruppel-Like Transcription Factors , Longevity , Animals , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Longevity/genetics , Killer Cells, Natural/immunology , Neoplasms/genetics , Genetic Engineering , Bone Marrow Transplantation , Female , Gene Expression Profiling , Male , Mice, Transgenic
4.
Bioorg Med Chem ; 95: 117502, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37866089

ABSTRACT

A structure-activity relationship (SAR) study of stimulator of interferon gene (STING) inhibition was performed using a series of indol-3-yl-N-phenylcarbamic amides and indol-2-yl-N-phenylcarbamic amides. Among these analogs, compounds 10, 13, 15, 19, and 21 inhibited the phosphorylation of STING and interferon regulatory factor 3 (IRF3) to a greater extent than the reference compound, H-151. All five analogs showed stronger STING inhibition than H-151 on the 2',3'-cyclic GMP-AMP-induced expression of interferon regulatory factors (IRFs) in a STINGR232 knock-in THP-1 reporter cell line. The half-maximal inhibitory concentration of the most potent compound, 21, was 11.5 nM. The molecular docking analysis of compound 21 and STING combined with the SAR study suggested that the meta- and para-positions of the benzene ring of the phenylcarbamic amide moiety could be structurally modified by introducing halides or alkyl substituents.


Subject(s)
Amides , Nucleotidyltransferases , Amides/pharmacology , Molecular Docking Simulation , Phosphorylation , Structure-Activity Relationship , Nucleotidyltransferases/metabolism
5.
Mol Cell ; 83(7): 1027-1029, 2023 04 06.
Article in English | MEDLINE | ID: mdl-37028414

ABSTRACT

Nassour et al.1 report that telomere dysfunction communicates with mitochondria via the ZBP1-TERRA-MAVS axis. This pathway activates a detrimental innate immune response that may promote the elimination of cells prone to oncogenic transformation during replicative crisis, thus serving as a telomere-dependent tumor-suppressive mechanism.


Subject(s)
Neoplasms , RNA, Long Noncoding , Humans , Telomere/metabolism , DNA Replication , Neoplasms/metabolism , Mitochondria/metabolism
6.
Structure ; 30(12): 1563-1564, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36459974

ABSTRACT

Emerging models of telomere and associated proteins in Tetrahymena have broadened our understanding of telomeric processes. In this issue of Structure, Ma et al. pinpoint a region in p50 that associates with the CST complex. The interaction mediated by p50 is crucial for the optimal positioning of TERT to maintain homeostasis at the chromosome ends.


Subject(s)
Telomerase , Tetrahymena , Catalytic Domain , Telomere , Homeostasis
7.
Int J Mol Sci ; 23(23)2022 Dec 02.
Article in English | MEDLINE | ID: mdl-36499545

ABSTRACT

Most cases of acquired aplastic anemia (AA) arise from autoimmune destruction of hematopoietic stem and progenitor cells. Human leukocyte antigen (HLA)-haploidentical nonmyeloablative hematopoietic stem cell transplantation (HSCT) plus post-transplantation cyclophosphamide (PTCy) is increasingly applied to salvage AA using bone marrow as graft and anti-thymocyte globulin (ATG) in conditioning. Herein, we characterize a cohort of twelve AA patients clinically and molecularly, six who possessed other immunological disorders (including two also carrying germline SAMD9L mutations). Each patient with SAMD9L mutation also carried an AA-related rare BCORL1 variant or CTLA4 p.T17A GG genotype, respectively, and both presented short telomere lengths. Six of the ten patients analyzed harbored AA-risky HLA polymorphisms. All patients recovered upon non-HSCT (n = 4) or HSCT (n = 8) treatments. Six of the eight HSCT-treated patients were subjected to a modified PTCy-based regimen involving freshly prepared peripheral blood stem cells (PBSC) as graft and exclusion of ATG. All patients were engrafted between post-transplantation days +13 and +18 and quickly reverted to normal life, displaying a sustained complete hematologic response and an absence of graft-versus-host disease. These outcomes indicate most AA cases, including of the SAMD9L-inherited subtype, are immune-mediated and the modified PTCy-based regimen we present is efficient and safe for salvage.


Subject(s)
Anemia, Aplastic , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Humans , Child , Antilymphocyte Serum/therapeutic use , Anemia, Aplastic/genetics , Anemia, Aplastic/therapy , Transplantation Conditioning , Graft vs Host Disease/etiology , Cyclophosphamide/therapeutic use , Hematopoietic Stem Cell Transplantation/adverse effects , HLA Antigens , Retrospective Studies
8.
Nat Commun ; 13(1): 5781, 2022 10 02.
Article in English | MEDLINE | ID: mdl-36184605

ABSTRACT

Alternative Lengthening of Telomeres (ALT) utilizes a recombination mechanism and break-induced DNA synthesis to maintain telomere length without telomerase, but it is unclear how cells initiate ALT. TERRA, telomeric repeat-containing RNA, forms RNA:DNA hybrids (R-loops) at ALT telomeres. We show that depleting TERRA using an RNA-targeting Cas9 system reduces ALT-associated PML bodies, telomere clustering, and telomere lengthening. TERRA interactome reveals that TERRA interacts with an extensive subset of DNA repair proteins in ALT cells. One of TERRA interacting proteins, the endonuclease XPF, is highly enriched at ALT telomeres and recruited by telomeric R-loops to induce DNA damage response (DDR) independent of CSB and SLX4, and thus triggers break-induced telomere synthesis and lengthening. The attraction of BRCA1 and RAD51 at telomeres requires XPF in FANCM-deficient cells that accumulate telomeric R-loops. Our results suggest that telomeric R-loops activate DDR via XPF to promote homologous recombination and telomere replication to drive ALT.


Subject(s)
Telomerase , DNA , Endonucleases/metabolism , RNA , Telomerase/genetics , Telomerase/metabolism , Telomere/genetics , Telomere/metabolism , Telomere Homeostasis
9.
Int J Mol Sci ; 23(16)2022 Aug 22.
Article in English | MEDLINE | ID: mdl-36012751

ABSTRACT

Graves' disease, characterized by hyperthyroidism resulting from loss of immune tolerance to thyroid autoantigens, may be attributable to both genetic and environmental factors. Allogeneic hematopoietic stem cell transplantation (HSCT) represents a means to induce immunotolerance via an artificial immune environment. We present a male patient with severe aplastic anemia arising from a germline SAMD9L missense mutation who successfully underwent HSCT from his HLA-haploidentical SAMD9L non-mutated father together with nonmyeloablative conditioning and post-transplant cyclophosphamide at 8 years of age. He did not suffer graft-versus-host disease, but Graves' disease evolved 10 months post-transplant when cyclosporine was discontinued for one month. Reconstitution of peripheral lymphocyte subsets was found to be transiently downregulated shortly after Graves' disease onset but recovered upon antithyroid treatment. Our investigation revealed the presence of genetic factors associated with Graves' disease, including HLA-B*46:01 and HLA-DRB1*09:01 haplotypes carried by the asymptomatic donor and germline FLT3 c.2500C>T mutation carried by both the patient and the donor. Given his current euthyroid state with normal hematopoiesis, the patient has returned to normal school life. This rare event of Graves' disease in a young boy arising from special HSCT circumstances indicates that both the genetic background and the HSCT environment can prompt the evolution of Graves' disease.


Subject(s)
Graft vs Host Disease , Graves Disease , Hematopoietic Stem Cell Transplantation , Immune Reconstitution , Peripheral Blood Stem Cell Transplantation , Germ Cells , Graft vs Host Disease/genetics , Graves Disease/genetics , Graves Disease/therapy , Hematopoietic Stem Cell Transplantation/methods , Humans , Male , fms-Like Tyrosine Kinase 3
10.
Pediatr Blood Cancer ; 69(8): e29781, 2022 08.
Article in English | MEDLINE | ID: mdl-35593015

ABSTRACT

Primary mediastinal non-seminomatous germ cell tumors (PMNSGCT) are rare but life-threatening thoracic cancers. We report our experience from eight patients with peri-treatment adverse events. By analyzing changes in tumor extent, serum tumor markers, and pathologies between diagnosis and transfer, those events could be attributed to postbiopsy respiratory insufficiency, growing teratoma syndrome, secondary histiocytic malignancy, and PMNSGCT progression. Subjecting patients to respiratory therapy, conventional or high-dose chemotherapy, and surgery controlled the disease, with five of the eight patients surviving disease free. These outcomes indicate that integrated appropriate and timely approaches are important in tackling peri-treatment adverse events.


Subject(s)
Mediastinal Neoplasms , Neoplasms, Germ Cell and Embryonal , Testicular Neoplasms , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Humans , Male , Mediastinal Neoplasms/pathology , Neoplasms, Germ Cell and Embryonal/drug therapy , Testicular Neoplasms/pathology
11.
Methods Mol Biol ; 2450: 467-478, 2022.
Article in English | MEDLINE | ID: mdl-35359323

ABSTRACT

Telomere shortening is a hallmark of aging and eventually constrains the proliferative capacity of cells. The protocols discussed here are used for monitoring telomeres comprehensively in Aeolosoma viride, a model system for regeneration studies. We present methods for analyzing the activity of telomerase enzyme in regenerating tissue by telomeric repeat amplification protocol (TRAP) assay, for comparing telomere length between existing tissue and newly regenerated tissue by telomere restriction fragment (TRF) assay, as well as for visualizing telomeres by fluorescence in situ hybridization (FISH).


Subject(s)
Annelida , Telomerase , Animals , In Situ Hybridization, Fluorescence , Telomerase/genetics , Telomerase/metabolism , Telomere/genetics , Telomere/metabolism , Telomere Homeostasis
12.
J Biomed Sci ; 28(1): 81, 2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34819065

ABSTRACT

BACKGROUND: RAD51-dependent homologous recombination (HR) is one of the most important pathways for repairing DNA double-strand breaks (DSBs), and its regulation is crucial to maintain genome integrity. Elp1 gene encodes IKAP/ELP1, a core subunit of the Elongator complex, which has been implicated in translational regulation. However, how ELP1 contributes to genome maintenance is unclear. METHODS: To investigate the function of Elp1, Elp1-deficient mouse embryonic fibroblasts (MEFs) were generated. Metaphase chromosome spreading, immunofluorescence, and comet assays were used to access chromosome abnormalities and DSB formation. Functional roles of Elp1 in MEFs were evaluated by cell viability, colony forming capacity, and apoptosis assays. HR-dependent DNA repair was assessed by reporter assay, immunofluorescence, and western blot. Polysome profiling was used to evaluate translational efficiency. Differentially expressed proteins and signaling pathways were identified using a label-free liquid chromatography-tandem mass spectrometry (LC-MS/MS) proteomics approach. RESULTS: Here, we report that Elp1 depletion enhanced genomic instability, manifested as chromosome breakage and genotoxic stress-induced genomic DNA fragmentation upon ionizing radiation (IR) exposure. Elp1-deficient cells were hypersensitive to DNA damage and exhibited impaired cell proliferation and defective HR repair. Moreover, Elp1 depletion reduced the formation of IR-induced RAD51 foci and decreased RAD51 protein levels. Polysome profiling analysis revealed that ELP1 regulated RAD51 expression by promoting its translation in response to DNA damage. Notably, the requirement for ELP1 in DSB repair could be partially rescued in Elp1-deficient cells by reintroducing RAD51, suggesting that Elp1-mediated HR-directed repair of DSBs is RAD51-dependent. Finally, using proteome analyses, we identified several proteins involved in cancer pathways and DNA damage responses as being differentially expressed upon Elp1 depletion. CONCLUSIONS: Our study uncovered a molecular mechanism underlying Elp1-mediated regulation of HR activity and provides a novel link between translational regulation and genome stability.


Subject(s)
Chromosome Breakage , DNA Damage/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Biosynthesis/genetics , Rad51 Recombinase/genetics , Recombinational DNA Repair/genetics , Animals , Fibroblasts , Genomic Instability , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Rad51 Recombinase/metabolism
13.
Int J Mol Sci ; 22(5)2021 Feb 27.
Article in English | MEDLINE | ID: mdl-33673424

ABSTRACT

Telomerase negative cancer cell types use the Alternative Lengthening of Telomeres (ALT) pathway to elongate telomeres ends. Here, we show that silencing human DNA polymerase (Pol λ) in ALT cells represses ALT activity and induces telomeric stress. In addition, replication stress in the absence of Pol λ, strongly affects the survival of ALT cells. In vitro, Pol λ can promote annealing of even a single G-rich telomeric repeat to its complementary strand and use it to prime DNA synthesis. The noncoding telomeric repeat containing RNA TERRA and replication protein A negatively regulate this activity, while the Protection of Telomeres protein 1 (POT1)/TPP1 heterodimer stimulates Pol λ. Pol λ associates with telomeres and colocalizes with TPP1 in cells. In summary, our data suggest a role of Pol λ in the maintenance of telomeres by the ALT mechanism.


Subject(s)
Aminopeptidases/metabolism , DNA Polymerase beta/metabolism , G-Quadruplexes , Serine Proteases/metabolism , Telomere Homeostasis , Telomere-Binding Proteins/metabolism , Cell Line, Tumor , Humans , Multiprotein Complexes , Replication Protein A/metabolism , Shelterin Complex , Telomere/chemistry , Telomere/metabolism
14.
Int J Mol Sci ; 20(13)2019 Jul 02.
Article in English | MEDLINE | ID: mdl-31269755

ABSTRACT

Hoyeraal-Hreidarsson syndrome (HHS), caused by several different germline mutations resulting in severe telomeropathy, presents with early-onset growth anomalies and neurologic/developmental disorders including characteristic cerebellar hypoplasia. Early mortalities may arise from immunodeficiency and bone marrow failure if not successfully salvaged by allogeneic hematopoietic stem cell transplantation (HSCT). Few reports have characterized the persistent somatic progression of HHS after successful HSCT. We present an HHS patient with an X-linked recessive DKC1 c.1058C > T; Ala353Val mutation who successfully underwent unrelated HSCT at 5 years of age. After months of early infections and organ toxicities immediately post-transplant, he had more than two years of excellent quality of life with correction of bone marrow failure and immunodeficiency. However, episodic massive variceal bleeding and progressive respiratory insufficiency, which were secondary to non-cirrhotic portal hypertension and pulmonary arteriovenous shunts, respectively, developed over 2 years after HSCT and resulted in his death from respiratory failure 4 years after HSCT. This outcome suggests that while HSCT can correct bone marrow failure and immunodeficiency, it may fail to prevent or even aggravate other fatal processes, such as portal hypertension and pulmonary arteriovenous shunting.


Subject(s)
Cell Cycle Proteins/genetics , Dyskeratosis Congenita/therapy , Fetal Growth Retardation/therapy , Intellectual Disability/therapy , Microcephaly/therapy , Nuclear Proteins/genetics , Peripheral Blood Stem Cell Transplantation , Child, Preschool , Dyskeratosis Congenita/complications , Dyskeratosis Congenita/genetics , Dyskeratosis Congenita/pathology , Fetal Growth Retardation/genetics , Fetal Growth Retardation/pathology , Humans , Intellectual Disability/complications , Intellectual Disability/genetics , Intellectual Disability/pathology , Male , Microcephaly/complications , Microcephaly/genetics , Microcephaly/pathology , Peripheral Blood Stem Cell Transplantation/adverse effects , Peripheral Blood Stem Cell Transplantation/methods , Point Mutation
15.
Sci Rep ; 8(1): 18078, 2018 12 24.
Article in English | MEDLINE | ID: mdl-30584242

ABSTRACT

Aging is a complex process involving declines in various cellular and physical functionalities, including regenerative ability. Telomere maintenance is thought to be necessary for regeneration, and telomere attrition is one mechanism that contributes to aging. However, it is unclear if aging affects regeneration owing to deterioration of telomeric maintenance. We introduce Aeolosoma viride-a freshwater annelid with strong regenerative abilities-as a new model for studying the effects of aging on telomere functions and regeneration. We show that the anterior regenerative ability of A. viride declines with age. We characterized the A. viride telomere sequence as being composed of TTAGGG repeats and identifyied the telomerase gene Avi-tert. In adult A. viride, telomerase was constantly active and telomere lengths were similar among different body sections and stably maintained with age. Notably, we found that regeneration did not result in telomere shortening at regenerating sites. Moreover, transient up-regulation of Avi-tert expression and telomerase activity was observed at regenerating sites, which might promote telomere lengthening to counteract telomere erosion resulting from cell proliferation. Our study suggests that although aging affects A. viride regeneration independent of steady-state telomere length, timely regulation of telomerase functions is critical for the regeneration process in A. viride.


Subject(s)
Aging/genetics , Annelida/genetics , Regeneration , Telomere Homeostasis , Animals , Annelida/growth & development , Annelida/physiology , Telomerase/genetics , Telomerase/metabolism
16.
EMBO Rep ; 19(12)2018 12.
Article in English | MEDLINE | ID: mdl-30446584

ABSTRACT

The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway mediates anti-microbial innate immunity by inducing the production of type I interferons (IFNs) and inflammatory cytokines upon recognition of microbial DNA. Recent studies reveal that self-DNA from tumors and by-products of genomic instability also activates the cGAS-STING pathway and either promotes or inhibits tumor development. This has led to the development of cancer therapeutics using STING agonists alone and in combination with conventional cancer treatment or immune checkpoint targeting. On the other hand, for cancers lacking the cGAS-STING pathway and thus a regular innate immunity response, oncolytic virus therapy has been shown to have therapeutic potential. We here review and discuss the dichotomous roles of the cGAS-STING pathway in cancer development and therapeutic approaches.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Membrane Proteins/metabolism , Neoplasms/therapy , Nucleotidyltransferases/metabolism , Signal Transduction , Animals , Humans , Telomere/metabolism
17.
Nat Struct Mol Biol ; 24(12): 1124-1131, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29106411

ABSTRACT

Extrachromosomal telomere repeat (ECTR) DNA is unique to cancer cells that maintain telomeres through the alternative lengthening of telomeres (ALT) pathway, but the role of ECTRs in ALT development remains elusive. We found that induction of ECTRs in normal human fibroblasts activated the cGAS-STING-TBK1-IRF3 signaling axis to trigger IFNß production and a type I interferon response, resulting in cell-proliferation defects. In contrast, ALT cancer cells are commonly defective in sensing cytosolic DNA. We found that STING expression was inhibited in ALT cancer cell lines and transformed ALT cells. Notably, the ALT suppressors histone H3.3 and the ATRX-Daxx histone chaperone complex were also required to activate the DNA-sensing pathway. Collectively, our data suggest that the loss of the cGAS-STING pathway may be required to evade ECTR-induced anti-proliferation effects and permit ALT development, and this requirement may be exploited for treatments specific to cancers utilizing the ALT pathway.


Subject(s)
Cell Proliferation/physiology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/biosynthesis , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Protein Serine-Threonine Kinases/metabolism , Telomere/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Co-Repressor Proteins , DNA/genetics , Humans , Molecular Chaperones , Neoplasms/genetics , Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/genetics , Telomere Homeostasis/genetics , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism
18.
Trends Biochem Sci ; 40(5): 275-85, 2015 May.
Article in English | MEDLINE | ID: mdl-25845889

ABSTRACT

Telomeres are nucleoprotein structures capping the natural termini of eukaryotic linear chromosomes. Telomeres possess an inherent ability to circumvent the activation of a full-blown DNA damage response (DDR), and hence fusion reactions, by limiting inappropriate double-strand break (DSB) repair and processing activities at eukaryotic chromosome ends. A telomere-specific protein complex, termed shelterin, has a crucial function in safeguarding and securing telomere integrity. Within this complex, TRF2 has emerged as the key player, dictating different states of telomere protection during the replicative lifespan of a cell. How TRF2 prevents activation of DSB repair activities at functional telomeres has now been extensively investigated. In this review we aim at exploring the complex and multi-faceted mechanisms underlying the TRF2-mediated protection of eukaryotic chromosome ends.


Subject(s)
Chromosomes/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Animals , Chromatin/metabolism , DNA Damage/genetics , DNA Damage/physiology , DNA Repair/genetics , DNA Repair/physiology , Humans , Telomere/genetics , Telomeric Repeat Binding Protein 2/genetics
19.
Genes Dev ; 27(19): 2099-108, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24115768

ABSTRACT

Mutations in CTC1 lead to the telomere syndromes Coats Plus and dyskeratosis congenita (DC), but the molecular mechanisms involved remain unknown. CTC1 forms with STN1 and TEN1 a trimeric complex termed CST, which binds ssDNA, promotes telomere DNA synthesis, and inhibits telomerase-mediated telomere elongation. Here we identify CTC1 disease mutations that disrupt CST complex formation, the physical interaction with DNA polymerase α-primase (polα-primase), telomeric ssDNA binding in vitro, accumulation in the nucleus, and/or telomere association in vivo. While having diverse molecular defects, CTC1 mutations commonly lead to the accumulation of internal single-stranded gaps of telomeric DNA, suggesting telomere DNA replication defects as a primary cause of the disease. Strikingly, mutations in CTC1 may also unleash telomerase repression and telomere length control. Hence, the telomere defect initiated by CTC1 mutations is distinct from the telomerase insufficiencies seen in classical forms of telomere syndromes, which cause short telomeres due to reduced maintenance of distal telomeric ends by telomerase. Our analysis provides molecular evidence that CST collaborates with DNA polα-primase to promote faithful telomere DNA replication.


Subject(s)
Genetic Diseases, Inborn/genetics , Mutation , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Telomere/genetics , Telomere/metabolism , Ataxia/genetics , Brain Neoplasms/genetics , Calcinosis/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Central Nervous System Cysts/genetics , Dyskeratosis Congenita/genetics , Gene Expression Regulation , Genes, myc/genetics , HEK293 Cells , Humans , Leukoencephalopathies/genetics , Muscle Spasticity/genetics , Retinal Diseases/genetics , Seizures/genetics , Syndrome , Telomere Homeostasis/genetics , Tubulin/genetics
20.
Nucleus ; 4(4): 277-82, 2013.
Article in English | MEDLINE | ID: mdl-23851344

ABSTRACT

Telomeric DNA at eukaryotic chromosome ends terminates with single stranded 3' G-rich overhangs. The overhang is generated by the interplay of several dynamic processes including semiconservative DNA replication, 3' end elongation by telomerase, C-strand fill-in synthesis and nucleolytic processing. The mammalian CST (CTC1-STN1-TEN1) complex is directly involved at several stages of telomere end formation. Elucidation of its structural organization and identification of interaction partners support the notion that mammalian CST is, as its yeast counterpart, a RPA-like complex. CST binding at mammalian telomere 3' overhangs increases upon their elongation by telomerase. Formation of a trimeric CST complex at telomeric 3'overhangs leads to telomerase inhibition and at the same time mediates a physical interaction with DNA polymerase-α. Thus CST seems to play critical roles in coordinating telomerase elongation and fill-in synthesis to complete telomere replication.


Subject(s)
Telomere-Binding Proteins/metabolism , Telomere/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...