Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38562717

ABSTRACT

Driver gene mutations can increase the metastatic potential of the primary tumor1-3, but their role in sustaining tumor growth at metastatic sites is poorly understood. A paradigm of such mutations is inactivation of SMAD4 - a transcriptional effector of TGFß signaling - which is a hallmark of multiple gastrointestinal malignancies4,5. SMAD4 inactivation mediates TGFß's remarkable anti- to pro-tumorigenic switch during cancer progression and can thus influence both tumor initiation and metastasis6-14. To determine whether metastatic tumors remain dependent on SMAD4 inactivation, we developed a mouse model of pancreatic ductal adenocarcinoma (PDAC) that enables Smad4 depletion in the pre-malignant pancreas and subsequent Smad4 reactivation in established metastases. As expected, Smad4 inactivation facilitated the formation of primary tumors that eventually colonized the liver and lungs. By contrast, Smad4 reactivation in metastatic disease had strikingly opposite effects depending on the tumor's organ of residence: suppression of liver metastases and promotion of lung metastases. Integrative multiomic analysis revealed organ-specific differences in the tumor cells' epigenomic state, whereby the liver and lungs harbored chromatin programs respectively dominated by the KLF and RUNX developmental transcription factors, with Klf4 depletion being sufficient to reverse Smad4's tumor-suppressive activity in liver metastases. Our results show how epigenetic states favored by the organ of residence can influence the function of driver genes in metastatic tumors. This organ-specific gene-chromatin interplay invites consideration of anatomical site in the interpretation of tumor genetics, with implications for the therapeutic targeting of metastatic disease.

2.
Nat Cancer ; 5(2): 315-329, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38177458

ABSTRACT

Metastatic gastric carcinoma is a highly lethal cancer that responds poorly to conventional and molecularly targeted therapies. Despite its clinical relevance, the mechanisms underlying the behavior and therapeutic response of this disease are poorly understood owing, in part, to a paucity of tractable models. Here we developed methods to somatically introduce different oncogenic lesions directly into the murine gastric epithelium. Genotypic configurations observed in patients produced metastatic gastric cancers that recapitulated the histological, molecular and clinical features of all nonviral molecular subtypes of the human disease. Applying this platform to both wild-type and immunodeficient mice revealed previously unappreciated links between the genotype, organotropism and immune surveillance of metastatic cells, which produced distinct patterns of metastasis that were mirrored in patients. Our results establish a highly portable platform for generating autochthonous cancer models with flexible genotypes and host backgrounds, which can unravel mechanisms of gastric tumorigenesis or test new therapeutic concepts.


Subject(s)
Stomach Neoplasms , Humans , Mice , Animals , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Disease Models, Animal , Gastric Mucosa/pathology , Genotype
3.
bioRxiv ; 2023 May 20.
Article in English | MEDLINE | ID: mdl-37292765

ABSTRACT

Overexpression of repetitive elements is an emerging hallmark of human cancers 1 . Diverse repeats can mimic viruses by replicating within the cancer genome through retrotransposition, or presenting pathogen-associated molecular patterns (PAMPs) to the pattern recognition receptors (PRRs) of the innate immune system 2-5 . Yet, how specific repeats affect tumor evolution and shape the tumor immune microenvironment (TME) in a pro- or anti-tumorigenic manner remains poorly defined. Here, we integrate whole genome and total transcriptome data from a unique autopsy cohort of multiregional samples collected in pancreatic ductal adenocarcinoma (PDAC) patients, into a comprehensive evolutionary analysis. We find that more recently evolved S hort I nterspersed N uclear E lements (SINE), a family of retrotransposable repeats, are more likely to form immunostimulatory double-strand RNAs (dsRNAs). Consequently, younger SINEs are strongly co-regulated with RIG-I like receptor associated type-I interferon genes but anti-correlated with pro-tumorigenic macrophage infiltration. We discover that immunostimulatory SINE expression in tumors is regulated by either L ong I nterspersed N uclear E lements 1 (LINE1/L1) mobility or ADAR1 activity in a TP53 mutation dependent manner. Moreover, L1 retrotransposition activity tracks with tumor evolution and is associated with TP53 mutation status. Altogether, our results suggest pancreatic tumors actively evolve to modulate immunogenic SINE stress and induce pro-tumorigenic inflammation. Our integrative, evolutionary analysis therefore illustrates, for the first time, how dark matter genomic repeats enable tumors to co-evolve with the TME by actively regulating viral mimicry to their selective advantage.

4.
Cancer Res ; 83(10): 1596-1610, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36912618

ABSTRACT

Cancer-associated fibroblasts (CAF) are a major cell type in the stroma of solid tumors and can exert both tumor-promoting and tumor-restraining functions. CAF heterogeneity is frequently observed in pancreatic ductal adenocarcinoma (PDAC), a tumor characterized by a dense and hypoxic stroma that features myofibroblastic CAFs (myCAF) and inflammatory CAFs (iCAF) that are thought to have opposing roles in tumor progression. While CAF heterogeneity can be driven in part by tumor cell-produced cytokines, other determinants shaping CAF identity and function are largely unknown. In vivo, we found that iCAFs displayed a hypoxic gene expression and biochemical profile and were enriched in hypoxic regions of PDAC tumors, while myCAFs were excluded from these regions. Hypoxia led fibroblasts to acquire an inflammatory gene expression signature and synergized with cancer cell-derived cytokines to promote an iCAF phenotype in a HIF1α-dependent fashion. Furthermore, HIF1α stabilization was sufficient to induce an iCAF phenotype in stromal cells introduced into PDAC organoid cocultures and to promote PDAC tumor growth. These findings indicate hypoxia-induced HIF1α as a regulator of CAF heterogeneity and promoter of tumor progression in PDAC. SIGNIFICANCE: Hypoxia in the tumor microenvironment of pancreatic cancer potentiates the cytokine-induced inflammatory CAF phenotype and promotes tumor growth. See related commentary by Fuentes and Taniguchi, p. 1560.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Cytokines/metabolism , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Fibroblasts/metabolism , Cancer-Associated Fibroblasts/metabolism , Phenotype , Tumor Microenvironment , Pancreatic Neoplasms
5.
Nat Cancer ; 3(11): 1367-1385, 2022 11.
Article in English | MEDLINE | ID: mdl-36344707

ABSTRACT

The most prominent homozygous deletions in cancer affect chromosome 9p21.3 and eliminate CDKN2A/B tumor suppressors, disabling a cell-intrinsic barrier to tumorigenesis. Half of 9p21.3 deletions, however, also encompass a type I interferon (IFN) gene cluster; the consequences of this co-deletion remain unexplored. To functionally dissect 9p21.3 and other large genomic deletions, we developed a flexible deletion engineering strategy, MACHETE (molecular alteration of chromosomes with engineered tandem elements). Applying MACHETE to a syngeneic mouse model of pancreatic cancer, we found that co-deletion of the IFN cluster promoted immune evasion, metastasis and immunotherapy resistance. Mechanistically, IFN co-deletion disrupted type I IFN signaling in the tumor microenvironment, leading to marked changes in infiltrating immune cells and escape from CD8+ T-cell surveillance, effects largely driven by the poorly understood interferon epsilon. These results reveal a chromosomal deletion that disables both cell-intrinsic and cell-extrinsic tumor suppression and provide a framework for interrogating large deletions in cancer and beyond.


Subject(s)
Interferons , Neoplasms , Animals , Mice , Chromosome Deletion , Chromosomes , Immune Evasion , Tumor Microenvironment/genetics , Tandem Repeat Sequences
6.
Rev Sci Instrum ; 93(8): 083514, 2022 Aug 01.
Article in English | MEDLINE | ID: mdl-36050046

ABSTRACT

We have developed a non-collective Thomson scattering diagnostic for measurements of electron density and temperature on the Large Plasma Device. A triple grating spectrometer with a tunable notch filter is used to discriminate the faint scattering signal from the stray light. In this paper, we describe the diagnostic and its calibration via Raman scattering and present the first measurements performed with the fully commissioned system. Depending on the discharge conditions, the measured densities and temperatures range from 4.0 × 1012 to 2.8 × 1013 cm-3 and from 1.2 to 6.8 eV, respectively. The variation of the measurement error with plasma parameters and discharges averaged is also discussed.

7.
Nature ; 608(7924): 795-802, 2022 08.
Article in English | MEDLINE | ID: mdl-35978189

ABSTRACT

Although p53 inactivation promotes genomic instability1 and presents a route to malignancy for more than half of all human cancers2,3, the patterns through which heterogenous TP53 (encoding human p53) mutant genomes emerge and influence tumorigenesis remain poorly understood. Here, in a mouse model of pancreatic ductal adenocarcinoma that reports sporadic p53 loss of heterozygosity before cancer onset, we find that malignant properties enabled by p53 inactivation are acquired through a predictable pattern of genome evolution. Single-cell sequencing and in situ genotyping of cells from the point of p53 inactivation through progression to frank cancer reveal that this deterministic behaviour involves four sequential phases-Trp53 (encoding mouse p53) loss of heterozygosity, accumulation of deletions, genome doubling, and the emergence of gains and amplifications-each associated with specific histological stages across the premalignant and malignant spectrum. Despite rampant heterogeneity, the deletion events that follow p53 inactivation target functionally relevant pathways that can shape genomic evolution and remain fixed as homogenous events in diverse malignant populations. Thus, loss of p53-the 'guardian of the genome'-is not merely a gateway to genetic chaos but, rather, can enable deterministic patterns of genome evolution that may point to new strategies for the treatment of TP53-mutant tumours.


Subject(s)
Carcinogenesis , Disease Progression , Genes, p53 , Genome , Loss of Heterozygosity , Pancreatic Neoplasms , Tumor Suppressor Protein p53 , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Evolution, Molecular , Gene Deletion , Genes, p53/genetics , Genome/genetics , Mice , Models, Genetic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Tumor Suppressor Protein p53/genetics
8.
Nat Biotechnol ; 40(6): 862-873, 2022 06.
Article in English | MEDLINE | ID: mdl-35165384

ABSTRACT

Base editing can be applied to characterize single nucleotide variants of unknown function, yet defining effective combinations of single guide RNAs (sgRNAs) and base editors remains challenging. Here, we describe modular base-editing-activity 'sensors' that link sgRNAs and cognate target sites in cis and use them to systematically measure the editing efficiency and precision of thousands of sgRNAs paired with functionally distinct base editors. By quantifying sensor editing across >200,000 editor-sgRNA combinations, we provide a comprehensive resource of sgRNAs for introducing and interrogating cancer-associated single nucleotide variants in multiple model systems. We demonstrate that sensor-validated tools streamline production of in vivo cancer models and that integrating sensor modules in pooled sgRNA libraries can aid interpretation of high-throughput base editing screens. Using this approach, we identify several previously uncharacterized mutant TP53 alleles as drivers of cancer cell proliferation and in vivo tumor development. We anticipate that the framework described here will facilitate the functional interrogation of cancer variants in cell and animal models.


Subject(s)
Gene Editing , Neoplasms , Animals , CRISPR-Cas Systems/genetics , Neoplasms/genetics , Nucleotides , RNA, Guide, Kinetoplastida/genetics
9.
J Clin Invest ; 131(22)2021 11 15.
Article in English | MEDLINE | ID: mdl-34779407

ABSTRACT

High expression of LIN28B is associated with aggressive malignancy and poor survival. Here, probing MYCN-amplified neuroblastoma as a model system, we showed that LIN28B expression was associated with enhanced cell migration in vitro and invasive and metastatic behavior in murine xenografts. Sequence analysis of the polyribosome fraction of LIN28B-expressing neuroblastoma cells revealed let-7-independent enrichment of transcripts encoding components of the translational and ribosomal apparatus and depletion of transcripts of neuronal developmental programs. We further observed that LIN28B utilizes both its cold shock and zinc finger RNA binding domains to preferentially interact with MYCN-induced transcripts of the ribosomal complex, enhancing their translation. These data demonstrated that LIN28B couples the MYCN-driven transcriptional program to enhanced ribosomal translation, thereby implicating LIN28B as a posttranscriptional driver of the metastatic phenotype.


Subject(s)
N-Myc Proto-Oncogene Protein/physiology , Neoplasm Metastasis , Neuroblastoma/pathology , RNA-Binding Proteins/physiology , Ribosomes/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Neuroblastoma/etiology
10.
Rev Sci Instrum ; 92(9): 093102, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34598480

ABSTRACT

We present optical Thomson scattering measurements of electron density and temperature in a large-scale (∼2 cm) exploding laser plasma produced by irradiating a solid target with a high-energy (5-10 J) laser pulse at a high repetition rate (1 Hz). The Thomson scattering diagnostic matches this high repetition rate. Unlike previous work performed in single shots at much higher energies, the instrument allows for point measurements anywhere inside the plasma by automatically translating the scattering volume using motorized stages as the experiment is repeated at 1 Hz. Measured densities around 4 × 1016 cm-3 and temperatures around 7 eV result in a scattering parameter near unity, depending on the distance from the target. The measured spectra show the transition from collective scattering close to the target to non-collective scattering at larger distances. Densities obtained by fitting the weakly collective spectra agree to within 10% with an irradiance calibration performed via Raman scattering in nitrogen.

11.
Cell Rep ; 36(3): 109408, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34289374

ABSTRACT

The molecular mechanisms that govern the choreographed timing of organ development remain poorly understood. Our investigation of the role of the Lin28a and Lin28b paralogs during the developmental process of branching morphogenesis establishes that dysregulation of Lin28a/b leads to abnormal branching morphogenesis in the lung and other tissues. Additionally, we find that the Lin28 paralogs, which regulate post-transcriptional processing of both mRNAs and microRNAs (miRNAs), predominantly control mRNAs during the initial phases of lung organogenesis. Target mRNAs include Sox2, Sox9, and Etv5, which coordinate lung development and differentiation. Moreover, we find that functional interactions between Lin28a and Sox9 are capable of bypassing branching defects in Lin28a/b mutant lungs. Here, we identify Lin28a and Lin28b as regulators of early embryonic lung development, highlighting the importance of the timing of post-transcriptional regulation of both miRNAs and mRNAs at distinct stages of organogenesis.


Subject(s)
Lung/embryology , Lung/metabolism , Morphogenesis , RNA-Binding Proteins/metabolism , Sequence Homology, Amino Acid , Embryo, Mammalian/metabolism , Feedback, Physiological , Fibroblast Growth Factor 10/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Biological , Morphogenesis/genetics , RNA-Binding Proteins/genetics , SOX9 Transcription Factor/metabolism , Signal Transduction/genetics
12.
Nature ; 590(7847): 642-648, 2021 02.
Article in English | MEDLINE | ID: mdl-33536616

ABSTRACT

Tissue damage increases the risk of cancer through poorly understood mechanisms1. In mouse models of pancreatic cancer, pancreatitis associated with tissue injury collaborates with activating mutations in the Kras oncogene to markedly accelerate the formation of early neoplastic lesions and, ultimately, adenocarcinoma2,3. Here, by integrating genomics, single-cell chromatin assays and spatiotemporally controlled functional perturbations in autochthonous mouse models, we show that the combination of Kras mutation and tissue damage promotes a unique chromatin state in the pancreatic epithelium that distinguishes neoplastic transformation from normal regeneration and is selected for throughout malignant evolution. This cancer-associated epigenetic state emerges within 48 hours of pancreatic injury, and involves an 'acinar-to-neoplasia' chromatin switch that contributes to the early dysregulation of genes that define human pancreatic cancer. Among the factors that are most rapidly activated after tissue damage in the pre-malignant pancreatic epithelium is the alarmin cytokine interleukin 33, which recapitulates the effects of injury in cooperating with mutant Kras to unleash the epigenetic remodelling program of early neoplasia and neoplastic transformation. Collectively, our study demonstrates how gene-environment interactions can rapidly produce gene-regulatory programs that dictate early neoplastic commitment, and provides a molecular framework for understanding the interplay between genetic and environmental cues in the initiation of cancer.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic , Gene-Environment Interaction , Pancreas/metabolism , Pancreas/pathology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/pathology , Chromatin/genetics , Chromatin/metabolism , Chromatin/pathology , Disease Models, Animal , Female , Genomics , Humans , Interleukin-33/metabolism , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Cancer Discov ; 10(7): 1038-1057, 2020 07.
Article in English | MEDLINE | ID: mdl-32376773

ABSTRACT

To study genetic factors influencing the progression and therapeutic responses of advanced prostate cancer, we developed a fast and flexible system that introduces genetic alterations relevant to human disease directly into the prostate glands of mice using tissue electroporation. These electroporation-based genetically engineered mouse models (EPO-GEMM) recapitulate features of traditional germline models and, by modeling genetic factors linked to late-stage human disease, can produce tumors that are metastatic and castration-resistant. A subset of tumors with Trp53 alterations acquired spontaneous WNT pathway alterations, which are also associated with metastatic prostate cancer in humans. Using the EPO-GEMM approach and an orthogonal organoid-based model, we show that WNT pathway activation drives metastatic disease that is sensitive to pharmacologic WNT pathway inhibition. Thus, by leveraging EPO-GEMMs, we reveal a functional role for WNT signaling in driving prostate cancer metastasis and validate the WNT pathway as therapeutic target in metastatic prostate cancer. SIGNIFICANCE: Our understanding of the factors driving metastatic prostate cancer is limited by the paucity of models of late-stage disease. Here, we develop EPO-GEMMs of prostate cancer and use them to identify and validate the WNT pathway as an actionable driver of aggressive metastatic disease.This article is highlighted in the In This Issue feature, p. 890.


Subject(s)
Prostatic Neoplasms/genetics , Tissue Engineering/methods , Wnt Signaling Pathway/genetics , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Neoplasm Metastasis
14.
Cell Stem Cell ; 22(4): 575-588.e7, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29625070

ABSTRACT

While gene expression dynamics have been extensively cataloged during hematopoietic differentiation in the adult, less is known about transcriptome diversity of human hematopoietic stem cells (HSCs) during development. To characterize transcriptional and post-transcriptional changes in HSCs during development, we leveraged high-throughput genomic approaches to profile miRNAs, lincRNAs, and mRNAs. Our findings indicate that HSCs manifest distinct alternative splicing patterns in key hematopoietic regulators. Detailed analysis of the splicing dynamics and function of one such regulator, HMGA2, identified an alternative isoform that escapes miRNA-mediated targeting. We further identified the splicing kinase CLK3 that, by regulating HMGA2 splicing, preserves HMGA2 function in the setting of an increase in let-7 miRNA levels, delineating how CLK3 and HMGA2 form a functional axis that influences HSC properties during development. Collectively, our study highlights molecular mechanisms by which alternative splicing and miRNA-mediated post-transcriptional regulation impact the molecular identity and stage-specific developmental features of human HSCs.


Subject(s)
Alternative Splicing/genetics , HMGA2 Protein/genetics , Hematopoietic Stem Cells/metabolism , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , HMGA2 Protein/metabolism , Hematopoietic Stem Cells/cytology , Humans , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , RNA Processing, Post-Transcriptional/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Nat Cell Biol ; 19(1): 60-67, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27992407

ABSTRACT

Signalling and post-transcriptional gene control are both critical for the regulation of pluripotency, yet how they are integrated to influence cell identity remains poorly understood. LIN28 (also known as LIN28A), a highly conserved RNA-binding protein, has emerged as a central post-transcriptional regulator of cell fate through blockade of let-7 microRNA biogenesis and direct modulation of mRNA translation. Here we show that LIN28 is phosphorylated by MAPK/ERK in pluripotent stem cells, which increases its levels via post-translational stabilization. LIN28 phosphorylation had little impact on let-7 but enhanced the effect of LIN28 on its direct mRNA targets, revealing a mechanism that uncouples LIN28's let-7-dependent and -independent activities. We have linked this mechanism to the induction of pluripotency by somatic cell reprogramming and the transition from naive to primed pluripotency. Collectively, our findings indicate that MAPK/ERK directly impacts LIN28, defining an axis that connects signalling, post-transcriptional gene control, and cell fate regulation.


Subject(s)
MAP Kinase Signaling System , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA-Binding Proteins/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Blotting, Western , HeLa Cells , Humans , Immunoprecipitation , Mass Spectrometry , Mice , Mice, Transgenic , MicroRNAs/metabolism , Mouse Embryonic Stem Cells/metabolism , Phosphorylation , Protein Domains , Protein Stability
17.
Nature ; 535(7611): 246-51, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27383785

ABSTRACT

Poor prognosis in neuroblastoma is associated with genetic amplification of MYCN. MYCN is itself a target of let-7, a tumour suppressor family of microRNAs implicated in numerous cancers. LIN28B, an inhibitor of let-7 biogenesis, is overexpressed in neuroblastoma and has been reported to regulate MYCN. Here we show, however, that LIN28B is dispensable in MYCN-amplified neuroblastoma cell lines, despite de-repression of let-7. We further demonstrate that MYCN messenger RNA levels in amplified disease are exceptionally high and sufficient to sponge let-7, which reconciles the dispensability of LIN28B. We found that genetic loss of let-7 is common in neuroblastoma, inversely associated with MYCN amplification, and independently associated with poor outcomes, providing a rationale for chromosomal loss patterns in neuroblastoma. We propose that let-7 disruption by LIN28B, MYCN sponging, or genetic loss is a unifying mechanism of neuroblastoma development with broad implications for cancer pathogenesis.


Subject(s)
Gene Amplification/genetics , MicroRNAs/genetics , Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , RNA-Binding Proteins/genetics , 3' Untranslated Regions/genetics , Animals , Chromosome Deletion , Female , Gene Deletion , Genes, Neoplasm/genetics , Humans , Mice , MicroRNAs/metabolism , Models, Genetic , N-Myc Proto-Oncogene Protein , Neuroblastoma/pathology , Xenograft Model Antitumor Assays
18.
Cell ; 155(4): 778-92, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24209617

ABSTRACT

Regeneration capacity declines with age, but why juvenile organisms show enhanced tissue repair remains unexplained. Lin28a, a highly conserved RNA-binding protein expressed during embryogenesis, plays roles in development, pluripotency, and metabolism. To determine whether Lin28a might influence tissue repair in adults, we engineered the reactivation of Lin28a expression in several models of tissue injury. Lin28a reactivation improved hair regrowth by promoting anagen in hair follicles and accelerated regrowth of cartilage, bone, and mesenchyme after ear and digit injuries. Lin28a inhibits let-7 microRNA biogenesis; however, let-7 repression was necessary but insufficient to enhance repair. Lin28a bound to and enhanced the translation of mRNAs for several metabolic enzymes, thereby increasing glycolysis and oxidative phosphorylation (OxPhos). Lin28a-mediated enhancement of tissue repair was negated by OxPhos inhibition, whereas a pharmacologically induced increase in OxPhos enhanced repair. Thus, Lin28a enhances tissue repair in some adult tissues by reprogramming cellular bioenergetics. PAPERCLIP:


Subject(s)
RNA-Binding Proteins/metabolism , Wound Healing , Animals , Embryo, Mammalian/metabolism , Energy Metabolism , Extremities/physiology , Hair Follicle/physiology , Humans , Mice , Mice, Transgenic , MicroRNAs/metabolism , Regeneration
19.
Genesis ; 50(5): 443-50, 2012 May.
Article in English | MEDLINE | ID: mdl-22083581

ABSTRACT

With the growing use of genome-wide screens for cis-regulatory elements, there is a pressing need for platforms that enable fast and cost-effective experimental validation of identified hits in relevant developmental and tissue contexts. Here, we describe a murine embryonic stem cell (ESC)-based system that facilitates rapid analysis of putative transcriptional enhancers. Candidate enhancers are targeted with high efficiency to a defined genomic locus via recombinase-mediated cassette exchange. Targeted ESCs are subsequently differentiated in vitro into desired cell types, where enhancer activity is monitored by reporter gene expression. As a proof of principle, we analyzed a previously characterized, Sonic hedgehog (Shh)-dependent, V3 interneuron progenitor (pV3)-specific enhancer for the Nkx2.2 gene, and observed highly specific enhancer activity. Given the broad potential of ESCs to generate a spectrum of cell types, this system can serve as an effective platform for the characterization of gene regulatory networks controlling cell fate specification and cell function.


Subject(s)
Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Base Sequence , Cell Differentiation , DNA Primers , Embryonic Stem Cells/cytology , Gene Targeting , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Immunohistochemistry , Nuclear Proteins , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...