47 results on '"Tabach Y"'
Search Results
2. Mutant p53R175H upregulates Twist1 expression and promotes epithelial–mesenchymal transition in immortalized prostate cells
- Author
-
Kogan-Sakin, I, primary, Tabach, Y, additional, Buganim, Y, additional, Molchadsky, A, additional, Solomon, H, additional, Madar, S, additional, Kamer, I, additional, Stambolsky, P, additional, Shelly, A, additional, Goldfinger, N, additional, Valsesia-Wittmann, S, additional, Puisieux, A, additional, Zundelevich, A, additional, Gal-Yam, E N, additional, Avivi, C, additional, Barshack, I, additional, Brait, M, additional, Sidransky, D, additional, Domany, E, additional, and Rotter, V, additional
- Published
- 2010
- Full Text
- View/download PDF
3. Mutant p53R175H upregulates Twist1 expression and promotes epithelial-mesenchymal transition in immortalized prostate cells.
- Author
-
Kogan-Sakin, I., Tabach, Y., Buganim, Y., Molchadsky, A., Solomon, H., Madar, S., Kamer, I., Stambolsky, P., Shelly, A., Goldfinger, N., Valsesia-Wittmann, S., Puisieux, A., Zundelevich, A., Gal-Yam, E. N., Avivi, C., Barshack, I., Brait, M., Sidransky, D., Domany, E., and Rotter, V.
- Subjects
- *
TUMOR suppressor genes , *PROSTATE cancer , *GENE expression , *CELL cycle regulation , *EPITHELIAL cells , *GENETIC markers , *GENETIC mutation - Abstract
A mutation within one allele of the p53 tumor suppressor gene can inactivate the remaining wild-type allele in a dominant-negative manner and in some cases can exert an additional oncogenic activity, known as mutant p53 'gain of function' (GOF). To study the role of p53 mutations in prostate cancer and to discriminate between the dominant-negative effect and the GOF activity of mutant p53, we measured, using microarrays, the expression profiles of three immortalized prostate epithelial cultures expressing wild-type, inactivated p53 or mutated p53. Analysis of these gene expression profiles showed that both inactivated p53 and p53R175H mutant expression resulted in the upregulation of cell cycle progression genes. A second group, which was upregulated exclusively by mutant p53R175H, was predominantly enriched in developmental genes. This group of genes included the Twist1, a regulator of metastasis and epithelial-mesenchymal transition (EMT). Twist1 levels were also elevated in metastatic prostate cancer-derived cell line DU145, in immortalized lung fibroblasts and in a subset of lung cancer samples, all in a mutant p53-dependent manner. p53R175H mutant bearing immortalized epithelial cells showed typical features of EMT, such as higher expression of mesenchymal markers, lower expression of epithelial markers and enhanced invasive properties in vitro. The mechanism by which p53R175H mutant induces Twist1 expression involves alleviation of the epigenetic repression. Our data suggest that Twist1 expression might be upregulated following p53 mutation in cancer cells. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
4. Mucosal Genes Encoding Clock, Inflammation and Their Mutual Regulators Are Disrupted in Pediatric Patients with Active Ulcerative Colitis.
- Author
-
Labes S, Froy O, Tabach Y, Shamir R, Shouval DS, and Weintraub Y
- Subjects
- Child, Humans, ARNTL Transcription Factors genetics, Circadian Rhythm physiology, Inflammation genetics, Interleukin-10, Interleukin-6, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha, PPAR alpha, PPAR gamma, Tumor Necrosis Factor-alpha, Period Circadian Proteins genetics, Period Circadian Proteins metabolism, Cryptochromes genetics, Cryptochromes metabolism, Colitis, Ulcerative genetics, CLOCK Proteins genetics, CLOCK Proteins metabolism
- Abstract
Patients with active ulcerative colitis (UC) display a misalignment of the circadian clock, which plays a vital role in various immune functions. Our aim was to characterize the expression of clock and inflammation genes, and their mutual regulatory genes in treatment-naïve pediatric patients with UC. Using the Inflammatory Bowel Disease Transcriptome and Metatranscriptome Meta-Analysis (IBD TaMMA) platform and R algorithms, we analyzed rectal biopsy transcriptomic data from two cohorts (206 patients with UC vs. 20 healthy controls from the GSE-109142 study, and 43 patients with UC vs. 55 healthy controls from the GSE-117993 study). We compared gene expression levels and correlation of clock genes ( BMAL1 , CLOCK , PER1 , PER2 , CRY1 , CRY2 ), inflammatory genes ( IκB , IL10 , NFκB1 , NFκB2 , IL6 , TNFα ) and their mutual regulatory genes ( RORα , RORγ , REV-ERBα , PGC1α , PPARα , PPARγ , AMPK , SIRT1 ) in patients with active UC and healthy controls. The clock genes BMAL1 , CLOCK , PER1 and CRY1 and the inflammatory genes IκB , IL10 , NFκB1 , NFκB2 , IL6 and TNFα were significantly upregulated in patients with active UC. The genes encoding the mutual regulators RORα , RORγ , PGC1α , PPARα and PPARγ were significantly downregulated in patients with UC. A uniform pattern of gene expression was found in healthy controls compared to the highly variable expression pattern in patients with UC. Among the healthy controls, inflammatory genes were positively correlated with clock genes and they all showed reduced expression. The difference in gene expression levels was associated with disease severity and endoscopic score but not with histological score. In patients with active UC, clock gene disruption is associated with abnormal mucosal immune response. Disrupted expression of genes encoding clock, inflammation and their mutual regulators together may play a role in active UC.
- Published
- 2024
- Full Text
- View/download PDF
5. Using multi-scale genomics to associate poorly annotated genes with rare diseases.
- Author
-
Canavati C, Sherill-Rofe D, Kamal L, Bloch I, Zahdeh F, Sharon E, Terespolsky B, Allan IA, Rabie G, Kawas M, Kassem H, Avraham KB, Renbaum P, Levy-Lahad E, Kanaan M, and Tabach Y
- Subjects
- Humans, Animals, Mice, Phylogeny, Phenotype, Exome, 1-Acylglycerophosphocholine O-Acyltransferase genetics, Rare Diseases genetics, Genomics methods
- Abstract
Background: Next-generation sequencing (NGS) has significantly transformed the landscape of identifying disease-causing genes associated with genetic disorders. However, a substantial portion of sequenced patients remains undiagnosed. This may be attributed not only to the challenges posed by harder-to-detect variants, such as non-coding and structural variations but also to the existence of variants in genes not previously associated with the patient's clinical phenotype. This study introduces EvORanker, an algorithm that integrates unbiased data from 1,028 eukaryotic genomes to link mutated genes to clinical phenotypes., Methods: EvORanker utilizes clinical data, multi-scale phylogenetic profiling, and other omics data to prioritize disease-associated genes. It was evaluated on solved exomes and simulated genomes, compared with existing methods, and applied to 6260 knockout genes with mouse phenotypes lacking human associations. Additionally, EvORanker was made accessible as a user-friendly web tool., Results: In the analyzed exomic cohort, EvORanker accurately identified the "true" disease gene as the top candidate in 69% of cases and within the top 5 candidates in 95% of cases, consistent with results from the simulated dataset. Notably, EvORanker outperformed existing methods, particularly for poorly annotated genes. In the case of the 6260 knockout genes with mouse phenotypes, EvORanker linked 41% of these genes to observed human disease phenotypes. Furthermore, in two unsolved cases, EvORanker successfully identified DLGAP2 and LPCAT3 as disease candidates for previously uncharacterized genetic syndromes., Conclusions: We highlight clade-based phylogenetic profiling as a powerful systematic approach for prioritizing potential disease genes. Our study showcases the efficacy of EvORanker in associating poorly annotated genes to disease phenotypes observed in patients. The EvORanker server is freely available at https://ccanavati.shinyapps.io/EvORanker/ ., (© 2023. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. Developmental and temporal changes in petunia petal transcriptome reveal scent-repressing plant-specific RING-kinase-WD40 protein.
- Author
-
Shor E, Skaliter O, Sharon E, Kitsberg Y, Bednarczyk D, Kerzner S, Vainstein D, Tabach Y, and Vainstein A
- Abstract
In moth-pollinated petunias, production of floral volatiles initiates when the flower opens and occurs rhythmically during the day, for optimal flower-pollinator interaction. To characterize the developmental transcriptomic response to time of day, we generated RNA-Seq databases for corollas of floral buds and mature flowers in the morning and in the evening. Around 70% of transcripts accumulating in petals demonstrated significant changes in expression levels in response to the flowers' transition from a 4.5-cm bud to a flower 1 day postanthesis (1DPA). Overall, 44% of the petal transcripts were differentially expressed in the morning vs. evening. Morning/evening changes were affected by flower developmental stage, with a 2.5-fold larger transcriptomic response to daytime in 1DPA flowers compared to buds. Analyzed genes known to encode enzymes in volatile organic compound biosynthesis were upregulated in 1DPA flowers vs. buds-in parallel with the activation of scent production. Based on analysis of global changes in the petal transcriptome, PhWD2 was identified as a putative scent-related factor. PhWD2 is a protein that is uniquely present in plants and has a three-domain structure: RING-kinase-WD40. Suppression of PhWD2 (termed UPPER - Unique Plant PhEnylpropanoid Regulator ) resulted in a significant increase in the levels of volatiles emitted from and accumulated in internal pools, suggesting that it is a negative regulator of petunia floral scent production., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Shor, Skaliter, Sharon, Kitsberg, Bednarczyk, Kerzner, Vainstein, Tabach and Vainstein.)
- Published
- 2023
- Full Text
- View/download PDF
7. An Exercise-Induced Metabolic Shield in Distant Organs Blocks Cancer Progression and Metastatic Dissemination.
- Author
-
Sheinboim D, Parikh S, Manich P, Markus I, Dahan S, Parikh R, Stubbs E, Cohen G, Zemser-Werner V, Bell RE, Ruiz SA, Percik R, Brenner R, Leibou S, Vaknine H, Arad G, Gerber Y, Keinan-Boker L, Shimony T, Bikovski L, Goldstein N, Constantini K, Labes S, Mordechai S, Doron H, Lonescu A, Ziv T, Nizri E, Choshen G, Eldar-Finkelman H, Tabach Y, Helman A, Ben-Eliyahu S, Erez N, Perlson E, Geiger T, Ben-Zvi D, Khaled M, Gepner Y, and Levy C
- Subjects
- Animals, Humans, Mice, Glucose metabolism, Prospective Studies, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases metabolism, Melanoma genetics, Melanoma metabolism, Melanoma pathology, Proteomics, Exercise physiology, Nutrients genetics, Nutrients metabolism
- Abstract
Exercise prevents cancer incidence and recurrence, yet the underlying mechanism behind this relationship remains mostly unknown. Here we report that exercise induces the metabolic reprogramming of internal organs that increases nutrient demand and protects against metastatic colonization by limiting nutrient availability to the tumor, generating an exercise-induced metabolic shield. Proteomic and ex vivo metabolic capacity analyses of murine internal organs revealed that exercise induces catabolic processes, glucose uptake, mitochondrial activity, and GLUT expression. Proteomic analysis of routinely active human subject plasma demonstrated increased carbohydrate utilization following exercise. Epidemiologic data from a 20-year prospective study of a large human cohort of initially cancer-free participants revealed that exercise prior to cancer initiation had a modest impact on cancer incidence in low metastatic stages but significantly reduced the likelihood of highly metastatic cancer. In three models of melanoma in mice, exercise prior to cancer injection significantly protected against metastases in distant organs. The protective effects of exercise were dependent on mTOR activity, and inhibition of the mTOR pathway with rapamycin treatment ex vivo reversed the exercise-induced metabolic shield. Under limited glucose conditions, active stroma consumed significantly more glucose at the expense of the tumor. Collectively, these data suggest a clash between the metabolic plasticity of cancer and exercise-induced metabolic reprogramming of the stroma, raising an opportunity to block metastasis by challenging the metabolic needs of the tumor., Significance: Exercise protects against cancer progression and metastasis by inducing a high nutrient demand in internal organs, indicating that reducing nutrient availability to tumor cells represents a potential strategy to prevent metastasis. See related commentary by Zerhouni and Piskounova, p. 4124., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
8. Systematic proximal mapping of the classical RAD51 paralogs unravel functionally and clinically relevant interactors for genome stability.
- Author
-
Simo Cheyou E, Boni J, Boulais J, Pinedo-Carpio E, Malina A, Sherill-Rofe D, Luo VM, Goncalves C, Bagci H, Maters A, Cuella-Martin R, Tabach Y, Del Rincon S, Côté JF, Rivera B, and Orthwein A
- Subjects
- Animals, Humans, Homologous Recombination genetics, DNA Breaks, Double-Stranded, RNA, DNA Repair genetics, Mammals genetics, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Rad51 Recombinase genetics, Rad51 Recombinase metabolism, Genomic Instability genetics
- Abstract
Homologous recombination (HR) plays an essential role in the maintenance of genome stability by promoting the repair of cytotoxic DNA double strand breaks (DSBs). More recently, the HR pathway has emerged as a core component of the response to replication stress, in part by protecting stalled replication forks from nucleolytic degradation. In that regard, the mammalian RAD51 paralogs (RAD51B, RAD51C, RAD51D, XRCC2, and XRCC3) have been involved in both HR-mediated DNA repair and collapsed replication fork resolution. Still, it remains largely obscure how they participate in both processes, thereby maintaining genome stability and preventing cancer development. To gain better insight into their contribution in cellulo, we mapped the proximal interactome of the classical RAD51 paralogs using the BioID approach. Aside from identifying the well-established BCDX2 and CX3 sub-complexes, the spliceosome machinery emerged as an integral component of our proximal mapping, suggesting a crosstalk between this pathway and the RAD51 paralogs. Furthermore, we noticed that factors involved RNA metabolic pathways are significantly modulated within the BioID of the classical RAD51 paralogs upon exposure to hydroxyurea (HU), pointing towards a direct contribution of RNA processing during replication stress. Importantly, several members of these pathways have prognostic potential in breast cancer (BC), where their RNA expression correlates with poorer patient outcome. Collectively, this study uncovers novel functionally relevant partners of the different RAD51 paralogs in the maintenance of genome stability that could be used as biomarkers for the prognosis of BC., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2022 Simo Cheyou et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2022
- Full Text
- View/download PDF
9. Transcriptome changes in DM1 patients' tissues are governed by the RNA interference pathway.
- Author
-
Braun M, Shoshani S, and Tabach Y
- Abstract
Myotonic dystrophy type 1 (DM1) is a multisystemic disease caused by pathogenic expansions of CTG repeats. The expanded repeats are transcribed to long RNA and induce cellular toxicity. Recent studies suggest that the CUG repeats are processed by the RNA interference (RNAi) pathway to generate small interfering repeated RNA (siRNA). However, the effects of the CTG repeat-derived siRNAs remain unclear. We hypothesize that the RNAi machinery in DM1 patients generates distinct gene expression patterns that determine the disease phenotype in the individual patient. The abundance of genes with complementary repeats that are targeted by siRNAs in each tissue determines the way that the tissue is affected in DM1. We integrated and analyzed published transcriptome data from muscle, heart, and brain biopsies of DM1 patients, and revealed shared, characteristic changes that correlated with disease phenotype. These signatures are overrepresented by genes and transcription factors bearing endogenous CTG/CAG repeats and are governed by aberrant activity of the RNAi machinery, miRNAs, and a specific gain-of-function of the CTG repeats. Computational analysis of the DM1 transcriptome enhances our understanding of the complex pathophysiology of the disease and may reveal a path for cure., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Braun, Shoshani and Tabach.)
- Published
- 2022
- Full Text
- View/download PDF
10. Cross-species identification of cancer resistance-associated genes that may mediate human cancer risk.
- Author
-
Nair NU, Cheng K, Naddaf L, Sharon E, Pal LR, Rajagopal PS, Unterman I, Aldape K, Hannenhalli S, Day CP, Tabach Y, and Ruppin E
- Subjects
- Animals, Humans, Loss of Function Mutation, Mammals, Mice, Genomics, Neoplasms genetics
- Abstract
Cancer is a predominant disease across animals. We applied a comparative genomics approach to systematically characterize genes whose conservation levels correlate positively (PC) or negatively (NC) with cancer resistance estimates across 193 vertebrates. Pathway analysis reveals that NC genes are enriched for metabolic functions and PC genes in cell cycle regulation, DNA repair, and immune response, pointing to their corresponding roles in mediating cancer risk. We find that PC genes are less tolerant to loss-of-function (LoF) mutations, are enriched in cancer driver genes, and are associated with germline mutations that increase human cancer risk. Their relevance to cancer risk is further supported via the analysis of mouse functional genomics and cancer mortality of zoo mammals' data. In sum, our study describes a cross-species genomic analysis pointing to candidate genes that may mediate human cancer risk.
- Published
- 2022
- Full Text
- View/download PDF
11. DEPCOD: a tool to detect and visualize co-evolution of protein domains.
- Author
-
Ji F, Bonilla G, Krykbaev R, Ruvkun G, Tabach Y, and Sadreyev RI
- Subjects
- Amino Acid Sequence, Phylogeny, Evolution, Molecular, Protein Domains, Proteins genetics, Software
- Abstract
Proteins with similar phylogenetic patterns of conservation or loss across evolutionary taxa are strong candidates to work in the same cellular pathways or engage in physical or functional interactions. Our previously published tools implemented our method of normalized phylogenetic sequence profiling to detect functional associations between non-homologous proteins. However, many proteins consist of multiple protein domains subjected to different selective pressures, so using protein domain as the unit of analysis improves the detection of similar phylogenetic patterns. Here we analyze sequence conservation patterns across the whole tree of life for every protein domain from a set of widely studied organisms. The resulting new interactive webserver, DEPCOD (DEtection of Phylogenetically COrrelated Domains), performs searches with either a selected pre-defined protein domain or a user-supplied sequence as a query to detect other domains from the same organism that have similar conservation patterns. Top similarities on two evolutionary scales (the whole tree of life or eukaryotic genomes) are displayed along with known protein interactions and shared complexes, pathway enrichment among the hits, and detailed visualization of sources of detected similarities. DEPCOD reveals functional relationships between often non-homologous domains that could not be detected using whole-protein sequences. The web server is accessible at http://genetics.mgh.harvard.edu/DEPCOD., (© The Author(s) 2022. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2022
- Full Text
- View/download PDF
12. Asymmetric inheritance of RNA toxicity in C. elegans expressing CTG repeats.
- Author
-
Braun M, Shoshani S, Teixeira J, Mellul Shtern A, Miller M, Granot Z, Fischer SEJ, Garcia SMDA, and Tabach Y
- Abstract
Nucleotide repeat expansions are a hallmark of over 40 neurodegenerative diseases and cause RNA toxicity and multisystemic symptoms that worsen with age. Through an unclear mechanism, RNA toxicity can trigger severe disease manifestation in infants if the repeats are inherited from their mother. Here we use Caenorhabditis elegans bearing expanded CUG repeats to show that this asymmetric intergenerational inheritance of toxicity contributes to disease pathogenesis. In addition, we show that this mechanism is dependent on small RNA pathways with maternal repeat-derived small RNAs causing transcriptomic changes in the offspring, reduced motility, and shortened lifespan. We rescued the toxicity phenotypes in the offspring by perturbing the RNAi machinery in the affected hermaphrodites. This points to a novel mechanism linking maternal bias and the RNAi machinery and suggests that toxic RNA is transmitted to offspring, causing disease phenotypes through intergenerational epigenetic inheritance., Competing Interests: The authors declare no competing interests., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
13. Multi-omics data integration analysis identifies the spliceosome as a key regulator of DNA double-strand break repair.
- Author
-
Sherill-Rofe D, Raban O, Findlay S, Rahat D, Unterman I, Samiei A, Yasmeen A, Kaiser Z, Kuasne H, Park M, Foulkes WD, Bloch I, Zick A, Gotlieb WH, Tabach Y, and Orthwein A
- Abstract
DNA repair by homologous recombination (HR) is critical for the maintenance of genome stability. Germline and somatic mutations in HR genes have been associated with an increased risk of developing breast (BC) and ovarian cancers (OvC). However, the extent of factors and pathways that are functionally linked to HR with clinical relevance for BC and OvC remains unclear. To gain a broader understanding of this pathway, we used multi-omics datasets coupled with machine learning to identify genes that are associated with HR and to predict their sub-function. Specifically, we integrated our phylogenetic-based co-evolution approach (CladePP) with 23 distinct genetic and proteomic screens that monitored, directly or indirectly, DNA repair by HR. This omics data integration analysis yielded a new database (HRbase) that contains a list of 464 predictions, including 76 gold standard HR genes. Interestingly, the spliceosome machinery emerged as one major pathway with significant cross-platform interactions with the HR pathway. We functionally validated 6 spliceosome factors, including the RNA helicase SNRNP200 and its co-factor SNW1. Importantly, their RNA expression correlated with BC/OvC patient outcome. Altogether, we identified novel clinically relevant DNA repair factors and delineated their specific sub-function by machine learning. Our results, supported by evolutionary and multi-omics analyses, suggest that the spliceosome machinery plays an important role during the repair of DNA double-strand breaks (DSBs)., (© The Author(s) 2022. Published by Oxford University Press on behalf of NAR Cancer.)
- Published
- 2022
- Full Text
- View/download PDF
14. Machine-learning of complex evolutionary signals improves classification of SNVs.
- Author
-
Labes S, Stupp D, Wagner N, Bloch I, Lotem M, L Lahad E, Polak P, Pupko T, and Tabach Y
- Abstract
Conservation is a strong predictor for the pathogenicity of single-nucleotide variants (SNVs). However, some positions that present complex conservation patterns across vertebrates stray from this paradigm. Here, we analyzed the association between complex conservation patterns and the pathogenicity of SNVs in the 115 disease-genes that had sufficient variant data. We show that conservation is not a one-rule-fits-all solution since its accuracy highly depends on the analyzed set of species and genes. For example, pairwise comparisons between the human and 99 vertebrate species showed that species differ in their ability to predict the clinical outcomes of variants among different genes using conservation. Furthermore, certain genes were less amenable for conservation-based variant prediction, while others demonstrated species that optimize prediction. These insights led to developing EvoDiagnostics, which uses the conservation against each species as a feature within a random-forest machine-learning classification algorithm. EvoDiagnostics outperformed traditional conservation algorithms, deep-learning based methods and most ensemble tools in every prediction-task, highlighting the strength of optimizing conservation analysis per-species and per-gene. Overall, we suggest a new and a more biologically relevant approach for analyzing conservation, which improves prediction of variant pathogenicity., (© The Author(s) 2022. Published by Oxford University Press on behalf of NAR Genomics and Bioinformatics.)
- Published
- 2022
- Full Text
- View/download PDF
15. Aurintricarboxylic Acid Decreases RNA Toxicity in a C. elegans Model of Repeat Expansions.
- Author
-
Braun M, Shoshani S, Mellul-Shtern A, and Tabach Y
- Subjects
- Animals, Caenorhabditis elegans, Drug Repositioning, Longevity drug effects, Motor Activity drug effects, Myotonic Dystrophy drug therapy, RNA Interference, Aurintricarboxylic Acid pharmacology, Gene Expression Regulation drug effects
- Abstract
Pathologic expansions of DNA nucleotide tandem repeats may generate toxic RNA that triggers disease phenotypes. RNA toxicity is the hallmark of multiple expansion repeat disorders, including myotonic dystrophy type 1 (DM1). To date, there are no available disease-modifying therapies for DM1. Our aim was to use drug repositioning to ameliorate the phenotype of affected individuals in a nematode model of DM1. As the RNA interference pathway plays a key role in mediating RNA toxicity, we investigated the effect of aurintricarboxylic acid. We demonstrated that by perturbing the RNA interference machinery using aurintricarboxylic acid, we could annihilate the RNA toxicity and ameliorate the phenotype. As our approach targets a universal disease mechanism, it is potentially relevant for more expansion repeat disorders.
- Published
- 2021
- Full Text
- View/download PDF
16. Co-evolution based machine-learning for predicting functional interactions between human genes.
- Author
-
Stupp D, Sharon E, Bloch I, Zitnik M, Zuk O, and Tabach Y
- Subjects
- DNA Repair genetics, DNA Repair physiology, Evolution, Molecular, Humans, Phylogeny, Sequence Analysis, DNA methods, Machine Learning
- Abstract
Over the next decade, more than a million eukaryotic species are expected to be fully sequenced. This has the potential to improve our understanding of genotype and phenotype crosstalk, gene function and interactions, and answer evolutionary questions. Here, we develop a machine-learning approach for utilizing phylogenetic profiles across 1154 eukaryotic species. This method integrates co-evolution across eukaryotic clades to predict functional interactions between human genes and the context for these interactions. We benchmark our approach showing a 14% performance increase (auROC) compared to previous methods. Using this approach, we predict functional annotations for less studied genes. We focus on DNA repair and verify that 9 of the top 50 predicted genes have been identified elsewhere, with others previously prioritized by high-throughput screens. Overall, our approach enables better annotation of function and functional interactions and facilitates the understanding of evolutionary processes underlying co-evolution. The manuscript is accompanied by a webserver available at: https://mlpp.cs.huji.ac.il ., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
17. Current gene panels account for nearly all homologous recombination repair-associated multiple-case breast cancer families.
- Author
-
Matis TS, Zayed N, Labraki B, de Ladurantaye M, Matis TA, Camacho Valenzuela J, Hamel N, Atayan A, Rivera B, Tabach Y, Tonin PN, Orthwein A, Mes-Masson AM, El Haffaf Z, Foulkes WD, and Polak P
- Abstract
It was hypothesized that variants in underexplored homologous recombination repair (HR) genes could explain unsolved multiple-case breast cancer (BC) families. We investigated HR deficiency (HRD)-associated mutational signatures and second hits in tumor DNA from familial BC cases. No candidates genes were associated with HRD in 38 probands previously tested negative with gene panels. We conclude it is unlikely that unknown HRD-associated genes explain a large fraction of unsolved familial BC., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
18. Expanding the MECP2 network using comparative genomics reveals potential therapeutic targets for Rett syndrome.
- Author
-
Unterman I, Bloch I, Cazacu S, Kazimirsky G, Ben-Zeev B, Berman BP, Brodie C, and Tabach Y
- Subjects
- Genomics, Humans, Rett Syndrome genetics, Methyl-CpG-Binding Protein 2 therapeutic use, Rett Syndrome therapy
- Abstract
Inactivating mutations in the Methyl-CpG Binding Protein 2 (MECP2) gene are the main cause of Rett syndrome (RTT). Despite extensive research into MECP2 function, no treatments for RTT are currently available. Here, we used an evolutionary genomics approach to construct an unbiased MECP2 gene network, using 1028 eukaryotic genomes to prioritize proteins with strong co-evolutionary signatures with MECP2. Focusing on proteins targeted by FDA-approved drugs led to three promising targets, two of which were previously linked to MECP2 function (IRAK, KEAP1) and one that was not (EPOR). The drugs targeting these three proteins (Pacritinib, DMF, and EPO) were able to rescue different phenotypes of MECP2 inactivation in cultured human neural cell types, and appeared to converge on Nuclear Factor Kappa B (NF-κB) signaling in inflammation. This study highlights the potential of comparative genomics to accelerate drug discovery, and yields potential new avenues for the treatment of RTT., Competing Interests: IU, IB, SC, GK, BB, BB, CB, YT No competing interests declared, (© 2021, Unterman et al.)
- Published
- 2021
- Full Text
- View/download PDF
19. Spatial patterning of scent in petunia corolla is discriminated by bees and involves the ABCG1 transporter.
- Author
-
Skaliter O, Kitsberg Y, Sharon E, Shklarman E, Shor E, Masci T, Yue Y, Arien Y, Tabach Y, Shafir S, and Vainstein A
- Subjects
- ATP Binding Cassette Transporter, Subfamily G, Member 1 genetics, Animals, Flowers metabolism, Plant Proteins genetics, Volatile Organic Compounds chemistry, Volatile Organic Compounds metabolism, ATP Binding Cassette Transporter, Subfamily G, Member 1 metabolism, Bees physiology, Flowers chemistry, Odorants analysis, Petunia physiology, Plant Proteins metabolism
- Abstract
Floral guides are patterned cues that direct the pollinator to the plant reproductive organs. The spatial distribution of showy visual and olfactory traits allows efficient plant-pollinator interactions. Data on the mechanisms underlying floral volatile patterns or their interactions with pollinators are lacking. Here we characterize the spatial emission patterns of volatiles from the corolla of the model plant Petunia × hybrida and reveal the ability of honeybees to distinguish these patterns. Along the adaxial epidermis, in correlation with cell density, the petal base adjacent to reproductive organs emitted significantly higher levels of volatiles than the distal petal rim. Volatile emission could also be differentiated between the two epidermal surfaces: emission from the adaxial side was significantly higher than that from the abaxial side. Similar emission patterns were also observed in other petunias, Dianthus caryophyllus (carnation) and Argyranthemum frutescens (Marguerite daisy). Analyses of transcripts involved in volatile production/emission revealed lower levels of the plasma-membrane transporter ABCG1 in the abaxial versus adaxial epidermis. Transient overexpression of ABCG1 enhanced emission from the abaxial epidermis to the level of the adaxial epidermis, suggesting its involvement in spatial emission patterns in the epidermal layers. Proboscis extension response experiments showed that differences in emission levels along the adaxial epidermis, that is, petal base versus rim, detected by GC-MS are also discernible by honeybees., (© 2021 Society for Experimental Biology and John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
20. CladeOScope: functional interactions through the prism of clade-wise co-evolution.
- Author
-
Tsaban T, Stupp D, Sherill-Rofe D, Bloch I, Sharon E, Schueler-Furman O, Wiener R, and Tabach Y
- Abstract
Mapping co-evolved genes via phylogenetic profiling (PP) is a powerful approach to uncover functional interactions between genes and to associate them with pathways. Despite many successful endeavors, the understanding of co-evolutionary signals in eukaryotes remains partial. Our hypothesis is that 'Clades', branches of the tree of life (e.g. primates and mammals), encompass signals that cannot be detected by PP using all eukaryotes. As such, integrating information from different clades should reveal local co-evolution signals and improve function prediction. Accordingly, we analyzed 1028 genomes in 66 clades and demonstrated that the co-evolutionary signal was scattered across clades. We showed that functionally related genes are frequently co-evolved in only parts of the eukaryotic tree and that clades are complementary in detecting functional interactions within pathways. We examined the non-homologous end joining pathway and the UFM1 ubiquitin-like protein pathway and showed that both demonstrated distinguished co-evolution patterns in specific clades. Our research offers a different way to look at co-evolution across eukaryotes and points to the importance of modular co-evolution analysis. We developed the 'CladeOScope' PP method to integrate information from 16 clades across over 1000 eukaryotic genomes and is accessible via an easy to use web server at http://cladeoscope.cs.huji.ac.il., (© The Author(s) 2021. Published by Oxford University Press on behalf of NAR Genomics and Bioinformatics.)
- Published
- 2021
- Full Text
- View/download PDF
21. Reversal of diet-induced hepatic steatosis by peripheral CB1 receptor blockade in mice is p53/miRNA-22/SIRT1/PPARα dependent.
- Author
-
Azar S, Udi S, Drori A, Hadar R, Nemirovski A, Vemuri KV, Miller M, Sherill-Rofe D, Arad Y, Gur-Wahnon D, Li X, Makriyannis A, Ben-Zvi D, Tabach Y, Ben-Dov IZ, and Tam J
- Subjects
- Animals, Diet, High-Fat, Fatty Acids metabolism, Fatty Liver genetics, Hep G2 Cells, Hepatocytes metabolism, Humans, Lipid Metabolism physiology, Lipids physiology, Liver metabolism, Male, Mice, Mice, Inbred C57BL, MicroRNAs genetics, Obesity metabolism, Oxidation-Reduction, PPAR alpha metabolism, Phylogeny, Receptor, Cannabinoid, CB1 antagonists & inhibitors, Signal Transduction, Sirtuin 1 metabolism, Tumor Suppressor Protein p53 metabolism, Fatty Liver metabolism, Receptor, Cannabinoid, CB1 metabolism
- Abstract
Objective: The endocannabinoid (eCB) system is increasingly recognized as being crucially important in obesity-related hepatic steatosis. By activating the hepatic cannabinoid-1 receptor (CB
1 R), eCBs modulate lipogenesis and fatty acid oxidation. However, the underlying molecular mechanisms are largely unknown., Methods: We combined unbiased bioinformatics techniques, mouse genetic manipulations, multiple pharmacological, molecular, and cellular biology approaches, and genomic sequencing to systematically decipher the role of the hepatic CB1 R in modulating fat utilization in the liver and explored the downstream molecular mechanisms., Results: Using an unbiased normalized phylogenetic profiling analysis, we found that the CB1 R evolutionarily coevolves with peroxisome proliferator-activated receptor-alpha (PPARα), a key regulator of hepatic lipid metabolism. In diet-induced obese (DIO) mice, peripheral CB1 R blockade (using AM6545) induced the reversal of hepatic steatosis and improved liver injury in WT, but not in PPARα-/- mice. The antisteatotic effect mediated by AM6545 in WT DIO mice was accompanied by increased hepatic expression and activity of PPARα as well as elevated hepatic levels of the PPARα-activating eCB-like molecules oleoylethanolamide and palmitoylethanolamide. Moreover, AM6545 was unable to rescue hepatic steatosis in DIO mice lacking liver sirtuin 1 (SIRT1), an upstream regulator of PPARα. Both of these signaling molecules were modulated by the CB1 R as measured in hepatocytes exposed to lipotoxic conditions or treated with CB1 R agonists in the absence/presence of AM6545. Furthermore, using microRNA transcriptomic profiling, we found that the CB1 R regulated the hepatic expression, acetylation, and transcriptional activity of p53, resulting in the enhanced expression of miR-22, which was found to specifically target SIRT1 and PPARα., Conclusions: We provide strong evidence for a functional role of the p53/miR-22/SIRT1/PPARα signaling pathway in potentially mediating the antisteatotic effect of peripherally restricted CB1 R blockade., (Copyright © 2020 The Authors. Published by Elsevier GmbH.. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
22. ACE2 Co-evolutionary Pattern Suggests Targets for Pharmaceutical Intervention in the COVID-19 Pandemic.
- Author
-
Braun M, Sharon E, Unterman I, Miller M, Shtern AM, Benenson S, Vainstein A, and Tabach Y
- Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spillover infection in December 2019 has caused an unprecedented pandemic. SARS-CoV-2, as other coronaviruses, binds its target cells through the angiotensin-converting enzyme 2 (ACE2) receptor. Accordingly, this makes ACE2 research essential for understanding the zoonotic nature of coronaviruses and identifying novel drugs. Here we present a systematic analysis of the ACE2 conservation and co-evolution protein network across 1,671 eukaryotes, revealing an unexpected conservation pattern in specific metazoans, plants, fungi, and protists. We identified the co-evolved protein network and pinpointed a list of drugs that target this network by using data integration from different sources. Our computational analysis found widely used drugs such as nonsteroidal anti-inflammatory drugs and vasodilators. These drugs are expected to perturb the ACE2 network affecting infectivity as well as the pathophysiology of the disease., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
23. Optimization of co-evolution analysis through phylogenetic profiling reveals pathway-specific signals.
- Author
-
Bloch I, Sherill-Rofe D, Stupp D, Unterman I, Beer H, Sharon E, and Tabach Y
- Subjects
- Genome, Phylogeny, Proteins, Genomics, Software
- Abstract
Summary: The exponential growth in available genomic data is expected to reach full sequencing of a million genomes in the coming decade. Improving and developing methods to analyze these genomes and to reveal their utility is of major interest in a wide variety of fields, such as comparative and functional genomics, evolution and bioinformatics. Phylogenetic profiling is an established method for predicting functional interactions between proteins based on similarities in their evolutionary patterns across species. Proteins that function together (i.e. generate complexes, interact in the same pathways or improve adaptation to environmental niches) tend to show coordinated evolution across the tree of life. The normalized phylogenetic profiling (NPP) method takes into account minute changes in proteins across species to identify protein co-evolution. Despite the success of this method, it is still not clear what set of parameters is required for optimal use of co-evolution in predicting functional interactions. Moreover, it is not clear if pathway evolution or function should direct parameter choice. Here, we create a reliable and usable NPP construction pipeline. We explore the effect of parameter selection on functional interaction prediction using NPP from 1028 genomes, both separately and in various value combinations. We identify several parameter sets that optimize performance for pathways with certain biological annotation. This work reveals the importance of choosing the right parameters for optimized function prediction based on a biological context., Availability and Implementation: Source code and documentation are available on GitHub: https://github.com/iditam/CompareNPPs., Contact: yuvaltab@ekmd.huji.ac.il., Supplementary Information: Supplementary data are available at Bioinformatics online., (© The Author(s) 2020. Published by Oxford University Press. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
24. Titanium Tackles the Endoplasmic Reticulum: A First Genomic Study on a Titanium Anticancer Metallodrug.
- Author
-
Miller M, Mellul A, Braun M, Sherill-Rofe D, Cohen E, Shpilt Z, Unterman I, Braitbard O, Hochman J, Tshuva EY, and Tabach Y
- Abstract
PhenolaTi is an advanced non-toxic anticancer chemotherapy; this inert bis(phenolato)bis(alkoxo) Ti(IV) complex demonstrates the intriguing combination of high and wide efficacy with no detected toxicity in animals. Here we unravel the cellular pathways involved in its mechanism of action by a first genome study on Ti(IV)-treated cells, using an attuned RNA sequencing-based available technology. First, phenolaTi induced apoptosis and cell-cycle arrest at the G2/M phase in MCF7 cells. Second, the transcriptome of the treated cells was analyzed, identifying alterations in pathways relating to protein translation, DNA damage, and mitochondrial eruption. Unlike for common metallodrugs, electrophoresis assay showed no inhibition of DNA polymerase activity. Reduced in vitro cytotoxicity with added endoplasmic reticulum (ER) stress inhibitor supported the ER as a putative cellular target. Altogether, this paper reveals a distinct ER-related mechanism by the Ti(IV) anticancer coordination complex, paving the way for wider applicability of related techniques in mechanistic analyses of metallodrugs., Competing Interests: Declaration of Interests The authors declare no competing interests. Relating patent: Tshuva EY and Hochman J; Cytotoxic titanium and vanadium complexes PCT/IL2013/05,069 filled 15/08/2013., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
25. A high-throughput screening and computation platform for identifying synthetic promoters with enhanced cell-state specificity (SPECS).
- Author
-
Wu MR, Nissim L, Stupp D, Pery E, Binder-Nissim A, Weisinger K, Enghuus C, Palacios SR, Humphrey M, Zhang Z, Maria Novoa E, Kellis M, Weiss R, Rabkin SD, Tabach Y, and Lu TK
- Subjects
- Breast Neoplasms, Cell Line, Tumor, Cell Separation methods, Female, Gene Expression Regulation, Gene Library, Glioblastoma, Humans, Induced Pluripotent Stem Cells, Lentivirus, Neoplastic Stem Cells, Organoids, Regulatory Elements, Transcriptional, Machine Learning, Promoter Regions, Genetic, Software
- Abstract
Cell state-specific promoters constitute essential tools for basic research and biotechnology because they activate gene expression only under certain biological conditions. Synthetic Promoters with Enhanced Cell-State Specificity (SPECS) can be superior to native ones, but the design of such promoters is challenging and frequently requires gene regulation or transcriptome knowledge that is not readily available. Here, to overcome this challenge, we use a next-generation sequencing approach combined with machine learning to screen a synthetic promoter library with 6107 designs for high-performance SPECS for potentially any cell state. We demonstrate the identification of multiple SPECS that exhibit distinct spatiotemporal activity during the programmed differentiation of induced pluripotent stem cells (iPSCs), as well as SPECS for breast cancer and glioblastoma stem-like cells. We anticipate that this approach could be used to create SPECS for gene therapies that are activated in specific cell states, as well as to study natural transcriptional regulatory networks.
- Published
- 2019
- Full Text
- View/download PDF
26. Mapping global and local coevolution across 600 species to identify novel homologous recombination repair genes.
- Author
-
Sherill-Rofe D, Rahat D, Findlay S, Mellul A, Guberman I, Braun M, Bloch I, Lalezari A, Samiei A, Sadreyev R, Goldberg M, Orthwein A, Zick A, and Tabach Y
- Subjects
- Animals, DNA Repair Enzymes genetics, Genetic Loci, Phylogeny, Plants genetics, Evolution, Molecular, Recombinational DNA Repair, Software
- Abstract
The homologous recombination repair (HRR) pathway repairs DNA double-strand breaks in an error-free manner. Mutations in HRR genes can result in increased mutation rate and genomic rearrangements, and are associated with numerous genetic disorders and cancer. Despite intensive research, the HRR pathway is not yet fully mapped. Phylogenetic profiling analysis, which detects functional linkage between genes using coevolution, is a powerful approach to identify factors in many pathways. Nevertheless, phylogenetic profiling has limited predictive power when analyzing pathways with complex evolutionary dynamics such as the HRR. To map novel HRR genes systematically, we developed clade phylogenetic profiling (CladePP). CladePP detects local coevolution across hundreds of genomes and points to the evolutionary scale (e.g., mammals, vertebrates, animals, plants) at which coevolution occurred. We found that multiscale coevolution analysis is significantly more biologically relevant and sensitive to detect gene function. By using CladePP, we identified dozens of unrecognized genes that coevolved with the HRR pathway, either globally across all eukaryotes or locally in different clades. We validated eight genes in functional biological assays to have a role in DNA repair at both the cellular and organismal levels. These genes are expected to play a role in the HRR pathway and might lead to a better understanding of missing heredity in HRR-associated cancers (e.g., heredity breast and ovarian cancer). Our platform presents an innovative approach to predict gene function, identify novel factors related to different diseases and pathways, and characterize gene evolution., (© 2019 Sherill-Rofe et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2019
- Full Text
- View/download PDF
27. MYORG is associated with recessive primary familial brain calcification.
- Author
-
Arkadir D, Lossos A, Rahat D, Abu Snineh M, Schueler-Furman O, Nitschke S, Minassian BA, Sadaka Y, Lerer I, Tabach Y, and Meiner V
- Subjects
- Adult, Asian People genetics, Brain Diseases, Metabolic complications, Brain Diseases, Metabolic pathology, Calcinosis complications, Female, Genes, Recessive, Humans, Male, Middle Aged, Middle East, Mutation, Pedigree, Exome Sequencing, Young Adult, Brain Diseases, Metabolic genetics, Calcinosis genetics, Glycoside Hydrolases genetics
- Abstract
Objective: To investigate the genetic basis of the recessive form of primary familial brain calcification and study pathways linking a novel gene with known dominant genes that cause the disease., Methods: Whole exome sequencing and Sanger-based segregation analysis were used to identify possible disease causing mutations. Mutation pathogenicity was validated by structural protein modeling. Functional associations between the candidate gene, MYORG , and genes previously implicated in the disease were examined through phylogenetic profiling., Results: We studied nine affected individuals from two unrelated families of Middle Eastern origin. The median age of symptom onset was 29.5 years (range 21-57 years) and dysarthria was the most common presenting symptom. We identified in the MYORG gene, a homozygous c.1233delC mutation in one family and c.1060_1062delGAC mutation in another. The first mutation results in protein truncation and the second in deletion of a highly conserved aspartic acid that is likely to disrupt binding of the protein with its substrate. Phylogenetic profiling analysis of the MYORG protein sequence suggests co-evolution with a number of calcium channels as well as other proteins related to regulation of anion transmembrane transport (False Discovery Rate, FDR < 10
-8 ) and with PDCD6IP, a protein interacting with PDGFR β which is known to be involved in the disease., Interpretation: MYORG mutations are linked to a recessive form of primary familial brain calcification. This association was recently described in patients of Chinese ancestry. We suggest the possibility that MYORG mutations lead to calcification in a PDGFR β -related pathway.- Published
- 2018
- Full Text
- View/download PDF
28. UV-Protection Timer Controls Linkage between Stress and Pigmentation Skin Protection Systems.
- Author
-
Malcov-Brog H, Alpert A, Golan T, Parikh S, Nordlinger A, Netti F, Sheinboim D, Dror I, Thomas L, Cosson C, Gonen P, Stanevsky Y, Brenner R, Perluk T, Frand J, Elgavish S, Nevo Y, Rahat D, Tabach Y, Khaled M, Shen-Orr SS, and Levy C
- Subjects
- Animals, Cell Line, Humans, Hypoxia-Inducible Factor 1, alpha Subunit physiology, Male, Melanocytes physiology, Melanocytes radiation effects, Mice, Mice, Inbred C57BL, MicroRNAs physiology, Microphthalmia-Associated Transcription Factor radiation effects, Primary Cell Culture, Skin Pigmentation radiation effects, Ultraviolet Rays adverse effects, Microphthalmia-Associated Transcription Factor metabolism, Skin metabolism, Skin radiation effects
- Abstract
Skin sun exposure induces two protection programs: stress responses and pigmentation, the former within minutes and the latter only hours afterward. Although serving the same physiological purpose, it is not known whether and how these programs are coordinated. Here, we report that UVB exposure every other day induces significantly more skin pigmentation than the higher frequency of daily exposure, without an associated increase in stress responses. Using mathematical modeling and empirical studies, we show that the melanocyte master regulator, MITF, serves to synchronize stress responses and pigmentation and, furthermore, functions as a UV-protection timer via damped oscillatory dynamics, thereby conferring a trade-off between the two programs. MITF oscillations are controlled by multiple negative regulatory loops, one at the transcriptional level involving HIF1α and another post-transcriptional loop involving microRNA-148a. These findings support trait linkage between the two skin protection programs, which, we speculate, arose during furless skin evolution to minimize skin damage., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
29. Mutated MITF-E87R in Melanoma Enhances Tumor Progression via S100A4.
- Author
-
Nordlinger A, Dror S, Elkahloun A, Del Rio J, Stubbs E, Golan T, Malcov H, Pricket TD, Cronin JC, Parikh S, Labes S, Thomas L, Yankovitz G, Tabach Y, Levy C, Samuels Y, and Khaled M
- Subjects
- DNA Mutational Analysis, Disease Progression, Humans, Immunoblotting, Melanoma metabolism, Melanoma pathology, Microphthalmia-Associated Transcription Factor metabolism, S100 Calcium-Binding Protein A4 biosynthesis, Skin Neoplasms metabolism, Skin Neoplasms pathology, Tumor Cells, Cultured, DNA, Neoplasm genetics, Gene Expression Regulation, Neoplastic, Melanoma genetics, Microphthalmia-Associated Transcription Factor genetics, Mutation, S100 Calcium-Binding Protein A4 genetics, Skin Neoplasms genetics
- Abstract
Melanoma, a melanocyte origin neoplasm, is the most lethal type of skin cancer, and incidence is increasing. Several familial and somatic mutations have been identified in the gene encoding the melanocyte lineage master regulator, MITF; however, the neoplastic mechanisms of these mutant MITF variants are mostly unknown. Here, by performing unbiased analysis of the transcriptomes in cells expressing mutant MITF, we identified calcium-binding protein S100A4 as a downstream target of MITF-E87R. By using wild-type and mutant MITF melanoma lines, we found that both endogenous wild-type and MITF-E87R variants occupy the S100A4 promoter. Remarkably, whereas wild-type MITF represses S100A4 expression, MITF-E87R activates its transcription. The opposite effects of wild-type and mutant MITF result in opposing cellular phenotypes, because MITF-E87R via S100A4 enhanced invasion and reduced adhesion in contrast to wild-type MITF activity. Finally, we found that melanoma patients with altered S100A4 expression have poor prognosis. These data show that a change in MITF transcriptional activity from repression to activation of S100A4 that results from a point mutation in MITF alters melanoma invasive ability. These data suggest new opportunities for diagnosis and treatment of metastatic melanoma., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
30. SHLD2/FAM35A co-operates with REV7 to coordinate DNA double-strand break repair pathway choice.
- Author
-
Findlay S, Heath J, Luo VM, Malina A, Morin T, Coulombe Y, Djerir B, Li Z, Samiei A, Simo-Cheyou E, Karam M, Bagci H, Rahat D, Grapton D, Lavoie EG, Dove C, Khaled H, Kuasne H, Mann KK, Klein KO, Greenwood CM, Tabach Y, Park M, Côté JF, Masson JY, Maréchal A, and Orthwein A
- Subjects
- Cell Cycle Proteins genetics, DNA-Binding Proteins genetics, G2 Phase genetics, HEK293 Cells, Humans, Mad2 Proteins genetics, S Phase genetics, Telomere-Binding Proteins genetics, Telomere-Binding Proteins metabolism, Tumor Suppressor p53-Binding Protein 1 genetics, Tumor Suppressor p53-Binding Protein 1 metabolism, Cell Cycle Proteins metabolism, DNA Breaks, Double-Stranded, DNA End-Joining Repair, DNA-Binding Proteins metabolism, Mad2 Proteins metabolism
- Abstract
DNA double-strand breaks (DSBs) can be repaired by two major pathways: non-homologous end-joining (NHEJ) and homologous recombination (HR). DNA repair pathway choice is governed by the opposing activities of 53BP1, in complex with its effectors RIF1 and REV7, and BRCA1. However, it remains unknown how the 53BP1/RIF1/REV7 complex stimulates NHEJ and restricts HR to the S/G2 phases of the cell cycle. Using a mass spectrometry (MS)-based approach, we identify 11 high-confidence REV7 interactors and elucidate the role of SHLD2 (previously annotated as FAM35A and RINN2) as an effector of REV7 in the NHEJ pathway. FAM35A depletion impairs NHEJ-mediated DNA repair and compromises antibody diversification by class switch recombination (CSR) in B cells. FAM35A accumulates at DSBs in a 53BP1-, RIF1-, and REV7-dependent manner and antagonizes HR by limiting DNA end resection. In fact, FAM35A is part of a larger complex composed of REV7 and SHLD1 (previously annotated as C20orf196 and RINN3), which promotes NHEJ and limits HR Together, these results establish SHLD2 as a novel effector of REV7 in controlling the decision-making process during DSB repair., (© 2018 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2018
- Full Text
- View/download PDF
31. Schlafen2 mutation in mice causes an osteopetrotic phenotype due to a decrease in the number of osteoclast progenitors.
- Author
-
Omar I, Guterman-Ram G, Rahat D, Tabach Y, Berger M, and Levaot N
- Subjects
- Animals, Mice, Cell Cycle Proteins deficiency, Cell Differentiation, Mutation, Myeloid Progenitor Cells physiology, Osteoclasts physiology, Osteopetrosis genetics, Osteopetrosis pathology
- Abstract
Osteoclasts are the bone resorbing cells that derive from myeloid progenitor cells. Although there have been recent advancements in the ability to identify osteoclast progenitors, very little is known about the molecular mechanisms governing their homeostasis. Here, by analyzing the normalized phylogenetic profiles of the Schlafen (Slfn) gene family, we found that it co-evolved with osteoclast-related genes. Following these findings, we used a Slfn2 loss-of-function mutant mouse, elektra, to study the direct role of Slfn2 in osteoclast development and function. Slfn2
eka/eka mice exhibited a profound increase in their cancellous bone mass and a significant reduction in osteoclast numbers. In addition, monocyte cultures from the bone marrow of Slfn2eka/eka mice showed a reduction in osteoclast number and total resorption area. Finally, we show that the bone marrow of Slfn2eka/eka mice have significantly less CD11b- Ly6Chi osteoclast precursors. Overall, our data suggest that Slfn2 is required for normal osteoclast differentiation and that loss of its function in mice results in an osteopetrotic phenotype.- Published
- 2018
- Full Text
- View/download PDF
32. Synthetic RNA-Based Immunomodulatory Gene Circuits for Cancer Immunotherapy.
- Author
-
Nissim L, Wu MR, Pery E, Binder-Nissim A, Suzuki HI, Stupp D, Wehrspaun C, Tabach Y, Sharp PA, and Lu TK
- Subjects
- Animals, Female, Humans, Immunomodulation, Mice, Ovarian Neoplasms immunology, Promoter Regions, Genetic, Receptors, Antigen, T-Cell metabolism, T-Lymphocytes, Cytotoxic immunology, Gene Regulatory Networks, Immunotherapy methods, Ovarian Neoplasms therapy
- Abstract
Despite its success in several clinical trials, cancer immunotherapy remains limited by the rarity of targetable tumor-specific antigens, tumor-mediated immune suppression, and toxicity triggered by systemic delivery of potent immunomodulators. Here, we present a proof-of-concept immunomodulatory gene circuit platform that enables tumor-specific expression of immunostimulators, which could potentially overcome these limitations. Our design comprised de novo synthetic cancer-specific promoters and, to enhance specificity, an RNA-based AND gate that generates combinatorial immunomodulatory outputs only when both promoters are mutually active. These outputs included an immunogenic cell-surface protein, a cytokine, a chemokine, and a checkpoint inhibitor antibody. The circuits triggered selective T cell-mediated killing of cancer cells, but not of normal cells, in vitro. In in vivo efficacy assays, lentiviral circuit delivery mediated significant tumor reduction and prolonged mouse survival. Our design could be adapted to drive additional immunomodulators, sense other cancers, and potentially treat other diseases that require precise immunological programming., (Copyright © 2017. Published by Elsevier Inc.)
- Published
- 2017
- Full Text
- View/download PDF
33. A continuum of mRNP complexes in embryonic microRNA-mediated silencing.
- Author
-
Wu E, Vashisht AA, Chapat C, Flamand MN, Cohen E, Sarov M, Tabach Y, Sonenberg N, Wohlschlegel J, and Duchaine TF
- Subjects
- Animals, Caenorhabditis elegans embryology, Caenorhabditis elegans enzymology, Caenorhabditis elegans genetics, Caenorhabditis elegans metabolism, Caenorhabditis elegans Proteins genetics, Caenorhabditis elegans Proteins metabolism, Cytoplasmic Granules metabolism, Embryo, Nonmammalian metabolism, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Intrinsically Disordered Proteins metabolism, RNA, Messenger metabolism, RNA-Induced Silencing Complex metabolism, Ribonucleases metabolism, Gene Expression Regulation, Developmental, MicroRNAs metabolism, RNA Interference, Ribonucleoproteins metabolism
- Abstract
MicroRNAs (miRNAs) impinge on the translation and stability of their target mRNAs, and play key roles in development, homeostasis and disease. The gene regulation mechanisms they instigate are largely mediated through the CCR4–NOT deadenylase complex, but the molecular events that occur on target mRNAs are poorly resolved. We observed a broad convergence of interactions of germ granule and P body mRNP components on AIN-1/GW182 and NTL-1/CNOT1 in Caenorhabditis elegans embryos. We show that the miRISC progressively matures on the target mRNA from a scanning form into an effector mRNP particle by sequentially recruiting the CCR4–NOT complex, decapping and decay, or germ granule proteins. Finally, we implicate intrinsically disordered proteins, key components in mRNP architectures, in the embryonic function of lsy-6 miRNA. Our findings define dynamic steps of effector mRNP assembly in miRNA-mediated silencing, and identify a functional continuum between germ granules and P bodies in the C. elegans embryo.
- Published
- 2017
- Full Text
- View/download PDF
34. Interactions of Melanoma Cells with Distal Keratinocytes Trigger Metastasis via Notch Signaling Inhibition of MITF.
- Author
-
Golan T, Messer AR, Amitai-Lange A, Melamed Z, Ohana R, Bell RE, Kapitansky O, Lerman G, Greenberger S, Khaled M, Amar N, Albrengues J, Gaggioli C, Gonen P, Tabach Y, Sprinzak D, Shalom-Feuerstein R, and Levy C
- Subjects
- Animals, Base Sequence, Binding Sites, Cell Communication, Cell Line, Tumor, Coculture Techniques, Gene Expression Regulation, Neoplastic, Melanoma, Experimental metabolism, Mice, Inbred NOD, Mice, SCID, MicroRNAs genetics, MicroRNAs metabolism, Neoplasm Invasiveness, Neoplasm Transplantation, Promoter Regions, Genetic, RNA Interference, Receptors, Notch metabolism, Signal Transduction, Skin Neoplasms metabolism, Keratinocytes physiology, Melanoma, Experimental secondary, Microphthalmia-Associated Transcription Factor metabolism, Skin Neoplasms pathology
- Abstract
The most critical stage in initiation of melanoma metastasis is the radial to vertical growth transition, yet the triggers of this transition remain elusive. We suggest that the microenvironment drives melanoma metastasis independently of mutation acquisition. Here we examined the changes in microenvironment that occur during melanoma radial growth. We show that direct contact of melanoma cells with the remote epidermal layer triggers vertical invasion via Notch signaling activation, the latter serving to inhibit MITF function. Briefly, within the native Notch ligand-free microenvironment, MITF, the melanocyte lineage master regulator, binds and represses miR-222/221 promoter in an RBPJK-dependent manner. However, when radial growth brings melanoma cells into contact with distal differentiated keratinocytes that express Notch ligands, the activated Notch intracellular domain impairs MITF binding to miR-222/221 promoter. This de-repression of miR-222/221 expression triggers initiation of invasion. Our findings may direct melanoma prevention opportunities via targeting specific microenvironments., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
35. PhyloGene server for identification and visualization of co-evolving proteins using normalized phylogenetic profiles.
- Author
-
Sadreyev IR, Ji F, Cohen E, Ruvkun G, and Tabach Y
- Subjects
- Algorithms, Animals, Computer Graphics, Genomics, Humans, Internet, Mice, Proteins chemistry, Proteins genetics, Phylogeny, Proteins classification, Sequence Homology, Amino Acid, Software
- Abstract
Proteins that function in the same pathways, protein complexes or the same environmental conditions can show similar patterns of sequence conservation across phylogenetic clades. In species that no longer require a specific protein complex or pathway, these proteins, as a group, tend to be lost or diverge. Analysis of the similarity in patterns of sequence conservation across a large set of eukaryotes can predict functional associations between different proteins, identify new pathway members and reveal the function of previously uncharacterized proteins. We used normalized phylogenetic profiling to predict protein function and identify new pathway members and disease genes. The phylogenetic profiles of tens of thousands conserved proteins in the human, mouse, Caenorhabditis elegans and Drosophila genomes can be queried on our new web server, PhyloGene. PhyloGene provides intuitive and user-friendly platform to query the patterns of conservation across 86 animal, fungal, plant and protist genomes. A protein query can be submitted either by selecting the name from whole-genome protein sets of the intensively studied species or by entering a protein sequence. The graphic output shows the profile of sequence conservation for the query and the most similar phylogenetic profiles for the proteins in the genome of choice. The user can also download this output in numerical form., (© The Author(s) 2015. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2015
- Full Text
- View/download PDF
36. Identification of genes in toxicity pathways of trinucleotide-repeat RNA in C. elegans.
- Author
-
Garcia SM, Tabach Y, Lourenço GF, Armakola M, and Ruvkun G
- Subjects
- 3' Untranslated Regions, Animals, Base Sequence, Caenorhabditis elegans cytology, Caenorhabditis elegans physiology, Caenorhabditis elegans Proteins genetics, Cell Nucleus metabolism, Cells, Cultured, Conserved Sequence, Fibroblasts metabolism, Humans, Locomotion, Muscles abnormalities, Mutation, Myotonic Dystrophy genetics, Nonsense Mediated mRNA Decay, RNA, Helminth metabolism, Caenorhabditis elegans genetics, Genes, Helminth, RNA, Helminth genetics, Trinucleotide Repeats
- Abstract
Myotonic dystrophy disorders are caused by expanded CUG repeats in noncoding regions. Here we used Caenorhabditis elegans expressing CUG repeats to identify genes that modulate the toxicity of such repeats. We identified 15 conserved genes that function as suppressors or enhancers of CUG repeat-induced toxicity and that modulate formation of nuclear foci by CUG-repeat RNA. These genes regulate CUG repeat-induced toxicity through distinct mechanisms including RNA export and clearance, thus suggesting that CUG-repeat toxicity is mediated by multiple pathways. A subset of the genes are also involved in other degenerative disorders. The nonsense-mediated mRNA decay (NMD) pathway has a conserved role in regulating CUG-repeat-RNA transcript levels and toxicity, and NMD recognition of toxic RNAs depends on 3'-untranslated-region GC-nucleotide content. Our studies suggest a broader surveillance role for NMD in which variations in this pathway influence multiple degenerative diseases.
- Published
- 2014
- Full Text
- View/download PDF
37. High-resolution mapping reveals a conserved, widespread, dynamic mRNA methylation program in yeast meiosis.
- Author
-
Schwartz S, Agarwala SD, Mumbach MR, Jovanovic M, Mertins P, Shishkin A, Tabach Y, Mikkelsen TS, Satija R, Ruvkun G, Carr SA, Lander ES, Fink GR, and Regev A
- Subjects
- Adenosine analogs & derivatives, Adenosine analysis, Adenosine metabolism, Cell Nucleolus metabolism, Genome, Fungal, Methylation, Nuclear Proteins metabolism, Saccharomyces cerevisiae Proteins metabolism, Transcription Factors metabolism, tRNA Methyltransferases metabolism, Meiosis, RNA, Fungal metabolism, RNA, Messenger metabolism, Saccharomyces cytology, Saccharomyces metabolism, Saccharomyces cerevisiae cytology, Saccharomyces cerevisiae metabolism
- Abstract
N(6)-methyladenosine (m(6)A) is the most ubiquitous mRNA base modification, but little is known about its precise location, temporal dynamics, and regulation. Here, we generated genomic maps of m(6)A sites in meiotic yeast transcripts at nearly single-nucleotide resolution, identifying 1,308 putatively methylated sites within 1,183 transcripts. We validated eight out of eight methylation sites in different genes with direct genetic analysis, demonstrated that methylated sites are significantly conserved in a related species, and built a model that predicts methylated sites directly from sequence. Sites vary in their methylation profiles along a dense meiotic time course and are regulated both locally, via predictable methylatability of each site, and globally, through the core meiotic circuitry. The methyltransferase complex components localize to the yeast nucleolus, and this localization is essential for mRNA methylation. Our data illuminate a conserved, dynamically regulated methylation program in yeast meiosis and provide an important resource for studying the function of this epitranscriptomic modification., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
38. Human disease locus discovery and mapping to molecular pathways through phylogenetic profiling.
- Author
-
Tabach Y, Golan T, Hernández-Hernández A, Messer AR, Fukuda T, Kouznetsova A, Liu JG, Lilienthal I, Levy C, and Ruvkun G
- Subjects
- Algorithms, Amino Acid Sequence, Animals, Bacteria genetics, Bacteria metabolism, Cell Line, Tumor, Chromosome Mapping, Databases, Genetic, Fungi genetics, Fungi metabolism, Gene Regulatory Networks, Genetic Loci, Humans, Immunoglobulin J Recombination Signal Sequence-Binding Protein classification, Immunoglobulin J Recombination Signal Sequence-Binding Protein metabolism, Metabolic Networks and Pathways, Microphthalmia-Associated Transcription Factor classification, Microphthalmia-Associated Transcription Factor metabolism, Microphthalmos metabolism, Microphthalmos pathology, Molecular Sequence Data, Sequence Alignment, Sequence Homology, Amino Acid, Evolution, Molecular, Genome, Immunoglobulin J Recombination Signal Sequence-Binding Protein genetics, Microphthalmia-Associated Transcription Factor genetics, Microphthalmos genetics, Phylogeny
- Abstract
Genes with common profiles of the presence and absence in disparate genomes tend to function in the same pathway. By mapping all human genes into about 1000 clusters of genes with similar patterns of conservation across eukaryotic phylogeny, we determined that sets of genes associated with particular diseases have similar phylogenetic profiles. By focusing on those human phylogenetic gene clusters that significantly overlap some of the thousands of human gene sets defined by their coexpression or annotation to pathways or other molecular attributes, we reveal the evolutionary map that connects molecular pathways and human diseases. The other genes in the phylogenetic clusters enriched for particular known disease genes or molecular pathways identify candidate genes for roles in those same disorders and pathways. Focusing on proteins coevolved with the microphthalmia-associated transcription factor (MITF), we identified the Notch pathway suppressor of hairless (RBP-Jk/SuH) transcription factor, and showed that RBP-Jk functions as an MITF cofactor.
- Published
- 2013
- Full Text
- View/download PDF
39. Identification of small RNA pathway genes using patterns of phylogenetic conservation and divergence.
- Author
-
Tabach Y, Billi AC, Hayes GD, Newman MA, Zuk O, Gabel H, Kamath R, Yacoby K, Chapman B, Garcia SM, Borowsky M, Kim JK, and Ruvkun G
- Subjects
- Animals, Caenorhabditis elegans classification, Caenorhabditis elegans Proteins genetics, Eukaryota classification, Eukaryota genetics, Genome genetics, MicroRNAs genetics, Proteome, RNA Splicing, Caenorhabditis elegans genetics, Genetic Variation, Phylogeny, RNA, Small Interfering genetics
- Abstract
Genetic and biochemical analyses of RNA interference (RNAi) and microRNA (miRNA) pathways have revealed proteins such as Argonaute and Dicer as essential cofactors that process and present small RNAs to their targets. Well-validated small RNA pathway cofactors such as these show distinctive patterns of conservation or divergence in particular animal, plant, fungal and protist species. We compared 86 divergent eukaryotic genome sequences to discern sets of proteins that show similar phylogenetic profiles with known small RNA cofactors. A large set of additional candidate small RNA cofactors have emerged from functional genomic screens for defects in miRNA- or short interfering RNA (siRNA)-mediated repression in Caenorhabditis elegans and Drosophila melanogaster, and from proteomic analyses of proteins co-purifying with validated small RNA pathway proteins. The phylogenetic profiles of many of these candidate small RNA pathway proteins are similar to those of known small RNA cofactor proteins. We used a Bayesian approach to integrate the phylogenetic profile analysis with predictions from diverse transcriptional coregulation and proteome interaction data sets to assign a probability for each protein for a role in a small RNA pathway. Testing high-confidence candidates from this analysis for defects in RNAi silencing, we found that about one-half of the predicted small RNA cofactors are required for RNAi silencing. Many of the newly identified small RNA pathway proteins are orthologues of proteins implicated in RNA splicing. In support of a deep connection between the mechanism of RNA splicing and small-RNA-mediated gene silencing, the presence of the Argonaute proteins and other small RNA components in the many species analysed strongly correlates with the number of introns in those species.
- Published
- 2013
- Full Text
- View/download PDF
40. Mutant p53(R175H) upregulates Twist1 expression and promotes epithelial-mesenchymal transition in immortalized prostate cells.
- Author
-
Kogan-Sakin I, Tabach Y, Buganim Y, Molchadsky A, Solomon H, Madar S, Kamer I, Stambolsky P, Shelly A, Goldfinger N, Valsesia-Wittmann S, Puisieux A, Zundelevich A, Gal-Yam EN, Avivi C, Barshack I, Brait M, Sidransky D, Domany E, and Rotter V
- Subjects
- Amino Acid Substitution, Cell Line, Transformed, Cell Line, Tumor, Epigenesis, Genetic, Histones metabolism, Humans, Male, Mutation, Nuclear Proteins genetics, Polycomb Repressive Complex 1, Promoter Regions, Genetic, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Proto-Oncogene Proteins metabolism, Repressor Proteins metabolism, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 physiology, Twist-Related Protein 1 genetics, Up-Regulation, Epithelial-Mesenchymal Transition, Nuclear Proteins metabolism, Prostatic Neoplasms metabolism, Tumor Suppressor Protein p53 metabolism, Twist-Related Protein 1 metabolism
- Abstract
A mutation within one allele of the p53 tumor suppressor gene can inactivate the remaining wild-type allele in a dominant-negative manner and in some cases can exert an additional oncogenic activity, known as mutant p53 'gain of function' (GOF). To study the role of p53 mutations in prostate cancer and to discriminate between the dominant-negative effect and the GOF activity of mutant p53, we measured, using microarrays, the expression profiles of three immortalized prostate epithelial cultures expressing wild-type, inactivated p53 or mutated p53. Analysis of these gene expression profiles showed that both inactivated p53 and p53(R175H) mutant expression resulted in the upregulation of cell cycle progression genes. A second group, which was upregulated exclusively by mutant p53(R175H), was predominantly enriched in developmental genes. This group of genes included the Twist1, a regulator of metastasis and epithelial-mesenchymal transition (EMT). Twist1 levels were also elevated in metastatic prostate cancer-derived cell line DU145, in immortalized lung fibroblasts and in a subset of lung cancer samples, all in a mutant p53-dependent manner. p53(R175H) mutant bearing immortalized epithelial cells showed typical features of EMT, such as higher expression of mesenchymal markers, lower expression of epithelial markers and enhanced invasive properties in vitro. The mechanism by which p53(R175H) mutant induces Twist1 expression involves alleviation of the epigenetic repression. Our data suggest that Twist1 expression might be upregulated following p53 mutation in cancer cells.
- Published
- 2011
- Full Text
- View/download PDF
41. Amplification of the 20q chromosomal arm occurs early in tumorigenic transformation and may initiate cancer.
- Author
-
Tabach Y, Kogan-Sakin I, Buganim Y, Solomon H, Goldfinger N, Hovland R, Ke XS, Oyan AM, Kalland KH, Rotter V, and Domany E
- Subjects
- Cell Line, Tumor, Computational Biology, Disease Progression, Genes, Neoplasm, Humans, Male, Neoplasms etiology, Neoplasms pathology, Prostatic Neoplasms, Signal Transduction, Cell Transformation, Neoplastic genetics, Chromosomes, Human, Pair 20 genetics, Gene Amplification, Neoplasms genetics
- Abstract
Duplication of chromosomal arm 20q occurs in prostate, cervical, colon, gastric, bladder, melanoma, pancreas and breast cancer, suggesting that 20q amplification may play a causal role in tumorigenesis. According to an alternative view, chromosomal imbalance is mainly a common side effect of cancer progression. To test whether a specific genomic aberration might serve as a cancer initiating event, we established an in vitro system that models the evolutionary process of early stages of prostate tumor formation; normal prostate cells were immortalized by the over-expression of human telomerase catalytic subunit hTERT, and cultured for 650 days till several transformation hallmarks were observed. Gene expression patterns were measured and chromosomal aberrations were monitored by spectral karyotype analysis at different times. Several chromosomal aberrations, in particular duplication of chromosomal arm 20q, occurred early in the process and were fixed in the cell populations, while other aberrations became extinct shortly after their appearance. A wide range of bioinformatic tools, applied to our data and to data from several cancer databases, revealed that spontaneous 20q amplification can promote cancer initiation. Our computational model suggests that 20q amplification induced deregulation of several specific cancer-related pathways including the MAPK pathway, the p53 pathway and Polycomb group factors. In addition, activation of Myc, AML, B-Catenin and the ETS family transcription factors was identified as an important step in cancer development driven by 20q amplification. Finally we identified 13 "cancer initiating genes", located on 20q13, which were significantly over-expressed in many tumors, with expression levels correlated with tumor grade and outcome suggesting that these genes induce the malignant process upon 20q amplification.
- Published
- 2011
- Full Text
- View/download PDF
42. Modulation of the vitamin D3 response by cancer-associated mutant p53.
- Author
-
Stambolsky P, Tabach Y, Fontemaggi G, Weisz L, Maor-Aloni R, Siegfried Z, Shiff I, Kogan I, Shay M, Kalo E, Blandino G, Simon I, Oren M, and Rotter V
- Subjects
- Apoptosis, Cell Line, Tumor, Cell Nucleus metabolism, Chromatin Immunoprecipitation, Gene Expression Regulation, Neoplastic, Humans, Oligonucleotide Array Sequence Analysis, Promoter Regions, Genetic, Transcriptional Activation, Cholecalciferol metabolism, Tumor Suppressor Protein p53 genetics, Vitamin D Response Element physiology
- Abstract
The p53 gene is mutated in many human tumors. Cells of such tumors often contain abundant mutant p53 (mutp53) protein, which may contribute actively to tumor progression via a gain-of-function mechanism. We applied ChIP-on-chip analysis and identified the vitamin D receptor (VDR) response element as overrepresented in promoter sequences bound by mutp53. We report that mutp53 can interact functionally and physically with VDR. Mutp53 is recruited to VDR-regulated genes and modulates their expression, augmenting the transactivation of some genes and relieving the repression of others. Furthermore, mutp53 increases the nuclear accumulation of VDR. Importantly, mutp53 converts vitamin D into an antiapoptotic agent. Thus, p53 status can determine the biological impact of vitamin D on tumor cells., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
43. Wide-scale analysis of human functional transcription factor binding reveals a strong bias towards the transcription start site.
- Author
-
Tabach Y, Brosh R, Buganim Y, Reiner A, Zuk O, Yitzhaky A, Koudritsky M, Rotter V, and Domany E
- Subjects
- Animals, Binding Sites, Cell Cycle, Humans, Mice, TATA Box, Transcription Factors chemistry, Promoter Regions, Genetic, Transcription Factors metabolism, Transcription Initiation Site
- Abstract
Background: Transcription factors (TF) regulate expression by binding to specific DNA sequences. A binding event is functional when it affects gene expression. Functionality of a binding site is reflected in conservation of the binding sequence during evolution and in over represented binding in gene groups with coherent biological functions. Functionality is governed by several parameters such as the TF-DNA binding strength, distance of the binding site from the transcription start site (TSS), DNA packing, and more. Understanding how these parameters control functionality of different TFs in different biological contexts is a must for identifying functional TF binding sites and for understanding regulation of transcription., Methodology/principal Findings: We introduce a novel method to screen the promoters of a set of genes with shared biological function (obtained from the functional Gene Ontology (GO) classification) against a precompiled library of motifs, and find those motifs which are statistically over-represented in the gene set. More than 8,000 human (and 23,000 mouse) genes, were assigned to one of 134 GO sets. Their promoters were searched (from 200 bp downstream to 1,000 bp upstream the TSS) for 414 known DNA motifs. We optimized the sequence similarity score threshold, independently for every location window, taking into account nucleotide heterogeneity along the promoters of the target genes. The method, combined with binding sequence and location conservation between human and mouse, identifies with high probability functional binding sites for groups of functionally-related genes. We found many location-sensitive functional binding events and showed that they clustered close to the TSS. Our method and findings were tested experimentally., Conclusions/significance: We identified reliably functional TF binding sites. This is an essential step towards constructing regulatory networks. The promoter region proximal to the TSS is of central importance for regulation of transcription in human and mouse, just as it is in bacteria and yeast.
- Published
- 2007
- Full Text
- View/download PDF
44. Gene expression analysis reveals a strong signature of an interferon-induced pathway in childhood lymphoblastic leukemia as well as in breast and ovarian cancer.
- Author
-
Einav U, Tabach Y, Getz G, Yitzhaky A, Ozbek U, Amariglio N, Izraeli S, Rechavi G, and Domany E
- Subjects
- Base Sequence, Child, DNA Primers, Female, Humans, Polymerase Chain Reaction, Breast Neoplasms genetics, Gene Expression Profiling, Interferons physiology, Ovarian Neoplasms genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
On the basis of epidemiological studies, infection was suggested to play a role in the etiology of human cancer. While for some cancers such a role was indeed demonstrated, there is no direct biological support for the role of viral pathogens in the pathogenesis of childhood leukemia. Using a novel bioinformatic tool that alternates between clustering and standard statistical methods of analysis, we performed a 'double-blind' search of published gene expression data of subjects with different childhood acute lymphoblastic leukemia (ALL) subtypes, looking for unanticipated partitions of patients, induced by unexpected groups of genes with correlated expression. We discovered a group of about 30 genes, related to the interferon response pathway, whose expression levels divide the ALL samples into two subgroups; high in 50, low in 285 patients. Leukemic subclasses prevalent in early childhood (the age most susceptible to infection) are over-represented in the high-expression subgroup. Similar partitions, induced by the same genes, were found also in breast and ovarian cancer but not in lung cancer, prostate cancer and lymphoma. About 40% of breast cancer samples expressed the 'interferon-related' signature. It is of interest that several studies demonstrated mouse mammary tumor virus-like sequences in about 40% of breast cancer samples. Our discovery of an unanticipated strong signature of an interferon-induced pathway provides molecular support for a role for either inflammation or viral infection in the pathogenesis of childhood leukemia as well as breast and ovarian cancer.
- Published
- 2005
- Full Text
- View/download PDF
45. Transcriptional programs following genetic alterations in p53, INK4A, and H-Ras genes along defined stages of malignant transformation.
- Author
-
Milyavsky M, Tabach Y, Shats I, Erez N, Cohen Y, Tang X, Kalis M, Kogan I, Buganim Y, Goldfinger N, Ginsberg D, Harris CC, Domany E, and Rotter V
- Subjects
- Cell Differentiation genetics, Cyclin-Dependent Kinase Inhibitor p16 biosynthesis, DNA-Binding Proteins, Fibroblasts cytology, Fibroblasts physiology, Gene Expression Profiling, Gene Silencing, Humans, Lung cytology, Lung physiology, Telomerase genetics, Cell Transformation, Neoplastic genetics, Cyclin-Dependent Kinase Inhibitor p16 genetics, Genes, p53 genetics, Genes, ras genetics, Transcription, Genetic genetics
- Abstract
The difficulty to dissect a complex phenotype of established malignant cells to several critical transcriptional programs greatly impedes our understanding of the malignant transformation. The genetic elements required to transform some primary human cells to a tumorigenic state were described in several recent studies. We took the advantage of the global genomic profiling approach and tried to go one step further in the dissection of the transformation network. We sought to identify the genetic signatures and key target genes, which underlie the genetic alterations in p53, Ras, INK4A locus, and telomerase, introduced in a stepwise manner into primary human fibroblasts. Here, we show that these are the minimally required genetic alterations for sarcomagenesis in vivo. A genome-wide expression profiling identified distinct genetic signatures corresponding to the genetic alterations listed above. Most importantly, unique transformation hallmarks, such as differentiation block, aberrant mitotic progression, increased angiogenesis, and invasiveness, were identified and coupled with genetic signatures assigned for the genetic alterations in the p53, INK4A locus, and H-Ras, respectively. Furthermore, a transcriptional program that defines the cellular response to p53 inactivation was an excellent predictor of metastasis development and bad prognosis in breast cancer patients. Deciphering these transformation fingerprints, which are affected by the most common oncogenic mutations, provides considerable insight into regulatory circuits controlling malignant transformation and will hopefully open new avenues for rational therapeutic decisions.
- Published
- 2005
- Full Text
- View/download PDF
46. The promoters of human cell cycle genes integrate signals from two tumor suppressive pathways during cellular transformation.
- Author
-
Tabach Y, Milyavsky M, Shats I, Brosh R, Zuk O, Yitzhaky A, Mantovani R, Domany E, Rotter V, and Pilpel Y
- Subjects
- Animals, Cell Cycle Proteins physiology, Cell Division, Cell Line, Transformed metabolism, Cell Line, Transformed transplantation, Computational Biology, DNA-Binding Proteins genetics, DNA-Binding Proteins physiology, Fibroblasts cytology, Fibroblasts metabolism, Gene Expression Regulation, Genes, p16, Genes, p53, Humans, Mice, Mice, Nude, Promoter Regions, Genetic genetics, Recombinant Fusion Proteins physiology, Regulatory Sequences, Nucleic Acid, Spindle Apparatus metabolism, Telomerase genetics, Telomerase physiology, Transcription, Genetic, Transplantation, Heterologous, Cell Cycle Proteins biosynthesis, Cell Transformation, Neoplastic genetics, Cyclin-Dependent Kinase Inhibitor p16 physiology, Gene Expression Profiling, Genes, Tumor Suppressor, Genes, cdc, Promoter Regions, Genetic physiology, Tumor Suppressor Protein p53 physiology
- Abstract
Deciphering regulatory events that drive malignant transformation represents a major challenge for systems biology. Here, we analyzed genome-wide transcription profiling of an in vitro cancerous transformation process. We focused on a cluster of genes whose expression levels increased as a function of p53 and p16(INK4A) tumor suppressors inactivation. This cluster predominantly consists of cell cycle genes and constitutes a signature of a diversity of cancers. By linking expression profiles of the genes in the cluster with the dynamic behavior of p53 and p16(INK4A), we identified a promoter architecture that integrates signals from the two tumor suppressive channels and that maps their activity onto distinct levels of expression of the cell cycle genes, which, in turn, correspond to different cellular proliferation rates. Taking components of the mitotic spindle as an example, we experimentally verified our predictions that p53-mediated transcriptional repression of several of these novel targets is dependent on the activities of p21, NFY, and E2F. Our study demonstrates how a well-controlled transformation process allows linking between gene expression, promoter architecture, and activity of upstream signaling molecules.
- Published
- 2005
- Full Text
- View/download PDF
47. Prolonged culture of telomerase-immortalized human fibroblasts leads to a premalignant phenotype.
- Author
-
Milyavsky M, Shats I, Erez N, Tang X, Senderovich S, Meerson A, Tabach Y, Goldfinger N, Ginsberg D, Harris CC, and Rotter V
- Subjects
- Cell Division physiology, Cell Transformation, Neoplastic metabolism, Contact Inhibition, Cyclin-Dependent Kinase Inhibitor p16 biosynthesis, Cyclin-Dependent Kinase Inhibitor p16 genetics, DNA-Binding Proteins, Fibroblasts cytology, Fibroblasts enzymology, Gene Silencing, Genes, p53 physiology, Genes, ras physiology, Humans, Precancerous Conditions pathology, Retinoblastoma Protein genetics, Retroviridae enzymology, Retroviridae genetics, Telomerase biosynthesis, Telomerase genetics, Time Factors, Transduction, Genetic, Tumor Suppressor Protein p14ARF biosynthesis, Tumor Suppressor Protein p14ARF genetics, Cell Transformation, Neoplastic pathology, Precancerous Conditions enzymology, Telomerase metabolism
- Abstract
Telomere shortening in primary human fibroblasts results in replicative senescence, which can be overcome by telomerase (hTERT) overexpression. However, because immortalization is one of the hallmarks of malignant transformation, careful analysis of hTERT-immortalized cells is of crucial importance for understanding both processes. To this end, we infected WI-38 fibroblasts with a retrovirus carrying the hTERT cDNA and analyzed their proliferative behavior during 600 days [ approximately 500 population doublings (PDLs)] of continuous culture. Growth of three independent mass cultures was uniform for approximately 150 PDLs after telomerase infection, followed by a progressive acceleration of growth in two of three cultures. Expression of p16(INK4A) was significantly elevated in the immortalized cells but gradually disappeared during the accelerated growth phase. This alteration correlated with loss of the contact inhibition response and conferred the cells with sensitivity to H-Ras-induced transformation. In contrast, the p53- and pRb-mediated checkpoints such as the DNA damage response, chromosomal stability and entry into quiescence remained intact, irrespective of INK4A locus expression. Importantly, detailed examination of one of the WI-38/hTERT cultures during the accelerated growth phase revealed overexpression of the c-myc and Bmi-1 oncogenes, as well as loss of p14(ARF) expression. Collectively, our results indicate that although hTERT-immortalized cells behave similarly to primary cells during the first 150 PDLs, long-term growth in culture may favor the appearance of clones carrying potentially malignant alterations.
- Published
- 2003
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.