Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 82(15): 2885-2899.e8, 2022 08 04.
Article in English | MEDLINE | ID: mdl-35841888

ABSTRACT

Translated small open reading frames (smORFs) can have important regulatory roles and encode microproteins, yet their genome-wide identification has been challenging. We determined the ribosome locations across six primary human cell types and five tissues and detected 7,767 smORFs with translational profiles matching those of known proteins. The human genome was found to contain highly cell-type- and tissue-specific smORFs and a subset that encodes highly conserved amino acid sequences. Changes in the translational efficiency of upstream-encoded smORFs (uORFs) and the corresponding main ORFs predominantly occur in the same direction. Integration with 456 mass-spectrometry datasets confirms the presence of 603 small peptides at the protein level in humans and provides insights into the subcellular localization of these small proteins. This study provides a comprehensive atlas of high-confidence translated smORFs derived from primary human cells and tissues in order to provide a more complete understanding of the translated human genome.


Subject(s)
Gene Expression Regulation , Ribosomes , Genome, Human/genetics , Humans , Open Reading Frames/genetics , Protein Biosynthesis , Proteins/metabolism , RNA/metabolism , Ribosomes/genetics , Ribosomes/metabolism
2.
Proc Natl Acad Sci U S A ; 116(19): 9622-9627, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31015293

ABSTRACT

White matter abnormalities are a nearly universal pathological feature of neurodegenerative disorders including Huntington disease (HD). A long-held assumption is that this white matter pathology is simply a secondary outcome of the progressive neuronal loss that manifests with advancing disease. Using a mouse model of HD, here we show that white matter and myelination abnormalities are an early disease feature appearing before the manifestation of any behavioral abnormalities or neuronal loss. We further show that selective inactivation of mutant huntingtin (mHTT) in the NG2+ oligodendrocyte progenitor cell population prevented myelin abnormalities and certain behavioral deficits in HD mice. Strikingly, the improvements in behavioral outcomes were seen despite the continued expression of mHTT in nonoligodendroglial cells including neurons, astrocytes, and microglia. Using RNA-seq and ChIP-seq analyses, we implicate a pathogenic mechanism that involves enhancement of polycomb repressive complex 2 (PRC2) activity by mHTT in the intrinsic oligodendroglial dysfunction and myelination deficits observed in HD. Our findings challenge the long-held dogma regarding the etiology of white matter pathology in HD and highlight the contribution of epigenetic mechanisms to the observed intrinsic oligodendroglial dysfunction. Our results further suggest that ameliorating white matter pathology and oligodendroglial dysfunction may be beneficial for HD.


Subject(s)
Behavior, Animal , Demyelinating Diseases , Huntingtin Protein , Huntington Disease , Mutation , Oligodendroglia , Animals , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Disease Models, Animal , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Mice , Mice, Mutant Strains , Oligodendroglia/metabolism , Oligodendroglia/pathology , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , White Matter/metabolism , White Matter/pathology
3.
RNA ; 25(12): 1696-1713, 2019 12.
Article in English | MEDLINE | ID: mdl-31506381

ABSTRACT

Differential gene isoform expression is a ubiquitous mechanism to enhance proteome diversity and maintain cell homeostasis. Mechanisms such as splicing that drive gene isoform variability are highly dynamic and responsive to changes in cell signaling pathways. Wnt/ß-catenin signaling has profound effects on cell activity and cell fate and is known to modify several splicing events by altering the expression of individual splicing factors. However, a global assessment of how extensively Wnt signaling regulates splicing and other mechanisms that determine mRNA isoform composition in cancer is lacking. We used deep time-resolved RNA-seq in two independent in vivo Wnt-addicted tumor models during treatment with the potent Wnt inhibitor ETC-159 and examined Wnt regulated splicing events and splicing regulators. We found 1025 genes that underwent Wnt regulated variable exon usage leading to isoform expression changes. This was accompanied by extensive Wnt regulated changes in the expression of splicing regulators. Many of these Wnt regulated events were conserved in multiple human cancers, and many were linked to previously defined cancer-associated splicing quantitative trait loci. This suggests that the Wnt regulated splicing events are components of fundamental oncogenic processes. These findings demonstrate the wide-ranging effects of Wnt signaling on the isoform composition of the cell and provides an extensive resource of expression changes of splicing regulators and gene isoforms regulated by Wnt signaling.


Subject(s)
Biomarkers, Tumor , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/metabolism , Wnt Signaling Pathway , Alternative Splicing , Biomarkers , Cell Line, Tumor , Exons , Gene Expression Profiling , Humans , Neoplasms/pathology , Protein Isoforms , Quantitative Trait Loci , RNA, Messenger/genetics
4.
J Am Soc Nephrol ; 31(2): 309-323, 2020 02.
Article in English | MEDLINE | ID: mdl-31919106

ABSTRACT

BACKGROUND: Several genetic susceptibility loci associated with diabetic nephropathy have been documented, but no causative variants implying novel pathogenetic mechanisms have been elucidated. METHODS: We carried out whole-genome sequencing of a discovery cohort of Finnish siblings with type 1 diabetes who were discordant for the presence (case) or absence (control) of diabetic nephropathy. Controls had diabetes without complications for 15-37 years. We analyzed and annotated variants at genome, gene, and single-nucleotide variant levels. We then replicated the associated variants, genes, and regions in a replication cohort from the Finnish Diabetic Nephropathy study that included 3531 unrelated Finns with type 1 diabetes. RESULTS: We observed protein-altering variants and an enrichment of variants in regions associated with the presence or absence of diabetic nephropathy. The replication cohort confirmed variants in both regulatory and protein-coding regions. We also observed that diabetic nephropathy-associated variants, when clustered at the gene level, are enriched in a core protein-interaction network representing proteins essential for podocyte function. These genes include protein kinases (protein kinase C isoforms ε and ι) and protein tyrosine kinase 2. CONCLUSIONS: Our comprehensive analysis of a diabetic nephropathy cohort of siblings with type 1 diabetes who were discordant for kidney disease points to variants and genes that are potentially causative or protective for diabetic nephropathy. This includes variants in two isoforms of the protein kinase C family not previously linked to diabetic nephropathy, adding support to previous hypotheses that the protein kinase C family members play a role in diabetic nephropathy and might be attractive therapeutic targets.


Subject(s)
Diabetes Mellitus, Type 1/complications , Diabetic Nephropathies/genetics , Whole Genome Sequencing/methods , Adolescent , Adult , Animals , Child , Child, Preschool , Diabetes Mellitus, Type 1/genetics , Female , HEK293 Cells , Humans , Male , Polymorphism, Single Nucleotide , Protein Kinase C/physiology , Siblings , Young Adult , Zebrafish
5.
J Cell Sci ; 130(9): 1532-1544, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28289266

ABSTRACT

Wnt ligands are involved in diverse signaling pathways that are active during development, maintenance of tissue homeostasis and in various disease states. While signaling regulated by individual Wnts has been extensively studied, Wnts are rarely expressed alone, and the consequences of Wnt gene co-expression are not well understood. Here, we studied the effect of co-expression of Wnts on the ß-catenin signaling pathway. While some Wnts are deemed 'non-canonical' due to their limited ability to activate ß-catenin when expressed alone, unexpectedly, we find that multiple Wnt combinations can synergistically activate ß-catenin signaling in multiple cell types. WNT1- and WNT7B-mediated synergistic Wnt signaling requires FZD5, FZD8 and LRP6, as well as the WNT7B co-receptors GPR124 (also known as ADGRA2) and RECK. Unexpectedly, this synergistic signaling occurs downstream of ß-catenin stabilization, and is correlated with increased lysine acetylation of ß-catenin. Wnt synergy provides a general mechanism to confer increased combinatorial control over this important regulatory pathway.


Subject(s)
Signal Transduction , Wnt Proteins/metabolism , beta Catenin/metabolism , Acetylation , Clone Cells , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Models, Biological , Phosphorylation , Protein Stability , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/genetics , Stomach Neoplasms/genetics , Up-Regulation/genetics
6.
Ann Rheum Dis ; 77(4): 596-601, 2018 04.
Article in English | MEDLINE | ID: mdl-29348297

ABSTRACT

OBJECTIVES: Several common and rare risk variants have been reported for systemic sclerosis (SSc), but the effector cell(s) mediating the function of these genetic variants remains to be elucidated. While innate immune cells have been proposed as the critical targets to interfere with the disease process underlying SSc, no studies have comprehensively established their effector role. Here we investigated the contribution of monocyte-derived macrophages (MDMs) in mediating genetic susceptibility to SSc. METHODS: We carried out RNA sequencing and genome-wide genotyping in MDMs from 57 patients with SSc and 15 controls. Our differential expression and expression quantitative trait locus (eQTL) analysis in SSc was further integrated with epigenetic, expression and eQTL data from skin, monocytes, neutrophils and lymphocytes. RESULTS: We identified 602 genes upregulated and downregulated in SSc macrophages that were significantly enriched for genes previously implicated in SSc susceptibility (P=5×10-4), and 270 cis-regulated genes in MDMs. Among these, GSDMA was reported to carry an SSc risk variant (rs3894194) regulating expression of neighbouring genes in blood. We show that GSDMA is upregulated in SSc MDMs (P=8.4×10-4) but not in the skin, and is a significant eQTL in SSc macrophages and lipopolysaccharide/interferon gamma (IFNγ)-stimulated monocytes. Furthermore, we identify an SSc macrophage transcriptome signature characterised by upregulation of glycolysis, hypoxia and mTOR signalling and a downregulation of IFNγ response pathways. CONCLUSIONS: Our data further establish the link between macrophages and SSc, and suggest that the contribution of the rs3894194 risk variant to SSc susceptibility can be mediated by GSDMA expression in macrophages.


Subject(s)
Genetic Predisposition to Disease , Macrophages/cytology , Neoplasm Proteins/genetics , Scleroderma, Systemic/genetics , Transcriptome/genetics , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Female , Genotyping Techniques , Humans , Male , Quantitative Trait Loci/genetics , Risk Factors , Scleroderma, Systemic/pathology , Signal Transduction/genetics , Skin/metabolism , Young Adult
7.
BMC Genomics ; 16: 963, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26576536

ABSTRACT

BACKGROUND: Precise quantitative and spatiotemporal control of gene expression is necessary to ensure proper cellular differentiation and the maintenance of homeostasis. The relationship between gene expression and the spatial organisation of chromatin is highly complex, interdependent and not completely understood. The development of experimental techniques to interrogate both the higher-order structure of chromatin and the interactions between regulatory elements has recently lead to important insights on how gene expression is controlled. The ability to gain these and future insights is critically dependent on computational tools for the analysis and visualisation of data produced by these techniques. RESULTS AND CONCLUSION: We have developed GenomicInteractions, a freely available R/Bioconductor package designed for processing, analysis and visualisation of data generated from various types of chromosome conformation capture experiments. The package allows the easy annotation and summarisation of large genome-wide datasets at both the level of individual interactions and sets of genomic features, and provides several different methods for interrogating and visualising this type of data. We demonstrate this package's utility by showing example analyses performed on interaction datasets generated using Hi-C and ChIA-PET.


Subject(s)
Chromatin/genetics , Genomics/methods , Software , Animals , Computer Graphics , Databases, Genetic , Humans , K562 Cells , Mice , Molecular Sequence Annotation , Thymocytes/metabolism
8.
Nucleic Acids Res ; 41(15): 7185-99, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23766291

ABSTRACT

The precise regulation of gene transcription during metazoan development is controlled by a complex system of interactions between transcription factors, histone modifications and modifying enzymes and chromatin conformation. Developments in chromosome conformation capture technologies have revealed that interactions between regions of chromatin are pervasive and highly cell-type specific. The movement of enhancers and promoters in and out of higher-order chromatin structures within the nucleus are associated with changes in expression and histone modifications. However, the factors responsible for mediating these changes and determining enhancer:promoter specificity are still not completely known. In this review, we summarize what is known about the patterns of epigenetic and chromatin features characteristic of elements involved in long-range interactions. In addition, we review the insights into both local and global patterns of chromatin interactions that have been revealed by the latest experimental and computational methods.


Subject(s)
Chromatin/metabolism , Epigenesis, Genetic , Gene Expression Regulation , Transcription, Genetic , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/genetics , Chromatin Assembly and Disassembly , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , Enhancer Elements, Genetic , Evolution, Molecular , Genome, Human , Histones/genetics , Histones/metabolism , Humans , Promoter Regions, Genetic
9.
MicroPubl Biol ; 20242024.
Article in English | MEDLINE | ID: mdl-38764944

ABSTRACT

Lifespan studies on fast-aging model organisms like C.elegans and D.melanogaster are conducted with multiple organisms per vial. Lifespan data results in a "one row, multiple individuals" format, which is incompatible with R packages that require a "one row, one individual" format. We present ggbulksurv , an R package for user-friendly survival analysis and highlight three key features. (1) pivot_prism converts data for PRISM, allowing biologists to plot survival curves without manually expanding each observation. (2) run_bulksurv() takes in a "one row, multiple individuals" table and plots a customizable survival curve. (3) Advanced users who require custom survival objects can specify a custom formula, facilitating complex survival analysis. We provide a time saving solution for lifespan data analysis.

10.
Cells ; 13(5)2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38474329

ABSTRACT

Wnt signaling is a highly conserved metazoan pathway that plays a crucial role in cell fate determination and morphogenesis during development. Wnt ligands can induce disparate cellular responses. The exact mechanism behind these different outcomes is not fully understood but may be due to interactions with different receptors on the cell membrane. PTK7/Otk is a transmembrane receptor that is implicated in various developmental and physiological processes including cell polarity, cell migration, and invasion. Here, we examine two roles of Otk-1 and Otk-2 in patterning and neurogenesis. We find that Otk-1 is a positive regulator of signaling and Otk-2 functions as its inhibitor. We propose that PTK7/Otk functions in signaling, cell migration, and polarity contributing to the diversity of cellular responses seen in Wnt-mediated processes.


Subject(s)
Body Patterning , Neurogenesis , Receptor Protein-Tyrosine Kinases , Wnt Signaling Pathway , Animals , Cell Differentiation , Cell Membrane/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Wnt Signaling Pathway/physiology
11.
J Clin Invest ; 134(6)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38488003

ABSTRACT

Wnts, cholesterol, and MAPK signaling are essential for development and adult homeostasis. Here, we report that fatty acid hydroxylase domain containing 2 (FAXDC2), a previously uncharacterized enzyme, functions as a methyl sterol oxidase catalyzing C4 demethylation in the Kandutsch-Russell branch of the cholesterol biosynthesis pathway. FAXDC2, a paralog of MSMO1, regulated the abundance of the specific C4-methyl sterols lophenol and dihydro-T-MAS. Highlighting its clinical relevance, FAXDC2 was repressed in Wnt/ß-catenin-high cancer xenografts, in a mouse genetic model of Wnt activation, and in human colorectal cancers. Moreover, in primary human colorectal cancers, the sterol lophenol, regulated by FAXDC2, accumulated in the cancerous tissues and not in adjacent normal tissues. FAXDC2 linked Wnts to RTK/MAPK signaling. Wnt inhibition drove increased recycling of RTKs and activation of the MAPK pathway, and this required FAXDC2. Blocking Wnt signaling in Wnt-high cancers caused both differentiation and senescence; and this was prevented by knockout of FAXDC2. Our data show the integration of 3 ancient pathways, Wnts, cholesterol synthesis, and RTK/MAPK signaling, in cellular proliferation and differentiation.


Subject(s)
Colorectal Neoplasms , beta Catenin , Adult , Humans , Mice , Animals , beta Catenin/genetics , beta Catenin/metabolism , Wnt Signaling Pathway , Cell Proliferation , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism
12.
Nat Commun ; 15(1): 2513, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38514625

ABSTRACT

In multiple myeloma, abnormal plasma cells establish oncogenic niches within the bone marrow by engaging the NF-κB pathway to nurture their survival while they accumulate pro-proliferative mutations. Under these conditions, many cases eventually develop genetic abnormalities endowing them with constitutive NF-κB activation. Here, we find that sustained NF-κB/p52 levels resulting from such mutations favours the recruitment of enhancers beyond the normal B-cell repertoire. Furthermore, through targeted disruption of p52, we characterise how such enhancers are complicit in the formation of super-enhancers and the establishment of cis-regulatory interactions with myeloma dependencies during constitutive activation of p52. Finally, we functionally validate the pathological impact of these cis-regulatory modules on cell and tumour phenotypes using in vitro and in vivo models, confirming RGS1 as a p52-dependent myeloma driver. We conclude that the divergent epigenomic reprogramming enforced by aberrant non-canonical NF-κB signalling potentiates transcriptional programs beneficial for multiple myeloma progression.


Subject(s)
Multiple Myeloma , NF-kappa B , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Transcriptome , Epigenome , Signal Transduction/genetics , NF-kappa B p52 Subunit/metabolism
13.
Bioinformatics ; 28(2): 254-60, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22135416

ABSTRACT

MOTIVATION: The scientific literature contains a wealth of information about biological systems. Manual curation lacks the scalability to extract this information due to the ever-increasing numbers of papers being published. The development and application of text mining technologies has been proposed as a way of dealing with this problem. However, the inter-species ambiguity of the genomic nomenclature makes mapping of gene mentions identified in text to their corresponding Entrez gene identifiers an extremely difficult task. We propose a novel method, which transforms a MEDLINE record into a mixture of adjacency matrices; by performing a random walkover the resulting graph, we can perform multi-class supervised classification allowing the assignment of taxonomy identifiers to individual gene mentions. The ability to achieve good performance at this task has a direct impact on the performance of normalizing gene mentions to Entrez gene identifiers. Such graph mixtures add flexibility and allow us to generate probabilistic classification schemes that naturally reflect the uncertainties inherent, even in literature-derived data. RESULTS: Our method performs well in terms of both micro- and macro-averaged performance, achieving micro-F(1) of 0.76 and macro-F(1) of 0.36 on the publicly available DECA corpus. Re-curation of the DECA corpus was performed, with our method achieving 0.88 micro-F(1) and 0.51 macro-F(1). Our method improves over standard classification techniques [such as support vector machines (SVMs)] in a number of ways: flexibility, interpretability and its resistance to the effects of class bias in the training data. Good performance is achieved without the need for computationally expensive parse tree generation or 'bag of words classification'.


Subject(s)
Data Mining , Genes , Terminology as Topic , Animals , Humans , MEDLINE , Software , Species Specificity , Support Vector Machine , United States
14.
Biol Open ; 11(4)2022 04 15.
Article in English | MEDLINE | ID: mdl-35377406

ABSTRACT

The expression of a large number of genes is regulated by regulatory elements that are located far away from their promoters. Identifying which gene is the target of a specific regulatory element or is affected by a non-coding mutation is often accomplished by assigning these regions to the nearest gene in the genome. However, this heuristic ignores key features of genome organisation and gene regulation; in that the genome is partitioned into regulatory domains, which at some loci directly coincide with the span of topologically associated domains (TADs), and that genes are regulated by enhancers located throughout these regions, even across intervening genes. In this review, we examine the results from genome-wide studies using chromosome conformation capture technologies and from those dissecting individual gene regulatory domains, to highlight that the phenomenon of enhancer skipping is pervasive and affects multiple types of genes. We discuss how simply assigning a genomic region of interest to its nearest gene is problematic and often leads to incorrect predictions and highlight that where possible information on both the conservation and topological organisation of the genome should be used to generate better hypotheses. The article has an associated Future Leader to Watch interview.


Subject(s)
Genome-Wide Association Study , Genome , Gene Expression Regulation , Genomics , Promoter Regions, Genetic
15.
J Clin Invest ; 132(12)2022 06 15.
Article in English | MEDLINE | ID: mdl-35536676

ABSTRACT

Wnt signaling regulates the balance between stemness and differentiation in multiple tissues and in cancer. RNF43-mutant pancreatic cancers are dependent on Wnt production, and pharmacologic blockade of the pathway, e.g., by PORCN inhibitors, leads to tumor differentiation. However, primary resistance to these inhibitors has been observed. To elucidate potential mechanisms, we performed in vivo CRISPR screens in PORCN inhibitor-sensitive RNF43-mutant pancreatic cancer xenografts. As expected, genes in the Wnt pathway whose loss conferred drug resistance were identified, including APC, AXIN1, and CTNNBIP1. Unexpectedly, the screen also identified the histone acetyltransferase EP300 (p300), but not its paralog, CREBBP (CBP). We found that EP300 is silenced due to genetic alterations in all the existing RNF43-mutant pancreatic cancer cell lines that are resistant to PORCN inhibitors. Mechanistically, loss of EP300 directly downregulated GATA6 expression, thereby silencing the GATA6-regulated differentiation program and leading to a phenotypic transition from the classical subtype to the dedifferentiated basal-like/squamous subtype of pancreatic cancer. EP300 mutation and loss of GATA6 function bypassed the antidifferentiation activity of Wnt signaling, rendering these cancer cells resistant to Wnt inhibition.


Subject(s)
Pancreatic Neoplasms , Acyltransferases/genetics , Cell Line, Tumor , E1A-Associated p300 Protein/metabolism , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Humans , Membrane Proteins/genetics , Mutation , Pancreatic Neoplasms/pathology , Wnt Signaling Pathway , Pancreatic Neoplasms
16.
Cells ; 11(2)2022 01 14.
Article in English | MEDLINE | ID: mdl-35053396

ABSTRACT

Patients with Alzheimer's disease suffer from a decrease in brain mass and a prevalence of amyloid-ß plaques. These plaques are thought to play a role in disease progression, but their exact role is not entirely established. We developed an optogenetic model to induce amyloid-ß intracellular oligomerization to model distinct disease etiologies. Here, we examine the effect of Wnt signaling on amyloid in an optogenetic, Drosophila gut stem cell model. We observe that Wnt activation rescues the detrimental effects of amyloid expression and oligomerization. We analyze the gene expression changes downstream of Wnt that contribute to this rescue and find changes in aging related genes, protein misfolding, metabolism, and inflammation. We propose that Wnt expression reduces inflammation through repression of Toll activating factors. We confirm that chronic Toll activation reduces lifespan, but a decrease in the upstream activator Persephone extends it. We propose that the protective effect observed for lithium treatment functions, at least in part, through Wnt activation and the inhibition of inflammation.


Subject(s)
Amyloid beta-Peptides/toxicity , Drosophila melanogaster/metabolism , Intestines/pathology , Stem Cells/pathology , Wnt Signaling Pathway , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/drug effects , Drosophila melanogaster/embryology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Gene Expression Regulation/drug effects , Longevity/drug effects , Optogenetics , Stem Cells/drug effects , Stem Cells/metabolism , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics
17.
Aging (Albany NY) ; 14(20): 8270-8291, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36287172

ABSTRACT

Why biological age is a major risk factor for many of the most important human diseases remains mysterious. We know that as organisms age, stem cell pools are exhausted while senescent cells progressively accumulate. Independently, induction of pluripotency via expression of Yamanaka factors (Oct4, Klf4, Sox2, c-Myc; OKSM) and clearance of senescent cells have each been shown to ameliorate cellular and physiological aspects of aging, suggesting that both processes are drivers of organismal aging. But stem cell exhaustion and cellular senescence likely interact in the etiology and progression of age-dependent diseases because both undermine tissue and organ homeostasis in different if not complementary ways. Here, we combine transient cellular reprogramming (stem cell rejuvenation) with targeted removal of senescent cells to test the hypothesis that simultaneously targeting both cell-fate based aging mechanisms will maximize life and health span benefits. We find that OKSM extends lifespan and show that both interventions protect the intestinal stem cell pool, lower inflammation, activate pro-stem cell signaling pathways, and synergistically improve health and lifespan. Our findings suggest that a combination therapy, simultaneously replacing lost stem cells and removing senescent cells, shows synergistic potential for anti-aging treatments. Our finding that transient expression of both is the most effective suggests that drug-based treatments in non-genetically tractable organisms will likely be the most translatable.


Subject(s)
Longevity , Rejuvenation , Humans , Longevity/physiology , Rejuvenation/physiology , Cellular Senescence/physiology , Aging/physiology , Stem Cells
18.
Sci Rep ; 12(1): 7684, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35538124

ABSTRACT

Proper embryonic development requires directional axes to pattern cells into embryonic structures. In Drosophila, spatially discrete expression of transcription factors determines the anterior to posterior organization of the early embryo, while the Toll and TGFß signalling pathways determine the early dorsal to ventral pattern. Embryonic MAPK/ERK signaling contributes to both anterior to posterior patterning in the terminal regions and to dorsal to ventral patterning during oogenesis and embryonic stages. Here we describe a novel loss of function mutation in the Raf kinase gene, which leads to loss of ventral cell fates as seen through the loss of the ventral furrow, the absence of Dorsal/NFκB nuclear localization, the absence of mesoderm determinants Twist and Snail, and the expansion of TGFß. Gene expression analysis showed cells adopting ectodermal fates much like loss of Toll signaling. Our results combine novel mutants, live imaging, optogenetics and transcriptomics to establish a novel role for Raf, that appears to be independent of the MAPK cascade, in embryonic patterning.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Body Patterning/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Oogenesis , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
19.
Hum Genomics ; 5(1): 17-29, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21106487

ABSTRACT

Keeping up with the rapidly growing literature has become virtually impossible for most scientists. This can have dire consequences. First, we may waste research time and resources on reinventing the wheel simply because we can no longer maintain a reliable grasp on the published literature. Second, and perhaps more detrimental, judicious (or serendipitous) combination of knowledge from different scientific disciplines, which would require following disparate and distinct research literatures, is rapidly becoming impossible for even the most ardent readers of research publications. Text mining - the automated extraction of information from (electronically) published sources - could potentially fulfil an important role - but only if we know how to harness its strengths and overcome its weaknesses. As we do not expect that the rate at which scientific results are published will decrease, text mining tools are now becoming essential in order to cope with, and derive maximum benefit from, this information explosion. In genomics, this is particularly pressing as more and more rare disease-causing variants are found and need to be understood. Not being conversant with this technology may put scientists and biomedical regulators at a severe disadvantage. In this review, we introduce the basic concepts underlying modern text mining and its applications in genomics and systems biology. We hope that this review will serve three purposes: (i) to provide a timely and useful overview of the current status of this field, including a survey of present challenges; (ii) to enable researchers to decide how and when to apply text mining tools in their own research; and (iii) to highlight how the research communities in genomics and systems biology can help to make text mining from biomedical abstracts and texts more straightforward.


Subject(s)
Data Mining/methods , Genomics/methods , Periodicals as Topic , Systems Biology/methods , Publication Bias , Terminology as Topic
20.
Cancer Res ; 81(2): 464-475, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33203702

ABSTRACT

Aberrant Wnt signaling drives a number of cancers through regulation of diverse downstream pathways. Wnt/ß-catenin signaling achieves this in part by increasing the expression of proto-oncogenes such as MYC and cyclins. However, global assessment of the Wnt-regulated transcriptome in vivo in genetically distinct cancers demonstrates that Wnt signaling suppresses the expression of as many genes as it activates. In this study, we examined the set of genes that are upregulated upon inhibition of Wnt signaling in Wnt-addicted pancreatic and colorectal cancer models. Decreasing Wnt signaling led to a marked increase in gene expression by activating ERK and JNK; these changes in gene expression could be mitigated in part by concurrent inhibition of MEK. These findings demonstrate that increased Wnt signaling in cancer represses MAPK activity, preventing RAS-mediated senescence while allowing cancer cells to proliferate. These results shift the paradigm from Wnt/ß-catenin primarily as an activator of transcription to a more nuanced view where Wnt/ß-catenin signaling drives both widespread gene repression and activation. SIGNIFICANCE: These findings show that Wnt/ß-catenin signaling causes widespread gene repression via inhibition of MAPK signaling, thus fine tuning the RAS-MAPK pathway to optimize proliferation in cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/pathology , Gene Expression Regulation, Neoplastic , Mitogen-Activated Protein Kinases/metabolism , Pancreatic Neoplasms/pathology , Wnt1 Protein/metabolism , beta Catenin/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Cell Proliferation , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mitogen-Activated Protein Kinases/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pyridones/pharmacology , Pyrimidinones/pharmacology , Tumor Cells, Cultured , Wnt1 Protein/genetics , Xenograft Model Antitumor Assays , beta Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL