Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
1.
J Cell Signal ; 2(1): 9-26, 2021.
Article in English | MEDLINE | ID: mdl-33554216

ABSTRACT

Forkhead transcription factors (TFs) often dimerize outside their extensive family, whereas bHLH transcription factors typically dimerize with E12/E47. Based on structural similarities, we predicted that a member of the former, Forkhead Box P1 (FOXP1), might heterodimerize with a member of the latter, MYOD1 (MyoD). Data shown here support this hypothesis and further demonstrate the specificity of this forkhead/myogenic interaction among other myogenic regulatory factors. We found that FOXP1-MyoD heterodimerization compromises the ability of MyoD to bind to E-boxes and to transactivate E box- containing promoters. We observed that FOXP1 is required for the full ability of MyoD to convert fibroblasts into myotubules. We provide a model in which FOXP1 displaces ID and E12/E47 to repress MyoD during the proliferative phase of myoblast differentiation. These data identify FOXP1 as a hitherto unsuspected transcriptional repressor of MyoD. We suggest that isolation of paired E-box and forkhead sites within 1 turn helical spacings provides potential for cooperative interactions among heretofore distinct classes of transcription factors.

2.
Exp Neurol ; 320: 113011, 2019 10.
Article in English | MEDLINE | ID: mdl-31306642

ABSTRACT

Aberrant expression of DUX4, a gene unique to humans and primates, causes Facioscapulohumeral Muscular Dystrophy-1 (FSHD), yet the pathogenic mechanism is unknown. As transgenic overexpression models have largely failed to replicate the genetic changes seen in FSHD, many studies of endogenously expressed DUX4 have been limited to patient biopsies and myogenic cell cultures, which never fully differentiate into mature muscle fibers. We have developed a method to xenograft immortalized human muscle precursor cells from patients with FSHD and first-degree relative controls into the tibialis anterior muscle compartment of immunodeficient mice, generating human muscle xenografts. We report that FSHD cells mature into organized and innervated human muscle fibers with minimal contamination of murine myonuclei. They also reconstitute the satellite cell niche within the xenografts. FSHD xenografts express DUX4 and DUX4 downstream targets, retain the 4q35 epigenetic signature of their original donors, and express a novel protein biomarker of FSHD, SLC34A2. Ours is the first scalable, mature in vivo human model of FSHD. It should be useful for studies of the pathogenic mechanism of the disease as well as for testing therapeutic strategies targeting DUX4 expression.


Subject(s)
Disease Models, Animal , Heterografts , Muscular Dystrophy, Facioscapulohumeral , Myoblasts/transplantation , Animals , Homeodomain Proteins/genetics , Humans , Mice , Muscle, Skeletal/pathology , Muscular Dystrophy, Facioscapulohumeral/genetics
3.
J Biol Chem ; 294(30): 11579-11596, 2019 07 26.
Article in English | MEDLINE | ID: mdl-31186347

ABSTRACT

Human telomerase maintains genome stability by adding telomeric repeats to the ends of linear chromosomes. Although previous studies have revealed profound insights into telomerase functions, the low cellular abundance of functional telomerase and the difficulties in quantifying its activity leave its thermodynamic and kinetic properties only partially characterized. Employing a stable cell line overexpressing both the human telomerase RNA component and the N-terminally biotinylated human telomerase reverse transcriptase and using a newly developed method to count individual extension products, we demonstrate here that human telomerase holoenzymes contain fast- and slow-acting catalytic sites. Surprisingly, both active sites became inactive after two consecutive rounds of catalysis, named single-run catalysis. The fast active sites turned off ∼40-fold quicker than the slow ones and exhibited higher affinities to DNA substrates. In a dimeric enzyme, the two active sites work in tandem, with the faster site functioning before the slower one, and in the monomeric enzyme, the active sites also perform single-run catalysis. Interestingly, inactive enzymes could be reactivated by intracellular telomerase-activating factors (iTAFs) from multiple cell types. We conclude that the single-run catalysis and the iTAF-triggered reactivation serve as an unprecedented control circuit for dynamic regulation of telomerase. They endow native telomerase holoenzymes with the ability to match their total number of active sites to the number of telomeres they extend. We propose that the exquisite kinetic control of telomerase activity may play important roles in both cell division and cell aging.


Subject(s)
Biological Factors/metabolism , Telomerase/antagonists & inhibitors , Catalysis , Catalytic Domain , Cell Line , Enzyme Activation , Humans , Telomerase/metabolism
4.
Genes Dev ; 33(13-14): 814-827, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31171703

ABSTRACT

Alternative lengthening of telomeres (ALT) is a telomerase-independent telomere maintenance mechanism that occurs in a subset of cancers. One of the hallmarks of ALT cancer is the excessively clustered telomeres in promyelocytic leukemia (PML) bodies, represented as large bright telomere foci. Here, we present a model system that generates telomere clustering in nuclear polySUMO (small ubiquitin-like modification)/polySIM (SUMO-interacting motif) condensates, analogous to PML bodies, and thus artificially engineered ALT-associated PML body (APB)-like condensates in vivo. We observed that the ALT-like phenotypes (i.e., a small fraction of heterogeneous telomere lengths and formation of C circles) are rapidly induced by introducing the APB-like condensates together with BLM through its helicase domain, accompanied by ssDNA generation and RPA accumulation at telomeres. Moreover, these events lead to mitotic DNA synthesis (MiDAS) at telomeres mediated by RAD52 through its highly conserved N-terminal domain. We propose that the clustering of large amounts of telomeres in human cancers promotes ALT that is mediated by MiDAS, analogous to Saccharomyces cerevisiae type II ALT survivors.


Subject(s)
Cell Nucleus/metabolism , DNA/biosynthesis , Leukemia, Promyelocytic, Acute/physiopathology , Mitosis , Rad52 DNA Repair and Recombination Protein/metabolism , RecQ Helicases/metabolism , Telomere Homeostasis/genetics , Amino Acid Motifs , Cell Line, Tumor , Gene Expression , Humans , Leukemia, Promyelocytic, Acute/genetics , Phenotype , Protein Transport , SUMO-1 Protein/metabolism , Telomere/genetics , Telomere/metabolism
5.
J Biol Chem ; 294(16): 6645-6656, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30819801

ABSTRACT

Linear chromosome ends are capped by telomeres that have been previously reported to adopt a t-loop structure. The lack of simple methods for detecting t-loops has hindered progress in understanding the dynamics of t-loop formation and its function in protecting chromosome ends. Here, we employed a classical two-dimensional agarose gel method (2D gel method) to innovatively apply to t-loop detection. Briefly, restriction fragments of genomic DNA were separated in a 2D gel, and the telomere sequence was detected by in-gel hybridization with telomeric probe. Using this method, we found that t-loops are present throughout the cell cycle, and t-loop formation tightly couples to telomere replication. We also observed that t-loop abundance positively correlates with chromatin condensation, i.e. cells with less compact telomeric chromatin (ALT cells and trichostatin A (TSA)-treated HeLa cells) exhibited fewer t-loops. Moreover, we observed that telomere dysfunction-induced foci, ALT-associated promyelocytic leukemia bodies, and telomere sister chromatid exchanges are activated upon TSA-induced loss of t-loops. These findings confirm the importance of the t-loop in protecting linear chromosomes from damage or illegitimate recombination.


Subject(s)
Cell Cycle/physiology , Chromatids/metabolism , Heterochromatin/metabolism , Telomere/metabolism , Chromatids/chemistry , Electrophoresis, Gel, Two-Dimensional , HeLa Cells , Heterochromatin/chemistry , Humans , Hydroxamic Acids/pharmacology , Telomere/chemistry
6.
Nat Rev Genet ; 20(5): 299-309, 2019 05.
Article in English | MEDLINE | ID: mdl-30760854

ABSTRACT

Many recent advances have emerged in the telomere and telomerase fields. This Timeline article highlights the key advances that have expanded our views on the mechanistic underpinnings of telomeres and telomerase and their roles in ageing and disease. Three decades ago, the classic view was that telomeres protected the natural ends of linear chromosomes and that telomerase was a specific telomere-terminal transferase necessary for the replication of chromosome ends in single-celled organisms. While this concept is still correct, many diverse fields associated with telomeres and telomerase have substantially matured. These areas include the discovery of most of the key molecular components of telomerase, implications for limits to cellular replication, identification and characterization of human genetic disorders that result in premature telomere shortening, the concept that inhibiting telomerase might be a successful therapeutic strategy and roles for telomeres in regulating gene expression. We discuss progress in these areas and conclude with challenges and unanswered questions in the field.


Subject(s)
Aging/genetics , Genomics/history , Neoplasms/genetics , Telomerase/genetics , Telomere/chemistry , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Aging/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA/chemistry , DNA/genetics , DNA/metabolism , Gene Expression Regulation , Genomics/methods , History, 20th Century , History, 21st Century , Humans , Molecular Chaperones , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Progeria/genetics , Progeria/metabolism , Progeria/pathology , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Ribonucleoproteins, Small Nucleolar/genetics , Ribonucleoproteins, Small Nucleolar/metabolism , Shelterin Complex , Telomerase/metabolism , Telomere/metabolism , Telomere Homeostasis , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism
7.
Aging Cell ; 18(1): e12859, 2019 02.
Article in English | MEDLINE | ID: mdl-30488553

ABSTRACT

It is generally recognized that the function of the immune system declines with increased age and one of the major immune changes is impaired T-cell responses upon antigen presentation/stimulation. Some "high-performing" centenarians (100+ years old) are remarkably successful in escaping, or largely postponing, major age-related diseases. However, the majority of centenarians ("low-performing") have experienced these pathologies and are forced to reside in long-term nursing facilities. Previous studies have pooled all centenarians examining heterogeneous populations of resting/unstimulated peripheral blood mononuclear cells (PBMCs). T cells represent around 60% of PBMC and are in a quiescence state when unstimulated. However, upon stimulation, T cells rapidly divide and exhibit dramatic changes in gene expression. We have compared stimulated T-cell responses and identified a set of transcripts expressed in vitro that are dramatically different in high- vs. low-performing centenarians. We have also identified several other measurements that are different between high- and low-performing centenarians: (a) The amount of proliferation following in vitro stimulation is dramatically greater in high-performing centenarians compared to 67- to 83-year-old controls and low-performing centenarians; (b) telomere length is greater in the high-performing centenarians; and (c) telomerase activity following stimulation is greater in the high-performing centenarians. In addition, we have validated a number of genes whose expression is directly related to telomere length and these are potential fundamental biomarkers of aging that may influence the risk and progression of multiple aging conditions.


Subject(s)
T-Lymphocytes/metabolism , Telomerase/metabolism , Telomere Homeostasis , Telomere/metabolism , Adult , Aged , Aged, 80 and over , Aging/genetics , Biomarkers/metabolism , Cell Proliferation , DNA Replication , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genome, Human , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Middle Aged , Young Adult
8.
Plast Reconstr Surg ; 143(2): 490-494, 2019 02.
Article in English | MEDLINE | ID: mdl-30531622

ABSTRACT

Keloids are benign fibroproliferative skin tumors that can cause disfigurement and disability. Although they frequently recur after excision or medical management and can affect 6 to 16 percent of African Americans, there is no gold standard therapy. Keloids are challenging to study because there are no animal or in vitro models of this disorder. This makes it very difficult to validate data from treated tissue samples or cells and develop targeted therapies for this disease. In this study, the authors demonstrate that intralesional 5-fluorouracil injection after keloid excision prevents recurrence for 2 years, with no reported adverse events. The authors analyze the expression of treated and untreated biopsy specimens of the same keloids in their native context to capture insights that may be missed by in vitro cell culture models and correct for intrakeloid variability. Random forest analysis of the microarray data dramatically increased the statistical power of the authors' results, permitting hypothesis-free creation of a gene expression profile of 5-fluorouracil-treated keloids. Through this analysis, the authors found a set of genes, including YAP1 and CCL-2, whose expression changes predict 5-fluorouracil therapy status and include genes that have not previously been associated with keloid biology and are of unknown function. The authors further describe keloid heterogeneity for the first time using multidimensional analysis of their microarray results. The methods and tools the authors developed in this research may overcome some of the challenges in studying keloids and developing effective treatments for this disease. CLINICAL QUESTION/LEVEL OF EVIDENCE:: Therapeutic, V.


Subject(s)
Dermis/pathology , Fluorouracil/administration & dosage , Keloid/drug therapy , Adaptor Proteins, Signal Transducing/metabolism , Biopsy , Chemokine CCL2/metabolism , Dermis/surgery , Follow-Up Studies , Gene Expression Profiling , Humans , Injections, Intralesional , Keloid/pathology , Keloid/surgery , Phosphoproteins/metabolism , Recurrence , Transcription Factors , Treatment Outcome , YAP-Signaling Proteins
9.
Oncogene ; 38(16): 2937-2952, 2019 04.
Article in English | MEDLINE | ID: mdl-30568224

ABSTRACT

Alternative splicing is dysregulated in cancer cells, driving the production of isoforms that allow tumor cells to survive and continuously proliferate. Part of the reactivation of telomerase involves the splicing of hTERT transcripts to produce full-length (FL) TERT. Very few splicing factors to date have been described to interact with hTERT and promote the production of FL TERT. We recently described one such splicing factor, NOVA1, that acts as an enhancer of FL hTERT splicing, increases telomerase activity, and promotes telomere maintenance in cancer cells. NOVA1 is expressed primarily in neurons and is involved in neurogenesis. In the present studies, we describe that polypyrimidine-tract binding proteins (PTBPs), which are also typically involved in neurogenesis, are also participating in the splicing of hTERT to FL in cancer. Knockdown experiments of PTBP1 in cancer cells indicate that PTBP1 reduces hTERT FL splicing and telomerase activity. Stable knockdown of PTBP1 results in progressively shortened telomere length in H1299 and H920 lung cancer cells. RNA pulldown experiments reveal that PTBP1 interacts with hTERT pre-mRNA in a NOVA1 dependent fashion. Knockdown of PTBP1 increases the expression of PTBP2 which also interacts with NOVA1, potentially preventing the association of NOVA1 with hTERT pre-mRNA. These new data highlight that splicing in cancer cells is regulated by competition for splice sites and that combinations of splicing factors interact at cis regulatory sites on pre-mRNA transcripts. By employing hTERT as a model gene, we show the coordination of the splicing factors NOVA1 and PTBP1 in cancer by regulating telomerase that is expressed in the vast majority of cancer cell types.


Subject(s)
Heterogeneous-Nuclear Ribonucleoproteins/genetics , Neoplasms/genetics , Polypyrimidine Tract-Binding Protein/genetics , RNA Precursors/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Telomerase/genetics , A549 Cells , Alternative Splicing/genetics , Cell Line , Cell Line, Tumor , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Neuro-Oncological Ventral Antigen , RNA Splicing/genetics
10.
Nat Commun ; 9(1): 3112, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30082712

ABSTRACT

Alternative splicing is dysregulated in cancer and the reactivation of telomerase involves the splicing of TERT transcripts to produce full-length (FL) TERT. Knowledge about the splicing factors that enhance or silence FL hTERT is lacking. We identified splicing factors that reduced telomerase activity and shortened telomeres using a siRNA minigene reporter screen and a lung cancer cell bioinformatics approach. A lead candidate, NOVA1, when knocked down resulted in a shift in hTERT splicing to non-catalytic isoforms, reduced telomerase activity, and progressive telomere shortening. NOVA1 knockdown also significantly altered cancer cell growth in vitro and in xenografts. Genome engineering experiments reveal that NOVA1 promotes the inclusion of exons in the reverse transcriptase domain of hTERT resulting in the production of FL hTERT transcripts. Utilizing hTERT splicing as a model splicing event in cancer may provide new insights into potentially targetable dysregulated splicing factors in cancer.


Subject(s)
Alternative Splicing , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , RNA-Binding Proteins/genetics , Telomerase/genetics , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Computational Biology , Gene Deletion , Gene Silencing , Genetic Engineering , Genome, Human , HeLa Cells , Humans , Lung Neoplasms/metabolism , Mice , Mutation , Neoplasm Transplantation , Neuro-Oncological Ventral Antigen , Phenotype , Protein Binding , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Telomerase/metabolism , Telomere/ultrastructure
11.
Neoplasia ; 20(8): 826-837, 2018 08.
Article in English | MEDLINE | ID: mdl-30015158

ABSTRACT

Standard and targeted cancer therapies for late-stage cancer patients almost universally fail due to tumor heterogeneity/plasticity and intrinsic or acquired drug resistance. We used the telomerase substrate nucleoside precursor, 6-thio-2'-deoxyguanosine (6-thio-dG), to target telomerase-expressing non-small cell lung cancer cells resistant to EGFR-inhibitors and commonly used chemotherapy combinations. Colony formation assays, human xenografts as well as syngeneic and genetically engineered immune competent mouse models of lung cancer were used to test the effect of 6-thio-dG on targeted therapy- and chemotherapy-resistant lung cancer human cells and mouse models. We observed that erlotinib-, paclitaxel/carboplatin-, and gemcitabine/cisplatin-resistant cells were highly sensitive to 6-thio-dG in cell culture and in mouse models. 6-thio-dG, with a known mechanism of action, is a potential novel therapeutic approach to prolong disease control of therapy-resistant lung cancer patients with minimal toxicities.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Telomerase/metabolism , Animals , Cell Line, Tumor , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/pharmacology , Female , Humans , Mice , Mice, Nude , Thionucleosides/pharmacology , Xenograft Model Antitumor Assays/methods
12.
Mol Cell Biol ; 38(17)2018 09 01.
Article in English | MEDLINE | ID: mdl-29866653

ABSTRACT

Adenomatous polyposis coli (APC) is a key molecule to maintain cellular homeostasis in colonic epithelium by regulating cell-cell adhesion, cell polarity, and cell migration through activating the APC-stimulated guanine nucleotide-exchange factor (Asef). The APC-activated Asef stimulates the small GTPase, which leads to decreased cell-cell adherence and cell polarity, and enhanced cell migration. In colorectal cancers, while truncated APC constitutively activates Asef and promotes cancer initiation and progression, regulation of Asef by full-length APC is still unclear. Here, we report the autoinhibition mechanism of full-length APC. We found that the armadillo repeats in full-length APC interact with the APC residues 1362 to 1540 (APC-2,3 repeats), and this interaction competes off and inhibits Asef. Deletion of APC-2,3 repeats permits Asef interactions leading to downstream signaling events, including the induction of Golgi fragmentation through the activation of the Asef-ROCK-MLC2. Truncated APC also disrupts protein trafficking and cholesterol homeostasis by inhibition of SREBP2 activity in a Golgi fragmentation-dependent manner. Our study thus uncovers the autoinhibition mechanism of full-length APC and a novel gain of function of truncated APC in regulating Golgi structure, as well as cholesterol homeostasis, which provides a potential target for pharmaceutical intervention against colon cancers.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Gain of Function Mutation , Genes, APC , Golgi Apparatus/metabolism , Adenomatous Polyposis Coli Protein/chemistry , Amino Acid Sequence , Armadillo Domain Proteins/chemistry , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Binding Sites , Cell Line, Tumor , Cell Movement , Cholesterol/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Golgi Apparatus/pathology , HCT116 Cells , HT29 Cells , Homeostasis , Humans , Models, Biological , Models, Molecular , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Sequence Deletion , Signal Transduction
13.
Methods Mol Biol ; 1768: 513-529, 2018.
Article in English | MEDLINE | ID: mdl-29717462

ABSTRACT

Telomerase is a cellular RNA template-dependent reverse transcriptase that adds telomere repeats to the 3' ends of chromosomes. Telomerase is expressed almost universally in tumor cells (>85%) to maintain telomere length, thus providing the ability of tumor cells to avoid senescence and to have unlimited replication ability, one of the key hallmarks of cancer. ddTRAP (droplet digital Telomere Repeat Amplification Protocol) is a two-step assay with whole cell lysates that utilizes a telomerase-mediated primer extension followed by droplet digital PCR (ddPCR) detection of extended products. The adoptation of the TRAP assay to ddPCR has resulted in improved throughput, increased sensitivity and better repeatability of the TRAP assay. The protocol described below details our procedures for ddTRAP.


Subject(s)
Biomarkers, Tumor/isolation & purification , DNA, Neoplasm/isolation & purification , Neoplasms/diagnosis , Polymerase Chain Reaction/methods , Telomerase/metabolism , Biomarkers, Tumor/genetics , Cell Line, Tumor , DNA Primers/genetics , DNA, Neoplasm/genetics , Enzyme Assays/instrumentation , Enzyme Assays/methods , HEK293 Cells , Humans , Neoplasms/genetics , Neoplasms/pathology , Polymerase Chain Reaction/instrumentation , Telomere/genetics , Telomere/metabolism
14.
Article in English | MEDLINE | ID: mdl-29335378

ABSTRACT

The strengths and limitations of the major methods developed to measure telomere lengths (TLs) in cells and tissues are presented in this review. These include Q-PCR (Quantitative Polymerase Chain Reaction), TRF (Terminal Restriction Fragment) analysis, a variety of Q-FISH (Quantitative Fluorescence In Situ Hybridization) methods, STELA (Single TElomere Length Analysis) and TeSLA (Telomere Shortest Length Assay). For each method, we will cover information about validation studies, including reproducibility in independent laboratories, accuracy, reliability and sensitivity for measuring not only the average but also the shortest telomeres. There is substantial evidence that it is the shortest telomeres that trigger DNA damage responses leading to replicative senescence in mammals. However, the most commonly used TL measurement methods generally provide information on average or relative TL, but it is the shortest telomeres that leads to telomere dysfunction (identified by TIF, Telomere dysfunction Induced Foci) and limit cell proliferation in the absence of a telomere maintenance mechanism, such as telomerase. As the length of the shortest telomeres is a key biomarker determining cell fate and the onset of senescence, a new technique (TeSLA) that provides quantitative information about all the shortest telomeres will be highlighted.This article is part of the theme issue 'Understanding diversity in telomere dynamics'.


Subject(s)
In Situ Hybridization, Fluorescence , Real-Time Polymerase Chain Reaction , Telomere Homeostasis , Telomere/genetics , Animals , Cellular Senescence/genetics , Humans , Reproducibility of Results , Sensitivity and Specificity , Validation Studies as Topic
15.
Tissue Eng Part A ; 24(7-8): 559-568, 2018 04.
Article in English | MEDLINE | ID: mdl-28726588

ABSTRACT

We developed methods for conditionally reprogramming (CR) primary human bronchial epithelial cells (HBECs) to extend their functional lifespan and permit their differentiation into both upper and lower airway lung epithelium. We also developed a bioreactor to support vascular perfusion and rhythmic breathing of decellularized mouse lungs reconstituted with CR HBECs isolated from patients with and without cystic fibrosis (CF). While conditionally reprogrammed cells only differentiate into an upper airway epithelium after 35 days at the air-liquid interface, in reconstituted lungs these cells differentiate into upper airway bronchial epithelium and lower airway alveolar structures after 12 days. Rapid scale-up and the ability to obtain clonal derivatives of primary patient-derived HBECs without the need for genetic manipulation may permit rapid reconstitution of the lung epithelium; facilitating the study of lung disease in tissue-engineered models.


Subject(s)
Bronchi/cytology , Epithelial Cells/cytology , Lung/cytology , Animals , Cell Differentiation/physiology , Cells, Cultured , Cystic Fibrosis/metabolism , Epithelial Cells/metabolism , Mice , Tissue Engineering
16.
Nat Commun ; 8(1): 1356, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29116081

ABSTRACT

Improved methods to measure the shortest (not just average) telomere lengths (TLs) are needed. We developed Telomere Shortest Length Assay (TeSLA), a technique that detects telomeres from all chromosome ends from <1 kb to 18 kb using small amounts of input DNA. TeSLA improves the specificity and efficiency of TL measurements that is facilitated by user friendly image-processing software to automatically detect and annotate band sizes, calculate average TL, as well as the percent of the shortest telomeres. Compared with other TL measurement methods, TeSLA provides more information about the shortest telomeres. The length of telomeres was measured longitudinally in peripheral blood mononuclear cells during human aging, in tissues during colon cancer progression, in telomere-related diseases such as idiopathic pulmonary fibrosis, as well as in mice and other organisms. The results indicate that TeSLA is a robust method that provides a better understanding of the shortest length of telomeres.


Subject(s)
Aging/genetics , Neoplasms/genetics , Nucleic Acid Amplification Techniques/methods , Telomere/genetics , Adult , Aged , Animals , Blotting, Southern , Female , HeLa Cells , Humans , Image Processing, Computer-Assisted , In Situ Hybridization, Fluorescence/methods , Male , Mice , Middle Aged , NIH 3T3 Cells , Software
17.
Mol Cell Biol ; 37(20)2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28760773

ABSTRACT

Alternative lengthening of telomeres (ALT) is a telomerase-independent telomere maintenance mechanism that occurs in a subset of cancers. By analyzing telomerase-positive cells and their human TERC knockout-derived ALT human cell lines, we show that ALT cells harbor more fragile telomeres representing telomere replication problems. ALT-associated replication defects trigger mitotic DNA synthesis (MiDAS) at telomeres in a RAD52-dependent, but RAD51-independent, manner. Telomeric MiDAS is a conservative DNA synthesis process, potentially mediated by break-induced replication, similar to type II ALT survivors in Saccharomyces cerevisiae Replication stresses induced by ectopic oncogenic expression of cyclin E, G-quadruplexes, or R-loop formation facilitate the ALT pathway and lead to telomere clustering, a hallmark of ALT cancers. The TIMELESS/TIPIN complex suppresses telomere clustering and telomeric MiDAS, whereas the SMC5/6 complex promotes them. In summary, ALT cells exhibit more telomere replication defects that result in persistent DNA damage responses at telomeres, leading to the engagement of telomeric MiDAS (spontaneous mitotic telomere synthesis) that is triggered by DNA replication stress, a potential driver of genomic duplications in cancer.


Subject(s)
DNA Repair/physiology , DNA/metabolism , Rad52 DNA Repair and Recombination Protein/metabolism , Telomerase/metabolism , Telomere Homeostasis/physiology , Telomere/metabolism , Cell Line , DNA Replication/physiology , Humans , Recombination, Genetic/genetics
18.
Sci Rep ; 7(1): 6785, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28754961

ABSTRACT

Telomerase activity is not readily detected in resting human T lymphocytes, however upon antigen presentation, telomerase is transiently upregulated. Presently, it is not known if telomerase activation is necessary for the proliferation of T cells or for the maintenance of telomere lengths. In this study, we found that telomerase activation is not required for the short- term proliferation of T cells and that telomeres progressively shorten in a heterogeneous population of T cells, even if telomerase is detected. By measuring telomerase activity at the single-cell level using quantitative ddPCR techniques (ddTRAP) and by monitoring changes in the shortest telomeres with more sensitive telomere length measurement assays, we show that only a subset of CD28+ T-cells have robust telomerase activity upon stimulation and are capable of maintaining their telomere lengths during induced proliferation. The study of this T-cell subset may lead to a better understanding on how telomerase is regulated and functions in immune cells.


Subject(s)
Lymphocyte Activation , T-Lymphocytes/immunology , Telomere Homeostasis , Adult , CD28 Antigens/genetics , CD28 Antigens/metabolism , Cells, Cultured , Humans , Telomerase/metabolism
19.
Methods Mol Biol ; 1587: 55-62, 2017.
Article in English | MEDLINE | ID: mdl-28324497

ABSTRACT

Telomeres terminate in 3' single-stranded G-rich overhangs that function in telomere end protection and telomerase action. An accurate measurement of overhang length is challenging due to the presence of many kilobases of double-stranded telomere DNA. Here, a simple method is described that utilizes duplex specific nuclease (DSN) to digest all double-stranded DNA including telomeres to fragments of <10 bp, leaving the single-stranded overhangs intact. Their length can then be determined by Southern blot using super-sensitive telomere C-rich probes.


Subject(s)
DNA Replication/genetics , Deoxyribonucleases/genetics , Telomere/genetics , Animals , Blotting, Southern/methods , Cells, Cultured , DNA/genetics , DNA, Single-Stranded/genetics , Mammals , Telomerase/genetics
20.
Methods Mol Biol ; 1587: 71-82, 2017.
Article in English | MEDLINE | ID: mdl-28324499

ABSTRACT

Telomerase is present in most human cancers, and proliferative stem cells including germline cells. Telomerase plays an essential role in tumorigenesis by maintaining/elongating telomeric DNA, and thus preventing the telomere shortening that results in replicative senescence. Understanding telomerase action in vivo has important implication for both cancer and aging, but there are not robust methods for monitoring telomerase action. By combining a series of cell biological and biochemical approaches, and taking advantage of the enzyme DSN that specifically cuts double-stranded DNA and releases the telomeric overhangs, we have developed a method to monitor telomerase action during one cell cycle. Here, we describe this method using Hela carcinoma cells as an example.


Subject(s)
DNA/genetics , Telomere/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cellular Senescence/genetics , HeLa Cells , Humans , Telomerase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...