Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.986
Filter
Add more filters

Publication year range
1.
Cell ; 180(5): 984-1001.e22, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32109414

ABSTRACT

Aging causes a functional decline in tissues throughout the body that may be delayed by caloric restriction (CR). However, the cellular profiles and signatures of aging, as well as those ameliorated by CR, remain unclear. Here, we built comprehensive single-cell and single-nucleus transcriptomic atlases across various rat tissues undergoing aging and CR. CR attenuated aging-related changes in cell type composition, gene expression, and core transcriptional regulatory networks. Immune cells were increased during aging, and CR favorably reversed the aging-disturbed immune ecosystem. Computational prediction revealed that the abnormal cell-cell communication patterns observed during aging, including the excessive proinflammatory ligand-receptor interplay, were reversed by CR. Our work provides multi-tissue single-cell transcriptional landscapes associated with aging and CR in a mammal, enhances our understanding of the robustness of CR as a geroprotective intervention, and uncovers how metabolic intervention can act upon the immune system to modify the process of aging.


Subject(s)
Aging/genetics , Caloric Restriction , Immune System/metabolism , Transcriptome/genetics , Aging/metabolism , Aging/pathology , Animals , Cellular Reprogramming/genetics , Gene Expression Regulation/genetics , Gene Regulatory Networks/genetics , Humans , Rats , Single-Cell Analysis
2.
Cell ; 183(6): 1617-1633.e22, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33259802

ABSTRACT

Histone H3.3 glycine 34 to arginine/valine (G34R/V) mutations drive deadly gliomas and show exquisite regional and temporal specificity, suggesting a developmental context permissive to their effects. Here we show that 50% of G34R/V tumors (n = 95) bear activating PDGFRA mutations that display strong selection pressure at recurrence. Although considered gliomas, G34R/V tumors actually arise in GSX2/DLX-expressing interneuron progenitors, where G34R/V mutations impair neuronal differentiation. The lineage of origin may facilitate PDGFRA co-option through a chromatin loop connecting PDGFRA to GSX2 regulatory elements, promoting PDGFRA overexpression and mutation. At the single-cell level, G34R/V tumors harbor dual neuronal/astroglial identity and lack oligodendroglial programs, actively repressed by GSX2/DLX-mediated cell fate specification. G34R/V may become dispensable for tumor maintenance, whereas mutant-PDGFRA is potently oncogenic. Collectively, our results open novel research avenues in deadly tumors. G34R/V gliomas are neuronal malignancies where interneuron progenitors are stalled in differentiation by G34R/V mutations and malignant gliogenesis is promoted by co-option of a potentially targetable pathway, PDGFRA signaling.


Subject(s)
Brain Neoplasms/genetics , Carcinogenesis/genetics , Glioma/genetics , Histones/genetics , Interneurons/metabolism , Mutation/genetics , Neural Stem Cells/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain Neoplasms/pathology , Carcinogenesis/pathology , Cell Lineage , Cellular Reprogramming/genetics , Chromatin/metabolism , Embryo, Mammalian/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Silencing , Glioma/pathology , Histones/metabolism , Lysine/metabolism , Mice, Inbred C57BL , Models, Biological , Neoplasm Grading , Oligodendroglia/metabolism , Promoter Regions, Genetic/genetics , Prosencephalon/embryology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Transcription, Genetic , Transcriptome/genetics
3.
Cell ; 176(4): 928-943.e22, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30712874

ABSTRACT

Understanding the molecular programs that guide differentiation during development is a major challenge. Here, we introduce Waddington-OT, an approach for studying developmental time courses to infer ancestor-descendant fates and model the regulatory programs that underlie them. We apply the method to reconstruct the landscape of reprogramming from 315,000 single-cell RNA sequencing (scRNA-seq) profiles, collected at half-day intervals across 18 days. The results reveal a wider range of developmental programs than previously characterized. Cells gradually adopt either a terminal stromal state or a mesenchymal-to-epithelial transition state. The latter gives rise to populations related to pluripotent, extra-embryonic, and neural cells, with each harboring multiple finer subpopulations. The analysis predicts transcription factors and paracrine signals that affect fates and experiments validate that the TF Obox6 and the cytokine GDF9 enhance reprogramming efficiency. Our approach sheds light on the process and outcome of reprogramming and provides a framework applicable to diverse temporal processes in biology.


Subject(s)
Cellular Reprogramming/genetics , Gene Expression Profiling/methods , Single-Cell Analysis/methods , Animals , Cell Differentiation/genetics , Cells, Cultured , Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Gene Expression , Gene Expression Regulation, Developmental/genetics , Induced Pluripotent Stem Cells/metabolism , Mice , Sequence Analysis, RNA/methods , Transcription Factors/metabolism
4.
Nat Rev Mol Cell Biol ; 22(6): 410-424, 2021 06.
Article in English | MEDLINE | ID: mdl-33619373

ABSTRACT

The reprogramming of somatic cells with defined factors, which converts cells from one lineage into cells of another, has greatly reshaped our traditional views on cell identity and cell fate determination. Direct reprogramming (also known as transdifferentiation) refers to cell fate conversion without transitioning through an intermediary pluripotent state. Given that the number of cell types that can be generated by direct reprogramming is rapidly increasing, it has become a promising strategy to produce functional cells for therapeutic purposes. This Review discusses the evolution of direct reprogramming from a transcription factor-based method to a small-molecule-driven approach, the recent progress in enhancing reprogrammed cell maturation, and the challenges associated with in vivo direct reprogramming for translational applications. It also describes our current understanding of the molecular mechanisms underlying direct reprogramming, including the role of transcription factors, epigenetic modifications, non-coding RNAs, and the function of metabolic reprogramming, and highlights novel insights gained from single-cell omics studies.


Subject(s)
Cell Differentiation/physiology , Cellular Reprogramming/physiology , Epigenesis, Genetic/genetics , Animals , Cell Differentiation/genetics , Cell Transdifferentiation/genetics , Cell Transdifferentiation/physiology , Cellular Reprogramming/genetics , Humans
5.
Cell ; 172(5): 993-1006.e13, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29456083

ABSTRACT

The fate and function of epigenetic marks during the germline-to-embryo transition is a key issue in developmental biology, with relevance to stem cell programming and transgenerational inheritance. In zebrafish, DNA methylation patterns are programmed in transcriptionally quiescent cleavage embryos; paternally inherited patterns are maintained, whereas maternal patterns are reprogrammed to match the paternal. Here, we provide the mechanism by demonstrating that "Placeholder" nucleosomes, containing histone H2A variant H2A.Z(FV) and H3K4me1, virtually occupy all regions lacking DNA methylation in both sperm and cleavage embryos and reside at promoters encoding housekeeping and early embryonic transcription factors. Upon genome-wide transcriptional onset, genes with Placeholder become either active (H3K4me3) or silent (H3K4me3/K27me3). Notably, perturbations causing Placeholder loss confer DNA methylation accumulation, whereas acquisition/expansion of Placeholder confers DNA hypomethylation and improper gene activation. Thus, during transcriptionally quiescent gametic and embryonic stages, an H2A.Z(FV)/H3K4me1-containing Placeholder nucleosome deters DNA methylation, poising parental genes for either gene-specific activation or facultative repression.


Subject(s)
Cellular Reprogramming/genetics , DNA Methylation/genetics , Embryo, Nonmammalian/metabolism , Germ Cells/metabolism , Nucleosomes/metabolism , Animals , Histones/metabolism , Male , Mutation/genetics , Spermatozoa/metabolism , Zebrafish/genetics , Zebrafish Proteins/metabolism
6.
Annu Rev Cell Dev Biol ; 35: 433-452, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31340126

ABSTRACT

Cellular reprogramming experiments from somatic cell types have demonstrated the plasticity of terminally differentiated cell states. Recent efforts in understanding the mechanisms of cellular reprogramming have begun to elucidate the differentiation trajectories along the reprogramming processes. In this review, we focus mainly on direct reprogramming strategies by transcription factors and highlight the variables that contribute to cell fate conversion outcomes. We review key studies that shed light on the cellular and molecular mechanisms by investigating differentiation trajectories and alternative cell states as well as transcription factor regulatory activities during cell fate reprogramming. Finally, we highlight a few concepts that we believe require attention, particularly when measuring the success of cell reprogramming experiments.


Subject(s)
Cell Transdifferentiation/physiology , Cellular Reprogramming/genetics , Epigenesis, Genetic/genetics , Transcription Factors/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Transdifferentiation/genetics , Epigenesis, Genetic/physiology , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Humans , Signal Transduction/genetics , Transcription Factors/genetics
7.
Nat Immunol ; 21(3): 309-320, 2020 03.
Article in English | MEDLINE | ID: mdl-31953534

ABSTRACT

Tissue-resident memory T cells (TRM cells) are critical for cellular immunity to respiratory pathogens and reside in both the airways and the interstitium. In the present study, we found that the airway environment drove transcriptional and epigenetic changes that specifically regulated the cytolytic functions of airway TRM cells and promoted apoptosis due to amino acid starvation and activation of the integrated stress response. Comparison of airway TRM cells and splenic effector-memory T cells transferred into the airways indicated that the environment was necessary to activate these pathways, but did not induce TRM cell lineage reprogramming. Importantly, activation of the integrated stress response was reversed in airway TRM cells placed in a nutrient-rich environment. Our data defined the genetic programs of distinct lung TRM cell populations and show that local environmental cues altered airway TRM cells to limit cytolytic function and promote cell death, which ultimately leads to fewer TRM cells in the lung.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Epigenesis, Genetic/immunology , Immunologic Memory/genetics , Lung/immunology , Animals , Apoptosis/immunology , CD8-Positive T-Lymphocytes/cytology , Cell Survival/genetics , Cell Survival/immunology , Cellular Microenvironment/genetics , Cellular Microenvironment/immunology , Female , Lung/cytology , Male , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology
8.
Nat Immunol ; 21(11): 1397-1407, 2020 11.
Article in English | MEDLINE | ID: mdl-32989328

ABSTRACT

Antiviral CD8+ T cell responses are characterized by an initial activation/priming of T lymphocytes followed by a massive proliferation, subset differentiation, population contraction and the development of a stable memory pool. The transcription factor BATF3 has been shown to play a central role in the development of conventional dendritic cells, which in turn are critical for optimal priming of CD8+ T cells. Here we show that BATF3 was expressed transiently within the first days after T cell priming and had long-lasting T cell-intrinsic effects. T cells that lacked Batf3 showed normal expansion and differentiation, yet succumbed to an aggravated contraction and had a diminished memory response. Vice versa, BATF3 overexpression in CD8+ T cells promoted their survival and transition to memory. Mechanistically, BATF3 regulated T cell apoptosis and longevity via the proapoptotic factor BIM. By programing CD8+ T cell survival and memory, BATF3 is a promising molecule to optimize adoptive T cell therapy in patients.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cellular Reprogramming/genetics , Immunologic Memory/genetics , Repressor Proteins/genetics , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation , Cell Survival/genetics , Gene Expression , Humans , Immunophenotyping , Mice , Mice, Knockout , Mice, Transgenic , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Nat Immunol ; 21(11): 1430-1443, 2020 11.
Article in English | MEDLINE | ID: mdl-32839607

ABSTRACT

Macrophages demonstrate remarkable plasticity that is essential for host defense and tissue repair. The tissue niche imprints macrophage identity, phenotype and function. The role of vascular endothelial signals in tailoring the phenotype and function of tissue macrophages remains unknown. The lung is a highly vascularized organ and replete with a large population of resident macrophages. We found that, in response to inflammatory injury, lung endothelial cells release the Wnt signaling modulator Rspondin3, which activates ß-catenin signaling in lung interstitial macrophages and increases mitochondrial respiration by glutaminolysis. The generated tricarboxylic acid cycle intermediate α-ketoglutarate, in turn, serves as the cofactor for the epigenetic regulator TET2 to catalyze DNA hydroxymethylation. Notably, endothelial-specific deletion of Rspondin3 prevented the formation of anti-inflammatory interstitial macrophages in endotoxemic mice and induced unchecked severe inflammatory injury. Thus, the angiocrine-metabolic-epigenetic signaling axis specified by the endothelium is essential for reprogramming interstitial macrophages and dampening inflammatory injury.


Subject(s)
Cellular Reprogramming , Energy Metabolism , Epigenesis, Genetic , Inflammation/etiology , Inflammation/metabolism , Macrophages/immunology , Macrophages/metabolism , Thrombospondins/genetics , Animals , Biomarkers , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Disease Models, Animal , Disease Susceptibility , Fluorescent Antibody Technique , Inflammation/pathology , Mice , Mice, Knockout , Mice, Transgenic , Thrombospondins/metabolism
10.
Genes Dev ; 38(7-8): 308-321, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38719541

ABSTRACT

The transcription factor Oct4/Pou5f1 is a component of the regulatory circuitry governing pluripotency and is widely used to induce pluripotency from somatic cells. Here we used domain swapping and mutagenesis to study Oct4's reprogramming ability, identifying a redox-sensitive DNA binding domain, cysteine residue (Cys48), as a key determinant of reprogramming and differentiation. Oct4 Cys48 sensitizes the protein to oxidative inhibition of DNA binding activity and promotes oxidation-mediated protein ubiquitylation. Pou5f1 C48S point mutation has little effect on undifferentiated embryonic stem cells (ESCs) but upon retinoic acid (RA) treatment causes retention of Oct4 expression, deregulated gene expression, and aberrant differentiation. Pou5f1 C48S ESCs also form less differentiated teratomas and contribute poorly to adult somatic tissues. Finally, we describe Pou5f1 C48S (Janky) mice, which in the homozygous condition are severely developmentally restricted after E4.5. Rare animals bypassing this restriction appear normal at birth but are sterile. Collectively, these findings uncover a novel Oct4 redox mechanism involved in both entry into and exit from pluripotency.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Octamer Transcription Factor-3 , Oxidation-Reduction , Octamer Transcription Factor-3/metabolism , Octamer Transcription Factor-3/genetics , Animals , Mice , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Tretinoin/pharmacology , Tretinoin/metabolism , Gene Expression Regulation, Developmental/genetics , Humans
11.
Nat Immunol ; 19(3): 279-290, 2018 03.
Article in English | MEDLINE | ID: mdl-29434353

ABSTRACT

Deletion of master regulators of the B cell lineage reprograms B cells into T cells. Here we found that the transcription factor Hoxb5, which is expressed in uncommitted hematopoietic progenitor cells but is not present in cells committed to the B cell or T cell lineage, was able to reprogram pro-pre-B cells into functional early T cell lineage progenitors. This reprogramming started in the bone marrow and was completed in the thymus and gave rise to T lymphocytes with transcriptomes, hierarchical differentiation, tissue distribution and immunological functions that closely resembled those of their natural counterparts. Hoxb5 repressed B cell 'master genes', activated regulators of T cells and regulated crucial chromatin modifiers in pro-pre-B cells and ultimately drove the B cell fate-to-T cell fate conversion. Our results provide a de novo paradigm for the generation of functional T cells through reprogramming in vivo.


Subject(s)
B-Lymphocytes/cytology , Cell Lineage/immunology , Cellular Reprogramming/immunology , Homeodomain Proteins/immunology , T-Lymphocytes/cytology , Animals , Cell Differentiation , Cell Lineage/genetics , Cellular Reprogramming/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Precursor Cells, B-Lymphoid/cytology
12.
Nature ; 631(8019): 170-178, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38768632

ABSTRACT

Epigenetic reprogramming resets parental epigenetic memories and differentiates primordial germ cells (PGCs) into mitotic pro-spermatogonia or oogonia. This process ensures sexually dimorphic germ cell development for totipotency1. In vitro reconstitution of epigenetic reprogramming in humans remains a fundamental challenge. Here we establish a strategy for inducing epigenetic reprogramming and differentiation of pluripotent stem-cell-derived human PGC-like cells (hPGCLCs) into mitotic pro-spermatogonia or oogonia, coupled with their extensive amplification (about >1010-fold). Bone morphogenetic protein (BMP) signalling is a key driver of these processes. BMP-driven hPGCLC differentiation involves attenuation of the MAPK (ERK) pathway and both de novo and maintenance DNA methyltransferase activities, which probably promote replication-coupled, passive DNA demethylation. hPGCLCs deficient in TET1, an active DNA demethylase abundant in human germ cells2,3, differentiate into extraembryonic cells, including amnion, with de-repression of key genes that bear bivalent promoters. These cells fail to fully activate genes vital for spermatogenesis and oogenesis, and their promoters remain methylated. Our study provides a framework for epigenetic reprogramming in humans and an important advance in human biology. Through the generation of abundant mitotic pro-spermatogonia and oogonia-like cells, our results also represent a milestone for human in vitro gametogenesis research and its potential translation into reproductive medicine.


Subject(s)
Cellular Reprogramming , Epigenesis, Genetic , Germ Cells , In Vitro Techniques , Female , Humans , Male , Amnion/cytology , Bone Morphogenetic Proteins/metabolism , Cellular Reprogramming/genetics , DNA Methylation/genetics , Germ Cells/metabolism , Germ Cells/cytology , MAP Kinase Signaling System , Mitosis/genetics , Mixed Function Oxygenases/deficiency , Oogenesis/genetics , Oogonia/cytology , Oogonia/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic/genetics , Spermatogenesis/genetics , Spermatogonia/cytology , Spermatogonia/metabolism , Gene Expression Regulation, Developmental
13.
Nature ; 631(8022): 876-883, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38987605

ABSTRACT

Advancements in precision oncology over the past decades have led to new therapeutic interventions, but the efficacy of such treatments is generally limited by an adaptive process that fosters drug resistance1. In addition to genetic mutations2, recent research has identified a role for non-genetic plasticity in transient drug tolerance3 and the acquisition of stable resistance4,5. However, the dynamics of cell-state transitions that occur in the adaptation to cancer therapies remain unknown and require a systems-level longitudinal framework. Here we demonstrate that resistance develops through trajectories of cell-state transitions accompanied by a progressive increase in cell fitness, which we denote as the 'resistance continuum'. This cellular adaptation involves a stepwise assembly of gene expression programmes and epigenetically reinforced cell states underpinned by phenotypic plasticity, adaptation to stress and metabolic reprogramming. Our results support the notion that epithelial-to-mesenchymal transition or stemness programmes-often considered a proxy for phenotypic plasticity-enable adaptation, rather than a full resistance mechanism. Through systematic genetic perturbations, we identify the acquisition of metabolic dependencies, exposing vulnerabilities that can potentially be exploited therapeutically. The concept of the resistance continuum highlights the dynamic nature of cellular adaptation and calls for complementary therapies directed at the mechanisms underlying adaptive cell-state transitions.


Subject(s)
Adaptation, Physiological , Cell Plasticity , Drug Resistance, Neoplasm , Neoplasms , Female , Humans , Mice , Adaptation, Physiological/drug effects , Adaptation, Physiological/genetics , Cell Line, Tumor , Cell Plasticity/drug effects , Cell Plasticity/genetics , Cellular Reprogramming/drug effects , Cellular Reprogramming/genetics , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Epigenesis, Genetic , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Phenotype
14.
Immunity ; 52(6): 1057-1074.e7, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32362324

ABSTRACT

Tissue-resident and recruited macrophages contribute to both host defense and pathology. Multiple macrophage phenotypes are represented in diseased tissues, but we lack deep understanding of mechanisms controlling diversification. Here, we investigate origins and epigenetic trajectories of hepatic macrophages during diet-induced non-alcoholic steatohepatitis (NASH). The NASH diet induced significant changes in Kupffer cell enhancers and gene expression, resulting in partial loss of Kupffer cell identity, induction of Trem2 and Cd9 expression, and cell death. Kupffer cell loss was compensated by gain of adjacent monocyte-derived macrophages that exhibited convergent epigenomes, transcriptomes, and functions. NASH-induced changes in Kupffer cell enhancers were driven by AP-1 and EGR that reprogrammed LXR functions required for Kupffer cell identity and survival to instead drive a scar-associated macrophage phenotype. These findings reveal mechanisms by which disease-associated environmental signals instruct resident and recruited macrophages to acquire distinct gene expression programs and corresponding functions.


Subject(s)
Cellular Microenvironment/genetics , Cellular Reprogramming/genetics , Epigenesis, Genetic , Gene Expression Regulation , Myeloid Cells/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Biomarkers , Chromatin Immunoprecipitation Sequencing , Diet , Disease Models, Animal , Gene Expression Profiling , Gene Ontology , High-Throughput Nucleotide Sequencing , Kupffer Cells/immunology , Kupffer Cells/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Non-alcoholic Fatty Liver Disease/pathology , Organ Specificity/genetics , Organ Specificity/immunology , Protein Binding , Signal Transduction , Single-Cell Analysis
15.
Nature ; 619(7971): 860-867, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37468622

ABSTRACT

Many cancers originate from stem or progenitor cells hijacked by somatic mutations that drive replication, exemplified by adenomatous transformation of pulmonary alveolar epithelial type II (AT2) cells1. Here we demonstrate a different scenario: expression of KRAS(G12D) in differentiated AT1 cells reprograms them slowly and asynchronously back into AT2 stem cells that go on to generate indolent tumours. Like human lepidic adenocarcinoma, the tumour cells slowly spread along alveolar walls in a non-destructive manner and have low ERK activity. We find that AT1 and AT2 cells act as distinct cells of origin and manifest divergent responses to concomitant WNT activation and KRAS(G12D) induction, which accelerates AT2-derived but inhibits AT1-derived adenoma proliferation. Augmentation of ERK activity in KRAS(G12D)-induced AT1 cells increases transformation efficiency, proliferation and progression from lepidic to mixed tumour histology. Overall, we have identified a new cell of origin for lung adenocarcinoma, the AT1 cell, which recapitulates features of human lepidic cancer. In so doing, we also uncover a capacity for oncogenic KRAS to reprogram a differentiated and quiescent cell back into its parent stem cell en route to adenomatous transformation. Our work further reveals that irrespective of a given cancer's current molecular profile and driver oncogene, the cell of origin exerts a pervasive and perduring influence on its subsequent behaviour.


Subject(s)
Adenocarcinoma of Lung , Cellular Reprogramming , Lung Neoplasms , Proto-Oncogene Proteins p21(ras) , Stem Cells , Humans , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Cellular Reprogramming/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Stem Cells/metabolism , Stem Cells/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism
16.
Annu Rev Genet ; 54: 367-385, 2020 11 23.
Article in English | MEDLINE | ID: mdl-32886547

ABSTRACT

Pioneer transcription factors have the intrinsic biochemical ability to scan partial DNA sequence motifs that are exposed on the surface of a nucleosome and thus access silent genes that are inaccessible to other transcription factors. Pioneer factors subsequently enable other transcription factors, nucleosome remodeling complexes, and histone modifiers to engage chromatin, thereby initiating the formation of an activating or repressive regulatory sequence. Thus, pioneer factors endow the competence for fate changes in embryonic development, are essential for cellular reprogramming, and rewire gene networks in cancer cells. Recent studies with reconstituted nucleosomes in vitro and chromatin binding in vivo reveal that pioneer factors can directly perturb nucleosome structure and chromatin accessibility in different ways. This review focuses on our current understanding of the mechanisms by which pioneer factors initiate gene network changes and will ultimately contribute to our ability to control cell fates at will.


Subject(s)
Gene Regulatory Networks/genetics , Transcription Factors/genetics , Animals , Cellular Reprogramming/genetics , Chromatin/genetics , Embryonic Development/genetics , Histones/genetics , Humans , Nucleosomes/genetics
17.
Mol Cell ; 80(6): 1013-1024.e6, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33338401

ABSTRACT

Impaired DNA crosslink repair leads to Fanconi anemia (FA), characterized by a unique manifestation of bone marrow failure and pancytopenia among diseases caused by DNA damage response defects. As a germline disorder, why the hematopoietic hierarchy is specifically affected is not fully understood. We find that reprogramming transcription during hematopoietic differentiation results in an overload of genotoxic stress, which causes aborted differentiation and depletion of FA mutant progenitor cells. DNA damage onset most likely arises from formaldehyde, an obligate by-product of oxidative protein demethylation during transcription regulation. Our results demonstrate that rapid and extensive transcription reprogramming associated with hematopoietic differentiation poses a major threat to genome stability and cell viability in the absence of the FA pathway. The connection between differentiation and DNA damage accumulation reveals a novel mechanism of genome scarring and is critical to exploring therapies to counteract the aplastic anemia for the treatment of FA patients.


Subject(s)
Cell Differentiation/drug effects , Cellular Reprogramming/genetics , Fanconi Anemia/genetics , Formaldehyde/toxicity , DNA Damage/drug effects , DNA Repair/genetics , Fanconi Anemia/blood , Fanconi Anemia/pathology , Formaldehyde/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Genomic Instability/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Humans , K562 Cells , Transcription, Genetic
18.
Development ; 151(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38984586

ABSTRACT

In the injured zebrafish retina, Müller glial cells (MG) reprogram to adopt retinal stem cell properties and regenerate damaged neurons. The strongest zebrafish reprogramming factors might be good candidates for stimulating a similar regenerative response by mammalian MG. Myc proteins are potent reprogramming factors that can stimulate cellular plasticity in differentiated cells; however, their role in MG reprogramming and retina regeneration remains poorly explored. Here, we report that retinal injury stimulates mycb and mych expression and that, although both Mycb and Mych stimulate MG reprogramming and proliferation, only Mych enhances retinal neuron apoptosis. RNA-sequencing analysis of wild-type, mychmut and mycbmut fish revealed that Mycb and Mych regulate ∼40% and ∼16%, respectively, of the genes contributing to the regeneration-associated transcriptome of MG. Of these genes, those that are induced are biased towards regulation of ribosome biogenesis, protein synthesis, DNA synthesis, and cell division, which are the top cellular processes affected by retinal injury, suggesting that Mycb and Mych are potent MG reprogramming factors. Consistent with this, forced expression of either of these proteins is sufficient to stimulate MG proliferation in the uninjured retina.


Subject(s)
Cell Proliferation , Cellular Reprogramming , Ependymoglial Cells , Retina , Zebrafish Proteins , Zebrafish , Animals , Apoptosis/genetics , Cellular Reprogramming/genetics , Ependymoglial Cells/metabolism , Ependymoglial Cells/cytology , Retina/metabolism , Retina/cytology , Retinal Neurons/metabolism , Transcriptome/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics
19.
Nat Rev Genet ; 22(3): 169-184, 2021 03.
Article in English | MEDLINE | ID: mdl-33318706

ABSTRACT

The encoded biosynthesis of proteins provides the ultimate paradigm for high-fidelity synthesis of long polymers of defined sequence and composition, but it is limited to polymerizing the canonical amino acids. Recent advances have built on genetic code expansion - which commonly permits the cellular incorporation of one type of non-canonical amino acid into a protein - to enable the encoded incorporation of several distinct non-canonical amino acids. Developments include strategies to read quadruplet codons, use non-natural DNA base pairs, synthesize completely recoded genomes and create orthogonal translational components with reprogrammed specificities. These advances may enable the genetically encoded synthesis of non-canonical biopolymers and provide a platform for transforming the discovery and evolution of new materials and therapeutics.


Subject(s)
Cellular Reprogramming/genetics , Genetic Code/genetics , Amino Acids/genetics , Animals , Codon/genetics , DNA/genetics , Humans , Protein Biosynthesis/genetics , Proteins/genetics
20.
Mol Cell ; 74(4): 651-663.e8, 2019 05 16.
Article in English | MEDLINE | ID: mdl-30954402

ABSTRACT

Accumulating evidence supports the role of the DNA damage response (DDR) in the negative regulation of tumorigenesis. Here, we found that DDR signaling poises a series of epigenetic events, resulting in activation of pro-tumorigenic genes but can go as far as reactivation of the pluripotency gene OCT4. Loss of DNA methylation appears to be a key initiating event in DDR-dependent OCT4 locus reactivation although full reactivation required the presence of a driving oncogene, such as Myc and macroH2A downregulation. Using genetic-lineage-tracing experiments and an in situ labeling approach, we show that DDR-induced epigenetic reactivation of OCT4 regulates the resistance to chemotherapy and contributes to tumor relapse both in mouse and primary human cancers. In turn, deletion of OCT4 reverses chemoresistance and delays the relapse. Here, we uncovered an unexpected tumor-promoting role of DDR in cancer cell reprogramming, providing novel therapeutic entry points for cancer intervention strategies.


Subject(s)
Carcinogenesis/genetics , DNA Methylation/genetics , Neoplasms/genetics , Octamer Transcription Factor-3/genetics , Animals , Cellular Reprogramming/genetics , DNA Damage/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic , Histones/genetics , Humans , Mice , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics , Recurrence , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL