Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Publication year range
1.
J Immunol ; 209(3): 606-620, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35817516

ABSTRACT

Despite recent therapeutic progress, advanced melanoma remains lethal for many patients. The composition of the immune tumor microenvironment (TME) has decisive impacts on therapy response and disease outcome, and high-dimensional analyses of patient samples reveal the heterogeneity of the immune TME. Macrophages infiltrate TMEs and generally associate with tumor progression, but the underlying mechanisms are incompletely understood. Because experimental systems are needed to elucidate the functional properties of these cells, we developed a humanized mouse model reconstituted with human immune cells and human melanoma. We used two strains of recipient mice, supporting or not supporting the development of human myeloid cells. We found that human myeloid cells favored metastatic spread of the primary tumor, thereby recapitulating the cancer-supportive role of macrophages. We next analyzed the transcriptome of human immune cells infiltrating tumors versus other tissues. This analysis identified a cluster of myeloid cells present in the TME, but not in other tissues, which do not correspond to canonical M2 cells. The transcriptome of these cells is characterized by high expression of glycolytic enzymes and multiple chemokines and by low expression of gene sets associated with inflammation and adaptive immunity. Compared with humanized mouse results, we found transcriptionally similar myeloid cells in patient-derived samples of melanoma and other cancer types. The humanized mouse model described here thus complements patient sample analyses, enabling further elucidation of fundamental principles in melanoma biology beyond M1/M2 macrophage polarization. The model can also support the development and evaluation of candidate antitumor therapies.


Subject(s)
Macrophages , Melanoma , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Macrophage Activation , Melanoma/pathology , Mice , Tumor Microenvironment
2.
Nat Methods ; 14(9): 891-896, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28737741

ABSTRACT

We developed a chemically inducible Cas9 (ciCas9) and a droplet digital PCR assay for double-strand breaks (DSB-ddPCR) to investigate the kinetics of Cas9-mediated generation and repair of DSBs in cells. ciCas9 is a rapidly activated, single-component Cas9 variant engineered by replacing the protein's REC2 domain with the BCL-xL protein and fusing an interacting BH3 peptide to the C terminus. ciCas9 can be tunably activated by a compound that disrupts the BCL-xL-BH3 interaction within minutes. DSB-ddPCR demonstrates time-resolved, highly quantitative, and targeted measurement of DSBs. Combining these tools facilitated an unprecedented exploration of the kinetics of Cas9-mediated DNA cleavage and repair. We find that sgRNAs targeting different sites generally induce cleavage within minutes and repair within 1 or 2 h. However, we observe distinct kinetic profiles, even for proximal sites, and this suggests that target sequence and chromatin state modulate cleavage and repair kinetics.


Subject(s)
Caspase 9/genetics , DNA Breaks, Double-Stranded , DNA Probes/genetics , Gene Editing/methods , Molecular Probe Techniques , Polymerase Chain Reaction/methods , Kinetics
3.
Nucleic Acids Res ; 44(18): 8513-8524, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27550180

ABSTRACT

The accumulation of somatic mitochondrial DNA (mtDNA) mutations contributes to the pathogenesis of human disease. Currently, mitochondrial mutations are largely considered results of inaccurate processing of its heavily damaged genome. However, mainly from a lack of methods to monitor mtDNA mutations with sufficient sensitivity and accuracy, a link between mtDNA damage and mutation has not been established. To test the hypothesis that mtDNA-damaging agents induce mtDNA mutations, we exposed MutaTMMouse mice to benzo[a]pyrene (B[a]P) or N-ethyl-N-nitrosourea (ENU), daily for 28 consecutive days, and quantified mtDNA point and deletion mutations in bone marrow and liver using our newly developed Digital Random Mutation Capture (dRMC) and Digital Deletion Detection (3D) assays. Surprisingly, our results demonstrate mutagen treatment did not increase mitochondrial point or deletion mutation frequencies, despite evidence both compounds increase nuclear DNA mutations and demonstrated B[a]P adduct formation in mtDNA. These findings contradict models of mtDNA mutagenesis that assert the elevated rate of mtDNA mutation stems from damage sensitivity and abridged repair capacity. Rather, our results demonstrate induced mtDNA damage does not readily convert into mutation. These findings suggest robust mitochondrial damage responses repress induced mutations after mutagen exposure.


Subject(s)
DNA, Mitochondrial/genetics , Point Mutation/genetics , Sequence Deletion/genetics , Animals , Benzo(a)pyrene , Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Nucleus/drug effects , Cell Nucleus/genetics , DNA Adducts/metabolism , Ethylnitrosourea , Liver/drug effects , Liver/metabolism , Male , Mice , Mutagenesis/drug effects , Mutagens/toxicity
4.
Nucleic Acids Res ; 44(3): e22, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26384417

ABSTRACT

Next-generation sequencing (NGS) technologies have transformed genomic research and have the potential to revolutionize clinical medicine. However, the background error rates of sequencing instruments and limitations in targeted read coverage have precluded the detection of rare DNA sequence variants by NGS. Here we describe a method, termed CypherSeq, which combines double-stranded barcoding error correction and rolling circle amplification (RCA)-based target enrichment to vastly improve NGS-based rare variant detection. The CypherSeq methodology involves the ligation of sample DNA into circular vectors, which contain double-stranded barcodes for computational error correction and adapters for library preparation and sequencing. CypherSeq is capable of detecting rare mutations genome-wide as well as those within specific target genes via RCA-based enrichment. We demonstrate that CypherSeq is capable of correcting errors incurred during library preparation and sequencing to reproducibly detect mutations down to a frequency of 2.4 × 10(-7) per base pair, and report the frequency and spectra of spontaneous and ethyl methanesulfonate-induced mutations across the Saccharomyces cerevisiae genome.


Subject(s)
DNA/genetics , High-Throughput Nucleotide Sequencing/methods , Mutation , Cell Line , Genes, p53 , Humans , Polymerase Chain Reaction/methods , Saccharomyces cerevisiae/genetics
5.
Nat Genet ; 39(4): 540-3, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17334366

ABSTRACT

Whether mitochondrial mutations cause mammalian aging, or are merely correlated with it, is an area of intense debate. Here, we use a new, highly sensitive assay to redefine the relationship between mitochondrial mutations and age. We measured the in vivo rate of change of the mitochondrial genome at a single-base pair level in mice, and we demonstrate that the mutation frequency in mouse mitochondria is more than ten times lower than previously reported. Although we observed an 11-fold increase in mitochondrial point mutations with age, we report that a mitochondrial mutator mouse was able to sustain a 500-fold higher mutation burden than normal mice, without any obvious features of rapidly accelerated aging. Thus, our results strongly indicate that mitochondrial mutations do not limit the lifespan of wild-type mice.


Subject(s)
Longevity/genetics , Mitochondria/genetics , Point Mutation/physiology , Aging/genetics , Animals , Cells, Cultured , DNA Polymerase gamma , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Mice , Mice, Transgenic , Mitochondria/enzymology , Mitochondria/metabolism
6.
PLoS Genet ; 8(6): e1002689, 2012.
Article in English | MEDLINE | ID: mdl-22685414

ABSTRACT

Genome instability is regarded as a hallmark of cancer. Human tumors frequently carry clonally expanded mutations in their mitochondrial DNA (mtDNA), some of which may drive cancer progression and metastasis. The high prevalence of clonal mutations in tumor mtDNA has commonly led to the assumption that the mitochondrial genome in cancer is genetically unstable, yet this hypothesis has not been experimentally tested. In this study, we directly measured the frequency of non-clonal (random) de novo single base substitutions in the mtDNA of human colorectal cancers. Remarkably, tumor tissue exhibited a decreased prevalence of these mutations relative to adjacent non-tumor tissue. The difference in mutation burden was attributable to a reduction in C:G to T:A transitions, which are associated with oxidative damage. We demonstrate that the lower random mutation frequency in tumor tissue was also coupled with a shift in glucose metabolism from oxidative phosphorylation to anaerobic glycolysis, as compared to non-neoplastic colon. Together these findings raise the intriguing possibility that fidelity of mitochondrial genome is, in fact, increased in cancer as a result of a decrease in reactive oxygen species-mediated mtDNA damage.


Subject(s)
Colorectal Neoplasms/genetics , DNA, Mitochondrial/genetics , Mutagenesis , Point Mutation/genetics , Aged , Aged, 80 and over , DNA Damage/genetics , Female , Genomic Instability , Glucose/metabolism , Glycolysis/genetics , Humans , Male , Middle Aged , Oxidative Stress , Reactive Oxygen Species/metabolism
7.
J Pathol ; 231(4): 433-440, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24027095

ABSTRACT

The cellular adaptive immune system mounts a response to many solid tumours mediated by tumour-infiltrating T lymphocytes (TILs). Basic measurements of these TILs, including total count, show promise as prognostic markers for a variety of cancers, including ovarian and colorectal. In addition, recent therapeutic advances are thought to exploit this immune response to effectively fight melanoma, with promising studies showing efficacy in additional cancers. However, many of the basic properties of TILs are poorly understood, including specificity, clonality, and spatial heterogeneity of the T-cell response. We utilize deep sequencing of rearranged T-cell receptor beta (TCRB) genes to characterize the basic properties of TILs in ovarian carcinoma. Due to somatic rearrangement during T-cell development, the TCR beta chain sequence serves as a molecular tag for each T-cell clone. Using these sequence tags, we assess similarities and differences between infiltrating T cells in discretely sampled sections of large tumours and compare to T cells from peripheral blood. Within the limits of sensitivity of our assay, the TIL repertoires show strong similarity throughout each tumour and are distinct from the circulating T-cell repertoire. We conclude that the cellular adaptive immune response within ovarian carcinomas is spatially homogeneous and distinct from the T-cell compartment of peripheral blood.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Ovarian Neoplasms/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Adaptive Immunity , Cluster Analysis , Complementarity Determining Regions/genetics , Female , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/immunology , High-Throughput Nucleotide Sequencing/methods , Humans , Omentum , Ovarian Neoplasms/genetics , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/secondary , Sequence Analysis, DNA/methods
8.
Nucleic Acids Res ; 39(11): e73, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21459851

ABSTRACT

Herein, a detailed protocol for a random mutation capture (RMC) assay to measure nuclear point mutation frequency in mouse tissue is described. This protocol is a simplified version of the original method developed for human tissue that is easier to perform, yet retains a high sensitivity of detection. In contrast to assays relying on phenotypic selection of reporter genes in transgenic mice, the RMC assay allows direct detection of mutations in endogenous genes in any mouse strain. Measuring mutation frequency within an intron of a transcribed gene, we show this assay to be highly reproducible. We analyzed mutation frequencies from the liver tissue of animals with a mutation within the intrinsic exonuclease domains of the two major DNA polymerases, δ and ε. These mice exhibited significantly higher mutation frequencies than did wild-type animals. A comparison with a previous analysis of these genotypes in Big Blue mice revealed the RMC assay to be more sensitive than the Big Blue assay for this application. As RMC does not require analysis of a particular gene, simultaneous analysis of mutation frequency at multiple genetic loci is feasible. This assay provides a versatile alternative to transgenic mouse models for the study of mutagenesis in vivo.


Subject(s)
DNA Mutational Analysis , Point Mutation , Animals , DNA Polymerase II/genetics , DNA Polymerase III/genetics , Genome , Mice , Mice, Inbred C57BL , Mutagenesis , Polymerase Chain Reaction , Reproducibility of Results
9.
Nucleic Acids Res ; 39(16): 7020-33, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21622658

ABSTRACT

DNA polymerase I (pol I) processes RNA primers during lagging-strand synthesis and fills small gaps during DNA repair reactions. However, it is unclear how pol I and pol III work together during replication and repair or how extensive pol I processing of Okazaki fragments is in vivo. Here, we address these questions by analyzing pol I mutations generated through error-prone replication of ColE1 plasmids. The data were obtained by direct sequencing, allowing an accurate determination of the mutation spectrum and distribution. Pol I's mutational footprint suggests: (i) during leading-strand replication pol I is gradually replaced by pol III over at least 1.3 kb; (ii) pol I processing of Okazaki fragments is limited to ∼20 nt and (iii) the size of Okazaki fragments is short (∼250 nt). While based on ColE1 plasmid replication, our findings are likely relevant to other pol I replicative processes such as chromosomal replication and DNA repair, which differ from ColE1 replication mostly at the recruitment steps. This mutation footprinting approach should help establish the role of other prokaryotic or eukaryotic polymerases in vivo, and provides a tool to investigate how sequence topology, DNA damage, or interactions with protein partners may affect the function of individual DNA polymerases.


Subject(s)
DNA Polymerase I/metabolism , DNA Replication , Mutation , Plasmids/biosynthesis , Base Sequence , DNA/metabolism , DNA Footprinting , DNA Polymerase I/genetics , DNA Polymerase I/physiology , Databases, Nucleic Acid , Plasmids/chemistry
10.
J Biol Chem ; 286(44): 38592-38601, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-21900249

ABSTRACT

Although neurogenesis occurs in discrete areas of the adult mammalian brain, neural progenitor cells (NPCs) produce fewer new neurons with age. To characterize the molecular changes that occur during aging, we performed a proteomic comparison between primary-cultured NPCs from the young adult and aged mouse forebrain. This analysis yielded changes in proteins necessary for cellular metabolism. Mitochondrial quantity and oxygen consumption rates decrease with aging, although mitochondrial DNA in aged NPCs does not have increased mutation rates. In addition, aged cells are resistant to the mitochondrial inhibitor rotenone and proliferate in response to lowered oxygen conditions. These results demonstrate that aging NPCs display an altered metabolic phenotype, characterized by a coordinated shift in protein expression, subcellular structure, and metabolic physiology.


Subject(s)
Aging , Mitochondria/metabolism , Neurogenesis/physiology , Neurons/pathology , Oxygen/metabolism , Stem Cells/cytology , Animals , Cell Proliferation , Female , Hypoxia , Mice , Mice, Inbred C57BL , Microscopy, Confocal/methods , Models, Biological , Oxygen Consumption , Proteomics/methods , Regeneration
11.
Nat Cancer ; 3(1): 25-42, 2022 01.
Article in English | MEDLINE | ID: mdl-35121993

ABSTRACT

Although dormancy is thought to play a key role in the metastasis of breast tumor cells to the brain, our knowledge of the molecular mechanisms regulating disseminated tumor cell (DTC) dormancy in this organ is limited. Here using serial intravital imaging of dormant and metastatic triple-negative breast cancer lines, we identify escape from the single-cell or micrometastatic state as the rate-limiting step towards brain metastasis. We show that every DTC occupies a vascular niche, with quiescent DTCs residing on astrocyte endfeet. At these sites, astrocyte-deposited laminin-211 drives DTC quiescence by inducing the dystroglycan receptor to associate with yes-associated protein, thereby sequestering it from the nucleus and preventing its prometastatic functions. These findings identify a brain-specific mechanism of DTC dormancy and highlight the need for a more thorough understanding of tumor dormancy to develop therapeutic approaches that prevent brain metastasis.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Astrocytes/metabolism , Brain/metabolism , Breast Neoplasms/drug therapy , Female , Humans , Laminin/metabolism , Tumor Microenvironment
12.
Elife ; 112022 12 13.
Article in English | MEDLINE | ID: mdl-36511483

ABSTRACT

Advanced prostate malignancies are a leading cause of cancer-related deaths in men, in large part due to our incomplete understanding of cellular drivers of disease progression. We investigate prostate cancer cell dynamics at single-cell resolution from disease onset to the development of androgen independence in an in vivo murine model. We observe an expansion of a castration-resistant intermediate luminal cell type that correlates with treatment resistance and poor prognosis in human patients. Moreover, transformed epithelial cells and associated fibroblasts create a microenvironment conducive to pro-tumorigenic immune infiltration, which is partially androgen responsive. Androgen-independent prostate cancer leads to significant diversification of intermediate luminal cell populations characterized by a range of androgen signaling activity, which is inversely correlated with proliferation and mRNA translation. Accordingly, distinct epithelial populations are exquisitely sensitive to translation inhibition, which leads to epithelial cell death, loss of pro-tumorigenic signaling, and decreased tumor heterogeneity. Our findings reveal a complex tumor environment largely dominated by castration-resistant luminal cells and immunosuppressive infiltrates.


Subject(s)
Androgens , Prostatic Neoplasms , Male , Humans , Mice , Animals , Prostate/metabolism , Prostatic Neoplasms/pathology , Orchiectomy , Population Dynamics , Receptors, Androgen/metabolism , Disease Progression , Tumor Microenvironment
13.
Nat Biotechnol ; 39(11): 1375-1384, 2021 11.
Article in English | MEDLINE | ID: mdl-34083791

ABSTRACT

Recent spatial gene expression technologies enable comprehensive measurement of transcriptomic profiles while retaining spatial context. However, existing analysis methods do not address the limited resolution of the technology or use the spatial information efficiently. Here, we introduce BayesSpace, a fully Bayesian statistical method that uses the information from spatial neighborhoods for resolution enhancement of spatial transcriptomic data and for clustering analysis. We benchmark BayesSpace against current methods for spatial and non-spatial clustering and show that it improves identification of distinct intra-tissue transcriptional profiles from samples of the brain, melanoma, invasive ductal carcinoma and ovarian adenocarcinoma. Using immunohistochemistry and an in silico dataset constructed from scRNA-seq data, we show that BayesSpace resolves tissue structure that is not detectable at the original resolution and identifies transcriptional heterogeneity inaccessible to histological analysis. Our results illustrate BayesSpace's utility in facilitating the discovery of biological insights from spatial transcriptomic datasets.


Subject(s)
Single-Cell Analysis , Transcriptome , Bayes Theorem , Cluster Analysis , Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcriptome/genetics
14.
JCI Insight ; 6(15)2021 08 09.
Article in English | MEDLINE | ID: mdl-34156975

ABSTRACT

The ectocervix is part of the lower female reproductive tract (FRT), which is susceptible to sexually transmitted infections (STIs). Comprehensive knowledge of the phenotypes and T cell receptor (TCR) repertoire of tissue-resident memory T cells (TRMs) in the human FRT is lacking. We took single-cell RNA-Seq approaches to simultaneously define gene expression and TCR clonotypes of the human ectocervix. There were significantly more CD8+ than CD4+ T cells. Unsupervised clustering and trajectory analysis identified distinct populations of CD8+ T cells with IFNGhiGZMBloCD69hiCD103lo or IFNGloGZMBhiCD69medCD103hi phenotypes. Little overlap was seen between their TCR repertoires. Immunofluorescence staining showed that CD103+CD8+ TRMs were preferentially localized in the epithelium, whereas CD69+CD8+ TRMs were distributed evenly in the epithelium and stroma. Ex vivo assays indicated that up to 14% of cervical CD8+ TRM clonotypes were HSV-2 reactive in HSV-2-seropositive persons, reflecting physiologically relevant localization. Our studies identified subgroups of CD8+ TRMs in the human ectocervix that exhibited distinct expression of antiviral defense and tissue residency markers, anatomic locations, and TCR repertoires that target anatomically relevant viral antigens. Optimization of the location, number, and function of FRT TRMs is an important approach for improving host defenses to STIs.


Subject(s)
Antigens, CD/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , CD8-Positive T-Lymphocytes/immunology , Cervix Uteri , Herpesvirus 2, Human , Integrin alpha Chains/analysis , Lectins, C-Type/analysis , Adaptive Immunity , CD4-Positive T-Lymphocytes/immunology , Cervix Uteri/immunology , Cervix Uteri/pathology , Cervix Uteri/virology , Female , Genes, T-Cell Receptor/immunology , Herpesvirus 2, Human/immunology , Herpesvirus 2, Human/isolation & purification , Humans , Immunologic Memory , Immunophenotyping/methods , Memory T Cells/immunology , Mucous Membrane/immunology , Mucous Membrane/pathology , Mucous Membrane/virology
15.
Trends Genet ; 23(4): 154-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17328987

ABSTRACT

Cancers are thought to originate in stem cells through the accumulation of multiple mutations. Some of these mutations result in a loss of heterozygosity (LOH). A recent report demonstrates that exposure of mouse embryonic stem cells to nontoxic amounts of mutagens triggers a marked increase in the frequency of LOH. Thus, mutagen induction of LOH in embryonic stem cells suggests a new pathway to account for the multiple homozygous mutations in human tumors. This induction could mimic early mutagenic events that generate cancers in human tissue stem cells.


Subject(s)
Embryonic Stem Cells , Loss of Heterozygosity , Neoplastic Stem Cells , Animals , Humans , Models, Biological
16.
Methods ; 46(4): 263-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18948200

ABSTRACT

Mitochondrial DNA (mtDNA) mutations contribute to the pathology of a number of age-related disorders, including Parkinson disease [A. Bender et al., Nat. Genet. 38 (2006) 515,Y. Kraytsberg et al., Nat. Genet. 38 (2006) 518], muscle-wasting [J. Wanagat, Z. Cao, P. Pathare, J.M. Aiken, FASEB J. 15 (2001) 322], and the metastatic potential of cancers [K. Ishikawa et al., Science 320 (2008) 661]. The impact of mitochondrial DNA mutations on a wide variety of human diseases has made it increasingly important to understand the mechanisms that drive mitochondrial mutagenesis. In order to provide new insight into the etiology and natural history of mtDNA mutations, we have developed an assay that can detect mitochondrial mutations in a variety of tissues and experimental settings [M. Vermulst et al., Nat. Genet. 40 (2008) 4, M. Vermulst et al., Nat. Genet. 39 (2007) 540]. This methodology, termed the Random Mutation Capture assay, relies on single-molecule amplification to detect rare mutations among millions of wild-type bases [J.H. Bielas, L.A. Loeb, Nat. Methods 2 (2005) 285], and can be used to analyze mitochondrial mutagenesis to a single base pair level in mammals.


Subject(s)
DNA, Mitochondrial/genetics , Genome, Mitochondrial , Mutation , Cell Fractionation/methods , Humans , Mitochondria/genetics , Mutagenesis , Polymerase Chain Reaction/methods , Sequence Deletion , Taq Polymerase/metabolism
17.
Article in English | MEDLINE | ID: mdl-31699343

ABSTRACT

We live in an era of 'big data', where the volume, velocity, and variety of the data being generated is increasingly influencing the way toxicological sciences are practiced. With this in mind, a workgroup was formed for the 2017 International Workshops on Genotoxicity Testing (IWGT) to consider the use of high information content data in genetic toxicology assessments. Presentations were given on adductomics, global transcriptional profiling, error-reduced single-molecule sequencing, and cellular phenotype-based assays, which were identified as methodologies that are relevant to present-day genetic toxicology assessments. Presenters and workgroup members discussed the state of the science for these methodologies, their potential use in genetic toxicology, current limitations, and the future work necessary to advance their utility and application. The session culminated with audience-assisted SWOT (strength, weakness, opportunities, and threats) analyses. The summary report described herein is structured similarly. A major conclusion of the workgroup is that while conventional regulatory genetic toxicology testing has served the public well over the last several decades, it does not provide the throughput that has become necessary in modern times, and it does not generate the mechanistic information that risk assessments ideally take into consideration. The high information content assay platforms that were discussed in this session, as well as others under development, have the potential to address aspect(s) of these issues and to meet new expectations in the field of genetic toxicology.


Subject(s)
Mutagenicity Tests/methods , Animals , Big Data , Cell Line , DNA Adducts/analysis , DNA Barcoding, Taxonomic/methods , DNA Damage , Data Mining , Drug Evaluation, Preclinical , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing , Humans , Image Processing, Computer-Assisted , Mass Spectrometry/methods , Meta-Analysis as Topic , Mice , Mutagenicity Tests/standards , Phenotype , Single Molecule Imaging , Toxicology/methods , Transcriptome
18.
Neurobiol Aging ; 73: 161-170, 2019 01.
Article in English | MEDLINE | ID: mdl-30359878

ABSTRACT

Accumulation of oxidative mitochondrial DNA (mtDNA) damage and impaired base excision repair (BER) in brains have been associated with Alzheimer's disease (AD). However, it is still not clear how these affect mtDNA stability, as reported levels of mtDNA mutations in AD are conflicting. Thus, we investigated whether alterations in BER correlate with mtDNA instability in AD using postmortem brain samples from cognitively normal AD subjects and individuals who show neuropathological features of AD, but remained cognitively normal (high-pathology control). To date, no data on DNA repair and mtDNA stability are available for these individuals. BER activities, mtDNA mutations, and mtDNA copy number were measured in the nuclear and mitochondrial extracts. Significantly lower uracil DNA glycosylase activity was detected in nuclear and mitochondrial extracts from AD subjects, while apurinic/apyrimidinic endonuclease activity was similar in all groups. Although mtDNA mutation frequency was similar in all groups, mtDNA copy number was significantly decreased in the temporal cortex of AD brains but not of high-pathology control subjects. Our results show that lower mitochondrial uracil DNA glycosylase activity does not result in increased mutagenesis, but rather in depletion of mtDNA in early-affected brain regions during AD development.


Subject(s)
Alzheimer Disease/genetics , Brain/metabolism , DNA Repair/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Female , Gene Dosage , Humans , Male , Middle Aged , Mutation , Oxidative Stress/genetics , Temporal Lobe/metabolism , Uracil-DNA Glycosidase/metabolism
19.
Cell Rep ; 22(12): 3115-3125, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29562168

ABSTRACT

Genetic instability of the mitochondrial genome (mtDNA) plays an important role in human aging and disease. Thus far, it has proven difficult to develop successful treatment strategies for diseases that are caused by mtDNA instability. To address this issue, we developed a model of mtDNA disease in the nematode C. elegans, an animal model that can rapidly be screened for genes and biological pathways that reduce mitochondrial pathology. These worms recapitulate all the major hallmarks of mtDNA disease in humans, including increased mtDNA instability, loss of respiration, reduced neuromuscular function, and a shortened lifespan. We found that these phenotypes could be rescued by intervening in numerous biological pathways, including IGF-1/insulin signaling, mitophagy, and the mitochondrial unfolded protein response, suggesting that it may be possible to ameliorate mtDNA disease through multiple molecular mechanisms.


Subject(s)
Caenorhabditis elegans/metabolism , DNA, Mitochondrial/genetics , Mitochondria/metabolism , Animals , Disease Progression , Mice , Models, Animal
20.
DNA Repair (Amst) ; 5(3): 294-302, 2006 Mar 07.
Article in English | MEDLINE | ID: mdl-16359931

ABSTRACT

Mutations are rare in normal cells. In contrast, multiple mutations are characteristic in most tumors. Previously we proposed a "mutator phenotype" hypothesis to explain how pre-cancer cells may acquire large number of mutations during carcinogenesis. Here we extend the "mutator phenotype" hypothesis considering recently discovered biochemical activities whose aberrant expression may result in genome-wide random mutations. The scope of this article is to emphasize that simple random point mutations can drive carcinogenesis and highlight new emerging pathways that generate these mutations. We focus specifically on random point mutations generated by replication errors, oxidative base damage, covalent base modifications by enzymes, and spontaneously generated abasic sites as a source of mutator mutants.


Subject(s)
Mutation , Neoplasms/genetics , Animals , Cell Transformation, Neoplastic , Humans , Microsatellite Repeats , Mutagenesis
SELECTION OF CITATIONS
SEARCH DETAIL