Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
Nat Commun ; 11(1): 5312, 2020 10 20.
Article En | MEDLINE | ID: mdl-33082319

Evidence is lacking as to how developing neurons integrate mitogenic signals with microenvironment cues to control proliferation and differentiation. We determine that the Siah2 E3 ubiquitin ligase functions in a coincidence detection circuit linking responses to the Shh mitogen and the extracellular matrix to control cerebellar granule neurons (CGN) GZ occupancy. We show that Shh signaling maintains Siah2 expression in CGN progenitors (GNPs) in a Ras/Mapk-dependent manner. Siah2 supports ciliogenesis in a feed-forward fashion by restraining cilium disassembly. Efforts to identify sources of the Ras/Mapk signaling led us to discover that GNPs respond to laminin, but not vitronectin, in the GZ microenvironment via integrin ß1 receptors, which engages the Ras/Mapk cascade with Shh, and that this niche interaction is essential for promoting GNP ciliogenesis. As GNPs leave the GZ, differentiation is driven by changing extracellular cues that diminish Siah2-activity leading to primary cilia shortening and attenuation of the mitogenic response.


Cilia/metabolism , Extracellular Matrix/metabolism , Neurons/cytology , Nuclear Proteins/metabolism , Stem Cells/cytology , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Cerebellum/cytology , Cerebellum/metabolism , Cilia/genetics , Extracellular Matrix/genetics , Humans , Mice , Mice, Inbred C57BL , Neurons/metabolism , Nuclear Proteins/genetics , Signal Transduction , Stem Cells/metabolism , Ubiquitin-Protein Ligases/genetics
2.
PLoS Pathog ; 16(3): e1008364, 2020 03.
Article En | MEDLINE | ID: mdl-32150572

Innate immunity responds to pathogens by producing alarm signals and activating pathways that make host cells inhospitable for pathogen replication. The intracellular bacterium Burkholderia thailandensis invades the cytosol, hijacks host actin, and induces cell fusion to spread to adjacent cells, forming multinucleated giant cells (MNGCs) which promote bacterial replication. We show that type I interferon (IFN) restricts macrophage MNGC formation during B. thailandensis infection. Guanylate-binding proteins (GBPs) expressed downstream of type I IFN were required to restrict MNGC formation through inhibition of bacterial Arp2/3-dependent actin motility during infection. GTPase activity and the CAAX prenylation domain were required for GBP2 recruitment to B. thailandensis, which restricted bacterial actin polymerization required for MNGC formation. Consistent with the effects in in vitro macrophages, Gbp2-/-, Gbp5-/-, GbpChr3-KO mice were more susceptible to intranasal infection with B. thailandensis than wildtype mice. Our findings reveal that IFN and GBPs play a critical role in restricting cell-cell fusion and bacteria-induced pathology during infection.


Burkholderia Infections/immunology , Burkholderia/immunology , GTP-Binding Proteins/immunology , Giant Cells/immunology , Macrophages/immunology , Nose Diseases/immunology , Protein Prenylation/immunology , Animals , Burkholderia Infections/genetics , Burkholderia Infections/pathology , Cell Fusion , GTP-Binding Proteins/genetics , Giant Cells/microbiology , Giant Cells/pathology , Interferon Type I/genetics , Interferon Type I/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Nose Diseases/genetics , Nose Diseases/microbiology , Nose Diseases/pathology
3.
Neuron ; 104(3): 512-528.e11, 2019 11 06.
Article En | MEDLINE | ID: mdl-31493975

More than 8,000 genes are turned on or off as progenitor cells produce the 7 classes of retinal cell types during development. Thousands of enhancers are also active in the developing retinae, many having features of cell- and developmental stage-specific activity. We studied dynamic changes in the 3D chromatin landscape important for precisely orchestrated changes in gene expression during retinal development by ultra-deep in situ Hi-C analysis on murine retinae. We identified developmental-stage-specific changes in chromatin compartments and enhancer-promoter interactions. We developed a machine learning-based algorithm to map euchromatin and heterochromatin domains genome-wide and overlaid it with chromatin compartments identified by Hi-C. Single-cell ATAC-seq and RNA-seq were integrated with our Hi-C and previous ChIP-seq data to identify cell- and developmental-stage-specific super-enhancers (SEs). We identified a bipolar neuron-specific core regulatory circuit SE upstream of Vsx2, whose deletion in mice led to the loss of bipolar neurons.


Euchromatin/metabolism , Gene Expression Regulation, Developmental/physiology , Heterochromatin/metabolism , Retina/embryology , Retinal Bipolar Cells/metabolism , Animals , Chromatin/metabolism , Chromatin Immunoprecipitation Sequencing , Enhancer Elements, Genetic , Gene Regulatory Networks , Homeodomain Proteins/genetics , Machine Learning , Mice , Nuclear Lamina/metabolism , Promoter Regions, Genetic , RNA-Seq , Receptors, Cytoplasmic and Nuclear/genetics , Retina/cytology , Retina/metabolism , Retina/ultrastructure , Retinal Bipolar Cells/cytology , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/metabolism , Single-Cell Analysis , Transcription Factors/genetics , Lamin B Receptor
4.
Cell Rep ; 28(9): 2275-2287.e5, 2019 08 27.
Article En | MEDLINE | ID: mdl-31461645

Necroptosis is a form of programmed cell death that is defined by activation of the kinase RIPK3 and subsequent cell membrane permeabilization by the effector MLKL. RIPK3 activation can also promote immune responses via production of cytokines and chemokines. How active cytokine production is coordinated with the terminal process of necroptosis is unclear. Here, we report that cytokine production continues within necroptotic cells even after they have lost cell membrane integrity and irreversibly committed to death. This continued cytokine production is dependent on mRNA translation and requires maintenance of endoplasmic reticulum integrity that remains after plasma membrane integrity is lost. The continued translation of cytokines by cellular corpses contributes to necroptotic cell uptake by innate immune cells and priming of adaptive immune responses to antigens associated with necroptotic corpses. These findings imply that cell death and production of inflammatory mediators are coordinated to optimize the immunogenicity of necroptotic cells.


Cell Membrane/metabolism , Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , 3T3 Cells , Animals , Endoplasmic Reticulum/metabolism , Female , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
6.
Exp Cell Res ; 379(1): 55-64, 2019 06 01.
Article En | MEDLINE | ID: mdl-30922922

Metabolic studies of human pluripotent stem cells (hPSCs) have focused on how the cells produce energy through the catabolic pathway. The less-studied anabolic pathway, by which hPSCs expend energy in the form of adenosine triphosphate (ATP), is not yet fully understood. Compared to fully differentiated somatic cells, hPSCs undergo significant changes not only in their gene expression but also in their production and/or expenditure of ATP. Here, we investigate how hPSCs tightly control their energy homeostasis by studying the main energy-consuming process, mRNA translation. In addition, change of subcellular organelles regarding energy homeostasis has been investigated. Lysosomes are organelles that play an important role in the elimination of unnecessary cellular materials by digestion and in the recycling system of the cell. We have found that hPSCs control their lysosome numbers in part by regulating lysosomal gene/protein expression. Thus, because the levels of mRNA translation rate are lower in hPSCs than in somatic cells, not only the global translational machinery but also the lysosomal recycling machinery is suppressed in hPSCs. Overall, the results of our study suggest that hPSCs reprogram gene expression and signaling to regulate energy-consuming processes and energy-controlling organelles.


Energy Metabolism/physiology , Organelles/metabolism , Pluripotent Stem Cells/metabolism , Adenosine Triphosphate/metabolism , Cell Differentiation/physiology , Cells, Cultured , Gene Expression/physiology , Homeostasis/physiology , Humans , Lysosomes/metabolism , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , Signal Transduction/physiology
7.
Dis Model Mech ; 11(5)2018 05 10.
Article En | MEDLINE | ID: mdl-29666155

Mutations in SIL1, a cofactor for the endoplasmic reticulum (ER)-localized Hsp70 chaperone, BiP, cause Marinesco-Sjögren syndrome (MSS), an autosomal recessive disorder. Using a mouse model, we characterized molecular aspects of the progressive myopathy associated with MSS. Proteomic profiling of quadriceps at the onset of myopathy revealed that SIL1 deficiency affected multiple pathways critical to muscle physiology. We observed an increase in ER chaperones prior to the onset of muscle weakness, which was complemented by upregulation of multiple components of cellular protein degradation pathways. These responses were inadequate to maintain normal expression of secretory pathway proteins, including insulin and IGF-1 receptors. There was a paradoxical enhancement of downstream PI3K-AKT-mTOR signaling and glucose uptake in SIL1-disrupted skeletal muscles, all of which were insufficient to maintain skeletal muscle mass. Together, these data reveal a disruption in ER homeostasis upon SIL1 loss, which is countered by multiple compensatory responses that are ultimately unsuccessful, leading to trans-organellar proteostasis collapse and myopathy.


Endoplasmic Reticulum/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Heat-Shock Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Proteostasis , Aging/pathology , Animals , Disease Progression , Endoplasmic Reticulum Chaperone BiP , Insulin/metabolism , Male , Mice , Models, Biological , Muscle Strength , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Muscular Diseases/metabolism , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Proteome/metabolism , Receptor, Insulin/metabolism , Signal Transduction
8.
Cell Rep ; 22(10): 2601-2614, 2018 03 06.
Article En | MEDLINE | ID: mdl-29514090

Diverse cell types can be reprogrammed into pluripotent stem cells by ectopic expression of Oct4 (Pou5f1), Klf4, Sox3, and Myc. Many of these induced pluripotent stem cells (iPSCs) retain memory, in terms of DNA methylation and histone modifications (epigenetic memory), of their cellular origins, and this may bias subsequent differentiation. Neurons are difficult to reprogram, and there has not been a systematic side-by-side characterization of reprogramming efficiency or epigenetic memory across different neuronal subtypes. Here, we compare reprogramming efficiency of five different retinal cell types at two different stages of development. Retinal differentiation from each iPSC line was measured using a quantitative standardized scoring system called STEM-RET and compared to the epigenetic memory. Neurons with the lowest reprogramming efficiency produced iPSC lines with the best retinal differentiation and were more likely to retain epigenetic memory of their cellular origins. In addition, we identified biomarkers of iPSCs that are predictive of retinal differentiation.


Cellular Reprogramming , DNA Methylation , Histones/metabolism , Organogenesis , Organoids/growth & development , Protein Processing, Post-Translational , Retina/cytology , Retina/metabolism , Animals , Biomarkers/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Nucleus/metabolism , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Mice , Promoter Regions, Genetic/genetics
9.
Autophagy ; 14(5): 796-811, 2018.
Article En | MEDLINE | ID: mdl-29099309

Mammalian ULK1 (unc-51 like kinase 1) and ULK2, Caenorhabditis elegans UNC-51, and Drosophila melanogaster Atg1 are serine/threonine kinases that regulate flux through the autophagy pathway in response to various types of cellular stress. C. elegans UNC-51 and D. melanogaster Atg1 also promote axonal growth and defasciculation; disruption of these genes results in defective axon guidance in invertebrates. Although disrupting ULK1/2 function impairs normal neurite outgrowth in vitro, the role of ULK1 and ULK2 in the developing brain remains poorly characterized. Here, we show that ULK1 and ULK2 are required for proper projection of axons in the forebrain. Mice lacking Ulk1 and Ulk2 in their central nervous systems showed defects in axonal pathfinding and defasciculation affecting the corpus callosum, anterior commissure, corticothalamic axons and thalamocortical axons. These defects impaired the midline crossing of callosal axons and caused hypoplasia of the anterior commissure and disorganization of the somatosensory cortex. The axon guidance defects observed in ulk1/2 double-knockout mice and central nervous system-specific (Nes-Cre) Ulk1/2-conditional double-knockout mice were not recapitulated in mice lacking other autophagy genes (i.e., Atg7 or Rb1cc1 [RB1-inducible coiled-coil 1]). The brains of Ulk1/2-deficient mice did not show stem cell defects previously attributed to defective autophagy in ambra1 (autophagy/Beclin 1 regulator 1)- and Rb1cc1-deficient mice or accumulation of SQSTM1 (sequestosome 1)+ or ubiquitin+ deposits. Together, these data demonstrate that ULK1 and ULK2 regulate axon guidance during mammalian brain development via a noncanonical (i.e., autophagy-independent) pathway.


Autophagy-Related Protein-1 Homolog/metabolism , Autophagy , Axon Guidance , Prosencephalon/embryology , Prosencephalon/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Animals, Newborn , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Autophagy-Related Protein 7/metabolism , Autophagy-Related Protein-1 Homolog/deficiency , Autophagy-Related Proteins , Axons/metabolism , Axons/ultrastructure , Hippocampus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/metabolism , Protein Serine-Threonine Kinases/deficiency , Somatosensory Cortex/metabolism , Ubiquitinated Proteins/metabolism
10.
Neuron ; 94(3): 550-568.e10, 2017 May 03.
Article En | MEDLINE | ID: mdl-28472656

In the developing retina, multipotent neural progenitors undergo unidirectional differentiation in a precise spatiotemporal order. Here we profile the epigenetic and transcriptional changes that occur during retinogenesis in mice and humans. Although some progenitor genes and cell cycle genes were epigenetically silenced during retinogenesis, the most dramatic change was derepression of cell-type-specific differentiation programs. We identified developmental-stage-specific super-enhancers and showed that most epigenetic changes are conserved in humans and mice. To determine how the epigenome changes during tumorigenesis and reprogramming, we performed integrated epigenetic analysis of murine and human retinoblastomas and induced pluripotent stem cells (iPSCs) derived from murine rod photoreceptors. The retinoblastoma epigenome mapped to the developmental stage when retinal progenitors switch from neurogenic to terminal patterns of cell division. The epigenome of retinoblastomas was more similar to that of the normal retina than that of retina-derived iPSCs, and we identified retina-specific epigenetic memory.


Carcinogenesis/genetics , Cell Differentiation/genetics , Cellular Reprogramming/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Histone Code/genetics , Retina/metabolism , Retinoblastoma/genetics , Animals , Animals, Genetically Modified , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Retina/embryology , Retinal Rod Photoreceptor Cells/cytology , Retinoblastoma Protein/genetics
11.
Mol Genet Metab ; 120(4): 350-362, 2017 04.
Article En | MEDLINE | ID: mdl-28189602

Coenzyme A (CoA) is a cofactor that is central to energy metabolism and CoA synthesis is controlled by the enzyme pantothenate kinase (PanK). A transgenic mouse strain expressing human PANK2 was derived to determine the physiological impact of PANK overexpression and elevated CoA levels. The Tg(PANK2) mice expressed high levels of the transgene in skeletal muscle and heart; however, CoA was substantially elevated only in skeletal muscle, possibly associated with the comparatively low endogenous levels of acetyl-CoA, a potent feedback inhibitor of PANK2. Tg(PANK2) mice were smaller, had less skeletal muscle mass and displayed significantly impaired exercise tolerance and grip strength. Skeletal myofibers were characterized by centralized nuclei and aberrant mitochondria. Both the content of fully assembled complex I of the electron transport chain and ATP levels were reduced, while markers of oxidative stress were elevated in Tg(PANK2) skeletal muscle. These abnormalities were not detected in the Tg(PANK2) heart muscle, with the exception of spotty loss of cristae organization in the mitochondria. The data demonstrate that excessively high CoA may be detrimental to skeletal muscle function.


Coenzyme A/metabolism , Hand Strength/physiology , Mitochondria/metabolism , Muscle, Skeletal/physiology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Animals , Electron Transport Complex I/metabolism , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Myocardium/metabolism , Oxidative Stress , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Up-Regulation
12.
Cell ; 167(2): 382-396.e17, 2016 Oct 06.
Article En | MEDLINE | ID: mdl-27693356

The inflammasome is an intracellular signaling complex, which on recognition of pathogens and physiological aberration, drives activation of caspase-1, pyroptosis, and the release of the pro-inflammatory cytokines IL-1ß and IL-18. Bacterial ligands must secure entry into the cytoplasm to activate inflammasomes; however, the mechanisms by which concealed ligands are liberated in the cytoplasm have remained unclear. Here, we showed that the interferon-inducible protein IRGB10 is essential for activation of the DNA-sensing AIM2 inflammasome by Francisella novicida and contributed to the activation of the LPS-sensing caspase-11 and NLRP3 inflammasome by Gram-negative bacteria. IRGB10 directly targeted cytoplasmic bacteria through a mechanism requiring guanylate-binding proteins. Localization of IRGB10 to the bacterial cell membrane compromised bacterial structural integrity and mediated cytosolic release of ligands for recognition by inflammasome sensors. Overall, our results reveal IRGB10 as part of a conserved signaling hub at the interface between cell-autonomous immunity and innate immune sensing pathways.


DNA-Binding Proteins/metabolism , Francisella/immunology , GTP Phosphohydrolases/metabolism , Gram-Negative Bacterial Infections/immunology , Host-Pathogen Interactions/immunology , Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , B-Lymphocytes/immunology , Caspases/metabolism , Caspases, Initiator , Cytosol/immunology , Cytosol/microbiology , GTP Phosphohydrolases/genetics , Gram-Negative Bacterial Infections/microbiology , Immunity, Cellular , Immunity, Innate , Inflammasomes/metabolism , Ligands , Mice , Mice, Mutant Strains , Myeloid Cells/immunology , T-Lymphocytes/immunology
13.
Sci Rep ; 6: 30757, 2016 08 01.
Article En | MEDLINE | ID: mdl-27476972

A feature in patients with constitutional DNA-mismatch repair deficiency is agenesis of the corpus callosum, the cause of which has not been established. Here we report a previously unrecognized consequence of deficiency in MSH2, a protein known primarily for its function in correcting nucleotide mismatches or insertions and deletions in duplex DNA caused by errors in DNA replication or recombination. We documented that Msh2 deficiency causes dysmyelination of the axonal projections in the corpus callosum. Evoked action potentials in the myelinated corpus callosum projections of Msh2-null mice were smaller than wild-type mice, whereas unmyelinated axons showed no difference. Msh2-null mice were also impaired in locomotive activity and had an abnormal response to heat. These findings reveal a novel pathogenic consequence of MSH2 deficiency, providing a new mechanistic hint to previously recognized neurological disorders in patients with inherited DNA-mismatch repair deficiency.


Corpus Callosum , DNA Mismatch Repair , Demyelinating Diseases , Evoked Potentials , Locomotion , MutS Homolog 2 Protein/deficiency , Animals , Corpus Callosum/metabolism , Corpus Callosum/pathology , Corpus Callosum/physiopathology , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Demyelinating Diseases/physiopathology , Mice , Mice, Knockout , MutS Homolog 2 Protein/metabolism
14.
Nat Commun ; 7: 12353, 2016 08 10.
Article En | MEDLINE | ID: mdl-27507172

Hereditary porphyrias are caused by mutations in genes that encode haem biosynthetic enzymes with resultant buildup of cytotoxic metabolic porphyrin intermediates. A long-standing open question is why the same causal porphyria mutations exhibit widely variable penetrance and expressivity in different individuals. Here we show that severely affected porphyria patients harbour variant alleles in the ABCB6 gene, also known as Lan, which encodes an ATP-binding cassette (ABC) transporter. Plasma membrane ABCB6 exports a variety of disease-related porphyrins. Functional studies show that most of these ABCB6 variants are expressed poorly and/or have impaired function. Accordingly, homozygous disruption of the Abcb6 gene in mice exacerbates porphyria phenotypes in the Fech(m1Pas) mouse model, as evidenced by increased porphyrin accumulation, and marked liver injury. Collectively, these studies support ABCB6 role as a genetic modifier of porphyria and suggest that porphyrin-inducing drugs may produce excessive toxicities in individuals with the rare Lan(-) blood type.


ATP-Binding Cassette Transporters/genetics , Isoantigens/genetics , Porphyrias/genetics , Porphyrins/metabolism , ATP-Binding Cassette Transporters/metabolism , Alleles , Animals , Biological Transport/genetics , Cell Membrane/metabolism , Cohort Studies , Disease Models, Animal , Female , Heme/biosynthesis , Heme/metabolism , Humans , Isoantigens/blood , Isoantigens/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mutation , Porphyrias/metabolism , Porphyrias/urine , Porphyrins/urine , Sequence Homology, Amino Acid , Severity of Illness Index , Exome Sequencing
15.
Sci Rep ; 6: 27814, 2016 06 13.
Article En | MEDLINE | ID: mdl-27291827

Food poisoning is one of the leading causes of morbidity and mortality in the world. Citrobacter rodentium is an enteric pathogen which attaches itself to enterocytes and induces attachment and effacing (A/E) lesions. The ability of the bacterium to cause infection requires subversion of the host actin cytoskeleton. Rac-dependent actin polymerization is activated by a guanine nucleotide exchange factor known as Dedicator of cytokinesis 2 (DOCK2). However, the role of DOCK2 in infectious disease is largely unexplored. Here, we found that mice lacking DOCK2 were susceptible to C. rodentium infection. These mice harbored increased levels of C. rodentium bacteria, showed more pronounced weight loss and inflammation-associated pathology, and were prone to bacterial dissemination to the systemic organs compared with wild-type mice. We found that mice lacking DOCK2 were more susceptible to C. rodentium attachment to intestinal epithelial cells. Therefore, our results underscored an important role of DOCK2 for gastrointestinal immunity to C. rodentium infection.


Citrobacter rodentium/physiology , Disease Resistance/immunology , Enterobacteriaceae Infections/immunology , GTPase-Activating Proteins/genetics , Intestinal Diseases/immunology , Animals , Antimicrobial Cationic Peptides/metabolism , Colon/immunology , Colon/metabolism , Colon/pathology , Cytokines/analysis , Cytokines/metabolism , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/mortality , Enterobacteriaceae Infections/pathology , Enzyme-Linked Immunosorbent Assay , GTPase-Activating Proteins/deficiency , Guanine Nucleotide Exchange Factors , Intestinal Diseases/microbiology , Intestinal Diseases/mortality , Intestinal Diseases/pathology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Neutrophil Infiltration , Neutrophils/cytology , Neutrophils/immunology
16.
Nat Commun ; 7: 11876, 2016 06 23.
Article En | MEDLINE | ID: mdl-27336173

Maintenance of epithelial cell polarity and epithelial barrier relies on the spatial organization of the actin cytoskeleton and proper positioning/assembly of intercellular junctions. However, how these processes are regulated is poorly understood. Here we reveal a key role for the multifunctional protein Alix in both processes. In a knockout mouse model of Alix, we identified overt structural changes in the epithelium of the choroid plexus and in the ependyma, such as asymmetrical cell shape and size, misplacement and abnormal beating of cilia, blebbing of the microvilli. These defects culminate in excessive cell extrusion, enlargement of the lateral ventricles and hydrocephalus. Mechanistically, we find that by interacting with F-actin, the Par complex and ZO-1, Alix ensures the formation and maintenance of the apically restricted actomyosin-tight junction complex. We propose that in this capacity Alix plays a role in the establishment of apical-basal polarity and in the maintenance of the epithelial barrier.


Actomyosin/metabolism , Blood-Brain Barrier , Calcium-Binding Proteins/physiology , Choroid Plexus/metabolism , Tight Junctions/metabolism , Actins/metabolism , Animals , Cell Polarity , Choroid Plexus/ultrastructure , Ependyma/ultrastructure , Epithelial Cells/ultrastructure , Hydrocephalus/etiology , Mice , Mice, Knockout , Zonula Occludens-1 Protein/metabolism
17.
Mol Cell ; 61(4): 589-601, 2016 Feb 18.
Article En | MEDLINE | ID: mdl-26853145

Necroptosis is a cell death pathway regulated by the receptor interacting protein kinase 3 (RIPK3) and the mixed lineage kinase domain-like (MLKL) pseudokinase. How MLKL executes plasma membrane rupture upon phosphorylation by RIPK3 remains controversial. Here, we characterize the hierarchical transduction of structural changes in MLKL that culminate in necroptosis. The MLKL brace, proximal to the N-terminal helix bundle (NB), is involved in oligomerization to facilitate plasma membrane targeting through the low-affinity binding of NB to phosphorylated inositol polar head groups of phosphatidylinositol phosphate (PIP) phospholipids. At the membrane, the NB undergoes a "rolling over" mechanism to expose additional higher-affinity PIP-binding sites responsible for robust association to the membrane and displacement of the brace from the NB. PI(4,5)P2 is the preferred PIP-binding partner. We investigate the specific association of MLKL with PIPs and subsequent structural changes during necroptosis.


Fibroblasts/cytology , Phosphatidylinositol Phosphates/metabolism , Protein Kinases/chemistry , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Binding Sites , Cell Line , Cell Membrane/metabolism , Fibroblasts/metabolism , Humans , Mice , Models, Molecular , Phosphorylation , Protein Kinases/genetics , Protein Multimerization , Protein Structure, Tertiary , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
18.
Immunity ; 44(1): 88-102, 2016 Jan 19.
Article En | MEDLINE | ID: mdl-26795252

The role of apoptosis inducing factor (AIF) in promoting cell death versus survival remains controversial. We report that the loss of AIF in fibroblasts led to mitochondrial electron transport chain defects and loss of proliferation that could be restored by ectopic expression of the yeast NADH dehydrogenase Ndi1. Aif-deficiency in T cells led to decreased peripheral T cell numbers and defective homeostatic proliferation, but thymic T cell development was unaffected. In contrast, Aif-deficient B cells developed and functioned normally. The difference in the dependency of T cells versus B cells on AIF for function and survival correlated with their metabolic requirements. Ectopic Ndi1 expression rescued homeostatic proliferation of Aif-deficient T cells. Despite its reported roles in cell death, fibroblasts, thymocytes and B cells lacking AIF underwent normal death. These studies suggest that the primary role of AIF relates to complex I function, with differential effects on T and B cells.


Apoptosis Inducing Factor/metabolism , B-Lymphocytes/metabolism , Mitochondria/physiology , T-Lymphocytes/metabolism , Animals , Apoptosis , Cell Respiration/physiology , Electron Transport Complex I/metabolism , Fibroblasts/metabolism , Glycolysis/physiology , Mice , Mice, Knockout , Mice, Mutant Strains
19.
Sci Rep ; 5: 16488, 2015 Nov 18.
Article En | MEDLINE | ID: mdl-26576726

Leydig cells are crucial to the production of testosterone in males. It is unknown if the cancer chemotherapeutic drug, 6-mercaptopurine (6 MP), produces Leydig cell failure among adult survivors of childhood acute lymphoblastic leukemia. Moreover, it is not known whether Leydig cell failure is due to either a loss of cells or an impairment in their function. Herein, we show, in a subset of childhood cancer survivors, that Leydig cell failure is related to the dose of 6 MP. This was extended, in a murine model, to demonstrate that 6 MP exposure induced caspase 3 activation, and the loss of Leydig cells was independent of Bak and Bax activation. The death of these non-proliferating cells was triggered by 6 MP metabolism, requiring formation of both cytosolic reactive oxygen species and thiopurine nucleotide triphosphates. The thiopurine nucleotide triphosphates (with physiological amounts of dATP) uniquely activated the apoptosome. An ABC transporter (Abcc4/Mrp4) reduced the amount of thiopurines, thereby providing protection for Leydig cells. The studies reported here demonstrate that the apoptosome is uniquely activated by thiopurine nucleotides and suggest that 6 MP induced Leydig cell death is likely a cause of Leydig cell failure in some survivors of childhood cancer.


Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosomes/metabolism , Leydig Cells/drug effects , Leydig Cells/metabolism , Mercaptopurine/pharmacology , Animals , Antimetabolites, Antineoplastic/toxicity , Caspases/metabolism , Cell Line, Tumor , Disease Models, Animal , Humans , Male , Mercaptopurine/toxicity , Methotrexate/pharmacology , Methotrexate/toxicity , Mice, Transgenic , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Reactive Oxygen Species/metabolism
20.
PLoS Genet ; 11(9): e1005500, 2015 Sep.
Article En | MEDLINE | ID: mdl-26352669

Nature's fastest motors are the cochlear outer hair cells (OHCs). These sensory cells use a membrane protein, Slc26a5 (prestin), to generate mechanical force at high frequencies, which is essential for explaining the exquisite hearing sensitivity of mammalian ears. Previous studies suggest that Slc26a5 continuously diffuses within the membrane, but how can a freely moving motor protein effectively convey forces critical for hearing? To provide direct evidence in OHCs for freely moving Slc26a5 molecules, we created a knockin mouse where Slc26a5 is fused with YFP. These mice and four other strains expressing fluorescently labeled membrane proteins were used to examine their lateral diffusion in the OHC lateral wall. All five proteins showed minimal diffusion, but did move after pharmacological disruption of membrane-associated structures with a cholesterol-depleting agent and salicylate. Thus, our results demonstrate that OHC lateral wall structure constrains the mobility of plasma membrane proteins and that the integrity of such membrane-associated structures are critical for Slc26a5's active and structural roles. The structural constraint of membrane proteins may exemplify convergent evolution of cellular motors across species. Our findings also suggest a possible mechanism for disorders of cholesterol metabolism with hearing loss such as Niemann-Pick Type C diseases.


Hair Cells, Auditory, Outer/metabolism , Molecular Motor Proteins/metabolism , Animals , Bacterial Proteins/genetics , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Rhodopsin/metabolism , Salicylic Acid/pharmacology , beta-Cyclodextrins/pharmacology
...