Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
NAR Cancer ; 6(3): zcae029, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38989007

ABSTRACT

In the past few decades, our view of ribosomes has changed substantially. Rather than passive machines without significant variability, it is now acknowledged that they are heterogeneous, and have direct regulatory capacity. This 'ribosome heterogeneity' comes in many flavors, including in both the RNA and protein components of ribosomes, so there are many paths through which ribosome specialization could arise. It is easy to imagine that specialized ribosomes could have wide physiological roles, through the translation of specific mRNA populations, and there is now evidence for this in several contexts. Translation is highly dysregulated in cancer, needed to support oncogenic phenotypes and to overcome cellular stress. However, the role of ribosome specialization in this is not clear. In this review we focus on specialized ribosomes in cancer. Specifically, we assess the impact that post-translational modifications and differential ribosome incorporation of ribosomal proteins (RPs) have in this disease. We focus on studies that have shown a ribosome-mediated change in translation of specific mRNA populations, and hypothesize how such a process could be driving other phenotypes. We review the impact of RP-mediated heterogeneity in both intrinsic and extrinsic oncogenic processes, and consider how this knowledge could be leveraged to benefit patients.

2.
Cancer Cell ; 42(4): 623-645.e10, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38490212

ABSTRACT

Genes limiting T cell antitumor activity may serve as therapeutic targets. It has not been systematically studied whether there are regulators that uniquely or broadly contribute to T cell fitness. We perform genome-scale CRISPR-Cas9 knockout screens in primary CD8 T cells to uncover genes negatively impacting fitness upon three modes of stimulation: (1) intense, triggering activation-induced cell death (AICD); (2) acute, triggering expansion; (3) chronic, causing dysfunction. Besides established regulators, we uncover genes controlling T cell fitness either specifically or commonly upon differential stimulation. Dap5 ablation, ranking highly in all three screens, increases translation while enhancing tumor killing. Loss of Icam1-mediated homotypic T cell clustering amplifies cell expansion and effector functions after both acute and intense stimulation. Lastly, Ctbp1 inactivation induces functional T cell persistence exclusively upon chronic stimulation. Our results functionally annotate fitness regulators based on their unique or shared contribution to traits limiting T cell antitumor activity.


Subject(s)
CRISPR-Cas Systems , Neoplasms , Humans , CD8-Positive T-Lymphocytes , Neoplasms/genetics
3.
Mol Cell ; 84(6): 1078-1089.e4, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38340715

ABSTRACT

Aberrantly slow ribosomes incur collisions, a sentinel of stress that triggers quality control, signaling, and translation attenuation. Although each collision response has been studied in isolation, the net consequences of their collective actions in reshaping translation in cells is poorly understood. Here, we apply cryoelectron tomography to visualize the translation machinery in mammalian cells during persistent collision stress. We find that polysomes are compressed, with up to 30% of ribosomes in helical polysomes or collided disomes, some of which are bound to the stress effector GCN1. The native collision interface extends beyond the in vitro-characterized 40S and includes the L1 stalk and eEF2, possibly contributing to translocation inhibition. The accumulation of unresolved tRNA-bound 80S and 60S and aberrant 40S configurations identifies potentially limiting steps in collision responses. Our work provides a global view of the translation machinery in response to persistent collisions and a framework for quantitative analysis of translation dynamics in situ.


Subject(s)
Protein Biosynthesis , Ribosomes , Animals , Ribosomes/genetics , Ribosomes/metabolism , Polyribosomes/genetics , Polyribosomes/metabolism , Mammals
4.
Life Sci Alliance ; 6(11)2023 11.
Article in English | MEDLINE | ID: mdl-37643866

ABSTRACT

In development of colorectal cancer, mutations in APC are often followed by mutations in oncogene KRAS The latter changes cellular metabolism and is associated with the Warburg phenomenon. Glucose-regulated protein 78 (Grp78) is an important regulator of the protein-folding machinery, involved in processing and localization of transmembrane proteins. We hypothesize that targeting Grp78 in Apc and Kras (AK)-mutant intestines interferes with the metabolic phenotype imposed by Kras mutations. In mice with intestinal epithelial mutations in Apc, Kras G12D and heterozygosity for Grp78 (AK-Grp78 HET ) adenoma number and size is decreased compared with AK-Grp78 WT mice. Organoids from AK-Grp78 WT mice exhibited a glycolysis metabolism which was completely rescued by Grp78 heterozygosity. Expression and correct localization of glucose transporter GLUT1 was diminished in AK-Grp78 HET cells. GLUT1 inhibition restrained the increased growth observed in AK-mutant organoids, whereas AK-Grp78 HET organoids were unaffected. We identify Grp78 as a critical factor in Kras-mutated adenomagenesis. This can be attributed to a critical role for Grp78 in GLUT1 expression and localization, targeting glycolysis and the Warburg effect.


Subject(s)
Endoplasmic Reticulum Chaperone BiP , Animals , Mice , Cell Proliferation , Glucose , Glucose Transporter Type 1/genetics , Glycolysis/genetics , Intestines , Proto-Oncogene Proteins p21(ras)/genetics
5.
bioRxiv ; 2023 Mar 24.
Article in English | MEDLINE | ID: mdl-36993420

ABSTRACT

Aberrantly slow mRNA translation leads to ribosome stalling and subsequent collision with the trailing neighbor. Ribosome collisions have recently been shown to act as stress sensors in the cell, with the ability to trigger stress responses balancing survival and apoptotic cell-fate decisions depending on the stress level. However, we lack a molecular understanding of the reorganization of translation processes over time in mammalian cells exposed to an unresolved collision stress. Here we visualize the effect of a persistent collision stress on translation using in situ cryo electron tomography. We observe that low dose anisomycin collision stress leads to the stabilization of Z-site bound tRNA on elongating 80S ribosomes, as well as to the accumulation of an off-pathway 80S complex possibly resulting from collision splitting events. We visualize collided disomes in situ, occurring on compressed polysomes and revealing a stabilized geometry involving the Z-tRNA and L1 stalk on the stalled ribosome, and eEF2 bound to its collided rotated-2 neighbor. In addition, non-functional post-splitting 60S complexes accumulate in the stressed cells, indicating a limiting Ribosome associated Quality Control clearing rate. Finally, we observe the apparition of tRNA-bound aberrant 40S complexes shifting with the stress timepoint, suggesting a succession of different initiation inhibition mechanisms over time. Altogether, our work visualizes the changes of translation complexes under persistent collision stress in mammalian cells, indicating how perturbations in initiation, elongation and quality control processes contribute to an overall reduced protein synthesis.

6.
Mol Cell ; 83(3): 469-480, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36521491

ABSTRACT

mRNA translation is a highly conserved and tightly controlled mechanism for protein synthesis and is well known to be altered by oncogenes to promote cancer development. This distorted mRNA translation is accompanied by the vulnerability of cancer to inhibitors of key mRNA translation components. Novel studies also suggest that these alternations could be utilized for immunotherapy. Ribosome heterogeneity and alternative responses to nutrient shortages, which aid cancer growth and spread, are proposed to elicit aberrant protein production but may also result in previously unidentified therapeutic targets, such as the presentation of cancer-specific peptides at the surface of cancer cells (neoepitopes). This review will assess the driving forces in tRNA and ribosome function that underlie proteome diversification due to alterations in mRNA translation in cancer cells.


Subject(s)
Neoplasms , Proteome , Proteome/genetics , Proteome/metabolism , Protein Biosynthesis , Ribosomes/genetics , Ribosomes/metabolism , Peptides/genetics , RNA, Transfer/genetics , RNA, Transfer/metabolism , Neoplasms/genetics , Neoplasms/metabolism
7.
Nat Commun ; 13(1): 7923, 2022 12 23.
Article in English | MEDLINE | ID: mdl-36564381

ABSTRACT

Human melanocytic nevi (moles) result from a brief period of clonal expansion of melanocytes. As a cellular defensive mechanism against oncogene-induced hyperplasia, nevus-resident melanocytes enter a senescent state of stable cell cycle arrest. Senescent melanocytes can persist for months in mice and years in humans with a risk to escape the senescent state and progress to melanoma. The mechanisms providing prolonged survival of senescent melanocytes remain poorly understood. Here, we show that senescent melanocytes in culture and in nevi express high level of the anti-apoptotic BCL-2 family member BCL-W but remain insensitive to the pan-BCL-2 inhibitor ABT-263. We demonstrate that resistance to ABT-263 is driven by mTOR-mediated enhanced translation of another anti-apoptotic member, MCL-1. Strikingly, the combination of ABT-263 and MCL-1 inhibitors results in synthetic lethality to senescent melanocytes, and its topical application sufficient to eliminate nevi in male mice. These data highlight the important role of redundant anti-apoptotic mechanisms for the survival advantage of senescent melanocytes, and the proof-of-concept for a non-invasive combination therapy for nevi removal.


Subject(s)
Nevus, Pigmented , Nevus , Skin Neoplasms , Male , Humans , Animals , Mice , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Melanocytes/metabolism , Nevus/metabolism , Skin Neoplasms/metabolism
8.
Mol Cell ; 82(20): 3840-3855.e8, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36270248

ABSTRACT

The use of alternative promoters, splicing, and cleavage and polyadenylation (APA) generates mRNA isoforms that expand the diversity and complexity of the transcriptome. Here, we uncovered thousands of previously undescribed 5' uncapped and polyadenylated transcripts (5' UPTs). We show that these transcripts resist exonucleases due to a highly structured RNA and N6-methyladenosine modification at their 5' termini. 5' UPTs appear downstream of APA sites within their host genes and are induced upon APA activation. Strong enrichment in polysomal RNA fractions indicates 5' UPT translational potential. Indeed, APA promotes downstream translation initiation, non-canonical protein output, and consistent changes to peptide presentation at the cell surface. Lastly, we demonstrate the biological importance of 5' UPTs using Bcl2, a prominent anti-apoptotic gene whose entire coding sequence is a 5' UPT generated from 5' UTR-embedded APA sites. Thus, APA is not only accountable for terminating transcripts, but also for generating downstream uncapped RNAs with translation potential and biological impact.


Subject(s)
Polyadenylation , RNA Isoforms , RNA Isoforms/genetics , 5' Untranslated Regions , 3' Untranslated Regions/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Exonucleases/genetics
9.
J Cell Sci ; 135(19)2022 10 01.
Article in English | MEDLINE | ID: mdl-36052643

ABSTRACT

Cell size varies between cell types but is tightly regulated by cell intrinsic and extrinsic mechanisms. Cell size control is important for cell function, and changes in cell size are frequently observed in cancer. Here, we uncover a role for SETD2 in regulating cell size. SETD2 is a lysine methyltransferase and a tumor suppressor protein involved in transcription, RNA processing and DNA repair. At the molecular level, SETD2 is best known for associating with RNA polymerase II through its Set2-Rbp1 interacting (SRI) domain and methylating histone H3 on lysine 36 (H3K36) during transcription. Using multiple independent perturbation strategies, we identify SETD2 as a negative regulator of global protein synthesis rates and cell size. We provide evidence that overexpression of the H3K36 demethylase KDM4A or the oncohistone H3.3K36M also increase cell size. In addition, ectopic overexpression of a decoy SRI domain increased cell size, suggesting that the relevant substrate is engaged by SETD2 via its SRI domain. These data add a central role of SETD2 in regulating cellular physiology and warrant further studies on separating the different functions of SETD2 in cancer development.


Subject(s)
Histones , Neoplasms , Cell Size , Histone Methyltransferases/metabolism , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine , Neoplasms/metabolism , RNA Polymerase II/metabolism , Tumor Suppressor Proteins/metabolism
10.
Nat Commun ; 13(1): 4492, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35918345

ABSTRACT

The small intestine is a rapidly proliferating organ that is maintained by a small population of Lgr5-expressing intestinal stem cells (ISCs). However, several Lgr5-negative ISC populations have been identified, and this remarkable plasticity allows the intestine to rapidly respond to both the local environment and to damage. However, the mediators of such plasticity are still largely unknown. Using intestinal organoids and mouse models, we show that upon ribosome impairment (driven by Rptor deletion, amino acid starvation, or low dose cyclohexamide treatment) ISCs gain an Lgr5-negative, fetal-like identity. This is accompanied by a rewiring of metabolism. Our findings suggest that the ribosome can act as a sensor of nutrient availability, allowing ISCs to respond to the local nutrient environment. Mechanistically, we show that this phenotype requires the activation of ZAKɑ, which in turn activates YAP, via SRC. Together, our data reveals a central role for ribosome dynamics in intestinal stem cells, and identify the activation of ZAKɑ as a critical mediator of stem cell identity.


Subject(s)
Intestinal Mucosa , Stem Cells , Animals , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Intestines , Mice , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Ribosomes/metabolism , Stem Cells/metabolism
11.
Nucleic Acids Res ; 50(16): e95, 2022 09 09.
Article in English | MEDLINE | ID: mdl-35687114

ABSTRACT

Recent studies have revealed multiple mechanisms that can lead to heterogeneity in ribosomal composition. This heterogeneity can lead to preferential translation of specific panels of mRNAs, and is defined in large part by the ribosomal protein (RP) content, amongst other things. However, it is currently unknown to what extent ribosomal composition is heterogeneous across tissues, which is compounded by a lack of tools available to study it. Here we present dripARF, a method for detecting differential RP incorporation into the ribosome using Ribosome Profiling (Ribo-seq) data. We combine the 'waste' rRNA fragment data generated in Ribo-seq with the known 3D structure of the human ribosome to predict differences in the composition of ribosomes in the material being studied. We have validated this approach using publicly available data, and have revealed a potential role for eS25/RPS25 in development. Our results indicate that ribosome heterogeneity can be detected in Ribo-seq data, providing a new method to study this phenomenon. Furthermore, with dripARF, previously published Ribo-seq data provides a wealth of new information, allowing the identification of RPs of interest in many disease and normal contexts. dripARF is available as part of the ARF R package and can be accessed through https://github.com/fallerlab/ARF.


Subject(s)
Ribosomes/chemistry , Humans , RNA, Messenger , RNA, Ribosomal/analysis , Ribosomal Proteins/analysis , Ribosomes/genetics
12.
Elife ; 102021 12 13.
Article in English | MEDLINE | ID: mdl-34895463

ABSTRACT

Increased protein synthesis supports the rapid cell proliferation associated with cancer. The Rpl24Bst mutant mouse reduces the expression of the ribosomal protein RPL24 and has been used to suppress translation and limit tumorigenesis in multiple mouse models of cancer. Here, we show that Rpl24Bst also suppresses tumorigenesis and proliferation in a model of colorectal cancer (CRC) with two common patient mutations, Apc and Kras. In contrast to previous reports, Rpl24Bst mutation has no effect on ribosomal subunit abundance but suppresses translation elongation through phosphorylation of eEF2, reducing protein synthesis by 40% in tumour cells. Ablating eEF2 phosphorylation in Rpl24Bst mutant mice by inactivating its kinase, eEF2K, completely restores the rates of elongation and protein synthesis. Furthermore, eEF2K activity is required for the Rpl24Bst mutant to suppress tumorigenesis. This work demonstrates that elevation of eEF2 phosphorylation is an effective means to suppress colorectal tumorigenesis with two driver mutations. This positions translation elongation as a therapeutic target in CRC, as well as in other cancers where the Rpl24Bst mutation has a tumour suppressive effect in mouse models.


Subject(s)
Colorectal Neoplasms/genetics , Elongation Factor 2 Kinase/metabolism , Mutation , Ribosomal Proteins/genetics , Signal Transduction , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Transgenic , Phosphorylation , Ribosomal Proteins/metabolism
13.
Mol Cell ; 81(22): 4709-4721.e9, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34562372

ABSTRACT

mRNA translation is a highly conserved and tightly controlled mechanism for protein synthesis. Despite protein quality control mechanisms, amino acid shortage in melanoma induces aberrant proteins by ribosomal frameshifting. The extent and the underlying mechanisms related to this phenomenon are yet unknown. Here, we show that tryptophan depletion-induced ribosomal frameshifting is a widespread phenomenon in cancer. We termed this event sloppiness and strikingly observed its association with MAPK pathway hyperactivation. Sloppiness is stimulated by RAS activation in primary cells, suppressed by pharmacological inhibition of the oncogenic MAPK pathway in sloppy cells, and restored in cells with acquired resistance to MAPK pathway inhibition. Interestingly, sloppiness causes aberrant peptide presentation at the cell surface, allowing recognition and specific killing of drug-resistant cancer cells by T lymphocytes. Thus, while oncogenes empower cancer progression and aggressiveness, they also expose a vulnerability by provoking the production of aberrant peptides through sloppiness.


Subject(s)
Neoplasms/genetics , Oncogenes , Protein Biosynthesis , RNA, Messenger/metabolism , T-Lymphocytes/cytology , Animals , Carcinogenesis , Cell Membrane/metabolism , Disease Progression , Drug Resistance, Neoplasm , Frameshift Mutation , Frameshifting, Ribosomal , Humans , Immunotherapy/methods , MAP Kinase Signaling System , Melanoma/metabolism , Mice , Neoplasms/metabolism , Peptides/chemistry , Protein Kinase Inhibitors , Ribosomes/metabolism , T-Lymphocytes/metabolism , Tryptophan/chemistry , Tryptophan/metabolism
14.
Nat Commun ; 12(1): 4360, 2021 07 16.
Article in English | MEDLINE | ID: mdl-34272384

ABSTRACT

The glucocorticoid receptor (GR) regulates gene expression, governing aspects of homeostasis, but is also involved in cancer. Pharmacological GR activation is frequently used to alleviate therapy-related side-effects. While prior studies have shown GR activation might also have anti-proliferative action on tumours, the underpinnings of glucocorticoid action and its direct effectors in non-lymphoid solid cancers remain elusive. Here, we study the mechanisms of glucocorticoid response, focusing on lung cancer. We show that GR activation induces reversible cancer cell dormancy characterised by anticancer drug tolerance, and activation of growth factor survival signalling accompanied by vulnerability to inhibitors. GR-induced dormancy is dependent on a single GR-target gene, CDKN1C, regulated through chromatin looping of a GR-occupied upstream distal enhancer in a SWI/SNF-dependent fashion. These insights illustrate the importance of GR signalling in non-lymphoid solid cancer biology, particularly in lung cancer, and warrant caution for use of glucocorticoids in treatment of anticancer therapy related side-effects.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Chromatin/metabolism , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Glucocorticoids/pharmacology , Lung Neoplasms/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/drug effects , Chromatin/genetics , Chromatin Immunoprecipitation Sequencing , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cyclin-Dependent Kinase Inhibitor p57/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Imidazoles/pharmacology , Immunohistochemistry , Lung Neoplasms/genetics , Mice , Proteomics , Pyrazines/pharmacology , RNA, Small Interfering , RNA-Seq , Receptor, IGF Type 1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
15.
Nat Commun ; 12(1): 3464, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34103493

ABSTRACT

Right-sided (proximal) colorectal cancer (CRC) has a poor prognosis and a distinct mutational profile, characterized by oncogenic BRAF mutations and aberrations in mismatch repair and TGFß signalling. Here, we describe a mouse model of right-sided colon cancer driven by oncogenic BRAF and loss of epithelial TGFß-receptor signalling. The proximal colonic tumours that develop in this model exhibit a foetal-like progenitor phenotype (Ly6a/Sca1+) and, importantly, lack expression of Lgr5 and its associated intestinal stem cell signature. These features are recapitulated in human BRAF-mutant, right-sided CRCs and represent fundamental differences between left- and right-sided disease. Microbial-driven inflammation supports the initiation and progression of these tumours with foetal-like characteristics, consistent with their predilection for the microbe-rich right colon and their antibiotic sensitivity. While MAPK-pathway activating mutations drive this foetal-like signature via ERK-dependent activation of the transcriptional coactivator YAP, the same foetal-like transcriptional programs are also initiated by inflammation in a MAPK-independent manner. Importantly, in both contexts, epithelial TGFß-receptor signalling is instrumental in suppressing the tumorigenic potential of these foetal-like progenitor cells.


Subject(s)
Carcinogenesis/metabolism , Colonic Neoplasms/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinogenesis/pathology , Cell Differentiation , Cell Survival , Colon/pathology , Colonic Neoplasms/genetics , Epithelial Cells/metabolism , Fetus/pathology , Inflammation/pathology , Kaplan-Meier Estimate , MAP Kinase Signaling System , Mice, Inbred C57BL , Mutation , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , YAP-Signaling Proteins
16.
Cell Mol Gastroenterol Hepatol ; 12(3): 943-981, 2021.
Article in English | MEDLINE | ID: mdl-34102314

ABSTRACT

BACKGROUND & AIMS: The use of antibiotics (ABs) is a common practice during the first months of life. ABs can perturb the intestinal microbiota, indirectly influencing the intestinal epithelial cells (IECs), but can also directly affect IECs independent of the microbiota. Previous studies have focused mostly on the impact of AB treatment during adulthood. However, the difference between the adult and neonatal intestine warrants careful investigation of AB effects in early life. METHODS: Neonatal mice were treated with a combination of amoxicillin, vancomycin, and metronidazole from postnatal day 10 to 20. Intestinal permeability and whole-intestine gene and protein expression were analyzed. IECs were sorted by a fluorescence-activated cell sorter and their genome-wide gene expression was analyzed. Mouse fetal intestinal organoids were treated with the same AB combination and their gene and protein expression and metabolic capacity were determined. RESULTS: We found that in vivo treatment of neonatal mice led to decreased intestinal permeability and a reduced number of specialized vacuolated cells, characteristic of the neonatal period and necessary for absorption of milk macromolecules. In addition, the expression of genes typically present in the neonatal intestinal epithelium was lower, whereas the adult gene expression signature was higher. Moreover, we found altered epithelial defense and transepithelial-sensing capacity. In vitro treatment of intestinal fetal organoids with AB showed that part of the consequences observed in vivo is a result of the direct action of the ABs on IECs. Lastly, ABs reduced the metabolic capacity of intestinal fetal organoids. CONCLUSIONS: Our results show that early life AB treatment induces direct and indirect effects on IECs, influencing their maturation and functioning.


Subject(s)
Amoxicillin/administration & dosage , Anti-Bacterial Agents/administration & dosage , Gene Regulatory Networks/drug effects , Intestines/metabolism , Metronidazole/administration & dosage , Vancomycin/administration & dosage , Amoxicillin/adverse effects , Animals , Animals, Newborn , Anti-Bacterial Agents/adverse effects , Disease Models, Animal , Enterocytes/cytology , Enterocytes/drug effects , Enterocytes/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Intestines/cytology , Intestines/drug effects , Metronidazole/adverse effects , Mice , Oligonucleotide Array Sequence Analysis , Permeability/drug effects , Postnatal Care , Vacuoles/drug effects , Vacuoles/metabolism , Vancomycin/adverse effects
17.
Bioinformatics ; 37(17): 2659-2667, 2021 Sep 09.
Article in English | MEDLINE | ID: mdl-33720291

ABSTRACT

MOTIVATION: Ribosome Profiling (Ribo-seq) has revolutionized the study of RNA translation by providing information on ribosome positions across all translated RNAs with nucleotide-resolution. Yet several technical limitations restrict the sequencing depth of such experiments, the most common of which is the overabundance of rRNA fragments. Various strategies can be employed to tackle this issue, including the use of commercial rRNA depletion kits. However, as they are designed for more standardized RNAseq experiments, they may perform suboptimally in Ribo-seq. In order to overcome this, it is possible to use custom biotinylated oligos complementary to the most abundant rRNA fragments, however currently no computational framework exists to aid the design of optimal oligos. RESULTS: Here, we first show that a major confounding issue is that the rRNA fragments generated via Ribo-seq vary significantly with differing experimental conditions, suggesting that a 'one-size-fits-all' approach may be inefficient. Therefore we developed Ribo-ODDR, an oligo design pipeline integrated with a user-friendly interface that assists in oligo selection for efficient experiment-specific rRNA depletion. Ribo-ODDR uses preliminary data to identify the most abundant rRNA fragments, and calculates the rRNA depletion efficiency of potential oligos. We experimentally show that Ribo-ODDR designed oligos outperform commercially available kits and lead to a significant increase in rRNA depletion in Ribo-seq. AVAILABILITY AND IMPLEMENTATION: Ribo-ODDR is freely accessible at https://github.com/fallerlab/Ribo-ODDR. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

18.
Cancer Discov ; 11(5): 1228-1247, 2021 05.
Article in English | MEDLINE | ID: mdl-33328217

ABSTRACT

KRAS-mutant colorectal cancers are resistant to therapeutics, presenting a significant problem for ∼40% of cases. Rapalogs, which inhibit mTORC1 and thus protein synthesis, are significantly less potent in KRAS-mutant colorectal cancer. Using Kras-mutant mouse models and mouse- and patient-derived organoids, we demonstrate that KRAS with G12D mutation fundamentally rewires translation to increase both bulk and mRNA-specific translation initiation. This occurs via the MNK/eIF4E pathway culminating in sustained expression of c-MYC. By genetic and small-molecule targeting of this pathway, we acutely sensitize KRASG12D models to rapamycin via suppression of c-MYC. We show that 45% of colorectal cancers have high signaling through mTORC1 and the MNKs, with this signature correlating with a 3.5-year shorter cancer-specific survival in a subset of patients. This work provides a c-MYC-dependent cotargeting strategy with remarkable potency in multiple Kras-mutant mouse models and metastatic human organoids and identifies a patient population that may benefit from its clinical application. SIGNIFICANCE: KRAS mutation and elevated c-MYC are widespread in many tumors but remain predominantly untargetable. We find that mutant KRAS modulates translation, culminating in increased expression of c-MYC. We describe an effective strategy targeting mTORC1 and MNK in KRAS-mutant mouse and human models, pathways that are also commonly co-upregulated in colorectal cancer.This article is highlighted in the In This Issue feature, p. 995.


Subject(s)
Colorectal Neoplasms/genetics , Eukaryotic Initiation Factor-4E/drug effects , Intracellular Signaling Peptides and Proteins/drug effects , MTOR Inhibitors/pharmacology , Protein Serine-Threonine Kinases/drug effects , Animals , Colorectal Neoplasms/metabolism , Disease Models, Animal , Eukaryotic Initiation Factor-4E/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Serine-Threonine Kinases/metabolism
19.
FEBS J ; 288(23): 6635-6651, 2021 12.
Article in English | MEDLINE | ID: mdl-33382175

ABSTRACT

Protein synthesis is one of the most essential processes in every kingdom of life, and its dysregulation is a known driving force in cancer development. Multiple signaling pathways converge on the translation initiation machinery, and this plays a crucial role in regulating differential gene expression. In colorectal cancer, dysregulation of initiation results in translational reprogramming, which promotes the selective translation of mRNAs required for many oncogenic processes. The majority of upstream mutations found in colorectal cancer, including alterations in the WNT, MAPK, and PI3K\AKT pathways, have been demonstrated to play a significant role in translational reprogramming. Many translation initiation factors are also known to be dysregulated, resulting in translational reprogramming during tumor initiation and/or maintenance. In this review, we outline the role of translational reprogramming that occurs during colorectal cancer development and progression and highlight some of the most critical factors affecting the etiology of this disease.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Peptide Chain Initiation, Translational , RNA, Messenger/genetics , Ribosomes/genetics , Animals , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Models, Genetic , Peptide Initiation Factors/genetics , Peptide Initiation Factors/metabolism , Ribosomes/metabolism , Signal Transduction/genetics
20.
Oncogene ; 38(28): 5670-5685, 2019 07.
Article in English | MEDLINE | ID: mdl-31043706

ABSTRACT

The hypoxia-inducible transcription factor HIF-1 is appreciated as a promising target for cancer therapy. However, conditional deletion of HIF-1 and HIF-1 target genes in cells of the tumor microenvironment can result in accelerated tumor growth, calling for a detailed characterization of the cellular context to fully comprehend HIF-1's role in tumorigenesis. We dissected cell type-specific functions of HIF-1 for intestinal tumorigenesis by lineage-restricted deletion of the Hif1a locus. Intestinal epithelial cell-specific Hif1a loss reduced activation of Wnt/ß-catenin, tumor-specific metabolism and inflammation, significantly inhibiting tumor growth. Deletion of Hif1a in myeloid cells reduced the expression of fibroblast-activating factors in tumor-associated macrophages resulting in decreased abundance of tumor-associated fibroblasts (TAF) and robustly reduced tumor formation. Interestingly, hypoxia was detectable only sparsely and without spatial association with HIF-1α, arguing for an importance of hypoxia-independent, i.e., non-canonical, HIF-1 stabilization for intestinal tumorigenesis that has not been previously appreciated. This adds a further layer of complexity to the regulation of HIF-1 and suggests that hypoxia and HIF-1α stabilization can be uncoupled in cancer. Collectively, our data show that HIF-1 is a pivotal pro-tumorigenic factor for intestinal tumor formation, controlling key oncogenic programs in both the epithelial tumor compartment and the tumor microenvironment.


Subject(s)
Colorectal Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Oncogenes , Protein Stability , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...