Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Stem Cell ; 30(11): 1400-1402, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37922877

ABSTRACT

Engineered hematopoietic stem cells can be shielded from targeted immunotherapy. Recently published in Nature, Casirati et al. utilized single-base editing of epitopes implicated in acute myeloid leukemia and healthy hematopoiesis to alter their antibody and chimeric antigen receptor (CAR) T recognition while preserving their ligand binding and enzymatic function.


Subject(s)
Leukemia, Myeloid, Acute , Receptors, Chimeric Antigen , Humans , T-Lymphocytes , Gene Editing , Receptors, Chimeric Antigen/metabolism , Immunotherapy , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/metabolism , Hematopoietic Stem Cells/pathology , Immunotherapy, Adoptive
2.
Haematologica ; 108(10): 2570-2581, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37439336

ABSTRACT

Children with Down syndrome (DS, trisomy 21) are at a significantly higher risk of developing acute leukemia compared to the overall population. Many studies investigating the link between trisomy 21 and leukemia initiation and progression have been conducted over the last two decades. Despite improved treatment regimens and significant progress in iden - tifying genes on chromosome 21 and the mechanisms by which they drive leukemogenesis, there is still much that is unknown. A focused group of scientists and clinicians with expertise in leukemia and DS met in October 2022 at the Jérôme Lejeune Foundation in Paris, France for the 1st International Symposium on Down Syndrome and Leukemia. This meeting was held to discuss the most recent advances in treatment regimens and the biology underlying the initiation, progression, and relapse of acute lymphoblastic leukemia and acute myeloid leukemia in children with DS. This review provides a summary of what is known in the field, challenges in the management of DS patients with leukemia, and key questions in the field.


Subject(s)
Down Syndrome , Leukemia, Myeloid, Acute , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Child , Humans , Down Syndrome/complications , Down Syndrome/genetics , Leukemia, Myeloid, Acute/epidemiology , Acute Disease , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , France
3.
Leukemia ; 36(11): 2690-2704, 2022 11.
Article in English | MEDLINE | ID: mdl-36131042

ABSTRACT

Many cancers are organized as cellular hierarchies sustained by cancer stem cells (CSC), whose eradication is crucial for achieving long-term remission. Difficulties to isolate and undertake in vitro and in vivo experimental studies of rare CSC under conditions that preserve their original properties currently constitute a bottleneck for identifying molecular mechanisms involving coding and non-coding genomic regions that govern stemness. We focussed on acute myeloid leukemia (AML) as a paradigm of the CSC model and developed a patient-derived system termed OCI-AML22 that recapitulates the cellular hierarchy driven by leukemia stem cells (LSC). Through classical flow sorting and functional analyses, we established that a single phenotypic population is highly enriched for LSC. The LSC fraction can be easily isolated and serially expanded in culture or in xenografts while faithfully recapitulating functional, transcriptional and epigenetic features of primary LSCs. A novel non-coding regulatory element was identified with a new computational approach using functionally validated primary AML LSC fractions and its role in LSC stemness validated through efficient CRISPR editing using methods optimized for OCI-AML22 LSC. Collectively, OCI-AML22 constitutes a valuable resource to uncover mechanisms governing CSC driven malignancies.


Subject(s)
Leukemia, Myeloid, Acute , Neoplastic Stem Cells , Humans , Neoplastic Stem Cells/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology
4.
Cell Rep ; 38(10): 110481, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35263585

ABSTRACT

Gene expression profiling and proteome analysis of normal and malignant hematopoietic stem cells (HSCs) point to shared core stemness properties. However, discordance between mRNA and protein signatures highlights an important role for post-transcriptional regulation by microRNAs (miRNAs) in governing this critical nexus. Here, we identify miR-130a as a regulator of HSC self-renewal and differentiation. Enforced expression of miR-130a impairs B lymphoid differentiation and expands long-term HSCs. Integration of protein mass spectrometry and chimeric AGO2 crosslinking and immunoprecipitation (CLIP) identifies TBL1XR1 as a primary miR-130a target, whose loss of function phenocopies miR-130a overexpression. Moreover, we report that miR-130a is highly expressed in t(8;21) acute myeloid leukemia (AML), where it is critical for maintaining the oncogenic molecular program mediated by the AML1-ETO complex. Our study establishes that identification of the comprehensive miRNA targetome within primary cells enables discovery of genes and molecular networks underpinning stemness properties of normal and leukemic cells.


Subject(s)
Leukemia, Myeloid, Acute , MicroRNAs , Cell Line, Tumor , Cell Self Renewal/genetics , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/pathology , MicroRNAs/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
5.
Cell Stem Cell ; 28(10): 1838-1850.e10, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34343492

ABSTRACT

It is critical to understand how human quiescent long-term hematopoietic stem cells (LT-HSCs) sense demand from daily and stress-mediated cues and then transition into bioenergetically active progeny to differentiate and meet these cellular needs. However, the demand-adapted regulatory circuits of these early steps of hematopoiesis are largely unknown. Here we show that lysosomes, sophisticated nutrient-sensing and signaling centers, are regulated dichotomously by transcription factor EB (TFEB) and MYC to balance catabolic and anabolic processes required for activating LT-HSCs and guiding their lineage fate. TFEB-mediated induction of the endolysosomal pathway causes membrane receptor degradation, limiting LT-HSC metabolic and mitogenic activation, promoting quiescence and self-renewal, and governing erythroid-myeloid commitment. In contrast, MYC engages biosynthetic processes while repressing lysosomal catabolism, driving LT-HSC activation. Our study identifies TFEB-mediated control of lysosomal activity as a central regulatory hub for proper and coordinated stem cell fate determination.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Hematopoiesis , Hematopoietic Stem Cells , Cell Differentiation , Hematopoietic Stem Cells/cytology , Humans , Lysosomes , Signal Transduction
6.
Science ; 373(6551)2021 07 09.
Article in English | MEDLINE | ID: mdl-34244384

ABSTRACT

Children with Down syndrome have a 150-fold increased risk of developing myeloid leukemia, but the mechanism of predisposition is unclear. Because Down syndrome leukemogenesis initiates during fetal development, we characterized the cellular and developmental context of preleukemic initiation and leukemic progression using gene editing in human disomic and trisomic fetal hematopoietic cells and xenotransplantation. GATA binding protein 1 (GATA1) mutations caused transient preleukemia when introduced into trisomy 21 long-term hematopoietic stem cells, where a subset of chromosome 21 microRNAs affected predisposition to preleukemia. By contrast, progression to leukemia was independent of trisomy 21 and originated in various stem and progenitor cells through additional mutations in cohesin genes. CD117+/KIT proto-oncogene (KIT) cells mediated the propagation of preleukemia and leukemia, and KIT inhibition targeted preleukemic stem cells.


Subject(s)
Cell Cycle Proteins/genetics , Down Syndrome/genetics , GATA1 Transcription Factor/genetics , Hematopoietic Stem Cells/physiology , Leukemia, Myeloid/genetics , Preleukemia/genetics , Animals , Antigens, CD34/analysis , Cell Cycle Proteins/metabolism , Cell Lineage , Cell Proliferation , Cell Transformation, Neoplastic , Chromosomal Proteins, Non-Histone/genetics , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 21/metabolism , Disease Models, Animal , Disease Progression , Down Syndrome/complications , Female , GATA1 Transcription Factor/metabolism , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Heterografts , Humans , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Liver/embryology , Male , Megakaryocytes/physiology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Mutation , Preleukemia/metabolism , Preleukemia/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-kit/analysis , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Cohesins
7.
Blood Cancer Discov ; 2(1): 32-53, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33458693

ABSTRACT

Acute myeloid leukemia (AML) is a caricature of normal hematopoiesis, driven from leukemia stem cells (LSC) that share some hematopoietic stem cell (HSC) programs including responsiveness to inflammatory signaling. Although inflammation dysregulates mature myeloid cells and influences stemness programs and lineage determination in HSC by activating stress myelopoiesis, such roles in LSC are poorly understood. Here, we show that S1PR3, a receptor for the bioactive lipid sphingosine-1-phosphate, is a central regulator which drives myeloid differentiation and activates inflammatory programs in both HSC and LSC. S1PR3-mediated inflammatory signatures varied in a continuum from primitive to mature myeloid states across AML patient cohorts, each with distinct phenotypic and clinical properties. S1PR3 was high in LSC and blasts of mature myeloid samples with linkages to chemosensitivity, while S1PR3 activation in primitive samples promoted LSC differentiation leading to eradication. Our studies open new avenues for therapeutic target identification specific for each AML subset.


Subject(s)
Leukemia, Myeloid, Acute , Neoplastic Stem Cells , Sphingosine-1-Phosphate Receptors , Cell Differentiation , Hematopoietic Stem Cells , Humans , Leukemia, Myeloid, Acute/drug therapy , Sphingosine-1-Phosphate Receptors/metabolism
8.
Epigenetics Chromatin ; 13(1): 39, 2020 10 02.
Article in English | MEDLINE | ID: mdl-33008446

ABSTRACT

BACKGROUND: Partially methylated domains (PMDs) are a hallmark of epigenomes in reproducible and specific biological contexts, including cancer cells, the placenta, and cultured cell lines. Existing methods for deciding whether PMDs exist in a sample, as well as their identification, are few, often tailored to specific biological questions, and require high coverage samples for accurate identification. RESULTS: In this study, we outline a set of axioms that take a step towards a functional definition for PMDs, describe an improved method for comparable PMD detection across samples with substantially differing sequencing depths, and refine the decision criteria for whether a sample contains PMDs using a data-driven approach. Applying our method to 267 methylomes from 7 species, we corroborated recent results regarding the general association between replication timing and PMD state, and report identification of several reproducibly "escapee" genes within late-replicating domains that escape the reduced expression and hypomethylation of their immediate genomic neighborhood. We also explored the discordant PMD state of orthologous genes between human and mouse, and observed a directional association of PMD state with gene expression and local gene density. CONCLUSIONS: Our improved method makes low sequencing depth, population-level studies of PMD variation possible and our results further refine the model of PMD formation as one where sequence context and regional epigenomic features both play a role in gradual genome-wide hypomethylation.


Subject(s)
DNA Methylation , Epigenome , Animals , Cell Line, Tumor , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic , Humans , Lung/metabolism , Mammary Glands, Human/metabolism , Mice , Mice, Inbred C57BL , Organ Specificity , Placenta/metabolism , Pregnancy , Species Specificity
9.
Nat Commun ; 10(1): 4730, 2019 10 18.
Article in English | MEDLINE | ID: mdl-31628330

ABSTRACT

In the human hematopoietic system, rare self-renewing multipotent long-term hematopoietic stem cells (LT-HSCs) are responsible for the lifelong production of mature blood cells and are the rational target for clinical regenerative therapies. However, the heterogeneity in the hematopoietic stem cell compartment and variable outcomes of CRISPR/Cas9 editing make functional interrogation of rare LT-HSCs challenging. Here, we report high efficiency LT-HSC editing at single-cell resolution using electroporation of modified synthetic gRNAs and Cas9 protein. Targeted short isoform expression of the GATA1 transcription factor elicit distinct differentiation and proliferation effects in single highly purified LT-HSC when analyzed with functional in vitro differentiation and long-term repopulation xenotransplantation assays. Our method represents a blueprint for systematic genetic analysis of complex tissue hierarchies at single-cell resolution.


Subject(s)
CRISPR-Cas Systems , Cell Differentiation/genetics , Cell Proliferation/genetics , Gene Editing/methods , Hematopoietic Stem Cells/metabolism , Animals , Electroporation/methods , Female , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Hematopoietic Stem Cells/cytology , Humans , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Transplantation, Heterologous
10.
Cell Stem Cell ; 25(5): 639-653.e7, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31631013

ABSTRACT

Cellular stress responses serve as crucial decision points balancing persistence or culling of hematopoietic stem cells (HSCs) for lifelong blood production. Although strong stressors cull HSCs, the linkage between stress programs and self-renewal properties that underlie human HSC maintenance remains unknown, particularly at quiescence exit when HSCs must also dynamically shift metabolic state. Here, we demonstrate distinct wiring of the sphingolipidome across the human hematopoietic hierarchy and find that genetic or pharmacologic modulation of the sphingolipid enzyme DEGS1 regulates lineage differentiation. Inhibition of DEGS1 in hematopoietic stem and progenitor cells during the transition from quiescence to cellular activation with N-(4-hydroxyphenyl) retinamide activates coordinated stress pathways that coalesce on endoplasmic reticulum stress and autophagy programs to maintain immunophenotypic and functional HSCs. Thus, our work identifies a linkage between sphingolipid metabolism, proteostatic quality control systems, and HSC self-renewal and provides therapeutic targets for improving HSC-based cellular therapeutics.


Subject(s)
Cell Self Renewal/genetics , Fatty Acid Desaturases/antagonists & inhibitors , Fenretinide/pharmacology , Hematopoietic Stem Cells/metabolism , Proteostasis/genetics , Sphingolipids/metabolism , Animals , Autophagy/drug effects , Autophagy/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Self Renewal/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Fatty Acid Desaturases/genetics , Fatty Acid Desaturases/metabolism , Female , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Hematopoietic Stem Cells/enzymology , Humans , Male , Mass Spectrometry , Mice , Mice, Inbred NOD , Proteostasis/drug effects , RNA, Small Interfering , RNA-Seq , Single-Cell Analysis , Sphingolipids/chemistry , Transplantation, Heterologous
11.
Blood ; 133(20): 2198-2211, 2019 05 16.
Article in English | MEDLINE | ID: mdl-30796022

ABSTRACT

There is a growing body of evidence that the molecular properties of leukemia stem cells (LSCs) are associated with clinical outcomes in acute myeloid leukemia (AML), and LSCs have been linked to therapy failure and relapse. Thus, a better understanding of the molecular mechanisms that contribute to the persistence and regenerative potential of LSCs is expected to result in the development of more effective therapies. We therefore interrogated functionally validated data sets of LSC-specific genes together with their known protein interactors and selected 64 candidates for a competitive in vivo gain-of-function screen to identify genes that enhanced stemness in human cord blood hematopoietic stem and progenitor cells. A consistent effect observed for the top hits was the ability to restrain early repopulation kinetics while preserving regenerative potential. Overexpression (OE) of the most promising candidate, the orphan gene C3orf54/INKA1, in a patient-derived AML model (8227) promoted the retention of LSCs in a primitive state manifested by relative expansion of CD34+ cells, accumulation of cells in G0, and reduced output of differentiated progeny. Despite delayed early repopulation, at later times, INKA1-OE resulted in the expansion of self-renewing LSCs. In contrast, INKA1 silencing in primary AML reduced regenerative potential. Mechanistically, our multidimensional confocal analysis found that INKA1 regulates G0 exit by interfering with nuclear localization of its target PAK4, with concomitant reduction of global H4K16ac levels. These data identify INKA1 as a novel regulator of LSC latency and reveal a link between the regulation of stem cell kinetics and pool size during regeneration.


Subject(s)
Gene Expression Regulation, Leukemic , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Neoplastic Stem Cells/metabolism , Animals , Cell Cycle Checkpoints , Cell Line, Tumor , Female , Humans , Leukemia, Myeloid, Acute/pathology , Male , Mice, Inbred NOD , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/pathology , Up-Regulation , p21-Activated Kinases/analysis
12.
Cell Rep ; 25(5): 1109-1117.e5, 2018 10 30.
Article in English | MEDLINE | ID: mdl-30380403

ABSTRACT

Lifelong maintenance of the blood system requires equilibrium between clearance of damaged hematopoietic stem cells (HSCs) and long-term survival of the HSC pool. Severe perturbations of cellular homeostasis result in rapid HSC loss to maintain clonal purity. However, normal homeostatic processes can also generate lower-level stress; how HSCs survive these conditions remains unknown. Here we show that the integrated stress response (ISR) is uniquely active in HSCs and facilitates their persistence. Activating transcription factor 4 (ATF4) mediates the ISR and is highly expressed in HSCs due to scarcity of the eIF2 translation initiation complex. Amino acid deprivation results in eIF2α phosphorylation-dependent upregulation of ATF4, promoting HSC survival. Primitive acute myeloid leukemia (AML) cells also display eIF2 scarcity and ISR activity marks leukemia stem cells (LSCs) in primary AML samples. These findings identify a link between the ISR and stem cell survival in the normal and leukemic contexts.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells/metabolism , Leukemia/metabolism , Stress, Physiological , Activating Transcription Factor 4/metabolism , Animals , Cell Survival , Cytoprotection , Eukaryotic Initiation Factor-2/metabolism , Fetal Blood/cytology , Genes, Reporter , Humans , Male , Mice, Inbred NOD , Mice, SCID , Multipotent Stem Cells/metabolism , Phosphorylation , Up-Regulation , Valine/deficiency
13.
PLoS Biol ; 16(6): e2004049, 2018 06.
Article in English | MEDLINE | ID: mdl-29924804

ABSTRACT

The contribution of basal and luminal cells to cancer progression and metastasis is poorly understood. We report generation of reporter systems driven by either keratin-14 (K14) or keratin-8 (K8) promoter that not only express a fluorescent protein but also an inducible suicide gene. Transgenic mice express the reporter genes in the right cell compartments of mammary gland epithelia and respond to treatment with toxins. In addition, we engineered the reporters into 4T1 metastatic mouse tumor cell line and demonstrate that K14+ cells, but not K14- or K8+, are both highly invasive in three-dimensional (3D) culture and metastatic in vivo. Treatment of cells in culture, or tumors in mice, with reporter-targeting toxin inhibited both invasive behavior and metastasis in vivo. RNA sequencing (RNA-seq), secretome, and epigenome analysis of K14+ and K14- cells led to the identification of amphoterin-induced protein 2 (Amigo2) as a new cell invasion driver whose expression correlated with decreased relapse-free survival in patients with TP53 wild-type (WT) breast cancer.


Subject(s)
Genes, Reporter/genetics , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Animals , Cell Division/genetics , Cell Line, Tumor , Cell Movement/genetics , Epithelial Cells/pathology , Female , Green Fluorescent Proteins/genetics , Keratin-14/genetics , Keratin-8/genetics , Mammary Glands, Animal/cytology , Mammary Neoplasms, Animal/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasm Metastasis/pathology , Promoter Regions, Genetic/genetics
15.
Nature ; 554(7692): 378-381, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29414946

ABSTRACT

Using a functional model of breast cancer heterogeneity, we previously showed that clonal sub-populations proficient at generating circulating tumour cells were not all equally capable of forming metastases at secondary sites. A combination of differential expression and focused in vitro and in vivo RNA interference screens revealed candidate drivers of metastasis that discriminated metastatic clones. Among these, asparagine synthetase expression in a patient's primary tumour was most strongly correlated with later metastatic relapse. Here we show that asparagine bioavailability strongly influences metastatic potential. Limiting asparagine by knockdown of asparagine synthetase, treatment with l-asparaginase, or dietary asparagine restriction reduces metastasis without affecting growth of the primary tumour, whereas increased dietary asparagine or enforced asparagine synthetase expression promotes metastatic progression. Altering asparagine availability in vitro strongly influences invasive potential, which is correlated with an effect on proteins that promote the epithelial-to-mesenchymal transition. This provides at least one potential mechanism for how the bioavailability of a single amino acid could regulate metastatic progression.


Subject(s)
Asparagine/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Neoplasm Metastasis/pathology , Animals , Asparaginase/metabolism , Asparaginase/therapeutic use , Asparagine/deficiency , Aspartate-Ammonia Ligase/genetics , Aspartate-Ammonia Ligase/metabolism , Biological Availability , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Male , Mice , Neoplasm Invasiveness/pathology , Prognosis , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA Interference , Reproducibility of Results
16.
Nature ; 520(7547): 358-62, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25855289

ABSTRACT

Cancer metastasis requires that primary tumour cells evolve the capacity to intravasate into the lymphatic system or vasculature, and extravasate into and colonize secondary sites. Others have demonstrated that individual cells within complex populations show heterogeneity in their capacity to form secondary lesions. Here we develop a polyclonal mouse model of breast tumour heterogeneity, and show that distinct clones within a mixed population display specialization, for example, dominating the primary tumour, contributing to metastatic populations, or showing tropism for entering the lymphatic or vasculature systems. We correlate these stable properties to distinct gene expression profiles. Those clones that efficiently enter the vasculature express two secreted proteins, Serpine2 and Slpi, which were necessary and sufficient to program these cells for vascular mimicry. Our data indicate that these proteins not only drive the formation of extravascular networks but also ensure their perfusion by acting as anticoagulants. We propose that vascular mimicry drives the ability of some breast tumour cells to contribute to distant metastases while simultaneously satisfying a critical need of the primary tumour to be fed by the vasculature. Enforced expression of SERPINE2 and SLPI in human breast cancer cell lines also programmed them for vascular mimicry, and SERPINE2 and SLPI were overexpressed preferentially in human patients that had lung-metastatic relapse. Thus, these two secreted proteins, and the phenotype they promote, may be broadly relevant as drivers of metastatic progression in human cancer.


Subject(s)
Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Endothelium, Vascular/pathology , Neoplasm Metastasis/pathology , Animals , Anticoagulants/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Clone Cells/metabolism , Clone Cells/pathology , Disease Models, Animal , Disease Progression , Endothelium, Vascular/metabolism , Extracellular Matrix/metabolism , Female , Gene Expression Profiling , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Neoplasm Metastasis/genetics , Recurrence , Secretory Leukocyte Peptidase Inhibitor/metabolism , Sequence Analysis, DNA , Serpin E2/metabolism
17.
Mol Cell ; 56(6): 796-807, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25435137

ABSTRACT

The strength of conclusions drawn from RNAi-based studies is heavily influenced by the quality of tools used to elicit knockdown. Prior studies have developed algorithms to design siRNAs. However, to date, no established method has emerged to identify effective shRNAs, which have lower intracellular abundance than transfected siRNAs and undergo additional processing steps. We recently developed a multiplexed assay for identifying potent shRNAs and used this method to generate ∼250,000 shRNA efficacy data points. Using these data, we developed shERWOOD, an algorithm capable of predicting, for any shRNA, the likelihood that it will elicit potent target knockdown. Combined with additional shRNA design strategies, shERWOOD allows the ab initio identification of potent shRNAs that specifically target the majority of each gene's multiple transcripts. We validated the performance of our shRNA designs using several orthogonal strategies and constructed genome-wide collections of shRNAs for humans and mice based on our approach.


Subject(s)
RNA, Small Interfering/genetics , Software , Algorithms , Base Sequence , Cell Line, Tumor , Computer Simulation , Consensus Sequence , Gene Knockdown Techniques , Humans , MicroRNAs/genetics , Models, Genetic , Molecular Sequence Data
18.
Cell ; 157(2): 382-394, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24725405

ABSTRACT

Missense mutations in the p53 tumor suppressor inactivate its antiproliferative properties but can also promote metastasis through a gain-of-function activity. We show that sustained expression of mutant p53 is required to maintain the prometastatic phenotype of a murine model of pancreatic cancer, a highly metastatic disease that frequently displays p53 mutations. Transcriptional profiling and functional screening identified the platelet-derived growth factor receptor b (PDGFRb) as both necessary and sufficient to mediate these effects. Mutant p53 induced PDGFRb through a cell-autonomous mechanism involving inhibition of a p73/NF-Y complex that represses PDGFRb expression in p53-deficient, noninvasive cells. Blocking PDGFRb signaling by RNA interference or by small molecule inhibitors prevented pancreatic cancer cell invasion in vitro and metastasis formation in vivo. Finally, high PDGFRb expression correlates with poor disease-free survival in pancreatic, colon, and ovarian cancer patients, implicating PDGFRb as a prognostic marker and possible target for attenuating metastasis in p53 mutant tumors.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Neoplasm Metastasis , Pancreatic Neoplasms/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Carcinoma, Pancreatic Ductal/pathology , Disease Models, Animal , Gene Expression Profiling , Humans , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...