Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 176(5): 1083-1097.e18, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30739799

ABSTRACT

Cell size varies greatly between cell types, yet within a specific cell type and growth condition, cell size is narrowly distributed. Why maintenance of a cell-type specific cell size is important remains poorly understood. Here we show that growing budding yeast and primary mammalian cells beyond a certain size impairs gene induction, cell-cycle progression, and cell signaling. These defects are due to the inability of large cells to scale nucleic acid and protein biosynthesis in accordance with cell volume increase, which effectively leads to cytoplasm dilution. We further show that loss of scaling beyond a certain critical size is due to DNA becoming limiting. Based on the observation that senescent cells are large and exhibit many of the phenotypes of large cells, we propose that the range of DNA:cytoplasm ratio that supports optimal cell function is limited and that ratios outside these bounds contribute to aging.


Subject(s)
Cell Enlargement , Cellular Senescence/physiology , Cytoplasm/metabolism , Candida albicans/genetics , Candida albicans/growth & development , Cell Cycle , Cell Proliferation , Cell Size , Cellular Senescence/genetics , Fibroblasts/metabolism , HEK293 Cells , Humans , Primary Cell Culture , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomycetales/genetics , Saccharomycetales/growth & development , Saccharomycetales/metabolism , Signal Transduction
2.
Cell ; 175(1): 200-211.e13, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30146160

ABSTRACT

Much of our understanding of chromosome segregation is based on cell culture systems. Here, we examine the importance of the tissue environment for chromosome segregation by comparing chromosome segregation fidelity across several primary cell types in native and nonnative contexts. We discover that epithelial cells have increased chromosome missegregation outside of their native tissues. Using organoid culture systems, we show that tissue architecture, specifically integrin function, is required for accurate chromosome segregation. We find that tissue architecture enhances the correction of merotelic microtubule-kinetochore attachments, and this is especially important for maintaining chromosome stability in the polyploid liver. We propose that disruption of tissue architecture could underlie the widespread chromosome instability across epithelial cancers. Moreover, our findings highlight the extent to which extracellular context can influence intrinsic cellular processes and the limitations of cell culture systems for studying cells that naturally function within a tissue.


Subject(s)
Chromosomal Instability/physiology , Chromosome Segregation/physiology , Epithelium/physiology , Animals , Cell Aggregation/physiology , Cell Culture Techniques/methods , Chromosomes/physiology , Epithelial Cells/physiology , Female , Kinetochores/physiology , Male , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Mitosis , Organoids/physiology , Spindle Apparatus/metabolism , Spindle Apparatus/physiology
3.
Cell ; 169(2): 229-242.e21, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28388408

ABSTRACT

Phenotypic variability is a hallmark of diseases involving chromosome gains and losses, such as Down syndrome and cancer. Allelic variances have been thought to be the sole cause of this heterogeneity. Here, we systematically examine the consequences of gaining and losing single or multiple chromosomes to show that the aneuploid state causes non-genetic phenotypic variability. Yeast cell populations harboring the same defined aneuploidy exhibit heterogeneity in cell-cycle progression and response to environmental perturbations. Variability increases with degree of aneuploidy and is partly due to gene copy number imbalances, suggesting that subtle changes in gene expression impact the robustness of biological networks and cause alternate behaviors when they occur across many genes. As inbred trisomic mice also exhibit variable phenotypes, we further propose that non-genetic individuality is a universal characteristic of the aneuploid state that may contribute to variability in presentation and treatment responses of diseases caused by aneuploidy.


Subject(s)
Aneuploidy , Genetic Heterogeneity , Phenotype , Animals , Cell Cycle , Cell Division , DNA Damage , Gene Expression Regulation , Kinetics , Mice , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics
4.
Cell ; 163(2): 406-18, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26411291

ABSTRACT

Message-specific translational control is required for gametogenesis. In yeast, the RNA-binding protein Rim4 mediates translational repression of numerous mRNAs, including the B-type cyclin CLB3, which is essential for establishing the meiotic chromosome segregation pattern. Here, we show that Rim4 forms amyloid-like aggregates and that it is the amyloid-like form of Rim4 that is the active, translationally repressive form of the protein. Our data further show that Rim4 aggregation is a developmentally regulated process. Starvation induces the conversion of monomeric Rim4 into amyloid-like aggregates, thereby activating the protein to bring about repression of translation. At the onset of meiosis II, Rim4 aggregates are abruptly degraded allowing translation to commence. Although amyloids are best known for their role in the etiology of diseases such as Alzheimer's, Parkinson's, and diabetes by forming toxic protein aggregates, our findings show that cells can utilize amyloid-like protein aggregates to function as central regulators of gametogenesis.


Subject(s)
Gametogenesis , Protein Aggregates , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Amyloidogenic Proteins/chemistry , Amyloidogenic Proteins/metabolism , Animals , Cyclin B/genetics , Gene Expression Regulation , Male , Meiosis , Mice , Mice, Inbred C57BL , Protein Aggregates/drug effects , Protein Biosynthesis , RNA-Binding Proteins/chemistry , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sodium Dodecyl Sulfate/pharmacology
5.
Genes Dev ; 35(7-8): 556-572, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33766983

ABSTRACT

Aneuploidy, defined as whole-chromosome gain or loss, causes cellular stress but, paradoxically, is a frequent occurrence in cancers. Here, we investigate why ∼50% of Ewing sarcomas, driven by the EWS-FLI1 fusion oncogene, harbor chromosome 8 gains. Expression of the EWS-FLI1 fusion in primary cells causes replication stress that can result in cellular senescence. Using an evolution approach, we show that trisomy 8 mitigates EWS-FLI1-induced replication stress through gain of a copy of RAD21. Low-level ectopic expression of RAD21 is sufficient to dampen replication stress and improve proliferation in EWS-FLI1-expressing cells. Conversely, deleting one copy in trisomy 8 cells largely neutralizes the fitness benefit of chromosome 8 gain and reduces tumorgenicity of a Ewing sarcoma cancer cell line in soft agar assays. We propose that RAD21 promotes tumorigenesis through single gene copy gain. Such genes may explain some recurrent aneuploidies in cancer.


Subject(s)
Carcinogenesis/genetics , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Sarcoma, Ewing/genetics , Stress, Physiological/genetics , Trisomy/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Chromosomes, Human, Pair 8/genetics , DNA Replication/genetics , DNA-Binding Proteins/genetics , Gene Duplication/genetics , Gene Expression Regulation, Neoplastic , Humans
6.
Genes Dev ; 35(15-16): 1079-1092, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34266888

ABSTRACT

Chromosome gains and losses are a frequent feature of human cancers. However, how these aberrations can outweigh the detrimental effects of aneuploidy remains unclear. An initial comparison of existing chromosomal instability (CIN) mouse models suggests that aneuploidy accumulates to low levels in these animals. We therefore developed a novel mouse model that enables unprecedented levels of chromosome missegregation in the adult animal. At the earliest stages of T-cell development, cells with random chromosome gains and/or losses are selected against, but CIN eventually results in the expansion of progenitors with clonal chromosomal imbalances. Clonal selection leads to the development of T-cell lymphomas with stereotypic karyotypes in which chromosome 15, containing the Myc oncogene, is gained with high prevalence. Expressing human MYC from chromosome 6 (MYCChr6) is sufficient to change the karyotype of these lymphomas to include universal chromosome 6 gains. Interestingly, while chromosome 15 is still gained in MYCChr6 tumors after genetic ablation of the endogenous Myc locus, this chromosome is not efficiently gained after deletion of one copy of Rad21, suggesting a synergistic effect of both MYC and RAD21 in driving chromosome 15 gains. Our results show that the initial detrimental effects of random missegregation are outbalanced by clonal selection, which is dictated by the chromosomal location and nature of certain genes and is sufficient to drive cancer with high prevalence.


Subject(s)
Aneuploidy , Chromosomal Instability , Animals , Cell Transformation, Neoplastic/genetics , Chromosomal Instability/genetics , Chromosome Aberrations , Karyotype , Mice , Prevalence , Stem Cells
7.
Cell ; 152(3): 394-405, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23374337

ABSTRACT

Changes in DNA copy number, whether confined to specific genes or affecting whole chromosomes, have been identified as causes of diseases and developmental abnormalities and as sources of adaptive potential. Here, we discuss the costs and benefits of DNA copy-number alterations. Changes in DNA copy number are largely detrimental. Amplifications or deletions of specific genes can elicit discrete defects. Large-scale changes in DNA copy number can also cause detrimental phenotypes that are due to the cumulative effects of copy-number alterations of many genes simultaneously. On the other hand, studies in microorganisms show that DNA copy-number alterations can be beneficial, increasing survival under selective pressure. As DNA copy-number alterations underlie many human diseases, we will end with a discussion of gene copy-number changes as therapeutic targets.


Subject(s)
Aneuploidy , DNA Copy Number Variations , Polyploidy , Animals , Gene Dosage , Gene Expression , Genetic Fitness , Humans , Neoplasms/genetics , Plants/genetics , Yeasts/genetics , Yeasts/physiology
8.
Nat Rev Mol Cell Biol ; 16(8): 473-85, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26204159

ABSTRACT

Dividing cells that experience chromosome mis-segregation generate aneuploid daughter cells, which contain an incorrect number of chromosomes. Although aneuploidy interferes with the proliferation of untransformed cells, it is also, paradoxically, a hallmark of cancer, a disease defined by increased proliferative potential. These contradictory effects are also observed in mouse models of chromosome instability (CIN). CIN can inhibit and promote tumorigenesis. Recent work has provided insights into the cellular consequences of CIN and aneuploidy. Chromosome mis-segregation per se can alter the genome in many more ways than just causing the gain or loss of chromosomes. The short- and long-term effects of aneuploidy are caused by gene-specific effects and a stereotypic aneuploidy stress response. Importantly, these recent findings provide insights into the role of aneuploidy in tumorigenesis.


Subject(s)
Aneuploidy , Chromosome Segregation , Abnormal Karyotype , Animals , Carcinogenesis/genetics , DNA Damage , Gene Dosage , Humans , Neoplasms/genetics , Organ Specificity
9.
Cell ; 150(6): 1170-81, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22959267

ABSTRACT

The cell-fate decision leading to gametogenesis is essential for sexual reproduction. In S. cerevisiae, only diploid MATa/α but not haploid MATa or MATα cells undergo gametogenesis, known as sporulation. We find that transcription of two long noncoding RNAs (lncRNAs) mediates mating-type control of sporulation. In MATa or MATα haploids, expression of IME1, the central inducer of gametogenesis, is inhibited in cis by transcription of the lncRNA IRT1, located in the IME1 promoter. IRT1 transcription recruits the Set2 histone methyltransferase and the Set3 histone deacetylase complex to establish repressive chromatin at the IME1 promoter. Inhibiting expression of IRT1 and an antisense transcript that antagonizes the expression of the meiotic regulator IME4 allows cells expressing the haploid mating type to sporulate with kinetics that are indistinguishable from that of MATa/α diploids. Conversely, expression of the two lncRNAs abolishes sporulation in MATa/α diploids. Thus, transcription of two lncRNAs governs mating-type control of gametogenesis in yeast.


Subject(s)
Gene Expression Regulation, Fungal , Genes, Mating Type, Fungal , RNA, Fungal/genetics , RNA, Long Noncoding/genetics , Saccharomyces cerevisiae/genetics , Transcription, Genetic , Gametogenesis , Nuclear Proteins/genetics , Promoter Regions, Genetic , Repressor Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Spores, Fungal , Transcription Factors/genetics
10.
Proc Natl Acad Sci U S A ; 121(36): e2405543121, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39190349

ABSTRACT

Higher levels of aneuploidy, characterized by imbalanced chromosome numbers, are associated with lethal progression in prostate cancer. However, how aneuploidy contributes to prostate cancer aggressiveness remains poorly understood. In this study, we assessed in patients which genes on chromosome 8q, one of the most frequently gained chromosome arms in prostate tumors, were most strongly associated with long-term risk of cancer progression to metastases and death from prostate cancer (lethal disease) in 403 patients and found the strongest candidate was cohesin subunit gene, RAD21, with an odds ratio of 3.7 (95% CI 1.8, 7.6) comparing the highest vs. lowest tertiles of mRNA expression and adjusting for overall aneuploidy burden and Gleason score, both strong prognostic factors in primary prostate cancer. Studying prostate cancer driven by the TMPRSS2-ERG oncogenic fusion, found in about half of all prostate tumors, we found that increased RAD21 alleviated toxic oncogenic stress and DNA damage caused by oncogene expression. Data from both organoids and patients indicate that increased RAD21 thereby enables aggressive tumors to sustain tumor proliferation, and more broadly suggests one path through which tumors benefit from aneuploidy.


Subject(s)
Aneuploidy , Carcinogenesis , Cell Cycle Proteins , DNA-Binding Proteins , Disease Progression , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Carcinogenesis/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Chromosomes, Human, Pair 8/genetics , Gene Expression Regulation, Neoplastic , DNA Damage
11.
Genes Dev ; 33(15-16): 1031-1047, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31196865

ABSTRACT

Aneuploidy, a condition characterized by chromosome gains and losses, causes reduced fitness and numerous cellular stresses, including increased protein aggregation. Here, we identify protein complex stoichiometry imbalances as a major cause of protein aggregation in aneuploid cells. Subunits of protein complexes encoded on excess chromosomes aggregate in aneuploid cells, which is suppressed when expression of other subunits is coordinately altered. We further show that excess subunits are either degraded or aggregate and that protein aggregation is nearly as effective as protein degradation at lowering levels of excess proteins. Our study explains why proteotoxic stress is a universal feature of the aneuploid state and reveals protein aggregation as a form of dosage compensation to cope with disproportionate expression of protein complex subunits.


Subject(s)
Aneuploidy , Cytosol/metabolism , Dosage Compensation, Genetic/physiology , Protein Aggregates/genetics , Humans , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Aggregation, Pathological , Protein Subunits/metabolism , Proteolysis , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
12.
Nat Rev Genet ; 21(1): 44-62, 2020 01.
Article in English | MEDLINE | ID: mdl-31548659

ABSTRACT

Cancer is driven by multiple types of genetic alterations, which range in size from point mutations to whole-chromosome gains and losses, known as aneuploidy. Chromosome instability, the process that gives rise to aneuploidy, can promote tumorigenesis by increasing genetic heterogeneity and promoting tumour evolution. However, much less is known about how aneuploidy itself contributes to tumour formation and progression. Unlike some pan-cancer oncogenes and tumour suppressor genes that drive transformation in virtually all cell types and cellular contexts, aneuploidy is not a universal promoter of tumorigenesis. Instead, recent studies suggest that aneuploidy is a context-dependent, cancer-type-specific oncogenic event that may have clinical relevance as a prognostic marker and as a potential therapeutic target.


Subject(s)
Aneuploidy , Cell Transformation, Neoplastic/pathology , Chromosomal Instability , Neoplasms/genetics , Neoplasms/pathology , Animals , Humans , Phenotype
13.
Cell ; 144(4): 499-512, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21315436

ABSTRACT

Aneuploidy, an incorrect chromosome number, is a hallmark of cancer. Compounds that cause lethality in aneuploid, but not euploid, cells could therefore provide new cancer therapies. We have identified the energy stress-inducing agent AICAR, the protein folding inhibitor 17-AAG, and the autophagy inhibitor chloroquine as exhibiting this property. AICAR induces p53-mediated apoptosis in primary mouse embryonic fibroblasts (MEFs) trisomic for chromosome 1, 13, 16, or 19. AICAR and 17-AAG, especially when combined, also show efficacy against aneuploid human cancer cell lines. Our results suggest that compounds that interfere with pathways that are essential for the survival of aneuploid cells could serve as a new treatment strategy against a broad spectrum of human tumors.


Subject(s)
Aneuploidy , Antineoplastic Agents/isolation & purification , Drug Screening Assays, Antitumor , Neoplasms/genetics , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Benzoquinones/pharmacology , Cell Line , Cell Proliferation/drug effects , Chloroquine/pharmacology , Chromosome Segregation , Embryo, Mammalian/cytology , Fibroblasts/drug effects , Humans , Lactams, Macrocyclic/pharmacology , Mice , Neoplasms/drug therapy , Ribonucleotides/pharmacology , Trisomy
14.
Annu Rev Cell Dev Biol ; 28: 189-214, 2012.
Article in English | MEDLINE | ID: mdl-22804579

ABSTRACT

Deviation from a balanced genome by either gain or loss of entire chromosomes is generally tolerated poorly in all eukaryotic systems studied to date. Errors in mitotic or meiotic cell division lead to aneuploidy, which places a burden of additional or insufficient gene products from the missegregated chromosomes on the daughter cells. The burden of aneuploidy often manifests itself as impaired fitness of individual cells and whole organisms, in which abnormal development is also characteristic. However, most human cancers, noted for their rapid growth, also display various levels of aneuploidy. Here we discuss the detrimental, potentially beneficial, and sometimes puzzling effects of aneuploidy on cellular and organismal fitness and tissue function as well as its role in diseases such as cancer and neurodegeneration.


Subject(s)
Aneuploidy , Abnormal Karyotype , Adaptation, Biological , Animals , Chromosome Segregation , Gene Dosage , Gene Expression , Humans , Mitosis , Neoplasms/genetics , Neurodegenerative Diseases/genetics , Phenotype
15.
Genes Dev ; 32(15-16): 1075-1084, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30042134

ABSTRACT

Budding yeast cells produce a finite number of daughter cells before they die. Why old yeast cells stop dividing and die is unclear. We found that age-induced accumulation of the G1/S-phase inhibitor Whi5 and defects in G1/S cyclin transcription cause cell cycle delays and genomic instability that result in cell death. We further identified extrachromosomal rDNA (ribosomal DNA) circles (ERCs) to cause the G1/S cyclin expression defect in old cells. Spontaneous segregation of Whi5 and ERCs into daughter cells rejuvenates old mothers, but daughters that inherit these aging factors die rapidly. Our results identify deregulation of the G1/S-phase transition as the proximal cause of age-induced proliferation decline and cell death in budding yeast.


Subject(s)
G1 Phase Cell Cycle Checkpoints , Aneuploidy , Cell Division , Cyclin G1/genetics , Cyclin G1/metabolism , DNA Damage , DNA, Ribosomal/chemistry , Fungal Proteins/metabolism , Gene Expression , Saccharomycetales/cytology , Saccharomycetales/genetics , Saccharomycetales/metabolism , Transcription, Genetic
16.
Cell ; 142(4): 556-67, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20723757

ABSTRACT

The monopolin complex regulates different types of kinetochore-microtubule attachments in fungi, ensuring sister chromatid co-orientation in Saccharomyces cerevisiae meiosis I and inhibiting merotelic attachment in Schizosaccharomyces pombe mitosis. In addition, the monopolin complex maintains the integrity and silencing of ribosomal DNA (rDNA) repeats in the nucleolus. We show here that the S. cerevisiae Csm1/Lrs4 monopolin subcomplex has a distinctive V-shaped structure, with two pairs of protein-protein interaction domains positioned approximately 10 nm apart. Csm1 presents a conserved hydrophobic surface patch that binds two kinetochore proteins: Dsn1, a subunit of the outer-kinetochore MIND/Mis12 complex, and Mif2/CENP-C. Csm1 point-mutations that disrupt kinetochore-subunit binding also disrupt sister chromatid co-orientation in S. cerevisiae meiosis I. We further show that the same Csm1 point-mutations affect rDNA silencing, probably by disrupting binding to the rDNA-associated protein Tof2. We propose that Csm1/Lrs4 functions as a molecular clamp, crosslinking kinetochore components to enforce sister chromatid co-orientation in S. cerevisiae meiosis I and to suppress merotelic attachment in S. pombe mitosis, and crosslinking rDNA repeats to aid rDNA silencing.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomes, Fungal/metabolism , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Kinetochores/metabolism , Meiosis , Mitosis , Models, Molecular , Point Mutation , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Schizosaccharomyces/cytology , Schizosaccharomyces/genetics
17.
Cell ; 143(1): 71-83, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20850176

ABSTRACT

Aneuploidy causes a proliferative disadvantage in all normal cells analyzed to date, yet this condition is associated with a disease characterized by unabated proliferative potential, cancer. The mechanisms that allow cancer cells to tolerate the adverse effects of aneuploidy are not known. To probe this question, we identified aneuploid yeast strains with improved proliferative abilities. Their molecular characterization revealed strain-specific genetic alterations as well as mutations shared between different aneuploid strains. Among the latter, a loss-of-function mutation in the gene encoding the deubiquitinating enzyme Ubp6 improves growth rates in four different aneuploid yeast strains by attenuating the changes in intracellular protein composition caused by aneuploidy. Our results demonstrate the existence of aneuploidy-tolerating mutations that improve the fitness of multiple different aneuploidies and highlight the importance of ubiquitin-proteasomal degradation in suppressing the adverse effects of aneuploidy.


Subject(s)
Aneuploidy , Saccharomyces cerevisiae/genetics , Cell Proliferation , Chromosome Aberrations , Endopeptidases/genetics , Gene Deletion , Humans , Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Ubiquitin/metabolism
18.
PLoS Genet ; 17(10): e1009808, 2021 10.
Article in English | MEDLINE | ID: mdl-34665800

ABSTRACT

Faithful inheritance of mitochondrial DNA (mtDNA) is crucial for cellular respiration/oxidative phosphorylation and mitochondrial membrane potential. However, how mtDNA is transmitted to progeny is not fully understood. We utilized hypersuppressive mtDNA, a class of respiratory deficient Saccharomyces cerevisiae mtDNA that is preferentially inherited over wild-type mtDNA (rho+), to uncover the factors governing mtDNA inheritance. We found that some regions of rho+ mtDNA persisted while others were lost after a specific hypersuppressive takeover indicating that hypersuppressive preferential inheritance may partially be due to active destruction of rho+ mtDNA. From a multicopy suppression screen, we found that overexpression of putative mitochondrial RNA exonuclease PET127 reduced biased inheritance of a subset of hypersuppressive genomes. This suppression required PET127 binding to the mitochondrial RNA polymerase RPO41 but not PET127 exonuclease activity. A temperature-sensitive allele of RPO41 improved rho+ mtDNA inheritance over a specific hypersuppressive mtDNA at semi-permissive temperatures revealing a previously unknown role for rho+ transcription in promoting hypersuppressive mtDNA inheritance.


Subject(s)
DNA, Fungal/genetics , DNA, Mitochondrial/genetics , DNA-Directed RNA Polymerases/genetics , Mitochondria/genetics , RNA, Mitochondrial/genetics , DNA Replication/genetics , Genes, Fungal/genetics , Replication Origin/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription, Genetic/genetics
19.
Genes Dev ; 30(12): 1395-408, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27313317

ABSTRACT

Aneuploidy, an imbalanced karyotype, is a widely observed feature of cancer cells that has long been hypothesized to promote tumorigenesis. Here we evaluate the fitness of cells with constitutional trisomy or chromosomal instability (CIN) in vivo using hematopoietic reconstitution experiments. We did not observe cancer but instead found that aneuploid hematopoietic stem cells (HSCs) exhibit decreased fitness. This reduced fitness is due at least in part to the decreased proliferative potential of aneuploid hematopoietic cells. Analyses of mice with CIN caused by a hypomorphic mutation in the gene Bub1b further support the finding that aneuploidy impairs cell proliferation in vivo. Whereas nonregenerating adult tissues are highly aneuploid in these mice, HSCs and other regenerative adult tissues are largely euploid. These findings indicate that, in vivo, mechanisms exist to select against aneuploid cells.


Subject(s)
Aneuploidy , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Regeneration/genetics , Aging/genetics , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/genetics , Cells, Cultured , Chromosomal Instability/genetics , Leukocytes , Mice , Mice, Inbred C57BL , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Survival Analysis
20.
Genes Dev ; 30(20): 2259-2271, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27807036

ABSTRACT

Aneuploidy-or an unbalanced karyotype in which whole chromosomes are gained or lost-causes reduced fitness at both the cellular and organismal levels but is also a hallmark of human cancers. Aneuploidy causes a variety of cellular stresses, including genomic instability, proteotoxic and oxidative stresses, and impaired protein trafficking. The deubiquitinase Ubp3, which was identified by a genome-wide screen for gene deletions that impair the fitness of aneuploid yeast, is a key regulator of aneuploid cell homeostasis. We show that deletion of UBP3 exacerbates both karyotype-specific phenotypes and global stresses of aneuploid cells, including oxidative and proteotoxic stress. Indeed, Ubp3 is essential for proper proteasome function in euploid cells, and deletion of this deubiquitinase leads to further proteasome-mediated proteotoxicity in aneuploid yeast. Notably, the importance of UBP3 in aneuploid cells is conserved. Depletion of the human homolog of UBP3, USP10, is detrimental to the fitness of human cells upon chromosome missegregation, and this fitness defect is accompanied by autophagy inhibition. We thus used a genome-wide screen in yeast to identify a guardian of aneuploid cell fitness conserved across species. We propose that interfering with Ubp3/USP10 function could be a productive avenue in the development of novel cancer therapeutics.


Subject(s)
Aneuploidy , Endopeptidases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Ubiquitin Thiolesterase/metabolism , Animals , Autophagy/genetics , Cell Line , Cell Proliferation/genetics , Endopeptidases/genetics , Gene Deletion , Humans , Mice , Proteasome Endopeptidase Complex/genetics , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/genetics , Stress, Physiological/genetics , Ubiquitin Thiolesterase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL