Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Genetics ; 223(3)2023 03 02.
Article in English | MEDLINE | ID: mdl-36563016

ABSTRACT

Telomeres are ribonucleoproteins that cap chromosome-ends and their DNA length is controlled by counteracting elongation and shortening processes. The budding yeast Saccharomyces cerevisiae has been a leading model to study telomere DNA length control and dynamics. Its telomeric DNA is maintained at a length that slightly varies between laboratory strains, but little is known about its variation at the species level. The recent publication of the genomes of over 1,000 S. cerevisiae strains enabled us to explore telomere DNA length variation at an unprecedented scale. Here, we developed a bioinformatic pipeline (YeaISTY) to estimate telomere DNA length from whole-genome sequences and applied it to the sequenced S. cerevisiae collection. Our results revealed broad natural telomere DNA length variation among the isolates. Notably, telomere DNA length is shorter in those derived from wild rather than domesticated environments. Moreover, telomere DNA length variation is associated with mitochondrial metabolism, and this association is driven by wild strains. Overall, these findings reveal broad variation in budding yeast's telomere DNA length regulation, which might be shaped by its different ecological life-styles.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Telomere/genetics , Telomere/metabolism , Saccharomyces cerevisiae Proteins/genetics , Telomere-Binding Proteins/genetics , Base Sequence
2.
Nat Ecol Evol ; 6(4): 448-460, 2022 04.
Article in English | MEDLINE | ID: mdl-35210580

ABSTRACT

Domestication of plants and animals is the foundation for feeding the world human population but can profoundly alter the biology of the domesticated species. Here we investigated the effect of domestication on one of our prime model organisms, the yeast Saccharomyces cerevisiae, at a species-wide level. We tracked the capacity for sexual and asexual reproduction and the chronological life span across a global collection of 1,011 genome-sequenced yeast isolates and found a remarkable dichotomy between domesticated and wild strains. Domestication had systematically enhanced fermentative and reduced respiratory asexual growth, altered the tolerance to many stresses and abolished or impaired the sexual life cycle. The chronological life span remained largely unaffected by domestication and was instead dictated by clade-specific evolution. We traced the genetic origins of the yeast domestication syndrome using genome-wide association analysis and genetic engineering and disclosed causative effects of aneuploidy, gene presence/absence variations, copy number variations and single-nucleotide polymorphisms. Overall, we propose domestication to be the most dramatic event in budding yeast evolution, raising questions about how much domestication has distorted our understanding of the natural biology of this key model species.


Subject(s)
Domestication , Saccharomycetales , Animals , DNA Copy Number Variations , Genome-Wide Association Study , Life Cycle Stages , Saccharomyces cerevisiae/genetics , Saccharomycetales/genetics
3.
Nat Commun ; 12(1): 6564, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34772931

ABSTRACT

Hybrids between diverged lineages contain novel genetic combinations but an impaired meiosis often makes them evolutionary dead ends. Here, we explore to what extent an aborted meiosis followed by a return-to-growth (RTG) promotes recombination across a panel of 20 Saccharomyces cerevisiae and S. paradoxus diploid hybrids with different genomic structures and levels of sterility. Genome analyses of 275 clones reveal that RTG promotes recombination and generates extensive regions of loss-of-heterozygosity in sterile hybrids with either a defective meiosis or a heavily rearranged karyotype, whereas RTG recombination is reduced by high sequence divergence between parental subgenomes. The RTG recombination preferentially arises in regions with low local heterozygosity and near meiotic recombination hotspots. The loss-of-heterozygosity has a profound impact on sexual and asexual fitness, and enables genetic mapping of phenotypic differences in sterile lineages where linkage analysis would fail. We propose that RTG gives sterile yeast hybrids access to a natural route for genome recombination and adaptation.


Subject(s)
Diploidy , Hybridization, Genetic , Infertility/genetics , Meiosis , Saccharomyces cerevisiae/genetics , Chromosome Mapping , Evolution, Molecular , Genome, Fungal , Homologous Recombination , Phenotype , Saccharomyces cerevisiae Proteins/metabolism
4.
Elife ; 102021 11 09.
Article in English | MEDLINE | ID: mdl-34751131

ABSTRACT

To understand the genetic basis and selective forces acting on longevity, it is useful to examine lifespan variation among closely related species, or ecologically diverse isolates of the same species, within a controlled environment. In particular, this approach may lead to understanding mechanisms underlying natural variation in lifespan. Here, we analyzed 76 ecologically diverse wild yeast isolates and discovered a wide diversity of replicative lifespan (RLS). Phylogenetic analyses pointed to genes and environmental factors that strongly interact to modulate the observed aging patterns. We then identified genetic networks causally associated with natural variation in RLS across wild yeast isolates, as well as genes, metabolites, and pathways, many of which have never been associated with yeast lifespan in laboratory settings. In addition, a combined analysis of lifespan-associated metabolic and transcriptomic changes revealed unique adaptations to interconnected amino acid biosynthesis, glutamate metabolism, and mitochondrial function in long-lived strains. Overall, our multiomic and lifespan analyses across diverse isolates of the same species shows how gene-environment interactions shape cellular processes involved in phenotypic variation such as lifespan.


Subject(s)
Gene Regulatory Networks , Genes, Fungal , Saccharomyces cerevisiae/physiology , Saccharomyces/physiology , Saccharomyces/genetics , Saccharomyces cerevisiae/genetics
5.
Nature ; 587(7834): 420-425, 2020 11.
Article in English | MEDLINE | ID: mdl-33177709

ABSTRACT

Genome introgressions drive evolution across the animal1, plant2 and fungal3 kingdoms. Introgressions initiate from archaic admixtures followed by repeated backcrossing to one parental species. However, how introgressions arise in reproductively isolated species, such as yeast4, has remained unclear. Here we identify a clonal descendant of the ancestral yeast hybrid that founded the extant Saccharomyces cerevisiae Alpechin lineage5, which carries abundant Saccharomyces paradoxus introgressions. We show that this clonal descendant, hereafter defined as a 'living ancestor', retained the ancestral genome structure of the first-generation hybrid with contiguous S. cerevisiae and S. paradoxus subgenomes. The ancestral first-generation hybrid underwent catastrophic genomic instability through more than a hundred mitotic recombination events, mainly manifesting as homozygous genome blocks generated by loss of heterozygosity. These homozygous sequence blocks rescue hybrid fertility by restoring meiotic recombination and are the direct origins of the introgressions present in the Alpechin lineage. We suggest a plausible route for introgression evolution through the reconstruction of extinct stages and propose that genome instability allows hybrids to overcome reproductive isolation and enables introgressions to emerge.


Subject(s)
Evolution, Molecular , Genetic Introgression/genetics , Genome, Fungal/genetics , Genomics , Phylogeny , Saccharomyces cerevisiae/genetics , Saccharomyces/genetics , Crosses, Genetic , Fertility/genetics , Genetic Fitness/genetics , Genomic Instability/genetics , Homologous Recombination/genetics , Loss of Heterozygosity/genetics , Meiosis/genetics , Mitosis/genetics , Reproduction, Asexual/genetics , Saccharomyces/classification , Saccharomyces/cytology , Saccharomyces cerevisiae/classification , Saccharomyces cerevisiae/cytology
6.
BMC Biol ; 18(1): 49, 2020 05 11.
Article in English | MEDLINE | ID: mdl-32393264

ABSTRACT

BACKGROUND: Mitochondria are essential organelles partially regulated by their own genomes. The mitochondrial genome maintenance and inheritance differ from the nuclear genome, potentially uncoupling their evolutionary trajectories. Here, we analysed mitochondrial sequences obtained from the 1011 Saccharomyces cerevisiae strain collection and identified pronounced differences with their nuclear genome counterparts. RESULTS: In contrast with pre-whole genome duplication fungal species, S. cerevisiae mitochondrial genomes show higher genetic diversity compared to the nuclear genomes. Strikingly, mitochondrial genomes appear to be highly admixed, resulting in a complex interconnected phylogeny with a weak grouping of isolates, whereas interspecies introgressions are very rare. Complete genome assemblies revealed that structural rearrangements are nearly absent with rare inversions detected. We tracked intron variation in COX1 and COB to infer gain and loss events throughout the species evolutionary history. Mitochondrial genome copy number is connected with the nuclear genome and linearly scale up with ploidy. We observed rare cases of naturally occurring mitochondrial DNA loss, petite, with a subset of them that do not suffer the expected growth defect in fermentable rich media. CONCLUSIONS: Overall, our results illustrate how differences in the biology of two genomes coexisting in the same cells can lead to discordant evolutionary histories.


Subject(s)
Cell Nucleus/genetics , Evolution, Molecular , Genetic Variation , Genome, Fungal , Genome, Mitochondrial , Saccharomyces cerevisiae/genetics , Phylogeny
7.
Genome Res ; 30(5): 697-710, 2020 05.
Article in English | MEDLINE | ID: mdl-32277013

ABSTRACT

Aging varies among individuals due to both genetics and environment, but the underlying molecular mechanisms remain largely unknown. Using a highly recombined Saccharomyces cerevisiae population, we found 30 distinct quantitative trait loci (QTLs) that control chronological life span (CLS) in calorie-rich and calorie-restricted environments and under rapamycin exposure. Calorie restriction and rapamycin extended life span in virtually all genotypes but through different genetic variants. We tracked the two major QTLs to the cell wall glycoprotein genes FLO11 and HPF1 We found that massive expansion of intragenic tandem repeats within the N-terminal domain of HPF1 was sufficient to cause pronounced life span shortening. Life span impairment by HPF1 was buffered by rapamycin but not by calorie restriction. The HPF1 repeat expansion shifted yeast cells from a sedentary to a buoyant state, thereby increasing their exposure to surrounding oxygen. The higher oxygenation altered methionine, lipid, and purine metabolism, and inhibited quiescence, which explains the life span shortening. We conclude that fast-evolving intragenic repeat expansions can fundamentally change the relationship between cells and their environment with profound effects on cellular lifestyle and longevity.


Subject(s)
DNA Repeat Expansion , Saccharomyces cerevisiae Proteins/genetics , Cell Wall , Genes, Fungal , Lipid Metabolism , Membrane Glycoproteins/genetics , Methionine/metabolism , Purines/metabolism , Quantitative Trait Loci , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sirolimus/pharmacology
8.
Mol Biol Evol ; 36(4): 691-708, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30657986

ABSTRACT

Pre-existing and de novo genetic variants can both drive adaptation to environmental changes, but their relative contributions and interplay remain poorly understood. Here we investigated the evolutionary dynamics in drug-treated yeast populations with different levels of pre-existing variation by experimental evolution coupled with time-resolved sequencing and phenotyping. We found a doubling of pre-existing variation alone boosts the adaptation by 64.1% and 51.5% in hydroxyurea and rapamycin, respectively. The causative pre-existing and de novo variants were selected on shared targets: RNR4 in hydroxyurea and TOR1, TOR2 in rapamycin. Interestingly, the pre-existing and de novo TOR variants map to different functional domains and act via distinct mechanisms. The pre-existing TOR variants from two domesticated strains exhibited opposite rapamycin resistance effects, reflecting lineage-specific functional divergence. This study provides a dynamic view on how pre-existing and de novo variants interactively drive adaptation and deepens our understanding of clonally evolving populations.


Subject(s)
Biological Evolution , Drug Resistance, Fungal/genetics , Saccharomyces cerevisiae/genetics , Cell Cycle Proteins/genetics , Hydroxyurea , Mutation , Phosphatidylinositol 3-Kinases/genetics , Quantitative Trait Loci , Saccharomyces cerevisiae Proteins/genetics , Selection, Genetic , Sirolimus
9.
Nature ; 556(7701): 339-344, 2018 04.
Article in English | MEDLINE | ID: mdl-29643504

ABSTRACT

Large-scale population genomic surveys are essential to explore the phenotypic diversity of natural populations. Here we report the whole-genome sequencing and phenotyping of 1,011 Saccharomyces cerevisiae isolates, which together provide an accurate evolutionary picture of the genomic variants that shape the species-wide phenotypic landscape of this yeast. Genomic analyses support a single 'out-of-China' origin for this species, followed by several independent domestication events. Although domesticated isolates exhibit high variation in ploidy, aneuploidy and genome content, genome evolution in wild isolates is mainly driven by the accumulation of single nucleotide polymorphisms. A common feature is the extensive loss of heterozygosity, which represents an essential source of inter-individual variation in this mainly asexual species. Most of the single nucleotide polymorphisms, including experimentally identified functional polymorphisms, are present at very low frequencies. The largest numbers of variants identified by genome-wide association are copy-number changes, which have a greater phenotypic effect than do single nucleotide polymorphisms. This resource will guide future population genomics and genotype-phenotype studies in this classic model system.


Subject(s)
Evolution, Molecular , Genetic Variation , Genome, Fungal/genetics , Saccharomyces cerevisiae/classification , Saccharomyces cerevisiae/genetics , Alleles , Aneuploidy , China , DNA Copy Number Variations , Genetic Association Studies , Genome-Wide Association Study , Genomics , Loss of Heterozygosity , Phenotype , Phylogeny , Phylogeography , Ploidies , Polymorphism, Single Nucleotide , Saccharomyces cerevisiae/isolation & purification , Sequence Analysis, DNA
10.
Cell Rep ; 21(3): 732-744, 2017 Oct 17.
Article in English | MEDLINE | ID: mdl-29045840

ABSTRACT

The joint contribution of pre-existing and de novo genetic variation to clonal adaptation is poorly understood but essential to designing successful antimicrobial or cancer therapies. To address this, we evolve genetically diverse populations of budding yeast, S. cerevisiae, consisting of diploid cells with unique haplotype combinations. We study the asexual evolution of these populations under selective inhibition with chemotherapeutic drugs by time-resolved whole-genome sequencing and phenotyping. All populations undergo clonal expansions driven by de novo mutations but remain genetically and phenotypically diverse. The clones exhibit widespread genomic instability, rendering recessive de novo mutations homozygous and refining pre-existing variation. Finally, we decompose the fitness contributions of pre-existing and de novo mutations by creating a large recombinant library of adaptive mutations in an ensemble of genetic backgrounds. Both pre-existing and de novo mutations substantially contribute to fitness, and the relative fitness of pre-existing variants sets a selective threshold for new adaptive mutations.


Subject(s)
Mutation/genetics , Saccharomyces cerevisiae/genetics , Clone Cells , Gene Frequency/genetics , Genetic Fitness , Genome, Fungal , Genomic Instability , Loss of Heterozygosity , Selection, Genetic
11.
Mol Biol Evol ; 31(4): 872-88, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24425782

ABSTRACT

The question of how genetic variation in a population influences phenotypic variation and evolution is of major importance in modern biology. Yet much is still unknown about the relative functional importance of different forms of genome variation and how they are shaped by evolutionary processes. Here we address these questions by population level sequencing of 42 strains from the budding yeast Saccharomyces cerevisiae and its closest relative S. paradoxus. We find that genome content variation, in the form of presence or absence as well as copy number of genetic material, is higher within S. cerevisiae than within S. paradoxus, despite genetic distances as measured in single-nucleotide polymorphisms being vastly smaller within the former species. This genome content variation, as well as loss-of-function variation in the form of premature stop codons and frameshifting indels, is heavily enriched in the subtelomeres, strongly reinforcing the relevance of these regions to functional evolution. Genes affected by these likely functional forms of variation are enriched for functions mediating interaction with the external environment (sugar transport and metabolism, flocculation, metal transport, and metabolism). Our results and analyses provide a comprehensive view of genomic diversity in budding yeast and expose surprising and pronounced differences between the variation within S. cerevisiae and that within S. paradoxus. We also believe that the sequence data and de novo assemblies will constitute a useful resource for further evolutionary and population genomics studies.


Subject(s)
Genes, Fungal , Saccharomyces cerevisiae/genetics , Arsenites/pharmacology , DNA Copy Number Variations , Drug Resistance, Fungal/genetics , Evolution, Molecular , Genetic Linkage , Genetic Speciation , Genome, Fungal , Molecular Sequence Annotation , Multigene Family , Phylogeny , Polymorphism, Single Nucleotide , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/growth & development , Sequence Analysis, DNA , Sodium Compounds/pharmacology
12.
JAKSTAT ; 2(3): e24353, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-24069557

ABSTRACT

Signal transducers and activators of transcription 3 (STAT3) proteins are cytoplasmic transcription factors that translocate into the nucleus to induce transcription following growth factor or cytokine stimulation. Besides their normal functions, these proteins play an important role in cancer cells through the abnormal activation of cell cycle progression and the deregulation of survival and senescence pathways. New data obtained from the laboratory of Guido Kroemer identifies STAT3 as a new autophagy regulator. In the cytoplasm, in the absence of conventional phosphorylation on the tyrosine 705 residue, STAT3 interacts with the PKR kinase to inhibit eIF2A phosphorylation and so reduce autophagic pathways. This new and nonconventional function of STAT3 has an important role in normal cells but we suggest that it might also affect cancer cells and the response to chemotherapy treatment.

13.
JAKSTAT ; 2(3): e24716, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-24069560

ABSTRACT

Aberrant activation of the STAT3 transcription factor has been reported in a large group of tumors and a strong biological basis now defines this protein as an oncogenic driver. Consequently, STAT3 is considered to be a promising target in the field of cancer therapy. For its inhibition to result in a successful therapeutic approach, the definition of a target tumor population identified by specific and detectable alterations is critical. The canonical activation model of STAT3 relies on a constitutive phosphorylation on its 705 tyrosine site, resulting in its dimerization, nuclear translocation, and the consequent activation of cancer genes. Therefore, it is expected that tumors expressing this phosphorylated form are addicted to STAT3 and will be sensitive to existing drugs which are targeting this dimeric form. However, recent results have shown that STAT3 can function as an oncogene in the absence of this tyrosine phosphorylation. This indicates that different forms of the transcription factor also play an important role in tumor growth and chemotherapy resistance. This complicates the definition of STAT3 as an oncogene and as a potential prognosis and predictive biomarker. The obligation to target a defined tumor type implies that future clinical trials should use a precise definition of STAT3 activation. This will allow tumors addicted to this oncogene to be identified correctly, leading to a strong rationale for patient stratification.

14.
JAKSTAT ; 2(1): e22996, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-24058790

ABSTRACT

Cell communication is well known to rely on direct contacts or on secreted factors that bind to receptors located on the surface of their target cells. In addition to this classical pathway, recent results have shown that cells produce microvesicles that contain functional DNA, RNA and proteins that can be directly transferred to recipient cells. This induces proliferation, differentiation or cell death to the same extent as classical soluble factors. New data obtained from the laboratory of Napoleone Ferrara show that these microvesicles also contain miRNAs that can induce angiogenic activities in neighboring endothelial cells. When secreted from cancer cells, these miRNA-loaded vesicles penetrate recipient cells where they activate the JAK-STAT pathway. This represents a new type of intercellular signaling and a new way of activating the STAT transcription factors that could be of interest for the design of cancer treatments.

15.
PLoS Biol ; 10(9): e1001395, 2012.
Article in English | MEDLINE | ID: mdl-23055831

ABSTRACT

Interleukin-26 (IL-26), a member of the IL-10 cytokine family, induces the production of proinflammatory cytokines by epithelial cells. IL-26 has been also reported overexpressed in Crohn's disease, suggesting that it may be involved in the physiopathology of chronic inflammatory disorders. Here, we have analyzed the expression and role of IL-26 in rheumatoid arthritis (RA), a chronic inflammatory disorder characterized by joint synovial inflammation. We report that the concentrations of IL-26 are higher in the serums of RA patients than of healthy subjects and dramatically elevated in RA synovial fluids compared to RA serums. Immunohistochemistry reveals that synoviolin(+) fibroblast-like synoviocytes and CD68(+) macrophage-like synoviocytes are the main IL-26-producing cells in RA joints. Fibroblast-like synoviocytes from RA patients constitutively produce IL-26 and this production is upregulated by IL-1-beta and IL-17A. We have therefore investigated the role of IL-26 in the inflammatory process. Results show that IL-26 induces the production of the proinflammatory cytokines IL-1-beta, IL-6, and tumor necrosis factor (TNF)-alpha by human monocytes and also upregulates the expression of numerous chemokines (mainly CCL20). Interestingly, IL-26-stimulated monocytes selectively promote the generation of RORgamma t(+) Th17 cells, through IL-1-beta secretion by monocytes. More precisely, IL-26-stimulated monocytes switch non-Th17 committed (IL-23R(-) or CCR6(-) CD161(-)) CD4(+) memory T cells into Th17 cells. Finally, synovial fluids from RA patients also induce Th17 cell generation and this effect is reduced after IL-26 depletion. These findings show that IL-26 is constitutively produced by RA synoviocytes, induces proinflammatory cytokine secretion by myeloid cells, and favors Th17 cell generation. IL-26 thereby appears as a novel proinflammatory cytokine, located upstream of the proinflammatory cascade, that may constitute a promising target to treat RA and chronic inflammatory disorders.


Subject(s)
Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Cytokines/biosynthesis , Inflammation Mediators/metabolism , Interleukins/metabolism , Th17 Cells/immunology , Arthritis, Rheumatoid/blood , Cytokines/metabolism , Demography , Female , Fibroblasts/immunology , Fibroblasts/pathology , Humans , Immunohistochemistry , Immunologic Memory , Interleukin-17/metabolism , Interleukin-1beta/metabolism , Interleukins/blood , Joints/immunology , Joints/pathology , Male , Middle Aged , Models, Immunological , Monocytes/metabolism , Myeloid Cells/metabolism , Synovial Fluid/metabolism , Synovial Membrane/metabolism , Synovial Membrane/pathology
16.
Mol Cell Proteomics ; 10(12): M111.009712, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21986994

ABSTRACT

Expression profiles represent new molecular tools that are useful to characterize the successive steps of tumor progression and the prediction of recurrence or chemotherapy response. In this study, we have used quantitative proteomic analysis to compare different stages of colorectal cancer. A combination of laser microdissection, OFFGEL separation, iTRAQ labeling, and MALDI-TOF/TOF MS was used to explore the proteome of 28 colorectal cancer tissues. Two software packages were used for identification and quantification of differentially expressed proteins: Protein Pilot and iQuantitator. Based on ∼1,190,702 MS/MS spectra, a total of 3138 proteins were identified, which represents the largest database of colorectal cancer realized to date and demonstrates the value of our quantitative proteomic approach. In this way, individual protein expression and variation have been identified for each patient and for each colorectal dysplasia and cancer stage (stages I-IV). A total of 555 proteins presenting a significant fold change were quantified in the different stages, and this differential expression correlated with immunohistochemistry results reported in the Human Protein Atlas database. To identify a candidate biomarker of the early stages of colorectal cancer, we focused our study on secreted proteins. In this way, we identified olfactomedin-4, which was overexpressed in adenomas and in early stages of colorectal tumors. This early stage overexpression was confirmed by immunohistochemistry in 126 paraffin-embedded tissues. Our results also indicate that OLFM4 is regulated by the Ras-NF-κB2 pathway, one of the main oncogenic pathways deregulated in colorectal tumors.


Subject(s)
Adenocarcinoma/pathology , Adenoma/pathology , Biomarkers, Tumor/metabolism , Carcinoma/pathology , Colorectal Neoplasms/pathology , Granulocyte Colony-Stimulating Factor/metabolism , Adenocarcinoma/metabolism , Adenoma/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Carcinoma/metabolism , Colorectal Neoplasms/metabolism , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Glycosylation , Granulocyte Colony-Stimulating Factor/genetics , HT29 Cells , Humans , Laser Capture Microdissection , Male , Middle Aged , Neoplasm Staging , Proteome/metabolism , Proteomics , Reproducibility of Results , ras Proteins/genetics , ras Proteins/metabolism
17.
Mol Cancer ; 10: 110, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21899728

ABSTRACT

BACKGROUND: Anti-apoptotic signals induced downstream of HER2 are known to contribute to the resistance to current treatments of breast cancer cells that overexpress this member of the EGFR family. Whether or not some of these signals are also involved in tumor maintenance by counteracting constitutive death signals is much less understood. To address this, we investigated what role anti- and pro-apoptotic Bcl-2 family members, key regulators of cancer cell survival, might play in the viability of HER2 overexpressing breast cancer cells. METHODS: We used cell lines as an in vitro model of HER2-overexpressing cells in order to evaluate how anti-apoptotic Bcl-2, Bcl-xL and Mcl-1, and pro-apoptotic Puma and Bim impact on their survival, and to investigate how the constitutive expression of these proteins is regulated. Expression of the proteins of interest was confirmed using lysates from HER2-overexpressing tumors and through analysis of publicly available RNA expression data. RESULTS: We show that the depletion of Mcl-1 is sufficient to induce apoptosis in HER2-overexpressing breast cancer cells. This Mcl-1 dependence is due to Bim expression and it directly results from oncogenic signaling, as depletion of the oncoprotein c-Myc, which occupies regions of the Bim promoter as evaluated in ChIP assays, decreases Bim levels and mitigates Mcl-1 dependence. Consistently, a reduction of c-Myc expression by inhibition of mTORC1 activity abrogates occupancy of the Bim promoter by c-Myc, decreases Bim expression and promotes tolerance to Mcl-1 depletion. Western blot analysis confirms that naïve HER2-overexpressing tumors constitutively express detectable levels of Mcl-1 and Bim, while expression data hint on enrichment for Mcl-1 transcripts in these tumors. CONCLUSIONS: This work establishes that, in HER2-overexpressing tumors, it is necessary, and maybe sufficient, to therapeutically impact on the Mcl-1/Bim balance for efficient induction of cancer cell death.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, ErbB-2/metabolism , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Breast Neoplasms , Cell Aggregation , Cell Line, Tumor , Cell Survival , Everolimus , Female , Gene Expression , Gene Knockdown Techniques , Humans , Mechanistic Target of Rapamycin Complex 1 , Membrane Proteins/genetics , Multiprotein Complexes , Myeloid Cell Leukemia Sequence 1 Protein , Promoter Regions, Genetic , Proteins/antagonists & inhibitors , Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Interference , Signal Transduction , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , bcl-X Protein/genetics , bcl-X Protein/metabolism
18.
Mol Biol Cell ; 22(17): 3032-40, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21737676

ABSTRACT

The NF-κB family of transcription factors is a well-established regulator of the immune and inflammatory responses and also plays a key role in other cellular processes, including cell death, proliferation, and migration. Conserved residues in the trans-activation domain of RelA, which can be posttranslationally modified, regulate divergent NF-κB functions in response to different cellular stimuli. Using rela(-/-) mouse embryonic fibroblasts reconstituted with RelA, we find that mutation of the threonine 505 (T505) phospho site to alanine has wide-ranging effects on NF-κB function. These include previously described effects on chemotherapeutic drug-induced apoptosis, as well as new roles for this modification in autophagy, cell proliferation, and migration. This last effect was associated with alterations in the actin cytoskeleton and expression of cellular migration-associated genes such as WAVE3 and α-actinin 4. We also define a new component of cisplatin-induced, RelA T505-dependent apoptosis, involving induction of NOXA gene expression, an effect explained at least in part through induction of the p53 homologue, p73. Therefore, in contrast to other RelA phosphorylation events, which positively regulate NF-κB function, we identified RelA T505 phosphorylation as a negative regulator of its ability to induce diverse cellular processes such as apoptosis, autophagy, proliferation, and migration.


Subject(s)
Cell Movement , Cell Proliferation , Cell Survival , Fibroblasts/physiology , Threonine/metabolism , Transcription Factor RelA/metabolism , Actin Cytoskeleton/metabolism , Actinin/genetics , Actinin/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line , Cisplatin/pharmacology , Fibroblasts/metabolism , Gene Expression , Gene Expression Regulation , Gene Knockout Techniques , Mice , Mutagenesis, Site-Directed , Mutation, Missense , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription Factor RelA/genetics , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/physiology , Wiskott-Aldrich Syndrome Protein Family/genetics , Wiskott-Aldrich Syndrome Protein Family/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
19.
J Biol Chem ; 286(15): 12825-38, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21292770

ABSTRACT

Oncogene-induced senescence (OIS) is a tumor suppressor response that induces permanent cell cycle arrest in response to oncogenic signaling. Through the combined activation of the p53-p21 and p16-Rb suppressor pathways, OIS leads to the transcriptional repression of proliferative genes. Although this protective mechanism has been essentially described in primary cells, we surprisingly observed in this study that the OIS program is conserved in established colorectal cell lines. In response to the RAS oncogene and despite the inactivation of p53 and p16(INK4), HT29 cells enter senescence, up-regulate p21(WAF1), and induce senescence-associated heterochromatin foci formation. The same effect was observed in response to B-RAF(v600E) in LS174T cells. We also observed that p21(WAF1) prevents the expression of the CDC25A and PLK1 genes to induce cell cycle arrest. Using ChIP and luciferase experiments, we have observed that p21(WAF1) binds to the PLK1 promoter to induce its down-regulation during OIS induction. Following 4-5 weeks, several clones were able to resume proliferation and escape this tumor suppressor pathway. Tumor progression was associated with p21(WAF1) down-regulation and CDC25A and PLK1 reexpression. In addition, OIS and p21(WAF1) escape was associated with an increase in DNA damage, an induction of the epithelial-mesenchymal transition program, and an increase in the proportion of cells expressing the CD24(low)/CD44(high) phenotype. Results also indicate that malignant cells having escaped OIS rely on survival pathways induced by Bcl-xL/MCL1 signaling. In light of these observations, it appears that the transcriptional functions of p21(WAF1) are active during OIS and that the inactivation of this protein is associated with cell dedifferentiation and enhanced survival.


Subject(s)
Cell Dedifferentiation , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Heterochromatin/genetics , Heterochromatin/metabolism , Humans , Mutation, Missense , Myeloid Cell Leukemia Sequence 1 Protein , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction/genetics , Time Factors , Transcription, Genetic/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation/genetics , bcl-X Protein/genetics , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism , Polo-Like Kinase 1
SELECTION OF CITATIONS
SEARCH DETAIL
...