Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Am J Hum Genet ; 110(10): 1787-1803, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37751738

ABSTRACT

Congenital diaphragmatic hernia (CDH) is a relatively common and genetically heterogeneous structural birth defect associated with high mortality and morbidity. We describe eight unrelated families with an X-linked condition characterized by diaphragm defects, variable anterior body-wall anomalies, and/or facial dysmorphism. Using linkage analysis and exome or genome sequencing, we found that missense variants in plastin 3 (PLS3), a gene encoding an actin bundling protein, co-segregate with disease in all families. Loss-of-function variants in PLS3 have been previously associated with X-linked osteoporosis (MIM: 300910), so we used in silico protein modeling and a mouse model to address these seemingly disparate clinical phenotypes. The missense variants in individuals with CDH are located within the actin-binding domains of the protein but are not predicted to affect protein structure, whereas the variants in individuals with osteoporosis are predicted to result in loss of function. A mouse knockin model of a variant identified in one of the CDH-affected families, c.1497G>C (p.Trp499Cys), shows partial perinatal lethality and recapitulates the key findings of the human phenotype, including diaphragm and abdominal-wall defects. Both the mouse model and one adult human male with a CDH-associated PLS3 variant were observed to have increased rather than decreased bone mineral density. Together, these clinical and functional data in humans and mice reveal that specific missense variants affecting the actin-binding domains of PLS3 might have a gain-of-function effect and cause a Mendelian congenital disorder.


Subject(s)
Hernias, Diaphragmatic, Congenital , Osteoporosis , Adult , Humans , Male , Animals , Mice , Hernias, Diaphragmatic, Congenital/genetics , Actins/genetics , Mutation, Missense/genetics , Osteoporosis/genetics
2.
Nature ; 471(7336): 104-9, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21368833

ABSTRACT

The effective use of targeted therapy is highly dependent on the identification of responder patient populations. Loss of FBW7, which encodes a tumour-suppressor protein, is frequently found in various types of human cancer, including breast cancer, colon cancer and T-cell acute lymphoblastic leukaemia (T-ALL). In line with these genomic data, engineered deletion of Fbw7 in mouse T cells results in T-ALL, validating FBW7 as a T-ALL tumour suppressor. Determining the precise molecular mechanisms by which FBW7 exerts antitumour activity is an area of intensive investigation. These mechanisms are thought to relate in part to FBW7-mediated destruction of key proteins relevant to cancer, including Jun, Myc, cyclin E and notch 1 (ref. 9), all of which have oncoprotein activity and are overexpressed in various human cancers, including leukaemia. In addition to accelerating cell growth, overexpression of Jun, Myc or notch 1 can also induce programmed cell death. Thus, considerable uncertainty surrounds how FBW7-deficient cells evade cell death in the setting of upregulated Jun, Myc and/or notch 1. Here we show that the E3 ubiquitin ligase SCF(FBW7) (a SKP1-cullin-1-F-box complex that contains FBW7 as the F-box protein) governs cellular apoptosis by targeting MCL1, a pro-survival BCL2 family member, for ubiquitylation and destruction in a manner that depends on phosphorylation by glycogen synthase kinase 3. Human T-ALL cell lines showed a close relationship between FBW7 loss and MCL1 overexpression. Correspondingly, T-ALL cell lines with defective FBW7 are particularly sensitive to the multi-kinase inhibitor sorafenib but resistant to the BCL2 antagonist ABT-737. On the genetic level, FBW7 reconstitution or MCL1 depletion restores sensitivity to ABT-737, establishing MCL1 as a therapeutically relevant bypass survival mechanism that enables FBW7-deficient cells to evade apoptosis. Therefore, our work provides insight into the molecular mechanism of direct tumour suppression by FBW7 and has implications for the targeted treatment of patients with FBW7-deficient T-ALL.


Subject(s)
Apoptosis , Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , SKP Cullin F-Box Protein Ligases/chemistry , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Amino Acid Sequence , Animals , Apoptosis/drug effects , Benzenesulfonates/pharmacology , Biphenyl Compounds/pharmacology , Cell Cycle Proteins/genetics , Cell Line, Tumor , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Glycogen Synthase Kinase 3/metabolism , Humans , Mice , Molecular Sequence Data , Myeloid Cell Leukemia Sequence 1 Protein , Niacinamide/analogs & derivatives , Nitrophenols/pharmacology , Phenylurea Compounds , Phosphorylation , Piperazines/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Binding/drug effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Pyridines/pharmacology , Sorafenib , Sulfonamides/pharmacology , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects
3.
Nature ; 470(7334): 359-65, 2011 Feb 17.
Article in English | MEDLINE | ID: mdl-21307849

ABSTRACT

Telomere dysfunction activates p53-mediated cellular growth arrest, senescence and apoptosis to drive progressive atrophy and functional decline in high-turnover tissues. The broader adverse impact of telomere dysfunction across many tissues including more quiescent systems prompted transcriptomic network analyses to identify common mechanisms operative in haematopoietic stem cells, heart and liver. These unbiased studies revealed profound repression of peroxisome proliferator-activated receptor gamma, coactivator 1 alpha and beta (PGC-1α and PGC-1ß, also known as Ppargc1a and Ppargc1b, respectively) and the downstream network in mice null for either telomerase reverse transcriptase (Tert) or telomerase RNA component (Terc) genes. Consistent with PGCs as master regulators of mitochondrial physiology and metabolism, telomere dysfunction is associated with impaired mitochondrial biogenesis and function, decreased gluconeogenesis, cardiomyopathy, and increased reactive oxygen species. In the setting of telomere dysfunction, enforced Tert or PGC-1α expression or germline deletion of p53 (also known as Trp53) substantially restores PGC network expression, mitochondrial respiration, cardiac function and gluconeogenesis. We demonstrate that telomere dysfunction activates p53 which in turn binds and represses PGC-1α and PGC-1ß promoters, thereby forging a direct link between telomere and mitochondrial biology. We propose that this telomere-p53-PGC axis contributes to organ and metabolic failure and to diminishing organismal fitness in the setting of telomere dysfunction.


Subject(s)
Mitochondria/metabolism , Mitochondria/pathology , Telomere/metabolism , Telomere/pathology , Adenosine Triphosphate/biosynthesis , Aging/metabolism , Aging/pathology , Animals , Cardiomyopathies/chemically induced , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Cell Proliferation , DNA, Mitochondrial/analysis , Doxorubicin/toxicity , Gluconeogenesis , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Liver/cytology , Liver/metabolism , Mice , Myocardium/cytology , Myocardium/metabolism , RNA/genetics , Reactive Oxygen Species/metabolism , Telomerase/deficiency , Telomerase/genetics , Telomere/enzymology , Telomere/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Proc Natl Acad Sci U S A ; 111(21): E2200-9, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24825892

ABSTRACT

The rhomboid 5 homolog 2 (Rhbdf2) gene encodes an inactive rhomboid (iRhom) protease, iRhom2, one of a family of enzymes containing a long cytosolic N terminus and a dormant peptidase domain of unknown function. iRhom2 has been implicated in epithelial regeneration and cancer growth through constitutive activation of epidermal growth factor receptor (EGFR) signaling. However, little is known about the physiological substrates for iRhom2 or the molecular mechanisms underlying these functions. We show that iRhom2 is a short-lived protein whose stability can be increased by select mutations in the N-terminal domain. In turn, these stable variants function to augment the secretion of EGF family ligands, including amphiregulin, independent of metalloprotease a disintegrin and metalloproteinase 17 (ADAM17) activity. In vivo, N-terminal iRhom2 mutations induce accelerated wound healing as well as accelerated tumorigenesis, but they do not drive spontaneous tumor development. This work underscores the physiological prominence of iRhom2 in controlling EGFR signaling events involved in wound healing and neoplastic growth, and yields insight into the function of key iRhom2 domains.


Subject(s)
Carrier Proteins/genetics , ErbB Receptors/metabolism , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Protein Stability , Signal Transduction/physiology , Amphiregulin , Animals , COS Cells , Chlorocebus aethiops , Cloning, Molecular , EGF Family of Proteins , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Kaplan-Meier Estimate , Mice , Mutagenesis , Mutation/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Wound Healing/genetics
5.
Nature ; 447(7147): 966-71, 2007 Jun 21.
Article in English | MEDLINE | ID: mdl-17515920

ABSTRACT

Highly rearranged and mutated cancer genomes present major challenges in the identification of pathogenetic events driving the neoplastic transformation process. Here we engineered lymphoma-prone mice with chromosomal instability to assess the usefulness of mouse models in cancer gene discovery and the extent of cross-species overlap in cancer-associated copy number aberrations. Along with targeted re-sequencing, our comparative oncogenomic studies identified FBXW7 and PTEN to be commonly deleted both in murine lymphomas and in human T-cell acute lymphoblastic leukaemia/lymphoma (T-ALL). The murine cancers acquire widespread recurrent amplifications and deletions targeting loci syntenic to those not only in human T-ALL but also in diverse human haematopoietic, mesenchymal and epithelial tumours. These results indicate that murine and human tumours experience common biological processes driven by orthologous genetic events in their malignant evolution. The highly concordant nature of genomic events encourages the use of genomically unstable murine cancer models in the discovery of biological driver events in the human oncogenome.


Subject(s)
Chromosomal Instability/genetics , Chromosome Aberrations , Conserved Sequence/genetics , Leukemia-Lymphoma, Adult T-Cell/genetics , Lymphoma, T-Cell/genetics , Animals , Genome/genetics , Humans , Mice , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Synteny/genetics
6.
Cancer Cell ; 3(1): 4-6, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12559169

ABSTRACT

The analysis of compound mouse mutants for nonhomologous end-joining DNA double-strand break repair and those deficient for the p53 checkpoint pathway has provided a fascinating look at the carcinogenic consequences of the failure to properly repair DNA damage and to elicit appropriate checkpoints.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA Damage , DNA Repair , Neoplasms/etiology , Animals , Chromosome Aberrations , Genes, p53/genetics , Humans , Mice , Mice, Transgenic , Models, Animal
7.
Cancer Cell ; 2(2): 149-55, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12204535

ABSTRACT

Telomere dysfunction and associated fusion-breakage in the mouse encourages epithelial carcinogenesis and a more humanized genomic profile that includes nonreciprocal translocations (NRTs). Here, array comparative genomic hybridization was used to determine the pathogenic significance of NRTs and to determine whether telomere dysfunction also drives amplifications and deletions of cancer-relevant loci. Compared to tumors arising in mice with intact telomeres, tumors with telomere dysfunction possessed higher levels of genomic instability and showed numerous amplifications and deletions in regions syntenic to human cancer hotspots. These observations suggest that telomere-based crisis provides a mechanism of chromosomal instability, including regional amplifications and deletions, that drives carcinogenesis. This model provides a platform for discovery of genes responsible for the major cancers affecting aged humans.


Subject(s)
Chromosomes, Mammalian/genetics , Gene Amplification , Gene Deletion , Neoplasms/genetics , Telomere/metabolism , Animals , Chromosome Aberrations , DNA, Neoplasm/genetics , Genes, p53 , Genome , Humans , Mice , RNA/genetics , Synteny , Telomerase/genetics , Telomere/genetics
8.
Mol Cell Biol ; 27(6): 2253-65, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17145779

ABSTRACT

Telomeres are key structural elements for the protection and maintenance of linear chromosomes, and they function to prevent recognition of chromosomal ends as DNA double-stranded breaks. Loss of telomere capping function brought about by telomerase deficiency and gradual erosion of telomere ends or by experimental disruption of higher-order telomere structure culminates in the fusion of defective telomeres and/or the activation of DNA damage checkpoints. Previous work has implicated the nonhomologous end-joining (NHEJ) DNA repair pathway as a critical mediator of these biological processes. Here, employing the telomerase-deficient mouse model, we tested whether the NHEJ component DNA-dependent protein kinase catalytic subunit (DNA-PKcs) was required for fusion of eroded/dysfunctional telomere ends and the telomere checkpoint responses. In late-generation mTerc(-/-) DNA-PKcs(-/-) cells and tissues, chromosomal end-to-end fusions and anaphase bridges were readily evident. Notably, nullizygosity for DNA Ligase4 (Lig4)--an additional crucial NHEJ component--was also permissive for chromosome fusions in mTerc(-/-) cells, indicating that, in contrast to results seen with experimental disruption of telomere structure, telomere dysfunction in the context of gradual telomere erosion can engage additional DNA repair pathways. Furthermore, we found that DNA-PKcs deficiency does not reduce apoptosis, tissue atrophy, or p53 activation in late-generation mTerc(-/-) tissues but rather moderately exacerbates germ cell apoptosis and testicular degeneration. Thus, our studies indicate that the NHEJ components, DNA-PKcs and Lig4, are not required for fusion of critically shortened telomeric ends and that DNA-PKcs is not required for sensing and executing the telomere checkpoint response, findings consistent with the consensus view of the limited role of DNA-PKcs in DNA damage signaling in general.


Subject(s)
Cell Cycle , DNA-Activated Protein Kinase/metabolism , Telomerase/deficiency , Telomerase/metabolism , Telomere/metabolism , Animals , Bone Marrow Cells/metabolism , Catalytic Domain , Chromosomes/genetics , DNA Ligase ATP , DNA Ligases/genetics , DNA-Activated Protein Kinase/genetics , Genomic Instability/genetics , Male , Mice , Mice, Knockout , Mutation/genetics , Protein Binding , Telomerase/genetics , Testis/cytology , Testis/metabolism
9.
Pain ; 160(8): 1740-1753, 2019 08.
Article in English | MEDLINE | ID: mdl-31335644

ABSTRACT

Identification of genetic variants that influence susceptibility to pain is key to identifying molecular mechanisms and targets for effective and safe therapeutic alternatives to opioids. To identify genes and variants associated with persistent pain, we measured late-phase response to formalin injection in 275 male and female Diversity Outbred mice genotyped for over 70,000 single nucleotide polymorphisms. One quantitative trait locus reached genome-wide significance on chromosome 1 with a support interval of 3.1 Mb. This locus, Nociq4 (nociceptive sensitivity quantitative trait locus 4; MGI: 5661503), harbors the well-known pain gene Trpa1 (transient receptor potential cation channel, subfamily A, member 1). Trpa1 is a cation channel known to play an important role in acute and chronic pain in both humans and mice. Analysis of Diversity Outbred founder strain allele effects revealed a significant effect of the CAST/EiJ allele at Trpa1, with CAST/EiJ carrier mice showing an early, but not late, response to formalin relative to carriers of the 7 other inbred founder alleles (A/J, C57BL/6J, 129S1/SvImJ, NOD/ShiLtJ, NZO/HlLtJ, PWK/PhJ, and WSB/EiJ). We characterized possible functional consequences of sequence variants in Trpa1 by assessing channel conductance, TRPA1-TRPV1 interactions, and isoform expression. The phenotypic differences observed in CAST/EiJ relative to C57BL/6J carriers were best explained by Trpa1 isoform expression differences, implicating a splice junction variant as the causal functional variant. This study demonstrates the utility of advanced, high-precision genetic mapping populations in resolving specific molecular mechanisms of variation in pain sensitivity.


Subject(s)
Genetic Variation , Genotype , Nociception/physiology , Pain/genetics , Phenotype , TRPA1 Cation Channel/genetics , Alleles , Animals , Collaborative Cross Mice , Female , Formaldehyde , Male , Mice , Quantitative Trait Loci
10.
Carcinogenesis ; 29(4): 747-53, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18283039

ABSTRACT

Current mouse models of lung cancer recapitulate signature genetic lesions and some phenotypic features of human lung cancer. However, because mice have long telomeres, models to date do not recapitulate the aspects of lung carcinogenesis-telomere attrition and the genomic instability that ensues-believed to serve as key mechanisms driving lung tumor initiation and progression. To explore the contributions of telomere dysfunction to lung cancer progression, we combined a telomerase catalytic subunit (mTerc) mutation with the well-characterized K-rasG12D mouse lung cancer model. K-ras(G12D) mTerc(-/-) mice with telomere dysfunction but intact p53 exhibited increased lung epithelial apoptosis, delayed tumor formation and increased life span relative to K-ras(G12D) mTerc(+/-) mice with intact telomere function. This demonstrates that by itself, telomere dysfunction acts in a tumor-suppressive mechanism. Introduction of a heterozygous p53 mutation exerted a marked histopathological, biological and genomic impact. K-ras(G12D) mTerc(-/-) p53(+/-) mice developed aggressive tumors with more chromosomal instabilities and high metastatic potential, leading to decreased overall survival. Thus, we have generated a murine model that more faithfully recapitulates key aspects of the human disease. Furthermore, these findings clearly demonstrate (in an in vivo model system) the dual nature of telomere shortening as both a tumor-suppressive and tumor-promoting mechanism in lung cancer, dependent on p53 status.


Subject(s)
Genes, ras , Genomic Instability , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Telomere/genetics , Tumor Suppressor Protein p53/genetics , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Mutation , Neoplasm Metastasis
11.
Diabetes ; 67(5): 923-935, 2018 05.
Article in English | MEDLINE | ID: mdl-29472249

ABSTRACT

Improved mouse models for type 1 diabetes (T1D) therapy development are needed. T1D susceptibility is restored to normally resistant NOD.ß2m-/- mice transgenically expressing human disease-associated HLA-A*02:01 or HLA-B*39:06 class I molecules in place of their murine counterparts. T1D is dependent on pathogenic CD8+ T-cell responses mediated by these human class I variants. NOD.ß2m-/--A2.1 mice were previously used to identify ß-cell autoantigens presented by this human class I variant to pathogenic CD8+ T cells and for testing therapies to attenuate such effectors. However, NOD.ß2m-/- mice also lack nonclassical MHC I family members, including FcRn, required for antigen presentation, and maintenance of serum IgG and albumin, precluding therapies dependent on these molecules. Hence, we used CRISPR/Cas9 to directly ablate the NOD H2-Kd and H2-Db classical class I variants either individually or in tandem (cMHCI-/-). Ablation of the H2-Ag7 class II variant in the latter stock created NOD mice totally lacking in classical murine MHC expression (cMHCI/II-/-). NOD-cMHCI-/- mice retained nonclassical MHC I molecule expression and FcRn activity. Transgenic expression of HLA-A2 or -B39 restored pathogenic CD8+ T-cell development and T1D susceptibility to NOD-cMHCI-/- mice. These next-generation HLA-humanized NOD models may provide improved platforms for T1D therapy development.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Disease Models, Animal , HLA-A2 Antigen/genetics , HLA-B Antigens/genetics , Mice , beta 2-Microglobulin/genetics , Animals , CRISPR-Cas Systems , Diabetes Mellitus, Type 1/therapy , Histocompatibility Antigens Class I/genetics , Humans , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic
12.
Biol Psychiatry ; 60(5): 432-5, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16581033

ABSTRACT

BACKGROUND: Little is known about the biological mechanisms underlying the excess medical morbidity and mortality associated with mood disorders. Substantial evidence supports abnormalities in stress-related biological systems in depression. Accelerated telomere shortening may reflect stress-related oxidative damage to cells and accelerated aging, and severe psychosocial stress has been linked to telomere shortening. We propose that chronic stress associated with mood disorders may contribute to excess vulnerability for diseases of aging such as cardiovascular disease and possibly some cancers through accelerated organismal aging. METHODS: Telomere length was measured by Southern Analysis in 44 individuals with chronic mood disorders and 44 nonpsychiatrically ill age-matched control subjects. RESULTS: Telomere length was significantly shorter in those with mood disorders, representing as much as 10 years of accelerated aging. CONCLUSIONS: These results provide preliminary evidence that mood disorders are associated with accelerated aging and may suggest a novel mechanism for mood disorder-associated morbidity and mortality.


Subject(s)
Aging/genetics , Chromosome Breakage , Mood Disorders/genetics , Stress, Psychological/genetics , Telomere , Adult , Anxiety/complications , Anxiety/genetics , Bipolar Disorder/complications , Bipolar Disorder/genetics , Case-Control Studies , Cellular Senescence/genetics , Chronic Disease , Female , Humans , Male , Matched-Pair Analysis , Middle Aged , Models, Genetic , Mood Disorders/complications , Reference Values , Statistics, Nonparametric , Stress, Psychological/etiology
14.
Curr Mol Med ; 5(2): 145-52, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15974867

ABSTRACT

The telomere-based model of cell aging has proven to among been among the most enduring hypotheses in cell biology. This model, suggesting that the gradual loss of telomere sequences during the proliferation of cultured human somatic cells imposes a barrier on cellular replicative potential, has been strongly supported by recent genetic and biochemical studies. In addition, evidence implicating telomere dynamics in organismal ageing and cancer progression in vivo suggest that such a process is likely to have considerable physiological relevance in homeostasis and disease. What is the sensing mechanism for shortened telomeres and what is the molecular basis for the ensuing checkpoint response? Moreover, what is the outcome when such failsafe mechanisms are lost? Here we will review the signaling pathways that are induced by alterations in telomere length and integrity and illustrate how these processes provoke downstream effects on cell proliferation and survival. In addition, we discuss how the telomere-induced pathways intersect with the DNA damage response and document how the failure in either process results in unrestrained chromosomal instability.


Subject(s)
Aging/genetics , Genomic Instability , Telomere/chemistry , Telomere/metabolism , Animals , Humans , Mice , Signal Transduction
15.
Curr Protoc Mouse Biol ; 6(1): 39-66, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26928663

ABSTRACT

The CRISPR-Cas9 system in bacteria and archaea has recently been exploited for genome editing in various model organisms, including mice. The CRISPR-Cas9 reagents can be delivered directly into the mouse zygote to derive a mutant animal carrying targeted genetic modifications. The major components of the system include the guide RNA, which provides target specificity, the Cas9 nuclease that creates the DNA double-strand break, and the donor oligonucleotide or plasmid carrying the intended mutation flanked by sequences homologous to the target site. Here we describe the general considerations and experimental protocols for creating genetically modified mice using the CRISPR-Cas9 system.


Subject(s)
CRISPR-Cas Systems/genetics , Genetic Engineering/methods , Genomics/methods , Models, Animal , Animals , Base Sequence , Genotyping Techniques , Mice , Microinjections , Oligonucleotides , Plasmids/genetics , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Zygote
16.
DNA Repair (Amst) ; 3(8-9): 979-88, 2004.
Article in English | MEDLINE | ID: mdl-15279784

ABSTRACT

DNA double strand breaks (DSBs) are repaired by an extensive network of proteins that recognize damaged DNA and catalyze its repair. By virtue of their similarity, the normal ends of linear chromosomes and internal DNA DSBs are both potential substrates for DSB repair enzymes. Thus, telomeres, specialized nucleo-protein complexes that cap chromosomal ends, serve a critical function to differentiate themselves from internal DNA strand breaks, and as a result prevent genomic instability that can result from their inappropriate involvement in repair reactions. Telomeres that become critically short due to failure of telomere maintenance mechanisms, or which become dysfunctional by loss of telomere binding proteins, elicit extensive checkpoint responses that in normal cells blocks proliferation. In this situation, the DNA DSB repair machinery plays a major role in responding to these "damaged" telomeres - creating chromosome fusions or capturing telomeres from other chromosomes in an effort to rid the cell of the perceived damage. However, a surprising aspect of telomere maintenance is that many of the same proteins that facilitate this repair of damaged telomeres are also necessary for their proper integrity. Here, we review recent work defining the roles for DSB repair machinery in telomere maintenance and in response to telomere dysfunction.


Subject(s)
DNA Damage , Recombination, Genetic , Saccharomyces cerevisiae/genetics , Telomere/ultrastructure , DNA Repair , Endodeoxyribonucleases/physiology , Exodeoxyribonucleases/physiology , Fungal Proteins/physiology , Humans , Intracellular Signaling Peptides and Proteins , Models, Biological , Models, Genetic , Mutation , Phenotype , Protein Serine-Threonine Kinases , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/physiology
17.
J Exp Med ; 204(13): 3059-66, 2007 Dec 24.
Article in English | MEDLINE | ID: mdl-18070937

ABSTRACT

Recent studies have demonstrated that the MYB oncogene is frequently duplicated in human T cell acute lymphoblastic leukemia (T-ALL). We find that the human MYB locus is flanked by 257-bp Alu repeats and that the duplication is mediated somatically by homologous recombination between the flanking Alu elements on sister chromatids. Nested long-range PCR analysis indicated a low frequency of homologous recombination leading to MYB tandem duplication in the peripheral blood mononuclear cells of approximately 50% of healthy individuals, none of whom had a MYB duplication in the germline. We conclude that Alu-mediated MYB tandem duplication occurs at low frequency during normal thymocyte development and is clonally selected during the molecular pathogenesis of human T-ALL.


Subject(s)
Alu Elements/genetics , Gene Duplication , Leukemia-Lymphoma, Adult T-Cell/genetics , Proto-Oncogene Proteins c-myb/genetics , Base Sequence , Humans , In Situ Hybridization, Fluorescence , Models, Biological , Models, Genetic , Molecular Sequence Data , Nucleic Acid Hybridization , Polymerase Chain Reaction , Recombination, Genetic
18.
Science ; 297(5581): 565-9, 2002 Jul 26.
Article in English | MEDLINE | ID: mdl-12142527

ABSTRACT

Cancer is a disease of impaired genome stability. The molecular forces that maintain genome integrity and sense altered chromosome structure are invariably subverted in cancer cells. Here, we explore the contrasting contributions of telomeres in the initiation and suppression of cancer and review the evidence supporting a role for telomere dysfunction as a mechanism driving the radical chromosomal aberrations that typify cancer genomes. Recent work suggests that passage of cells through crisis in the setting of deactivated DNA damage checkpoints provides a mutational mechanism that can generate the diverse genetic alterations required for cancer initiation. A greater understanding of telomere-induced crisis and the cell's crisis management mechanisms should guide the rational development of new therapeutics for cancer and other disorders.


Subject(s)
Cell Transformation, Neoplastic , Neoplasms/genetics , Neoplasms/physiopathology , Telomerase/metabolism , Telomere/physiology , Animals , Cell Cycle , Cell Division , Cells, Cultured , DNA Damage , DNA Repair , Disease Progression , Genetic Therapy , Humans , Neoplasms/pathology , Signal Transduction , Telomerase/antagonists & inhibitors , Telomere/ultrastructure
19.
Genes Dev ; 17(1): 88-100, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12514102

ABSTRACT

Telomerase activation is a common feature of most advanced human cancers and is postulated to restore genomic stability to a level permissive for cell viability and tumor progression. Here, we used genetically defined transformed mouse embryonic fibroblast (MEF) cultures derived from late generation mTerc(-/-) Ink4a/Arf(-/-) mice to explore more directly how telomere-based crisis relates to the evolution of cancer cell genomes and to tumor biology. An exhaustive serial analysis of cytogenetic profiles over extensive passage in culture revealed that the emergence of chromosomal fusions (including dicentrics) coincided with onset of deletions and complex nonreciprocal translocations (NRTs), whereas mTerc-transduced cultures maintained intact chromosomes and stable genomes. Despite a high degree of telomere dysfunction and genomic instability, transformed late passage mTerc(-/-) Ink4a/Arf(-/-) cultures retained the capacity to form subcutaneous tumors in immunocompromised mice. However, even moderate levels of telomere dysfunction completely abrogated the capacity of these cells to form lung metastases after tail-vein injection, whereas mTerc reconstitution alone conferred robust metastatic activity in these cells. Finally, serial subcutaneous tumor formation using late passage transformed mTerc(-/-) Ink4a/Arf(-/-) cultures revealed clear evidence of telomerase-independent alternative lengthening of telomeres (ALT). Significantly, despite a marked increase in telomere reserve, cells derived from the ALT+ subcutaneous tumors were unable to generate lung metastases, indicating in vivo functional differences in these principal mechanisms of telomere maintenance. Together, these results are consistent with the model that although telomere dysfunction provokes chromosomal aberrations that initiate carcinogenesis, telomerase-mediated telomere maintenance enables such initiated cells to efficiently achieve a fully malignant endpoint, including metastasis.


Subject(s)
Neoplasm Proteins/physiology , Telomerase/physiology , Telomere/physiology , Animals , Cell Line, Transformed/enzymology , Cell Line, Transformed/pathology , Cell Line, Transformed/transplantation , Chromosome Aberrations , Chromosome Painting , Chromosomes/ultrastructure , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Embryo, Mammalian/cytology , Fibroblasts/enzymology , Fibroblasts/pathology , Fibroblasts/transplantation , In Situ Hybridization, Fluorescence , Lung Neoplasms/secondary , Mice , Mice, Knockout , Mice, SCID , Neoplasm Transplantation , RNA/genetics , Telomerase/deficiency , Telomerase/genetics
20.
Nature ; 421(6923): 643-8, 2003 Feb 06.
Article in English | MEDLINE | ID: mdl-12540856

ABSTRACT

Ataxia-telangiectasia (A-T) results from the loss of ataxia-telangiectasia mutated (Atm) function and is characterized by accelerated telomere loss, genomic instability, progressive neurological degeneration, premature ageing and increased neoplasia incidence. Here we evaluate the functional interaction of Atm and telomeres in vivo. We examined the impact of Atm deficiency as a function of progressive telomere attrition at both the cellular and whole-organism level in mice doubly null for Atm and the telomerase RNA component (Terc). These compound mutants showed increased telomere erosion and genomic instability, yet they experienced a substantial elimination of T-cell lymphomas associated with Atm deficiency. A generalized proliferation defect was evident in all cell types and tissues examined, and this defect extended to tissue stem/progenitor cell compartments, thereby providing a basis for progressive multi-organ system compromise, accelerated ageing and premature death. We show that Atm deficiency and telomere dysfunction act together to impair cellular and whole-organism viability, thus supporting the view that aspects of A-T pathophysiology are linked to the functional state of telomeres and its adverse effects on stem/progenitor cell reserves.


Subject(s)
Aging/genetics , Homeostasis/genetics , Protein Serine-Threonine Kinases/genetics , RNA/genetics , Telomerase/genetics , Telomere/metabolism , Alopecia/genetics , Anaphase , Animals , Apoptosis , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , Cell Division , Cells, Cultured , DNA-Binding Proteins , Fibroblasts , Hair Color/genetics , Lymphoma, T-Cell/genetics , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/deficiency , Stem Cells/cytology , Stem Cells/metabolism , Survival Rate , Telomere/enzymology , Telomere/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins , Wound Healing/genetics
SELECTION OF CITATIONS
SEARCH DETAIL