Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30.327
Filter
Add more filters

Publication year range
1.
Cell ; 187(13): 3229-3230, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38906098

ABSTRACT

Dr. Shinya Yamanaka is recognized for his discovery of the induction of pluripotent stem cells from fibroblasts by a combination of defined factors. In this interview with Cell, he discusses the progress of the field, what's next for clinical applications of iPS cells, and the state of science in Japan and the rest of the world.


Subject(s)
Induced Pluripotent Stem Cells , Animals , Humans , Fibroblasts/cytology , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Japan , Cell- and Tissue-Based Therapy , Cell Separation , Cell Culture Techniques , Community Medicine
2.
Cell ; 184(3): 810-826.e23, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33406409

ABSTRACT

Development of the human intestine is not well understood. Here, we link single-cell RNA sequencing and spatial transcriptomics to characterize intestinal morphogenesis through time. We identify 101 cell states including epithelial and mesenchymal progenitor populations and programs linked to key morphogenetic milestones. We describe principles of crypt-villus axis formation; neural, vascular, mesenchymal morphogenesis, and immune population of the developing gut. We identify the differentiation hierarchies of developing fibroblast and myofibroblast subtypes and describe diverse functions for these including as vascular niche cells. We pinpoint the origins of Peyer's patches and gut-associated lymphoid tissue (GALT) and describe location-specific immune programs. We use our resource to present an unbiased analysis of morphogen gradients that direct sequential waves of cellular differentiation and define cells and locations linked to rare developmental intestinal disorders. We compile a publicly available online resource, spatio-temporal analysis resource of fetal intestinal development (STAR-FINDer), to facilitate further work.


Subject(s)
Intestines/cytology , Intestines/growth & development , Single-Cell Analysis , Endothelial Cells/cytology , Enteric Nervous System/cytology , Fetus/embryology , Fibroblasts/cytology , Humans , Immunity , Intestinal Diseases/congenital , Intestinal Diseases/pathology , Intestinal Mucosa/growth & development , Intestines/blood supply , Ligands , Mesoderm/cytology , Neovascularization, Physiologic , Pericytes/cytology , Stem Cells/cytology , Time Factors , Transcription Factors/metabolism
3.
Cell ; 184(12): 3299-3317.e22, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34019794

ABSTRACT

Organoids capable of forming tissue-like structures have transformed our ability to model human development and disease. With the notable exception of the human heart, lineage-specific self-organizing organoids have been reported for all major organs. Here, we established self-organizing cardioids from human pluripotent stem cells that intrinsically specify, pattern, and morph into chamber-like structures containing a cavity. Cardioid complexity can be controlled by signaling that instructs the separation of cardiomyocyte and endothelial layers and by directing epicardial spreading, inward migration, and differentiation. We find that cavity morphogenesis is governed by a mesodermal WNT-BMP signaling axis and requires its target HAND1, a transcription factor linked to developmental heart chamber defects. Upon cryoinjury, cardioids initiated a cell-type-dependent accumulation of extracellular matrix, an early hallmark of both regeneration and heart disease. Thus, human cardioids represent a powerful platform to mechanistically dissect self-organization, congenital heart defects and serve as a foundation for future translational research.


Subject(s)
Heart/embryology , Organogenesis , Organoids/embryology , Activins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Morphogenetic Proteins/metabolism , Calcium/metabolism , Cell Line , Cell Lineage , Chickens , Endothelial Cells/cytology , Extracellular Matrix Proteins/metabolism , Female , Fibroblasts/cytology , Homeobox Protein Nkx-2.5/metabolism , Humans , Male , Mesoderm/embryology , Models, Biological , Myocardium/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Wnt Proteins/metabolism
4.
Cell ; 177(3): 622-638.e22, 2019 04 18.
Article in English | MEDLINE | ID: mdl-31002797

ABSTRACT

DNA repair has been hypothesized to be a longevity determinant, but the evidence for it is based largely on accelerated aging phenotypes of DNA repair mutants. Here, using a panel of 18 rodent species with diverse lifespans, we show that more robust DNA double-strand break (DSB) repair, but not nucleotide excision repair (NER), coevolves with longevity. Evolution of NER, unlike DSB, is shaped primarily by sunlight exposure. We further show that the capacity of the SIRT6 protein to promote DSB repair accounts for a major part of the variation in DSB repair efficacy between short- and long-lived species. We dissected the molecular differences between a weak (mouse) and a strong (beaver) SIRT6 protein and identified five amino acid residues that are fully responsible for their differential activities. Our findings demonstrate that DSB repair and SIRT6 have been optimized during the evolution of longevity, which provides new targets for anti-aging interventions.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Longevity/genetics , Sirtuins/metabolism , Amino Acid Sequence , Animals , Body Weight , DNA Breaks, Double-Stranded/radiation effects , Evolution, Molecular , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Knockout Techniques , Humans , Kinetics , Male , Mutagenesis , Phylogeny , Rodentia/classification , Sequence Alignment , Sirtuins/chemistry , Sirtuins/genetics , Ultraviolet Rays
5.
Nat Immunol ; 22(8): 1042-1051, 2021 08.
Article in English | MEDLINE | ID: mdl-34267375

ABSTRACT

Pathogens and vaccines that produce persisting antigens can generate expanded pools of effector memory CD8+ T cells, described as memory inflation. While properties of inflating memory CD8+ T cells have been characterized, the specific cell types and tissue factors responsible for their maintenance remain elusive. Here, we show that clinically applied adenovirus vectors preferentially target fibroblastic stromal cells in cultured human tissues. Moreover, we used cell-type-specific antigen targeting to define critical cells and molecules that sustain long-term antigen presentation and T cell activity after adenovirus vector immunization in mice. While antigen targeting to myeloid cells was insufficient to activate antigen-specific CD8+ T cells, genetic activation of antigen expression in Ccl19-cre-expressing fibroblastic stromal cells induced inflating CD8+ T cells. Local ablation of vector-targeted cells revealed that lung fibroblasts support the protective function and metabolic fitness of inflating memory CD8+ T cells in an interleukin (IL)-33-dependent manner. Collectively, these data define a critical fibroblastic niche that underpins robust protective immunity operating in a clinically important vaccine platform.


Subject(s)
Adenoviridae/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Interleukin-33/immunology , Lymphocyte Activation/immunology , Stromal Cells/immunology , Adenoviridae/genetics , Animals , Cell Line, Tumor , Chemokine CCL19/metabolism , Chimera/genetics , Epitopes, T-Lymphocyte/immunology , Fibroblasts/cytology , Fibroblasts/metabolism , Genetic Vectors/immunology , Humans , Lung/cytology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Vaccination
6.
Cell ; 175(6): 1620-1633.e13, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30415836

ABSTRACT

Fibroblasts are an essential cellular and structural component of our organs. Despite several advances, the critical behaviors that fibroblasts utilize to maintain their homeostasis in vivo have remained unclear. Here, by tracking the same skin fibroblasts in live mice, we show that fibroblast position is stable over time and that this stability is maintained despite the loss of neighboring fibroblasts. In contrast, fibroblast membranes are dynamic during homeostasis and extend to fill the space of lost neighboring fibroblasts in a Rac1-dependent manner. Positional stability is sustained during aging despite a progressive accumulation of gaps in fibroblast nuclei organization, while membrane occupancy continues to be maintained. This work defines positional stability and cell occupancy as key principles of skin fibroblast homeostasis in vivo, throughout the lifespan of mice, and identifies membrane extension in the absence of migration as the core cellular mechanism to carry out these principles.


Subject(s)
Cell Membrane/metabolism , Cell Nucleus/metabolism , Fibroblasts/metabolism , Homeostasis/physiology , Skin/metabolism , Animals , Cell Membrane/genetics , Cell Nucleus/genetics , Cells, Cultured , Fibroblasts/cytology , Mice , Mice, Transgenic , Skin/cytology
7.
Cell ; 172(4): 744-757.e17, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29398113

ABSTRACT

Cell communication within tissues is mediated by multiple paracrine signals including growth factors, which control cell survival and proliferation. Cells and the growth factors they produce and receive constitute a circuit with specific properties that ensure homeostasis. Here, we used computational and experimental approaches to characterize the features of cell circuits based on growth factor exchange between macrophages and fibroblasts, two cell types found in most mammalian tissues. We found that the macrophage-fibroblast cell circuit is stable and robust to perturbations. Analytical screening of all possible two-cell circuit topologies revealed the circuit features sufficient for stability, including environmental constraint and negative-feedback regulation. Moreover, we found that cell-cell contact is essential for the stability of the macrophage-fibroblast circuit. These findings illustrate principles of cell circuit design and provide a quantitative perspective on cell interactions.


Subject(s)
Cell Communication/physiology , Cell Proliferation/physiology , Fibroblasts/metabolism , Macrophages/metabolism , Animals , Cell Survival/physiology , Female , Fibroblasts/cytology , Macrophages/cytology , Male , Mice , Mice, Transgenic
8.
Cell ; 174(2): 363-376.e16, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29887381

ABSTRACT

Visualization of the transcriptome and the nuclear organization in situ has been challenging for single-cell analysis. Here, we demonstrate a multiplexed single-molecule in situ method, intron seqFISH, that allows imaging of 10,421 genes at their nascent transcription active sites in single cells, followed by mRNA and lncRNA seqFISH and immunofluorescence. This nascent transcriptome-profiling method can identify different cell types and states with mouse embryonic stem cells and fibroblasts. The nascent sites of RNA synthesis tend to be localized on the surfaces of chromosome territories, and their organization in individual cells is highly variable. Surprisingly, the global nascent transcription oscillated asynchronously in individual cells with a period of 2 hr in mouse embryonic stem cells, as well as in fibroblasts. Together, spatial genomics of the nascent transcriptome by intron seqFISH reveals nuclear organizational principles and fast dynamics in single cells that are otherwise obscured.


Subject(s)
In Situ Hybridization, Fluorescence/methods , Transcriptome , Animals , Catalytic Domain , Cell Line , Chromosomes/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Introns , Mice , Microscopy, Fluorescence , Microscopy, Video , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Single-Cell Analysis
9.
Cell ; 169(1): 132-147.e16, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28340339

ABSTRACT

The accumulation of irreparable cellular damage restricts healthspan after acute stress or natural aging. Senescent cells are thought to impair tissue function, and their genetic clearance can delay features of aging. Identifying how senescent cells avoid apoptosis allows for the prospective design of anti-senescence compounds to address whether homeostasis can also be restored. Here, we identify FOXO4 as a pivot in senescent cell viability. We designed a FOXO4 peptide that perturbs the FOXO4 interaction with p53. In senescent cells, this selectively causes p53 nuclear exclusion and cell-intrinsic apoptosis. Under conditions where it was well tolerated in vivo, this FOXO4 peptide neutralized doxorubicin-induced chemotoxicity. Moreover, it restored fitness, fur density, and renal function in both fast aging XpdTTD/TTD and naturally aged mice. Thus, therapeutic targeting of senescent cells is feasible under conditions where loss of health has already occurred, and in doing so tissue homeostasis can effectively be restored.


Subject(s)
Aging/pathology , Antibiotics, Antineoplastic/adverse effects , Cell-Penetrating Peptides/pharmacology , Doxorubicin/adverse effects , Aging/drug effects , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Cell Cycle Proteins , Cell Line , Cell Survival , Cellular Senescence/drug effects , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Female , Fibroblasts/cytology , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/metabolism , Humans , Inclusion Bodies/drug effects , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Kidney/drug effects , Kidney/physiology , Liver/drug effects , Liver/physiology , Male , Mice , Trichothiodystrophy Syndromes/drug therapy , Tumor Suppressor Protein p53/metabolism
10.
Cell ; 160(4): 659-672, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25679760

ABSTRACT

The mesenchymal-amoeboid transition (MAT) was proposed as a mechanism for cancer cells to adapt their migration mode to their environment. While the molecular pathways involved in this transition are well documented, the role of the microenvironment in the MAT is still poorly understood. Here, we investigated how confinement and adhesion affect this transition. We report that, in the absence of focal adhesions and under conditions of confinement, mesenchymal cells can spontaneously switch to a fast amoeboid migration phenotype. We identified two main types of fast migration--one involving a local protrusion and a second involving a myosin-II-dependent mechanical instability of the cell cortex that leads to a global cortical flow. Interestingly, transformed cells are more prone to adopt this fast migration mode. Finally, we propose a generic model that explains migration transitions and predicts a phase diagram of migration phenotypes based on three main control parameters: confinement, adhesion, and contractility.


Subject(s)
Mesoderm/cytology , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Epithelial Cells/cytology , Fibroblasts/cytology , Focal Adhesions , HeLa Cells , Humans , Skin/cytology
11.
Nature ; 631(8019): 207-215, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38926576

ABSTRACT

Pyroptosis is a lytic cell death mode that helps limit the spread of infections and is also linked to pathology in sterile inflammatory diseases and autoimmune diseases1-4. During pyroptosis, inflammasome activation and the engagement of caspase-1 lead to cell death, along with the maturation and secretion of the inflammatory cytokine interleukin-1ß (IL-1ß). The dominant effect of IL-1ß in promoting tissue inflammation has clouded the potential influence of other factors released from pyroptotic cells. Here, using a system in which macrophages are induced to undergo pyroptosis without IL-1ß or IL-1α release (denoted Pyro-1), we identify unexpected beneficial effects of the Pyro-1 secretome. First, we noted that the Pyro-1 supernatants upregulated gene signatures linked to migration, cellular proliferation and wound healing. Consistent with this gene signature, Pyro-1 supernatants boosted migration of primary fibroblasts and macrophages, and promoted faster wound closure in vitro and improved tissue repair in vivo. In mechanistic studies, lipidomics and metabolomics of the Pyro-1 supernatants identified the presence of both oxylipins and metabolites, linking them to pro-wound-healing effects. Focusing specifically on the oxylipin prostaglandin E2 (PGE2), we find that its synthesis is induced de novo during pyroptosis, downstream of caspase-1 activation and cyclooxygenase-2 activity; further, PGE2 synthesis occurs late in pyroptosis, with its release dependent on gasdermin D pores opened during pyroptosis. As for the pyroptotic metabolites, they link to immune cell infiltration into the wounds, and polarization to CD301+ macrophages. Collectively, these data advance the concept that the pyroptotic secretome possesses oxylipins and metabolites with tissue repair properties that may be harnessed therapeutically.


Subject(s)
Macrophages , Oxylipins , Pyroptosis , Secretome , Wound Healing , Animals , Female , Humans , Mice , Caspase 1/metabolism , Cell Movement , Cell Proliferation , Cyclooxygenase 2/metabolism , Dinoprostone/biosynthesis , Dinoprostone/metabolism , Fibroblasts/metabolism , Fibroblasts/cytology , Gasdermins/metabolism , Inflammasomes/metabolism , Interleukin-1beta , Lipidomics , Macrophages/metabolism , Macrophages/cytology , Mice, Inbred C57BL , Oxylipins/metabolism , Phosphate-Binding Proteins/metabolism , Secretome/metabolism , Wound Healing/physiology
12.
Immunity ; 52(5): 767-781.e6, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32277911

ABSTRACT

The enzyme cyclic GMP-AMP synthase (cGAS) senses cytosolic DNA in infected and malignant cells and catalyzes the formation of 2'3'cGMP-AMP (cGAMP), which in turn triggers interferon (IFN) production via the STING pathway. Here, we examined the contribution of anion channels to cGAMP transfer and anti-viral defense. A candidate screen revealed that inhibition of volume-regulated anion channels (VRACs) increased propagation of the DNA virus HSV-1 but not the RNA virus VSV. Chemical blockade or genetic ablation of LRRC8A/SWELL1, a VRAC subunit, resulted in defective IFN responses to HSV-1. Biochemical and electrophysiological analyses revealed that LRRC8A/LRRC8E-containing VRACs transport cGAMP and cyclic dinucleotides across the plasma membrane. Enhancing VRAC activity by hypotonic cell swelling, cisplatin, GTPγS, or the cytokines TNF or interleukin-1 increased STING-dependent IFN response to extracellular but not intracellular cGAMP. Lrrc8e-/- mice exhibited impaired IFN responses and compromised immunity to HSV-1. Our findings suggest that cell-to-cell transmission of cGAMP via LRRC8/VRAC channels is central to effective anti-viral immunity.


Subject(s)
Fibroblasts/immunology , Interferons/immunology , Membrane Proteins/immunology , Nucleotides, Cyclic/immunology , Voltage-Dependent Anion Channels/immunology , Animals , Antiviral Agents/immunology , Antiviral Agents/metabolism , Bystander Effect , Cell Line , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , HeLa Cells , Herpes Simplex/immunology , Herpes Simplex/virology , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/physiology , Humans , Interferons/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/immunology , Nucleotidyltransferases/metabolism , Voltage-Dependent Anion Channels/metabolism
13.
Cell ; 158(3): 633-46, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25083873

ABSTRACT

ATR controls chromosome integrity and chromatin dynamics. We have previously shown that yeast Mec1/ATR promotes chromatin detachment from the nuclear envelope to counteract aberrant topological transitions during DNA replication. Here, we provide evidence that ATR activity at the nuclear envelope responds to mechanical stress. Human ATR associates with the nuclear envelope during S phase and prophase, and both osmotic stress and mechanical stretching relocalize ATR to nuclear membranes throughout the cell cycle. The ATR-mediated mechanical response occurs within the range of physiological forces, is reversible, and is independent of DNA damage signaling. ATR-defective cells exhibit aberrant chromatin condensation and nuclear envelope breakdown. We propose that mechanical forces derived from chromosome dynamics and torsional stress on nuclear membranes activate ATR to modulate nuclear envelope plasticity and chromatin association to the nuclear envelope, thus enabling cells to cope with the mechanical strain imposed by these molecular processes.


Subject(s)
Nuclear Envelope/metabolism , Stress, Mechanical , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Checkpoint Kinase 1 , Chromatin/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Osmosis , Protein Kinases/metabolism
14.
Cell ; 156(4): 649-62, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24486105

ABSTRACT

Reprogramming somatic cells to induced pluripotency by Yamanaka factors is usually slow and inefficient and is thought to be a stochastic process. We identified a privileged somatic cell state, from which acquisition of pluripotency could occur in a nonstochastic manner. Subsets of murine hematopoietic progenitors are privileged whose progeny cells predominantly adopt the pluripotent fate with activation of endogenous Oct4 locus after four to five divisions in reprogramming conditions. Privileged cells display an ultrafast cell cycle of ∼8 hr. In fibroblasts, a subpopulation cycling at a similar ultrafast speed is observed after 6 days of factor expression and is increased by p53 knockdown. This ultrafast cycling population accounts for >99% of the bulk reprogramming activity in wild-type or p53 knockdown fibroblasts. Our data demonstrate that the stochastic nature of reprogramming can be overcome in a privileged somatic cell state and suggest that cell-cycle acceleration toward a critical threshold is an important bottleneck for reprogramming. PAPERCLIP:


Subject(s)
Cellular Reprogramming , Granulocyte-Macrophage Progenitor Cells/cytology , Induced Pluripotent Stem Cells , Animals , Bone Marrow Cells , Cell Differentiation , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Knockdown Techniques , Genes, p53 , Granulocyte-Macrophage Progenitor Cells/metabolism , Mice
15.
Nature ; 623(7988): 792-802, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37968392

ABSTRACT

Optimal tissue recovery and organismal survival are achieved by spatiotemporal tuning of tissue inflammation, contraction and scar formation1. Here we identify a multipotent fibroblast progenitor marked by CD201 expression in the fascia, the deepest connective tissue layer of the skin. Using skin injury models in mice, single-cell transcriptomics and genetic lineage tracing, ablation and gene deletion models, we demonstrate that CD201+ progenitors control the pace of wound healing by generating multiple specialized cell types, from proinflammatory fibroblasts to myofibroblasts, in a spatiotemporally tuned sequence. We identified retinoic acid and hypoxia signalling as the entry checkpoints into proinflammatory and myofibroblast states. Modulating CD201+ progenitor differentiation impaired the spatiotemporal appearances of fibroblasts and chronically delayed wound healing. The discovery of proinflammatory and myofibroblast progenitors and their differentiation pathways provide a new roadmap to understand and clinically treat impaired wound healing.


Subject(s)
Endothelial Protein C Receptor , Fascia , Wound Healing , Animals , Mice , Cell Differentiation , Cell Hypoxia , Cell Lineage , Disease Models, Animal , Endothelial Protein C Receptor/metabolism , Fascia/cytology , Fascia/injuries , Fascia/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Inflammation/metabolism , Inflammation/pathology , Myofibroblasts/cytology , Myofibroblasts/metabolism , Signal Transduction , Single-Cell Gene Expression Analysis , Skin/cytology , Skin/injuries , Skin/metabolism , Tretinoin/metabolism
16.
Nature ; 619(7971): 801-810, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37438528

ABSTRACT

The function of a cell is defined by its intrinsic characteristics and its niche: the tissue microenvironment in which it dwells. Here we combine single-cell and spatial transcriptomics data to discover cellular niches within eight regions of the human heart. We map cells to microanatomical locations and integrate knowledge-based and unsupervised structural annotations. We also profile the cells of the human cardiac conduction system1. The results revealed their distinctive repertoire of ion channels, G-protein-coupled receptors (GPCRs) and regulatory networks, and implicated FOXP2 in the pacemaker phenotype. We show that the sinoatrial node is compartmentalized, with a core of pacemaker cells, fibroblasts and glial cells supporting glutamatergic signalling. Using a custom CellPhoneDB.org module, we identify trans-synaptic pacemaker cell interactions with glia. We introduce a druggable target prediction tool, drug2cell, which leverages single-cell profiles and drug-target interactions to provide mechanistic insights into the chronotropic effects of drugs, including GLP-1 analogues. In the epicardium, we show enrichment of both IgG+ and IgA+ plasma cells forming immune niches that may contribute to infection defence. Overall, we provide new clarity to cardiac electro-anatomy and immunology, and our suite of computational approaches can be applied to other tissues and organs.


Subject(s)
Cellular Microenvironment , Heart , Multiomics , Myocardium , Humans , Cell Communication , Fibroblasts/cytology , Glutamic Acid/metabolism , Heart/anatomy & histology , Heart/innervation , Ion Channels/metabolism , Myocardium/cytology , Myocardium/immunology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Neuroglia/cytology , Pericardium/cytology , Pericardium/immunology , Plasma Cells/immunology , Receptors, G-Protein-Coupled/metabolism , Sinoatrial Node/anatomy & histology , Sinoatrial Node/cytology , Sinoatrial Node/physiology , Heart Conduction System/anatomy & histology , Heart Conduction System/cytology , Heart Conduction System/metabolism
17.
Mol Cell ; 81(4): 859-869.e8, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33352108

ABSTRACT

Active DNA demethylation via ten-eleven translocation (TET) family enzymes is essential for epigenetic reprogramming in cell state transitions. TET enzymes catalyze up to three successive oxidations of 5-methylcytosine (5mC), generating 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), or 5-carboxycytosine (5caC). Although these bases are known to contribute to distinct demethylation pathways, the lack of tools to uncouple these sequential oxidative events has constrained our mechanistic understanding of the role of TETs in chromatin reprogramming. Here, we describe the first application of biochemically engineered TET mutants that unlink 5mC oxidation steps, examining their effects on somatic cell reprogramming. We show that only TET enzymes proficient for oxidation to 5fC/5caC can rescue the reprogramming potential of Tet2-deficient mouse embryonic fibroblasts. This effect correlated with rapid DNA demethylation at reprogramming enhancers and increased chromatin accessibility later in reprogramming. These experiments demonstrate that DNA demethylation through 5fC/5caC has roles distinct from 5hmC in somatic reprogramming to pluripotency.


Subject(s)
5-Methylcytosine/metabolism , Cellular Reprogramming , DNA-Binding Proteins/metabolism , Embryo, Mammalian/metabolism , Enhancer Elements, Genetic , Epigenesis, Genetic , Fibroblasts/metabolism , Proto-Oncogene Proteins/metabolism , Animals , DNA-Binding Proteins/genetics , Dioxygenases , Embryo, Mammalian/cytology , Fibroblasts/cytology , HEK293 Cells , Humans , Mice , Mice, Knockout , Mutation , NIH 3T3 Cells , Proto-Oncogene Proteins/genetics
18.
Mol Cell ; 81(11): 2428-2444.e6, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33882298

ABSTRACT

Repair pathway "choice" at stalled mammalian replication forks is an important determinant of genome stability; however, the underlying mechanisms are poorly understood. FANCM encodes a multi-domain scaffolding and motor protein that interacts with several distinct repair protein complexes at stalled forks. Here, we use defined mutations engineered within endogenous Fancm in mouse embryonic stem cells to study how Fancm regulates stalled fork repair. We find that distinct FANCM repair functions are enacted by molecularly separable scaffolding domains. These findings define FANCM as a key mediator of repair pathway choice at stalled replication forks and reveal its molecular mechanism. Notably, mutations that inactivate FANCM ATPase function disable all its repair functions and "trap" FANCM at stalled forks. We find that Brca1 hypomorphic mutants are synthetic lethal with Fancm null or Fancm ATPase-defective mutants. The ATPase function of FANCM may therefore represent a promising "druggable" target for therapy of BRCA1-linked cancer.


Subject(s)
BRCA1 Protein/genetics , DNA Helicases/genetics , DNA Repair , DNA Replication , Mouse Embryonic Stem Cells/metabolism , Synthetic Lethal Mutations , Animals , BRCA1 Protein/metabolism , Cell Cycle/genetics , Cell Line , Clone Cells , DNA Helicases/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Ubiquitination
19.
Mol Cell ; 81(5): 1074-1083.e5, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33453169

ABSTRACT

The RAD51 recombinase forms nucleoprotein filaments to promote double-strand break repair, replication fork reversal, and fork stabilization. The stability of these filaments is highly regulated, as both too little and too much RAD51 activity can cause genome instability. RADX is a single-strand DNA (ssDNA) binding protein that regulates DNA replication. Here, we define its mechanism of action. We find that RADX inhibits RAD51 strand exchange and D-loop formation activities. RADX directly and selectively interacts with ATP-bound RAD51, stimulates ATP hydrolysis, and destabilizes RAD51 nucleofilaments. The RADX interaction with RAD51, in addition to its ssDNA binding capability, is required to maintain replication fork elongation rates and fork stability. Furthermore, BRCA2 can overcome the RADX-dependent RAD51 inhibition. Thus, RADX functions in opposition to BRCA2 in regulating RAD51 nucleofilament stability to ensure the right level of RAD51 function during DNA replication.


Subject(s)
BRCA2 Protein/genetics , DNA Replication , DNA, Single-Stranded/genetics , DNA-Binding Proteins/genetics , RNA-Binding Proteins/genetics , Rad51 Recombinase/genetics , Adenosine Triphosphate/metabolism , BRCA2 Protein/metabolism , Cell Line, Tumor , DNA/genetics , DNA/metabolism , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Hydrolysis , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , RNA-Binding Proteins/metabolism , Rad51 Recombinase/metabolism , Signal Transduction , Single Molecule Imaging , Red Fluorescent Protein
20.
Mol Cell ; 81(5): 1027-1042.e4, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33453166

ABSTRACT

Alternative lengthening of telomeres (ALT) is mediated by break-induced replication (BIR), but how BIR is regulated at telomeres is poorly understood. Here, we show that telomeric BIR is a self-perpetuating process. By tethering PML-IV to telomeres, we induced telomere clustering in ALT-associated PML bodies (APBs) and a POLD3-dependent ATR response at telomeres, showing that BIR generates replication stress. Ablation of BLM helicase activity in APBs abolishes telomere synthesis but causes multiple chromosome bridges between telomeres, revealing a function of BLM in processing inter-telomere BIR intermediates. Interestingly, the accumulation of BLM in APBs requires its own helicase activity and POLD3, suggesting that BIR triggers a feedforward loop to further recruit BLM. Enhancing BIR induces PIAS4-mediated TRF2 SUMOylation, and PIAS4 loss deprives APBs of repair proteins and compromises ALT telomere synthesis. Thus, a BLM-driven and PIAS4-mediated feedforward loop operates in APBs to perpetuate BIR, providing a critical mechanism to extend ALT telomeres.


Subject(s)
Fanconi Anemia Complementation Group Proteins/genetics , Feedback, Physiological , Poly-ADP-Ribose Binding Proteins/genetics , Protein Inhibitors of Activated STAT/genetics , RNA Helicases/genetics , Telomere Homeostasis , Telomere/chemistry , Telomeric Repeat Binding Protein 2/metabolism , Cell Line , Cell Line, Tumor , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fanconi Anemia Complementation Group Proteins/antagonists & inhibitors , Fanconi Anemia Complementation Group Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Intranuclear Inclusion Bodies/genetics , Intranuclear Inclusion Bodies/metabolism , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , Poly-ADP-Ribose Binding Proteins/metabolism , Protein Inhibitors of Activated STAT/antagonists & inhibitors , Protein Inhibitors of Activated STAT/metabolism , RNA Helicases/antagonists & inhibitors , RNA Helicases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rad52 DNA Repair and Recombination Protein/genetics , Rad52 DNA Repair and Recombination Protein/metabolism , RecQ Helicases/genetics , RecQ Helicases/metabolism , Signal Transduction , Sumoylation , Telomere/metabolism , Telomeric Repeat Binding Protein 2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL