Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40.718
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 41: 229-254, 2023 04 26.
Article in English | MEDLINE | ID: mdl-36737597

ABSTRACT

Type 2 immunity mediates protective responses to helminths and pathological responses to allergens, but it also has broad roles in the maintenance of tissue integrity, including wound repair. Type 2 cytokines are known to promote fibrosis, an overzealous repair response, but their contribution to healthy wound repair is less well understood. This review discusses the evidence that the canonical type 2 cytokines, IL-4 and IL-13, are integral to the tissue repair process through two main pathways. First, essential for the progression of effective tissue repair, IL-4 and IL-13 suppress the initial inflammatory response to injury. Second, these cytokines regulate how the extracellular matrix is modified, broken down, and rebuilt for effective repair. IL-4 and/or IL-13 amplifies multiple aspects of the tissue repair response, but many of these pathways are highly redundant and can be induced by other signals. Therefore, the exact contribution of IL-4Rα signaling remains difficult to unravel.


Subject(s)
Interleukin-13 , Interleukin-4 , Animals , Humans , Cytokines/metabolism , Fibrosis , Helminths
2.
Cell ; 187(16): 4289-4304.e26, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-38942015

ABSTRACT

Cellular homeostasis is intricately influenced by stimuli from the microenvironment, including signaling molecules, metabolites, and pathogens. Functioning as a signaling hub within the cell, mitochondria integrate information from various intracellular compartments to regulate cellular signaling and metabolism. Multiple studies have shown that mitochondria may respond to various extracellular signaling events. However, it is less clear how changes in the extracellular matrix (ECM) can impact mitochondrial homeostasis to regulate animal physiology. We find that ECM remodeling alters mitochondrial homeostasis in an evolutionarily conserved manner. Mechanistically, ECM remodeling triggers a TGF-ß response to induce mitochondrial fission and the unfolded protein response of the mitochondria (UPRMT). At the organismal level, ECM remodeling promotes defense of animals against pathogens through enhanced mitochondrial stress responses. We postulate that this ECM-mitochondria crosstalk represents an ancient immune pathway, which detects infection- or mechanical-stress-induced ECM damage, thereby initiating adaptive mitochondria-based immune and metabolic responses.


Subject(s)
Extracellular Matrix , Homeostasis , Mitochondria , Unfolded Protein Response , Extracellular Matrix/metabolism , Animals , Mitochondria/metabolism , Humans , Transforming Growth Factor beta/metabolism , Mitochondrial Dynamics , Mice , Signal Transduction , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/immunology
3.
Cell ; 187(12): 3072-3089.e20, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38781967

ABSTRACT

Tissue folds are structural motifs critical to organ function. In the intestine, bending of a flat epithelium into a periodic pattern of folds gives rise to villi, finger-like protrusions that enable nutrient absorption. However, the molecular and mechanical processes driving villus morphogenesis remain unclear. Here, we identify an active mechanical mechanism that simultaneously patterns and folds the intestinal epithelium to initiate villus formation. At the cellular level, we find that PDGFRA+ subepithelial mesenchymal cells generate myosin II-dependent forces sufficient to produce patterned curvature in neighboring tissue interfaces. This symmetry-breaking process requires altered cell and extracellular matrix interactions that are enabled by matrix metalloproteinase-mediated tissue fluidization. Computational models, together with in vitro and in vivo experiments, revealed that these cellular features manifest at the tissue level as differences in interfacial tensions that promote mesenchymal aggregation and interface bending through a process analogous to the active dewetting of a thin liquid film.


Subject(s)
Extracellular Matrix , Intestinal Mucosa , Animals , Mice , Intestinal Mucosa/metabolism , Intestinal Mucosa/cytology , Extracellular Matrix/metabolism , Myosin Type II/metabolism , Mesoderm/metabolism , Mesoderm/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Morphogenesis , Matrix Metalloproteinases/metabolism
4.
Cell ; 185(11): 1960-1973.e11, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35551765

ABSTRACT

During vertebrate embryogenesis, cell collectives engage in coordinated behavior to form tissue structures of increasing complexity. In the avian skin, assembly into follicles depends on intrinsic mechanical forces of the dermis, but how cell mechanics initiate pattern formation is not known. Here, we reconstitute the initiation of follicle patterning ex vivo using only freshly dissociated avian dermal cells and collagen. We find that contractile cells physically rearrange the extracellular matrix (ECM) and that ECM rearrangement further aligns cells. This exchange transforms a mechanically unlinked collective of dermal cells into a continuum, with coherent, long-range order. Combining theory with experiment, we show that this ordered cell-ECM layer behaves as an active contractile fluid that spontaneously forms regular patterns. Our study illustrates a role for mesenchymal dynamics in generating cell-level ordering and tissue-level patterning through a fluid instability-processes that may be at play across morphological symmetry-breaking contexts.


Subject(s)
Extracellular Matrix , Hair Follicle , Animals , Collagen , Skin , Vertebrates
5.
Annu Rev Cell Dev Biol ; 39: 123-144, 2023 Oct 16.
Article in English | MEDLINE | ID: mdl-37315160

ABSTRACT

Multicellular organisms generate tissues of diverse shapes and functions from cells and extracellular matrices. Their adhesion molecules mediate cell-cell and cell-matrix interactions, which not only play crucial roles in maintaining tissue integrity but also serve as key regulators of tissue morphogenesis. Cells constantly probe their environment to make decisions: They integrate chemical and mechanical information from the environment via diffusible ligand- or adhesion-based signaling to decide whether to release specific signaling molecules or enzymes, to divide or differentiate, to move away or stay, or even whether to live or die. These decisions in turn modify their environment, including the chemical nature and mechanical properties of the extracellular matrix. Tissue morphology is the physical manifestation of the remodeling of cells and matrices by their historical biochemical and biophysical landscapes. We review our understanding of matrix and adhesion molecules in tissue morphogenesis, with an emphasis on key physical interactions that drive morphogenesis.

6.
Cell ; 184(23): 5740-5758.e17, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34735796

ABSTRACT

Biofilms are community architectures adopted by bacteria inclusive of a self-formed extracellular matrix that protects resident bacteria from diverse environmental stresses and, in many species, incorporates extracellular DNA (eDNA) and DNABII proteins for structural integrity throughout biofilm development. Here, we present evidence that this eDNA-based architecture relies on the rare Z-form. Z-form DNA accumulates as biofilms mature and, through stabilization by the DNABII proteins, confers structural integrity to the biofilm matrix. Indeed, substances known to drive B-DNA into Z-DNA promoted biofilm formation whereas those that drive Z-DNA into B-DNA disrupted extant biofilms. Importantly, we demonstrated that the universal bacterial DNABII family of proteins stabilizes both bacterial- and host-eDNA in the Z-form in situ. A model is proposed that incorporates the role of Z-DNA in biofilm pathogenesis, innate immune response, and immune evasion.


Subject(s)
Bacteria/genetics , Biofilms , DNA, Bacterial/chemistry , Extracellular Matrix/metabolism , Extracellular Space/chemistry , Animals , Antibody Specificity , Bacterial Proteins/metabolism , Cell Line , Chinchilla , DNA, Cruciform , Deoxyribonucleases/metabolism , Extracellular Traps/metabolism , Humans , Tetradecanoylphorbol Acetate/pharmacology
7.
Cell ; 184(14): 3702-3716.e30, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34133940

ABSTRACT

Many embryonic organs undergo epithelial morphogenesis to form tree-like hierarchical structures. However, it remains unclear what drives the budding and branching of stratified epithelia, such as in the embryonic salivary gland and pancreas. Here, we performed live-organ imaging of mouse embryonic salivary glands at single-cell resolution to reveal that budding morphogenesis is driven by expansion and folding of a distinct epithelial surface cell sheet characterized by strong cell-matrix adhesions and weak cell-cell adhesions. Profiling of single-cell transcriptomes of this epithelium revealed spatial patterns of transcription underlying these cell adhesion differences. We then synthetically reconstituted budding morphogenesis by experimentally suppressing E-cadherin expression and inducing basement membrane formation in 3D spheroid cultures of engineered cells, which required ß1-integrin-mediated cell-matrix adhesion for successful budding. Thus, stratified epithelial budding, the key first step of branching morphogenesis, is driven by an overall combination of strong cell-matrix adhesion and weak cell-cell adhesion by peripheral epithelial cells.


Subject(s)
Cell-Matrix Junctions/metabolism , Morphogenesis , Animals , Basement Membrane/metabolism , Cell Adhesion , Cell Division , Cell Movement , Cell Tracking , Embryo, Mammalian/cytology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Integrins/metabolism , Mice , Models, Biological , Salivary Glands/cytology , Salivary Glands/embryology , Salivary Glands/metabolism , Transcriptome/genetics
8.
Annu Rev Biochem ; 89: 695-715, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32569527

ABSTRACT

The zona pellucida (ZP) is an extracellular matrix that surrounds all mammalian oocytes, eggs, and early embryos and plays vital roles during oogenesis, fertilization, and preimplantation development. The ZP is composed of three or four glycosylated proteins, ZP1-4, that are synthesized, processed, secreted, and assembled into long, cross-linked fibrils by growing oocytes. ZP proteins have an immunoglobulin-like three-dimensional structure and a ZP domain that consists of two subdomains, ZP-N and ZP-C, with ZP-N of ZP2 and ZP3 required for fibril assembly. A ZP2-ZP3 dimer is located periodically along ZP fibrils that are cross-linked by ZP1, a protein with a proline-rich N terminus. Fibrils in the inner and outer regions of the ZP are oriented perpendicular and parallel to the oolemma, respectively, giving the ZP a multilayered appearance. Upon fertilization of eggs, modification of ZP2 and ZP3 results in changes in the ZP's physical and biological properties that have important consequences. Certain structural features of ZP proteins suggest that they may be amyloid-like proteins.


Subject(s)
Amyloidogenic Proteins/chemistry , Zona Pellucida Glycoproteins/chemistry , Zygote/metabolism , Amyloidogenic Proteins/genetics , Amyloidogenic Proteins/metabolism , Animals , Embryo, Mammalian/metabolism , Embryo, Mammalian/ultrastructure , Female , Gene Expression Regulation, Developmental , Humans , Oocytes/growth & development , Oocytes/metabolism , Oocytes/ultrastructure , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Zona Pellucida/metabolism , Zona Pellucida/ultrastructure , Zona Pellucida Glycoproteins/genetics , Zona Pellucida Glycoproteins/metabolism , Zygote/growth & development , Zygote/ultrastructure
9.
Cell ; 182(2): 388-403.e15, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32615087

ABSTRACT

Synapse remodeling is essential to encode experiences into neuronal circuits. Here, we define a molecular interaction between neurons and microglia that drives experience-dependent synapse remodeling in the hippocampus. We find that the cytokine interleukin-33 (IL-33) is expressed by adult hippocampal neurons in an experience-dependent manner and defines a neuronal subset primed for synaptic plasticity. Loss of neuronal IL-33 or the microglial IL-33 receptor leads to impaired spine plasticity, reduced newborn neuron integration, and diminished precision of remote fear memories. Memory precision and neuronal IL-33 are decreased in aged mice, and IL-33 gain of function mitigates age-related decreases in spine plasticity. We find that neuronal IL-33 instructs microglial engulfment of the extracellular matrix (ECM) and that its loss leads to impaired ECM engulfment and a concomitant accumulation of ECM proteins in contact with synapses. These data define a cellular mechanism through which microglia regulate experience-dependent synapse remodeling and promote memory consolidation.


Subject(s)
Extracellular Matrix/metabolism , Microglia/physiology , Neuronal Plasticity/physiology , Aging , Animals , Fear , Gene Expression Regulation , Hippocampus/metabolism , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/genetics , Interleukin-33/metabolism , Memory , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Signal Transduction
10.
Cell ; 182(4): 919-932.e19, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32763156

ABSTRACT

Redox cycling of extracellular electron shuttles can enable the metabolic activity of subpopulations within multicellular bacterial biofilms that lack direct access to electron acceptors or donors. How these shuttles catalyze extracellular electron transfer (EET) within biofilms without being lost to the environment has been a long-standing question. Here, we show that phenazines mediate efficient EET through interactions with extracellular DNA (eDNA) in Pseudomonas aeruginosa biofilms. Retention of pyocyanin (PYO) and phenazine carboxamide in the biofilm matrix is facilitated by eDNA binding. In vitro, different phenazines can exchange electrons in the presence or absence of DNA and can participate directly in redox reactions through DNA. In vivo, biofilm eDNA can also support rapid electron transfer between redox active intercalators. Together, these results establish that PYO:eDNA interactions support an efficient redox cycle with rapid EET that is faster than the rate of PYO loss from the biofilm.


Subject(s)
Biofilms/growth & development , DNA/chemistry , Pseudomonas aeruginosa/physiology , Pyocyanine/chemistry , DNA/metabolism , Electrochemical Techniques , Electrodes , Electron Transport/drug effects , Fluorescent Dyes/chemistry , Hydrogen-Ion Concentration , Oxidation-Reduction , Phenazines/chemistry , Phenazines/metabolism , Phenazines/pharmacology , Pyocyanine/metabolism
11.
Cell ; 177(4): 806-819, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31051105

ABSTRACT

Over the last several decades, an impressive array of advanced microscopic and analytical tools, such as single-particle tracking and nanoscopic fluorescence correlation spectroscopy, has been applied to characterize the lateral organization and mobility of components in the plasma membrane. Such analysis can tell researchers about the local dynamic composition and structure of membranes and is important for predicting the outcome of membrane-based reactions. However, owing to the unresolved complexity of the membrane and the structures peripheral to it, identification of the detailed molecular origin of the interactions that regulate the organization and mobility of the membrane has not proceeded quickly. This Perspective presents an overview of how cell-surface structure may give rise to the types of lateral mobility that are observed and some potentially fruitful future directions to elucidate the architecture of these structures in more molecular detail.


Subject(s)
Cell Membrane/metabolism , Membrane Microdomains/metabolism , Membrane Proteins/metabolism , Cell Membrane/physiology , Lipid Bilayers/chemistry , Membrane Lipids/metabolism , Membrane Microdomains/chemistry , Membrane Proteins/physiology
12.
Cell ; 179(6): 1306-1318.e18, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31761535

ABSTRACT

Cells have evolved complex mechanisms to maintain protein homeostasis, such as the UPRER, which are strongly associated with several diseases and the aging process. We performed a whole-genome CRISPR-based knockout (KO) screen to identify genes important for cells to survive ER-based protein misfolding stress. We identified the cell-surface hyaluronidase (HAase), Transmembrane Protein 2 (TMEM2), as a potent modulator of ER stress resistance. The breakdown of the glycosaminoglycan, hyaluronan (HA), by TMEM2 within the extracellular matrix (ECM) altered ER stress resistance independent of canonical UPRER pathways but dependent upon the cell-surface receptor, CD44, a putative HA receptor, and the MAPK cell-signaling components, ERK and p38. Last, and most surprisingly, ectopic expression of human TMEM2 in C. elegans protected animals from ER stress and increased both longevity and pathogen resistance independent of canonical UPRER activation but dependent on the ERK ortholog mpk-1 and the p38 ortholog pmk-1.


Subject(s)
Caenorhabditis elegans/physiology , Endoplasmic Reticulum/metabolism , Hyaluronoglucosaminidase/metabolism , Longevity/physiology , Membrane Proteins/metabolism , Unfolded Protein Response , Animals , Caenorhabditis elegans/immunology , Cell Line , Cell Proliferation , Disease Resistance , Endoplasmic Reticulum Stress , Fibroblasts/metabolism , Humans , Immunity, Innate , Models, Biological , Molecular Weight , Signal Transduction
13.
Cell ; 176(1-2): 113-126.e15, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30633902

ABSTRACT

Here, we describe a novel pathogenic entity, the activated PMN (polymorphonuclear leukocyte, i.e., neutrophil)-derived exosome. These CD63+/CD66b+ nanovesicles acquire surface-bound neutrophil elastase (NE) during PMN degranulation, NE being oriented in a configuration resistant to α1-antitrypsin (α1AT). These exosomes bind and degrade extracellular matrix (ECM) via the integrin Mac-1 and NE, respectively, causing the hallmarks of chronic obstructive pulmonary disease (COPD). Due to both ECM targeting and α1AT resistance, exosomal NE is far more potent than free NE. Importantly, such PMN-derived exosomes exist in clinical specimens from subjects with COPD but not healthy controls and are capable of transferring a COPD-like phenotype from humans to mice in an NE-driven manner. Similar findings were observed for another neutrophil-driven disease of ECM remodeling (bronchopulmonary dysplasia [BPD]). These findings reveal an unappreciated role for exosomes in the pathogenesis of disorders of ECM homeostasis such as COPD and BPD, providing a critical mechanism for proteolytic damage.


Subject(s)
Exosomes/physiology , Neutrophils/metabolism , Animals , Bronchoalveolar Lavage Fluid/cytology , Cells, Cultured , Extracellular Matrix/metabolism , Female , Humans , Inflammation , Integrins , Leukocyte Elastase/metabolism , Lung/metabolism , Lung/physiopathology , Male , Mice , Mice, Inbred C57BL , Neutrophils/physiology , Pulmonary Disease, Chronic Obstructive/physiopathology , alpha 1-Antitrypsin/metabolism
14.
Cell ; 175(1): 117-132.e21, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30197082

ABSTRACT

The metabolic state of a cell is influenced by cell-extrinsic factors, including nutrient availability and growth factor signaling. Here, we present extracellular matrix (ECM) remodeling as another fundamental node of cell-extrinsic metabolic regulation. Unbiased analysis of glycolytic drivers identified the hyaluronan-mediated motility receptor as being among the most highly correlated with glycolysis in cancer. Confirming a mechanistic link between the ECM component hyaluronan and metabolism, treatment of cells and xenografts with hyaluronidase triggers a robust increase in glycolysis. This is largely achieved through rapid receptor tyrosine kinase-mediated induction of the mRNA decay factor ZFP36, which targets TXNIP transcripts for degradation. Because TXNIP promotes internalization of the glucose transporter GLUT1, its acute decline enriches GLUT1 at the plasma membrane. Functionally, induction of glycolysis by hyaluronidase is required for concomitant acceleration of cell migration. This interconnection between ECM remodeling and metabolism is exhibited in dynamic tissue states, including tumorigenesis and embryogenesis.


Subject(s)
Carrier Proteins/physiology , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Carbohydrate Metabolism/physiology , Carrier Proteins/metabolism , Cell Line, Tumor , Glucose/metabolism , Glucose Transporter Type 1 , Glycolysis/physiology , Humans , Hyaluronic Acid/physiology , Hyaluronoglucosaminidase/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Signal Transduction , Tristetraprolin/metabolism , Tristetraprolin/physiology
15.
Immunity ; 56(2): 289-306.e7, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36750099

ABSTRACT

Granulomas are lumps of immune cells that can form in various organs. Most granulomas appear unstructured, yet they have some resemblance to lymphoid organs. To better understand granuloma formation, we performed single-cell sequencing and spatial transcriptomics on granulomas from patients with sarcoidosis and bioinformatically reconstructed the underlying gene regulatory networks. We discovered an immune stimulatory environment in granulomas that repurposes transcriptional programs associated with lymphoid organ development. Granuloma formation followed characteristic spatial patterns and involved genes linked to immunometabolism, cytokine and chemokine signaling, and extracellular matrix remodeling. Three cell types emerged as key players in granuloma formation: metabolically reprogrammed macrophages, cytokine-producing Th17.1 cells, and fibroblasts with inflammatory and tissue-remodeling phenotypes. Pharmacological inhibition of one of the identified processes attenuated granuloma formation in a sarcoidosis mouse model. We show that human granulomas adopt characteristic aspects of normal lymphoid organ development in aberrant combinations, indicating that granulomas constitute aberrant lymphoid organs.


Subject(s)
Sarcoidosis , Transcriptome , Animals , Mice , Humans , Cytokines/metabolism , Granuloma , Gene Expression Profiling
16.
Immunity ; 56(7): 1561-1577.e9, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37402364

ABSTRACT

Hypodermis is the predominant site of Staphylococcus aureus infections that cause cellulitis. Given the importance of macrophages in tissue remodeling, we examined the hypodermal macrophages (HDMs) and their impact on host susceptibility to infection. Bulk and single-cell transcriptomics uncovered HDM subsets with CCR2-dichotomy. HDM homeostasis required the fibroblast-derived growth factor CSF1, ablation of which abrogated HDMs from the hypodermal adventitia. Loss of CCR2- HDMs resulted in accumulation of the extracellular matrix component, hyaluronic acid (HA). HDM-mediated HA clearance required sensing by the HA receptor, LYVE-1. Cell-autonomous IGF1 was required for accessibility of AP-1 transcription factor motifs that controlled LYVE-1 expression. Remarkably, loss of HDMs or IGF1 limited Staphylococcus aureus expansion via HA and conferred protection against cellulitis. Our findings reveal a function for macrophages in the regulation of HA with an impact on infection outcomes, which may be harnessed to limit the establishment of infection in the hypodermal niche.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Humans , Staphylococcus aureus/physiology , Cellulitis/metabolism , Macrophages/metabolism , Extracellular Matrix
17.
Immunity ; 55(7): 1250-1267.e12, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35709757

ABSTRACT

The intestine harbors a large population of resident eosinophils, yet the function of intestinal eosinophils has not been explored. Flow cytometry and whole-mount imaging identified eosinophils residing in the lamina propria along the length of the intestine prior to postnatal microbial colonization. Microscopy, transcriptomic analysis, and mass spectrometry of intestinal tissue revealed villus blunting, altered extracellular matrix, decreased epithelial cell turnover, increased gastrointestinal motility, and decreased lipid absorption in eosinophil-deficient mice. Mechanistically, intestinal epithelial cells released IL-33 in a microbiota-dependent manner, which led to eosinophil activation. The colonization of germ-free mice demonstrated that eosinophil activation in response to microbes regulated villous size alterations, macrophage maturation, epithelial barrier integrity, and intestinal transit. Collectively, our findings demonstrate a critical role for eosinophils in facilitating the mutualistic interactions between the host and microbiota and provide a rationale for the functional significance of their early life recruitment in the small intestine.


Subject(s)
Communicable Diseases , Microbiota , Animals , Eosinophils , Homeostasis , Intestinal Mucosa , Intestine, Small , Mice
18.
Mol Cell ; 82(2): 447-462.e6, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34856123

ABSTRACT

Quantitative subcellular metabolomic measurements can explain the roles of metabolites in cellular processes but are subject to multiple confounding factors. We developed stable isotope labeling of essential nutrients in cell culture-subcellular fractionation (SILEC-SF), which uses isotope-labeled internal standard controls that are present throughout fractionation and processing to quantify acyl-coenzyme A (acyl-CoA) thioesters in subcellular compartments by liquid chromatography-mass spectrometry. We tested SILEC-SF in a range of sample types and examined the compartmentalized responses to oxygen tension, cellular differentiation, and nutrient availability. Application of SILEC-SF to the challenging analysis of the nuclear compartment revealed a nuclear acyl-CoA profile distinct from that of the cytosol, with notable nuclear enrichment of propionyl-CoA. Using isotope tracing, we identified the branched chain amino acid isoleucine as a major metabolic source of nuclear propionyl-CoA and histone propionylation, thus revealing a new mechanism of crosstalk between metabolism and the epigenome.


Subject(s)
Acyl Coenzyme A/metabolism , Cell Compartmentation , Cell Nucleus/metabolism , Energy Metabolism , Histones/metabolism , Metabolomics , Protein Processing, Post-Translational , Animals , Cell Differentiation , Chromatography, Liquid , Cytosol/metabolism , Epigenesis, Genetic , Hep G2 Cells , Humans , Isoleucine , Metabolome , Mice , Mitochondria/metabolism , Oxygen/metabolism , Spectrometry, Mass, Electrospray Ionization
19.
Physiol Rev ; 102(2): 1025-1151, 2022 04 01.
Article in English | MEDLINE | ID: mdl-33949874

ABSTRACT

The brain harbors a unique ability to, figuratively speaking, shift its gears. During wakefulness, the brain is geared fully toward processing information and behaving, while homeostatic functions predominate during sleep. The blood-brain barrier establishes a stable environment that is optimal for neuronal function, yet the barrier imposes a physiological problem; transcapillary filtration that forms extracellular fluid in other organs is reduced to a minimum in brain. Consequently, the brain depends on a special fluid [the cerebrospinal fluid (CSF)] that is flushed into brain along the unique perivascular spaces created by astrocytic vascular endfeet. We describe this pathway, coined the term glymphatic system, based on its dependency on astrocytic vascular endfeet and their adluminal expression of aquaporin-4 water channels facing toward CSF-filled perivascular spaces. Glymphatic clearance of potentially harmful metabolic or protein waste products, such as amyloid-ß, is primarily active during sleep, when its physiological drivers, the cardiac cycle, respiration, and slow vasomotion, together efficiently propel CSF inflow along periarterial spaces. The brain's extracellular space contains an abundance of proteoglycans and hyaluronan, which provide a low-resistance hydraulic conduit that rapidly can expand and shrink during the sleep-wake cycle. We describe this unique fluid system of the brain, which meets the brain's requisites to maintain homeostasis similar to peripheral organs, considering the blood-brain-barrier and the paths for formation and egress of the CSF.


Subject(s)
Glymphatic System , Amyloid beta-Peptides/metabolism , Biological Transport , Blood-Brain Barrier , Brain/metabolism , Cerebrospinal Fluid/metabolism , Glymphatic System/metabolism , Humans
20.
Mol Cell ; 81(17): 3509-3525.e5, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34320406

ABSTRACT

Nuclear chromosomes transcribe far more RNA than required to encode protein. Here we investigate whether non-coding RNA broadly contributes to cytological-scale chromosome territory architecture. We develop a procedure that depletes soluble proteins, chromatin, and most nuclear RNA from the nucleus but does not delocalize XIST, a known architectural RNA, from an insoluble chromosome "scaffold." RNA-seq analysis reveals that most RNA in the nuclear scaffold is repeat-rich, non-coding, and derived predominantly from introns of nascent transcripts. Insoluble, repeat-rich (C0T-1) RNA co-distributes with known scaffold proteins including scaffold attachment factor A (SAF-A), and distribution of these components inversely correlates with chromatin compaction in normal and experimentally manipulated nuclei. We further show that RNA is required for SAF-A to interact with chromatin and for enrichment of structurally embedded "scaffold attachment regions" prevalent in euchromatin. Collectively, the results indicate that long nascent transcripts contribute a dynamic structural role that promotes the open architecture of active chromosome territories.


Subject(s)
Chromatin/metabolism , Nuclear Matrix/metabolism , RNA, Untranslated/metabolism , Animals , Cell Line , Cell Nucleus/physiology , Chromatin/genetics , Chromosomes/genetics , Chromosomes/metabolism , Euchromatin/metabolism , Humans , Mice , Nuclear Matrix/genetics , RNA/genetics , RNA/metabolism , RNA, Long Noncoding/genetics , RNA, Untranslated/genetics , Transcription, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL