Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
J Biol Chem ; 299(6): 104774, 2023 06.
Article in English | MEDLINE | ID: mdl-37142218

ABSTRACT

Mitochondria are signaling organelles implicated in cancer, but the mechanisms are elusive. Here, we show that Parkin, an E3 ubiquitination (Ub) ligase altered in Parkinson's disease, forms a complex with the regulator of cell motility, Kindlin-2 (K2), at mitochondria of tumor cells. In turn, Parkin ubiquitinates Lys581 and Lys582 using Lys48 linkages, resulting in proteasomal degradation of K2 and shortened half-life from ∼5 h to ∼1.5 h. Loss of K2 inhibits focal adhesion turnover and ß1 integrin activation, impairs membrane lamellipodia size and frequency, and inhibits mitochondrial dynamics, altogether suppressing tumor cell-extracellular matrix interactions, migration, and invasion. Conversely, Parkin does not affect tumor cell proliferation, cell cycle transitions, or apoptosis. Expression of a Parkin Ub-resistant K2 Lys581Ala/Lys582Ala double mutant is sufficient to restore membrane lamellipodia dynamics, correct mitochondrial fusion/fission, and preserve single-cell migration and invasion. In a 3D model of mammary gland developmental morphogenesis, impaired K2 Ub drives multiple oncogenic traits of EMT, increased cell proliferation, reduced apoptosis, and disrupted basal-apical polarity. Therefore, deregulated K2 is a potent oncogene, and its Ub by Parkin enables mitochondria-associated metastasis suppression.


Subject(s)
Membrane Proteins , Ubiquitin-Protein Ligases , Cell Movement , Membrane Proteins/metabolism , Mitochondria/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Humans
2.
Mol Cell ; 59(5): 755-67, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26257282

ABSTRACT

Genome/chromosome organization is highly ordered and controls various nuclear events, although the molecular mechanisms underlying the functional organization remain largely unknown. Here, we show that the TATA box-binding protein (TBP) interacts with the Cnd2 kleisin subunit of condensin to mediate interphase and mitotic chromosomal organization in fission yeast. TBP recruits condensin onto RNA polymerase III-transcribed (Pol III) genes and highly transcribed Pol II genes; condensin in turn associates these genes with centromeres. Inhibition of the Cnd2-TBP interaction disrupts condensin localization across the genome and the proper assembly of mitotic chromosomes, leading to severe defects in chromosome segregation and eventually causing cellular lethality. We propose that the Cnd2-TBP interaction coordinates transcription with chromosomal architecture by linking dispersed gene loci with centromeres. This chromosome arrangement can contribute to the efficient transmission of physical force at the kinetochore to chromosomal arms, thereby supporting the fidelity of chromosome segregation.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/chemistry , Centromere/genetics , Centromere/metabolism , Chromosome Segregation , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genes, Fungal , Mitosis , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Point Mutation , Protein Interaction Domains and Motifs , Protein Subunits , RNA Polymerase III/genetics , RNA Polymerase III/metabolism , Schizosaccharomyces/cytology , Schizosaccharomyces pombe Proteins/chemistry , TATA-Box Binding Protein/chemistry
3.
PLoS Biol ; 17(4): e3000204, 2019 04.
Article in English | MEDLINE | ID: mdl-30951520

ABSTRACT

Telomerase, a unique reverse transcriptase that specifically extends the ends of linear chromosomes, is up-regulated in the vast majority of cancer cells. Here, we show that an indole nucleotide analog, 5-methylcarboxyl-indolyl-2'-deoxyriboside 5'-triphosphate (5-MeCITP), functions as an inhibitor of telomerase activity. The crystal structure of 5-MeCITP bound to the Tribolium castaneum telomerase reverse transcriptase reveals an atypical interaction, in which the nucleobase is flipped in the active site. In this orientation, the methoxy group of 5-MeCITP extends out of the canonical active site to interact with a telomerase-specific hydrophobic pocket formed by motifs 1 and 2 in the fingers domain and T-motif in the RNA-binding domain of the telomerase reverse transcriptase. In vitro data show that 5-MeCITP inhibits telomerase with a similar potency as the clinically administered nucleoside analog reverse transcriptase inhibitor azidothymidine (AZT). In addition, cell-based studies show that treatment with the cell-permeable nucleoside counterpart of 5-MeCITP leads to telomere shortening in telomerase-positive cancer cells, while resulting in significantly lower cytotoxic effects in telomerase-negative cell lines when compared with AZT treatment.


Subject(s)
Nucleosides/metabolism , Telomerase/antagonists & inhibitors , Telomerase/physiology , Animals , Catalytic Domain/drug effects , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Models, Molecular , Nucleosides/chemical synthesis , Nucleosides/physiology , Nucleotides/chemical synthesis , Nucleotides/metabolism , RNA/metabolism , Reverse Transcriptase Inhibitors/pharmacology , Telomere , Tribolium/genetics , Tribolium/metabolism , Zidovudine/metabolism , Zidovudine/pharmacology
4.
Int J Mol Sci ; 23(18)2022 Sep 15.
Article in English | MEDLINE | ID: mdl-36142669

ABSTRACT

The ribonucleoprotein telomerase contains two essential components: telomerase RNA (TER) and telomerase reverse transcriptase (TERT, Est2 in yeast). A small portion of TER, termed the template, is copied by TERT onto the chromosome ends, thus compensating for sequence loss due to incomplete DNA replication and nuclease action. Although telomerase RNA is highly divergent in sequence and length across fungi and mammals, structural motifs essential for telomerase function are conserved. Here, we show that Est2 from the budding yeast Kluyveromyces lactis (klEst2) binds specifically to an essential three-way junction (TWJ) structure in K. lactis TER, which shares a conserved structure and sequence features with the essential CR4-CR5 domain of vertebrate telomerase RNA. klEst2 also binds specifically to the template domain, independently and mutually exclusive of its interaction with TWJ. Furthermore, we present the high-resolution structure of the klEst2 telomerase RNA-binding domain (klTRBD). Mutations introduced in vivo in klTRBD based on the solved structure or in TWJ based on its predicted RNA structure caused severe telomere shortening. These results demonstrate the conservation and importance of these domains and the multiple protein-RNA interactions between Est2 and TER for telomerase function.


Subject(s)
Kluyveromyces , Telomerase , Animals , Base Sequence , Kluyveromyces/genetics , Kluyveromyces/metabolism , Mammals/metabolism , Nucleic Acid Conformation , RNA/metabolism , Telomerase/metabolism
5.
Nucleic Acids Res ; 46(2): 972-984, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29228254

ABSTRACT

The human CST (Ctc1, Stn1 and Ten1) complex binds the telomeric overhang and regulates telomere length by promoting C-strand replication and inhibiting telomerase-dependent G-strand synthesis. Structural and biochemical studies on the human Stn1 and Ten1 complex revealed its mechanism of assembly and nucleic acid binding. However, little is known about the structural organization of the multi-domain Ctc1 protein and how each of these domains contribute to telomere length regulation. Here, we report the structure of a central domain of human Ctc1. The structure reveals a canonical OB-fold with the two identified disease mutations (R840W and V871M) contributing to the fold of the protein. In vitro assays suggest that although this domain is not contributing directly to Ctc1's substrate binding properties, it affects full-length Ctc1 localization to telomeres and Stn1-Ten1 binding. Moreover, functional assays show that deletion of the entire OB-fold domain leads to significant increase in telomere length, frequency of internal single G-strands and fragile telomeres. Our findings demonstrate that a previously unknown OB-fold domain contributes to efficient Ctc1 telomere localization and chromosome end maintenance.


Subject(s)
Bone Marrow/metabolism , Protein Folding , Telomere Homeostasis , Telomere-Binding Proteins/chemistry , Telomere/metabolism , Amino Acid Sequence , Bone Marrow/pathology , Crystallography, X-Ray , HEK293 Cells , Humans , Models, Molecular , Mutation , Protein Binding , Protein Domains , Sequence Homology, Amino Acid , Syndrome , Telomere/genetics , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism
6.
J Biol Chem ; 292(11): 4593-4601, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28154186

ABSTRACT

Naturally occurring mutations in the ribonucleoprotein reverse transcriptase, telomerase, are associated with the bone marrow failure syndromes dyskeratosis congenita, aplastic anemia, and idiopathic pulmonary fibrosis. However, the mechanism by which these mutations impact telomerase function remains unknown. Here we present the structure of the human telomerase C-terminal extension (or thumb domain) determined by the method of single-wavelength anomalous diffraction to 2.31 Å resolution. We also used direct telomerase activity and nucleic acid binding assays to explain how naturally occurring mutations within this portion of telomerase contribute to human disease. The single mutations localize within three highly conserved regions of the telomerase thumb domain referred to as motifs E-I (thumb loop and helix), E-II, and E-III (the FVYL pocket, comprising the hydrophobic residues Phe-1012, Val-1025, Tyr-1089, and Leu-1092). Biochemical data show that the mutations associated with dyskeratosis congenita, aplastic anemia, and idiopathic pulmonary fibrosis disrupt the binding between the protein subunit reverse transcriptase of the telomerase and its nucleic acid substrates leading to loss of telomerase activity and processivity. Collectively our data show that although these mutations do not alter the overall stability or expression of telomerase reverse transcriptase, these rare genetic disorders are associated with an impaired telomerase holoenzyme that is unable to correctly assemble with its nucleic acid substrates, leading to incomplete telomere extension and telomere attrition, which are hallmarks of these diseases.


Subject(s)
Anemia, Aplastic/genetics , Bone Marrow Diseases/genetics , Hemoglobinuria, Paroxysmal/genetics , Point Mutation , Telomerase/genetics , Amino Acid Sequence , Anemia, Aplastic/metabolism , Bone Marrow/metabolism , Bone Marrow Diseases/metabolism , Bone Marrow Failure Disorders , Crystallography, X-Ray , Dyskeratosis Congenita/genetics , Dyskeratosis Congenita/metabolism , HEK293 Cells , Hemoglobinuria, Paroxysmal/metabolism , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Models, Molecular , Nucleic Acids/metabolism , Protein Conformation , Protein Domains , Telomerase/chemistry , Telomerase/metabolism , Telomere/metabolism
7.
J Biol Chem ; 288(27): 19882-99, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23689370

ABSTRACT

Unlike the core histones, which are incorporated into nucleosomes concomitant with DNA replication, histone H3.3 is synthesized throughout the cell cycle and utilized for replication-independent (RI) chromatin assembly. The RI incorporation of H3.3 into nucleosomes is highly conserved and occurs at both euchromatin and heterochromatin. However, neither the mechanism of H3.3 recruitment nor its essential function is well understood. Several different chaperones regulate H3.3 assembly at distinct sites. The H3.3 chaperone, Daxx, and the chromatin-remodeling factor, ATRX, are required for H3.3 incorporation and heterochromatic silencing at telomeres, pericentromeres, and the cytomegalovirus (CMV) promoter. By evaluating H3.3 dynamics at a CMV promoter-regulated transcription site in a genetic background in which RI chromatin assembly is blocked, we have been able to decipher the regulatory events upstream of RI nucleosomal deposition. We find that at the activated transcription site, H3.3 accumulates with sense and antisense RNA, suggesting that it is recruited through an RNA-mediated mechanism. Sense and antisense transcription also increases after H3.3 knockdown, suggesting that the RNA signal is amplified when chromatin assembly is blocked and attenuated by nucleosomal deposition. Additionally, we find that H3.3 is still recruited after Daxx knockdown, supporting a chaperone-independent recruitment mechanism. Sequences in the H3.3 N-terminal tail and αN helix mediate both its recruitment to RNA at the activated transcription site and its interaction with double-stranded RNA in vitro. Interestingly, the H3.3 gain-of-function pediatric glioblastoma mutations, G34R and K27M, differentially affect H3.3 affinity in these assays, suggesting that disruption of an RNA-mediated regulatory event could drive malignant transformation.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Cytomegalovirus/metabolism , Histones/metabolism , Promoter Regions, Genetic/physiology , RNA, Viral/biosynthesis , Transcription, Genetic/physiology , Cell Line , Cytomegalovirus/genetics , Histones/genetics , Humans , Nucleosomes/genetics , Nucleosomes/metabolism , Protein Structure, Secondary , RNA, Viral/genetics
8.
Nature ; 455(7213): 633-7, 2008 Oct 02.
Article in English | MEDLINE | ID: mdl-18758444

ABSTRACT

A common hallmark of human cancers is the overexpression of telomerase, a ribonucleoprotein complex that is responsible for maintaining the length and integrity of chromosome ends. Telomere length deregulation and telomerase activation is an early, and perhaps necessary, step in cancer cell evolution. Here we present the high-resolution structure of the Tribolium castaneum catalytic subunit of telomerase, TERT. The protein consists of three highly conserved domains, organized into a ring-like structure that shares common features with retroviral reverse transcriptases, viral RNA polymerases and B-family DNA polymerases. Domain organization places motifs implicated in substrate binding and catalysis in the interior of the ring, which can accommodate seven to eight bases of double-stranded nucleic acid. Modelling of an RNA-DNA heteroduplex in the interior of this ring demonstrates a perfect fit between the protein and the nucleic acid substrate, and positions the 3'-end of the DNA primer at the active site of the enzyme, providing evidence for the formation of an active telomerase elongation complex.


Subject(s)
Telomerase/chemistry , Tribolium/enzymology , Amino Acid Motifs , Animals , Binding Sites , Catalysis , Catalytic Domain , Conserved Sequence , Crystallization , Crystallography, X-Ray , Humans , Models, Molecular , Nucleotides/metabolism , Protein Structure, Tertiary , Telomerase/metabolism
9.
bioRxiv ; 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38464183

ABSTRACT

RTEL1 is an essential DNA helicase that plays multiple roles in genome stability and telomere length regulation. A variant of RTEL1 with a lysine at position 492 is associated with short telomeres in Mus spretus , while a conserved methionine at this position is found in M. musculus, which has ultra-long telomeres. In humans, a missense mutation at this position ( RTEL1 M492I ) causes a fatal telomere biology disease termed Hoyeraal-Hreidarsson syndrome (HHS). We previously described a M. musculus mouse model termed 'Telomouse', in which changing methionine 492 to a lysine (M492K) shortened the telomeres to their length in humans. Here, we report on the derivation of a mouse strain carrying the M492I mutation, termed 'HHS mouse'. The HHS mouse telomeres are not as short as those of Telomice but nevertheless they display higher levels of telomeric DNA damage, fragility and recombination, associated with anaphase bridges and micronuclei. These observations indicate that the two mutations separate critical functions of RTEL1: M492K mainly reduces the telomere length setpoint, while M492I predominantly disrupts telomere protection. The two mouse models enable dissecting the mechanistic roles of RTEL1 and the different contributions of short telomeres and DNA damage to telomere biology diseases, genomic instability, cancer, and aging.

10.
Oncogenesis ; 13(1): 4, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38191593

ABSTRACT

The essential G1-cyclin, CCND1, is frequently overexpressed in cancer, contributing to tumorigenesis by driving cell-cycle progression. D-type cyclins are rate-limiting regulators of G1-S progression in mammalian cells via their ability to bind and activate CDK4 and CDK6. In addition, cyclin D1 conveys kinase-independent transcriptional functions of cyclin D1. Here we report that cyclin D1 associates with H2BS14 via an intrinsically disordered domain (IDD). The same region of cyclin D1 was necessary for the induction of aneuploidy, induction of the DNA damage response, cyclin D1-mediated recruitment into chromatin, and CIN gene transcription. In response to DNA damage H2BS14 phosphorylation occurs, resulting in co-localization with γH2AX in DNA damage foci. Cyclin D1 ChIP seq and γH2AX ChIP seq revealed ~14% overlap. As the cyclin D1 IDD functioned independently of the CDK activity to drive CIN, the IDD domain may provide a rationale new target to complement CDK-extinction strategies.

11.
Nat Commun ; 14(1): 6708, 2023 10 23.
Article in English | MEDLINE | ID: mdl-37872177

ABSTRACT

Telomeres, the ends of eukaryotic chromosomes, protect genome integrity and enable cell proliferation. Maintaining optimal telomere length in the germline and throughout life limits the risk of cancer and enables healthy aging. Telomeres in the house mouse, Mus musculus, are about five times longer than human telomeres, limiting the use of this common laboratory animal for studying the contribution of telomere biology to aging and cancer. We identified a key amino acid variation in the helicase RTEL1, naturally occurring in the short-telomere mouse species M. spretus. Introducing this variation into M. musculus is sufficient to reduce the telomere length set point in the germline and generate mice with human-length telomeres. While these mice are fertile and appear healthy, the regenerative capacity of their colonic epithelium is compromised. The engineered Telomouse reported here demonstrates a dominant role of RTEL1 in telomere length regulation and provides a unique model for aging and cancer.


Subject(s)
Genome , Neoplasms , Humans , Mice , Animals , Disease Models, Animal , Telomere/genetics , Cell Proliferation , Neoplasms/genetics , DNA Helicases/genetics
12.
Hum Mol Genet ; 19(6): 1033-47, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20028791

ABSTRACT

Rab GTPases are molecular switches that orchestrate vesicular trafficking, maturation and fusion by cycling between an active, GTP-bound form, and an inactive, GDP-bound form. The activity cycle is coupled to GTP hydrolysis and is tightly controlled by regulatory proteins. Missense mutations of the GTPase Rab7 cause a dominantly inherited axonal degeneration known as Charcot-Marie-Tooth type 2B through an unknown mechanism. We present the 2.8 A crystal structure of GTP-bound L129F mutant Rab7 which reveals normal conformations of the effector binding regions and catalytic site, but an alteration to the nucleotide binding pocket that is predicted to alter GTP binding. Through extensive biochemical analysis, we demonstrate that disease-associated mutations in Rab7 do not lead to an intrinsic GTPase defect, but permit unregulated nucleotide exchange leading to both excessive activation and hydrolysis-independent inactivation. Consistent with augmented activity, mutant Rab7 shows significantly enhanced interaction with a subset of effector proteins. In addition, dynamic imaging demonstrates that mutant Rab7 is abnormally retained on target membranes. However, we show that the increased activation of mutant Rab7 is counterbalanced by unregulated, GTP hydrolysis-independent membrane cycling. Notably, disease mutations are able to rescue the membrane cycling of a GTPase-deficient mutant. Thus, we demonstrate that disease mutations uncouple Rab7 from the spatial and temporal control normally imposed by regulatory proteins and cause disease not by a gain of novel toxic function, but by misregulation of native Rab7 activity.


Subject(s)
Charcot-Marie-Tooth Disease/enzymology , Charcot-Marie-Tooth Disease/genetics , Guanine Nucleotide Exchange Factors/metabolism , Mutation/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , Amino Acid Substitution/genetics , Binding Sites , Cell Line , Cytoplasmic Vesicles/metabolism , Enzyme Activation , Guanosine Triphosphate/metabolism , Humans , Intracellular Membranes/metabolism , Models, Biological , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding , Protein Structure, Secondary , Protein Transport , rab GTP-Binding Proteins/chemistry , rab7 GTP-Binding Proteins
13.
Proc Natl Acad Sci U S A ; 106(51): 21573-8, 2009 Dec 22.
Article in English | MEDLINE | ID: mdl-19966225

ABSTRACT

Chromatin can be modified by posttranslational modifications of histones, ATP-dependent remodeling, and incorporation of histone variants. The Saccharomyces cerevisiae protein Yaf9 is a subunit of both the essential histone acetyltransferase complex NuA4 and the ATP-dependent chromatin remodeling complex SWR1-C, which deposits histone variant H2A.Z into euchromatin. Yaf9 contains a YEATS domain, found in proteins associated with multiple chromatin-modifying enzymes and transcription complexes across eukaryotes. Here, we established the conservation of YEATS domain function from yeast to human, and determined the structure of this region from Yaf9 by x-ray crystallography to 2.3 A resolution. The Yaf9 YEATS domain consisted of a beta-sandwich characteristic of the Ig fold and contained three distinct conserved structural features. The structure of the Yaf9 YEATS domain was highly similar to that of the histone chaperone Asf1, a similarity that extended to an ability of Yaf9 to bind histones H3 and H4 in vitro. Using structure-function analysis, we found that the YEATS domain was required for Yaf9 function, histone variant H2A.Z chromatin deposition at specific promoters, and H2A.Z acetylation.


Subject(s)
Cell Cycle Proteins/metabolism , Histone Acetyltransferases/metabolism , Histones/metabolism , Molecular Chaperones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Acetylation , Amino Acid Sequence , Cell Cycle Proteins/chemistry , Histone Acetyltransferases/chemistry , Models, Molecular , Molecular Chaperones/chemistry , Molecular Sequence Data , Protein Conformation , Saccharomyces cerevisiae Proteins/chemistry , Sequence Homology, Amino Acid
14.
Comput Struct Biotechnol J ; 20: 675-684, 2022.
Article in English | MEDLINE | ID: mdl-35140887

ABSTRACT

Telomeric POT1-TPP1 binding is critical to telomere maintenance and disruption of this complex may lead to cancer. Current data suggests a reduction of intracellular POT1 levels in the absence of TPP1. Here we provide evidence of POT1 plasticity that contributes to its lack of stability in the absence of TPP1 binding. Structural data reveals inter- and intramolecular POT1C domain flexibility in the absence of TPP1. Thermostability and proteolytic resistance assays show that POT1C and the mutant complex POT1C(Q623H)-TPP1(PBD) are less stable than the wild type POT1C-TPP1(PBD), suggesting that TPP1 binding to POT1 stabilizes POT1C and makes it less accessible to proteasomal degradation in the cell. Disruption of the POT1-TPP1 complex such as through cancer-associated mutations leads to a reduction of intracellular POT1, telomere uncapping, and telomere associated DNA damage response (DDR). DDR in turn leads to senescence or genomic instability and oncogenesis.

15.
J Exp Med ; 219(5)2022 05 02.
Article in English | MEDLINE | ID: mdl-35420632

ABSTRACT

Exonic sequencing identified a family with idiopathic pulmonary fibrosis (IPF) containing a previously unreported heterozygous mutation in POT1 p.(L259S). The family displays short telomeres and genetic anticipation. We found that POT1(L259S) is defective in binding the telomeric overhang, nuclear accumulation, negative regulation of telomerase, and lagging strand maintenance. Patient cells containing the mutation display telomere loss, lagging strand defects, telomere-induced DNA damage, and premature senescence with G1 arrest. Our data suggest POT1(L259S) is a pathogenic driver of IPF and provide insights into gene therapy options.


Subject(s)
Idiopathic Pulmonary Fibrosis , Telomerase , Humans , Idiopathic Pulmonary Fibrosis/genetics , Shelterin Complex , Telomerase/genetics , Telomerase/metabolism , Telomere/genetics , Telomere/metabolism , Telomere-Binding Proteins/genetics
16.
Commun Biol ; 4(1): 349, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33731801

ABSTRACT

The human CST complex composed of CTC1, STN1, and TEN1 is critically involved in telomere maintenance and homeostasis. Specifically, CST terminates telomere extension by inhibiting telomerase access to the telomeric overhang and facilitates lagging strand fill in by recruiting DNA Polymerase alpha primase (Pol α-primase) to the telomeric C-strand. Here we reveal that CST has a dynamic intracellular localization that is cell cycle dependent. We report an increase in nuclear CST several hours after the initiation of DNA replication, followed by exit from the nucleus prior to mitosis. We identify amino acids of CTC1 involved in Pol α-primase binding and nuclear localization. We conclude, the CST complex does not contain a nuclear localization signal (NLS) and suggest that its nuclear localization is reliant on Pol α-primase. Hypomorphic mutations affecting CST nuclear import are associated with telomere syndromes and cancer, emphasizing the important role of this process in health.


Subject(s)
Cell Nucleus/enzymology , DNA Polymerase I/metabolism , DNA Primase/metabolism , Telomere Homeostasis , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Cell Nucleus/genetics , DNA Polymerase I/genetics , DNA Primase/genetics , DNA Replication , HEK293 Cells , Humans , Mitosis , Multiprotein Complexes , Mutation , Protein Binding , Telomere/genetics , Telomere-Binding Proteins/genetics
17.
Cancer Res ; 81(5): 1252-1264, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33414169

ABSTRACT

Acetyl-CoA is a vitally important and versatile metabolite used for many cellular processes including fatty acid synthesis, ATP production, and protein acetylation. Recent studies have shown that cancer cells upregulate acetyl-CoA synthetase 2 (ACSS2), an enzyme that converts acetate to acetyl-CoA, in response to stresses such as low nutrient availability and hypoxia. Stressed cancer cells use ACSS2 as a means to exploit acetate as an alternative nutrient source. Genetic depletion of ACSS2 in tumors inhibits the growth of a wide variety of cancers. However, there are no studies on the use of an ACSS2 inhibitor to block tumor growth. In this study, we synthesized a small-molecule inhibitor that acts as a transition-state mimetic to block ACSS2 activity in vitro and in vivo. Pharmacologic inhibition of ACSS2 as a single agent impaired breast tumor growth. Collectively, our findings suggest that targeting ACSS2 may be an effective therapeutic approach for the treatment of patients with breast cancer. SIGNIFICANCE: These findings suggest that targeting acetate metabolism through ACSS2 inhibitors has the potential to safely and effectively treat a wide range of patients with cancer.


Subject(s)
Acetate-CoA Ligase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Acetate-CoA Ligase/genetics , Acetate-CoA Ligase/metabolism , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Drug Stability , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fatty Acids/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Mice, Inbred Strains , Molecular Docking Simulation , Molecular Targeted Therapy/methods , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
18.
Comput Struct Biotechnol J ; 18: 1939-1946, 2020.
Article in English | MEDLINE | ID: mdl-32774788

ABSTRACT

Telomeres are DNA repeats at the ends of linear chromosomes and are replicated by telomerase, a ribonucleoprotein reverse transcriptase. Telomere length regulation and chromosome end capping are essential for genome stability and are mediated primarily by the shelterin and CST complexes. POT1-TPP1, a subunit of shelterin, binds the telomeric overhang, suppresses ATR-dependent DNA damage response, and recruits telomerase to telomeres for DNA replication. POT1 localization to telomeres and chromosome end protection requires its interaction with TPP1. Therefore, the POT1-TPP1 complex is critical to telomere maintenance and full telomerase processivity. The aim of this mini-review is to summarize recent POT1-TPP1 structural studies and discuss how the complex contributes to telomere length regulation. In addition, we review how disruption of POT1-TPP1 function leads to human disease.

19.
Nat Commun ; 11(1): 2219, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32376827

ABSTRACT

Heterochromatin in the eukaryotic genome is rigorously controlled by the concerted action of protein factors and RNAs. Here, we investigate the RNA binding function of ATRX, a chromatin remodeler with roles in silencing of repetitive regions of the genome and in recruitment of the polycomb repressive complex 2 (PRC2). We identify ATRX RNA binding regions (RBRs) and discover that the major ATRX RBR lies within the N-terminal region of the protein, distinct from its PHD and helicase domains. Deletion of this ATRX RBR (ATRXΔRBR) compromises ATRX interactions with RNAs in vitro and in vivo and alters its chromatin binding properties. Genome-wide studies reveal that loss of RNA interactions results in a redistribution of ATRX on chromatin. Finally, our studies identify a role for ATRX-RNA interactions in regulating PRC2 localization to a subset of polycomb target genes.


Subject(s)
Chromatin/metabolism , Polycomb Repressive Complex 2/metabolism , RNA/metabolism , X-linked Nuclear Protein/genetics , Animals , Chromatin Assembly and Disassembly/genetics , Female , Fibroblasts/enzymology , Fibroblasts/metabolism , Heterochromatin/metabolism , Histones/chemistry , Histones/metabolism , Methylation , Mice , Protein Binding , Protein Domains/genetics , X-linked Nuclear Protein/metabolism
20.
Cell Rep ; 32(11): 108151, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32937140

ABSTRACT

Cyclin D1 encodes the regulatory subunit of a holoenzyme that phosphorylates RB and functions as a collaborative nuclear oncogene. The serine threonine kinase Akt plays a pivotal role in the control of cellular metabolism, survival, and mitogenic signaling. Herein, Akt1-mediated phosphorylation of downstream substrates in the mammary gland is reduced by cyclin D1 genetic deletion and is induced by mammary-gland-targeted cyclin D1 overexpression. Cyclin D1 is associated with Akt1 and augments the rate of onset and maximal cellular Akt1 activity induced by mitogens. Cyclin D1 is identified in a cytoplasmic-membrane-associated pool, and cytoplasmic-membrane-localized cyclin D1-but not nuclear-localized cyclin D1-recapitulates Akt1 transcriptional function. These studies identify a novel extranuclear function of cyclin D1 to enhance proliferative functions via augmenting Akt1 phosphorylation at Ser473.


Subject(s)
Cyclin D1/metabolism , Mitogens/metabolism , Phosphoserine/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , 3T3 Cells , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Membrane/metabolism , Cyclin D1/genetics , Cyclin-Dependent Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , MCF-7 Cells , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL