1
|
Listian SA, Mazur AC, Kol M, Ufelmann E, Eising S, Fröhlich F, Walter S, Holthuis JCM, Barisch C. Complex sphingolipid profiling and identification of an inositol-phosphorylceramide synthase in Dictyostelium discoideum. iScience 2024; 27:110609. [PMID: 39286488 PMCID: PMC11402645 DOI: 10.1016/j.isci.2024.110609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/12/2024] [Accepted: 07/26/2024] [Indexed: 09/19/2024] Open
Abstract
Dictyostelium discoideum is a professional phagocyte frequently used to study cellular processes underlying the recognition, engulfment, and infection course of microbial pathogens. Sphingolipids are abundant components of the plasma membrane that bind cholesterol, control membrane properties, participate in signal transmission, and serve as adhesion molecules in recognition processes relevant to immunity and infection. By combining lipidomics with a bioinformatics-based cloning strategy, we show here that D. discoideum produces phosphoinositol-containing sphingolipids with predominantly phytoceramide backbones. Cell-free expression of candidate inositol-phosphorylceramide (IPC) synthases from D. discoideum enabled identification of an enzyme that selectively catalyzes the transfer of phosphoinositol from phosphatidylinositol onto ceramide. The IPC synthase, DdIPCS1, shares multiple sequence motifs with yeast IPC and human sphingomyelin synthases and localizes to the Golgi apparatus as well as the contractile vacuole of D. discoideum. These findings open up important opportunities for exploring a role of sphingolipids in phagocytosis and infection across major evolutionary boundaries.
Collapse
Affiliation(s)
- Stevanus A Listian
- Division of Molecular Infection Biology, Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Anna-Carina Mazur
- Division of Molecular Infection Biology, Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
- Centre for Structural Systems Biology, Hamburg, Germany
- Division of Host-Microbe Interactome, Research Center Borstel (FZB) - Leibniz Lung Center, Borstel, Germany
- Department of Biology, University of Hamburg, Hamburg, Germany
| | - Matthijs Kol
- Division of Molecular Cell Biology, Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
- Center of Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
| | - Edwin Ufelmann
- Division of Molecular Infection Biology, Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Sebastian Eising
- Division of Molecular Membrane Biology, Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Florian Fröhlich
- Division of Molecular Membrane Biology, Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
- Center of Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
| | - Stefan Walter
- Center of Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
| | - Joost C M Holthuis
- Division of Molecular Cell Biology, Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
- Center of Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
| | - Caroline Barisch
- Division of Molecular Infection Biology, Department of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
- Center of Cellular Nanoanalytics (CellNanOs), University of Osnabrück, Osnabrück, Germany
- Centre for Structural Systems Biology, Hamburg, Germany
- Division of Host-Microbe Interactome, Research Center Borstel (FZB) - Leibniz Lung Center, Borstel, Germany
- Department of Biology, University of Hamburg, Hamburg, Germany
| |
Collapse
|
2
|
Wangler MF, Chao YH, Roth M, Welti R, McNew JA. Drosophila Models Uncover Substrate Channeling Effects on Phospholipids and Sphingolipids in Peroxisomal Biogenesis Disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.591192. [PMID: 38746221 PMCID: PMC11092477 DOI: 10.1101/2024.04.26.591192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Peroxisomal Biogenesis Disorders Zellweger Spectrum (PBD-ZSD) disorders are a group of autosomal recessive defects in peroxisome formation that produce a multi-systemic disease presenting at birth or in childhood. Well documented clinical biomarkers such as elevated very long chain fatty acids (VLCFA) are key biochemical diagnostic findings in these conditions. Additional, secondary biochemical alterations such as elevated very long chain lysophosphatidylcholines are allowing newborn screening for peroxisomal disease. In addition, a more widespread impact on metabolism and lipids is increasingly being documented by metabolomic and lipidomic studies. Here we utilize Drosophila models of pex2 and pex16 as well as human plasma from individuals with PEX1 mutations. We identify phospholipid abnormalities in Drosophila larvae and brain characterized by differences in the quantities of phosphatidylcholine (PC) and phosphatidylethanolamines (PE) with long chain lengths and reduced levels of intermediate chain lengths. For diacylglycerol (DAG) the precursor of PE and PC through the Kennedy pathway, the intermediate chain lengths are increased suggesting an imbalance between DAGs and PE and PC that suggests the two acyl chain pools are not in equilibrium. Altered acyl chain lengths are also observed in PE ceramides in the fly models. Interestingly, plasma from human subjects exhibit phospholipid alterations similar to the fly model. Moreover, human plasma shows reduced levels of sphingomyelin with 18 and 22 carbon lengths but normal levels of C24. Our results suggest that peroxisomal biogenesis defects alter shuttling of the acyl chains of multiple phospholipid and ceramide lipid classes, whereas DAG species with intermediate fatty acids are more abundant. These data suggest an imbalance between de novo synthesis of PC and PE through the Kennedy pathway and remodeling of existing PC and PE through the Lands cycle. This imbalance is likely due to overabundance of very long and long acyl chains in PBD and a subsequent imbalance due to substrate channeling effects. Given the fundamental role of phospholipid and sphingolipids in nervous system functions, these observations suggest PBD-ZSD are diseases characterized by widespread cell membrane lipid abnormalities.
Collapse
|
3
|
Mendoza-Grimau V, Pérez-Gálvez A, Busturia A, Fontecha J. Lipidomic profiling of Drosophila strains Canton-S and white 1118 reveals intraspecific lipid variations in basal metabolic rate. Prostaglandins Leukot Essent Fatty Acids 2024; 201:102618. [PMID: 38795635 DOI: 10.1016/j.plefa.2024.102618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/23/2024] [Accepted: 05/08/2024] [Indexed: 05/28/2024]
Abstract
Drosophila melanogaster is a well-established model system for studies on lipid metabolism and energy homeostasis. In this study, we identified and quantified the main components of the lipid profile of two widely utilized Drosophila strains, namely Canton-S and white1118, under identical experimental conditions. Differences observed between the strains can be attributed to inherent metabolic divergences, thus limiting the influence of confounding factors. Using the comprehensive lipid data acquired, we applied cluster analysis and PLS-DA techniques to ascertain whether the lipidome could effectively differentiate between the strains. Certain lipid features, such as triacylglycerols, polar lipids, and specific sterol components, could be distinguished between flies of both strains regardless of sex. Our results suggest that although Canton-S and white1118 have similar lipid profiles and distributions, a selected subset of lipids demonstrates clear discriminatory potential between strains, thereby bearing significant implications for planning biological studies using these strains as control references.
Collapse
Affiliation(s)
- Victor Mendoza-Grimau
- Food Lipid Biomarkers and Health Group, Institute of Food Science Research (CIAL, CSIC-UAM), Madrid 28049, Spain
| | - Antonio Pérez-Gálvez
- Group of Chemistry and Biochemistry of Pigments, Instituto de la Grasa, CSIC, Sevilla 41013, Spain
| | - Ana Busturia
- Tissue and organ homeostasis, Centro de Biología Molecular Severo Ochoa, (CBMSO, CSIC-UAM), Madrid 28049, Spain
| | - Javier Fontecha
- Food Lipid Biomarkers and Health Group, Institute of Food Science Research (CIAL, CSIC-UAM), Madrid 28049, Spain.
| |
Collapse
|
4
|
Tsarouhas V, Liu D, Tsikala G, Engström Y, Strigini M, Samakovlis C. A surfactant lipid layer of endosomal membranes facilitates airway gas filling in Drosophila. Curr Biol 2023; 33:5132-5146.e5. [PMID: 37992718 DOI: 10.1016/j.cub.2023.10.058] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 09/14/2023] [Accepted: 10/26/2023] [Indexed: 11/24/2023]
Abstract
The mechanisms underlying the construction of an air-liquid interface in respiratory organs remain elusive. Here, we use live imaging and genetic analysis to describe the morphogenetic events generating an extracellular lipid lining of the Drosophila airways required for their gas filing and animal survival. We show that sequential Rab39/Syx1A/Syt1-mediated secretion of lysosomal acid sphingomyelinase (Drosophila ASM [dASM]) and Rab11/35/Syx1A/Rop-dependent exosomal secretion provides distinct components for lipid film assembly. Tracheal inactivation of Rab11 or Rab35 or loss of Rop results in intracellular accumulation of exosomal, multi-vesicular body (MVB)-derived vesicles. On the other hand, loss of dASM or Rab39 causes luminal bubble-like accumulations of exosomal membranes and liquid retention in the airways. Inactivation of the exosomal secretion in dASM mutants counteracts this phenotype, arguing that the exosomal secretion provides the lipid vesicles and that secreted lysosomal dASM organizes them into a continuous film. Our results reveal the coordinated functions of extracellular vesicle and lysosomal secretions in generating a lipid layer crucial for airway gas filling and survival.
Collapse
Affiliation(s)
- Vasilios Tsarouhas
- Stockholm University, Department of Molecular Biosciences, The Wenner-Gren Institute, 10691 Stockholm, Sweden; Science for Life Laboratory, SciLifeLab, 171 65 Stockholm, Sweden.
| | - Dan Liu
- Stockholm University, Department of Molecular Biosciences, The Wenner-Gren Institute, 10691 Stockholm, Sweden
| | - Georgia Tsikala
- Stockholm University, Department of Molecular Biosciences, The Wenner-Gren Institute, 10691 Stockholm, Sweden; IMBB, 70013 Heraklion, Crete, Greece
| | - Ylva Engström
- Stockholm University, Department of Molecular Biosciences, The Wenner-Gren Institute, 10691 Stockholm, Sweden
| | | | - Christos Samakovlis
- Stockholm University, Department of Molecular Biosciences, The Wenner-Gren Institute, 10691 Stockholm, Sweden; Science for Life Laboratory, SciLifeLab, 171 65 Stockholm, Sweden; ECCPS, Justus Liebig University of Giessen, 35390 Giessen, Germany.
| |
Collapse
|
5
|
Shaheen A, Richter Gorey CL, Sghaier A, Dason JS. Cholesterol is required for activity-dependent synaptic growth. J Cell Sci 2023; 136:jcs261563. [PMID: 37902091 DOI: 10.1242/jcs.261563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/24/2023] [Indexed: 10/31/2023] Open
Abstract
Changes in cholesterol content of neuronal membranes occur during development and brain aging. Little is known about whether synaptic activity regulates cholesterol levels in neuronal membranes and whether these changes affect neuronal development and function. We generated transgenic flies that express the cholesterol-binding D4H domain of perfringolysin O toxin and found increased levels of cholesterol in presynaptic terminals of Drosophila larval neuromuscular junctions following increased synaptic activity. Reduced cholesterol impaired synaptic growth and largely prevented activity-dependent synaptic growth. Presynaptic knockdown of adenylyl cyclase phenocopied the impaired synaptic growth caused by reducing cholesterol. Furthermore, the effects of knocking down adenylyl cyclase and reducing cholesterol were not additive, suggesting that they function in the same pathway. Increasing cAMP levels using a dunce mutant with reduced phosphodiesterase activity failed to rescue this impaired synaptic growth, suggesting that cholesterol functions downstream of cAMP. We used a protein kinase A (PKA) sensor to show that reducing cholesterol levels reduced presynaptic PKA activity. Collectively, our results demonstrate that enhanced synaptic activity increased cholesterol levels in presynaptic terminals and that these changes likely activate the cAMP-PKA pathway during activity-dependent growth.
Collapse
Affiliation(s)
- Amber Shaheen
- Department of Biomedical Sciences, University of Windsor, Windsor, Ontario, N9B 3P4, Canada
| | - Claire L Richter Gorey
- Department of Biomedical Sciences, University of Windsor, Windsor, Ontario, N9B 3P4, Canada
| | - Adam Sghaier
- Department of Biomedical Sciences, University of Windsor, Windsor, Ontario, N9B 3P4, Canada
| | - Jeffrey S Dason
- Department of Biomedical Sciences, University of Windsor, Windsor, Ontario, N9B 3P4, Canada
| |
Collapse
|
6
|
Abstract
Ligands of the Hedgehog (HH) pathway are paracrine signaling molecules that coordinate tissue development in metazoans. A remarkable feature of HH signaling is the repeated use of cholesterol in steps spanning ligand biogenesis, secretion, dispersal, and reception on target cells. A cholesterol molecule covalently attached to HH ligands is used as a molecular baton by transfer proteins to guide their secretion, spread, and reception. On target cells, a signaling circuit composed of a cholesterol transporter and sensor regulates transmission of HH signals across the plasma membrane to the cytoplasm. The repeated use of cholesterol in signaling supports the view that the HH pathway likely evolved by coopting ancient systems to regulate the abundance or organization of sterol-like lipids in membranes.
Collapse
Affiliation(s)
- Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom;
| | - Rajat Rohatgi
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, California, USA;
| |
Collapse
|
7
|
Gehin C, Lone MA, Lee W, Capolupo L, Ho S, Adeyemi AM, Gerkes EH, Stegmann AP, López-Martín E, Bermejo-Sánchez E, Martínez-Delgado B, Zweier C, Kraus C, Popp B, Strehlow V, Gräfe D, Knerr I, Jones ER, Zamuner S, Abriata LA, Kunnathully V, Moeller BE, Vocat A, Rommelaere S, Bocquete JP, Ruchti E, Limoni G, Van Campenhoudt M, Bourgeat S, Henklein P, Gilissen C, van Bon BW, Pfundt R, Willemsen MH, Schieving JH, Leonardi E, Soli F, Murgia A, Guo H, Zhang Q, Xia K, Fagerberg CR, Beier CP, Larsen MJ, Valenzuela I, Fernández-Álvarez P, Xiong S, Śmigiel R, López-González V, Armengol L, Morleo M, Selicorni A, Torella A, Blyth M, Cooper NS, Wilson V, Oegema R, Herenger Y, Garde A, Bruel AL, Tran Mau-Them F, Maddocks AB, Bain JM, Bhat MA, Costain G, Kannu P, Marwaha A, Champaigne NL, Friez MJ, Richardson EB, Gowda VK, Srinivasan VM, Gupta Y, Lim TY, Sanna-Cherchi S, Lemaitre B, Yamaji T, Hanada K, Burke JE, Jakšić AM, McCabe BD, De Los Rios P, Hornemann T, D’Angelo G, Gennarino VA. CERT1 mutations perturb human development by disrupting sphingolipid homeostasis. J Clin Invest 2023; 133:e165019. [PMID: 36976648 PMCID: PMC10178846 DOI: 10.1172/jci165019] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Neural differentiation, synaptic transmission, and action potential propagation depend on membrane sphingolipids, whose metabolism is tightly regulated. Mutations in the ceramide transporter CERT (CERT1), which is involved in sphingolipid biosynthesis, are associated with intellectual disability, but the pathogenic mechanism remains obscure. Here, we characterize 31 individuals with de novo missense variants in CERT1. Several variants fall into a previously uncharacterized dimeric helical domain that enables CERT homeostatic inactivation, without which sphingolipid production goes unchecked. The clinical severity reflects the degree to which CERT autoregulation is disrupted, and inhibiting CERT pharmacologically corrects morphological and motor abnormalities in a Drosophila model of the disease, which we call ceramide transporter (CerTra) syndrome. These findings uncover a central role for CERT autoregulation in the control of sphingolipid biosynthetic flux, provide unexpected insight into the structural organization of CERT, and suggest a possible therapeutic approach for patients with CerTra syndrome.
Collapse
Affiliation(s)
- Charlotte Gehin
- Institute of Bioengineering (IBI), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Museer A. Lone
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Winston Lee
- Department of Genetics and Development and
- Department Ophthalmology, Columbia University Irving Medical Center, New York, New York, USA
| | - Laura Capolupo
- Institute of Bioengineering (IBI), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sylvia Ho
- Institute of Bioengineering (IBI), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Adekemi M. Adeyemi
- Department of Medical Genetics, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada
| | - Erica H. Gerkes
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, Netherlands
| | - Alexander P.A. Stegmann
- Department of Clinical Genetics and School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, Netherlands
| | - Estrella López-Martín
- Institute of Rare Diseases Research (IIER), Instituto de Salud Carlos III, Madrid, Spain
| | - Eva Bermejo-Sánchez
- Institute of Rare Diseases Research (IIER), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Christiane Zweier
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Human Genetics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Cornelia Kraus
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Bernt Popp
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Functional Genomics, Berlin, Germany
| | - Vincent Strehlow
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Daniel Gräfe
- Department of Pediatric Radiology, University Hospital Leipzig, Leipzig, Leipzig, Germany
| | - Ina Knerr
- National Centre for Inherited Metabolic Disorders, Children’s Health Ireland (CHI) at Temple Street, Dublin, Ireland
- UCD School of Medicine, Dublin, Ireland
| | - Eppie R. Jones
- Genuity Science, Cherrywood Business Park, Dublin, Ireland
| | - Stefano Zamuner
- Institute of Physics, School of Basic Sciences, École Polytechnique Féderale de Lausanne (EPFL), Lausanne, Switzerland
| | - Luciano A. Abriata
- Laboratory for Biomolecular Modeling and Protein Purification and Structure Facility, EPFL and Swiss Institute of Bioinformatics, Lausanne Switzerland
| | - Vidya Kunnathully
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Brandon E. Moeller
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada
| | - Anthony Vocat
- Institute of Bioengineering (IBI), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | | | | - Evelyne Ruchti
- Brain Mind Institute, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Greta Limoni
- Brain Mind Institute, School of Life Sciences, EPFL, Lausanne, Switzerland
| | | | - Samuel Bourgeat
- Brain Mind Institute, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Petra Henklein
- Berlin Institute of Health, Institut für Biochemie, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian Gilissen
- Radboud University Medical Center, Department of Human Genetics, Nijmegen, Netherlands
- Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Bregje W. van Bon
- Radboud University Medical Center, Department of Human Genetics, Nijmegen, Netherlands
| | - Rolph Pfundt
- Radboud University Medical Center, Department of Human Genetics, Nijmegen, Netherlands
- Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | | | - Jolanda H. Schieving
- Radboud University Medical Center, Department of Pediatric Neurology, Amalia Children’s Hospital and Donders Institute for Brain, Cognition and Behavior, Nijmegen, Netherlands
| | - Emanuela Leonardi
- Molecular Genetics of Neurodevelopment, Department of Woman and Child Health, University of Padova, Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica (IRP), Città della Speranza, Padova, Italy
| | - Fiorenza Soli
- Medical Genetics Department, APSS Trento, Trento, Italy
| | - Alessandra Murgia
- Fondazione Istituto di Ricerca Pediatrica (IRP), Città della Speranza, Padova, Italy
| | - Hui Guo
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Qiumeng Zhang
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Kun Xia
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Christina R. Fagerberg
- Department of Neurology, Odense University Hospital, and Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Christoph P. Beier
- Department of Neurology, Odense University Hospital, and Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Martin J. Larsen
- Department of Neurology, Odense University Hospital, and Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Irene Valenzuela
- Department of Clinical and Molecular Genetics, University Hospital Vall d′Hebron, Medicine Genetics Group, Valle Hebron Research Institute, Barcelona, Spain
| | - Paula Fernández-Álvarez
- Department of Clinical and Molecular Genetics, University Hospital Vall d′Hebron, Medicine Genetics Group, Valle Hebron Research Institute, Barcelona, Spain
| | - Shiyi Xiong
- Fetal Medicine Unit and Prenatal Diagnosis Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Robert Śmigiel
- Department of Family and Pediatric Nursing, Medical University, Wroclaw, Poland
| | - Vanesa López-González
- Sección de Genética Médica, Servicio de Pediatría, Hospital Clínico Universitario Virgen de la Arrixaca, IMIB-Arrixaca, CIBERER-ISCIII, Murcia, Spain
| | - Lluís Armengol
- Quantitative Genomic Medicine Laboratories, S.L., CSO & CEO, Esplugues del Llobregat, Barcelona, Catalunya, Spain
| | - Manuela Morleo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli,” Naples, Italy
| | - Angelo Selicorni
- Department of Pediatrics, ASST Lariana Sant’ Anna Hospital, San Fermo Della Battaglia, Como, Italy
| | - Annalaura Torella
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli,” Naples, Italy
| | - Moira Blyth
- North of Scotland Regional Genetics Service, Clinical Genetics Centre, Ashgrove House, Foresterhill, Aberdeen, United Kingdom
| | - Nicola S. Cooper
- W Midlands Clinical Genetics Service, Birmingham Women’s Hospital, Edgbaston Birmingham, United Kingdom
| | - Valerie Wilson
- Northern Regional Genetics Laboratory, Newcastle upon Tyne, United Kingdom
| | - Renske Oegema
- Department of Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Yvan Herenger
- Genetica AG, Humangenetisches Labor und Beratungsstelle, Zürich, Switzerland
| | - Aurore Garde
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d’Enfants, CHU Dijon, Dijon, France
- UMR1231 GAD, INSERM – Université Bourgogne-Franche Comté, Dijon, France
| | - Ange-Line Bruel
- UMR1231 GAD, INSERM – Université Bourgogne-Franche Comté, Dijon, France
- Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Frederic Tran Mau-Them
- UMR1231 GAD, INSERM – Université Bourgogne-Franche Comté, Dijon, France
- Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Alexis B.R. Maddocks
- Department of Radiology at Columbia University Irving Medical Center, New York, New York, USA
| | - Jennifer M. Bain
- Department of Neurology, Columbia University Irving Medical Center, New York Presbyterian Hospital, Columbia University Medical Center, New York, New York, USA
| | - Musadiq A. Bhat
- Institute of Pharmacology and Toxicology University of Zürich, Zürich, Switzerland
| | - Gregory Costain
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Peter Kannu
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Ashish Marwaha
- Department of Medical Genetics, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada
| | - Neena L. Champaigne
- Greenwood Genetic Center and the Medical University of South Carolina, Greenwood, South Carolina, USA
| | - Michael J. Friez
- Greenwood Genetic Center and the Medical University of South Carolina, Greenwood, South Carolina, USA
| | - Ellen B. Richardson
- Greenwood Genetic Center and the Medical University of South Carolina, Greenwood, South Carolina, USA
| | - Vykuntaraju K. Gowda
- Department of Pediatric Neurology, Indira Gandhi Institute of Child Health, Bangalore, India
| | | | - Yask Gupta
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York, USA
| | - Tze Y. Lim
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York, USA
| | - Simone Sanna-Cherchi
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York, USA
| | | | - Toshiyuki Yamaji
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kentaro Hanada
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - John E. Burke
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Ana Marjia Jakšić
- Brain Mind Institute, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Brian D. McCabe
- Brain Mind Institute, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Paolo De Los Rios
- Institute of Bioengineering (IBI), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Physics, School of Basic Sciences, École Polytechnique Féderale de Lausanne (EPFL), Lausanne, Switzerland
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Giovanni D’Angelo
- Institute of Bioengineering (IBI), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
- Global Health Institute, School of Life Sciences and
| | - Vincenzo A. Gennarino
- Department of Genetics and Development and
- Department of Pediatrics
- Department of Neurology
- Columbia Stem Cell Initiative, and
- Initiative for Columbia Ataxia and Tremor, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
8
|
Ehtezazi T, Rahman K, Davies R, Leach AG. The Pathological Effects of Circulating Hydrophobic Bile Acids in Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:173-211. [PMID: 36994114 PMCID: PMC10041467 DOI: 10.3233/adr-220071] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
Abstract
Recent clinical studies have revealed that the serum levels of toxic hydrophobic bile acids (deoxy cholic acid, lithocholic acid [LCA], and glycoursodeoxycholic acid) are significantly higher in patients with Alzheimer's disease (AD) and amnestic mild cognitive impairment (aMCI) when compared to control subjects. The elevated serum bile acids may be the result of hepatic peroxisomal dysfunction. Circulating hydrophobic bile acids are able to disrupt the blood-brain barrier and promote the formation of amyloid-β plaques through enhancing the oxidation of docosahexaenoic acid. Hydrophobic bile acid may find their ways into the neurons via the apical sodium-dependent bile acid transporter. It has been shown that hydrophobic bile acids impose their pathological effects by activating farnesoid X receptor and suppressing bile acid synthesis in the brain, blocking NMDA receptors, lowering brain oxysterol levels, and interfering with 17β-estradiol actions such as LCA by binding to E2 receptors (molecular modelling data exclusive to this paper). Hydrophobic bile acids may interfere with the sonic hedgehog signaling through alteration of cell membrane rafts and reducing brain 24(S)-hydroxycholesterol. This article will 1) analyze the pathological roles of circulating hydrophobic bile acids in the brain, 2) propose therapeutic approaches, and 3) conclude that consideration be given to reducing/monitoring toxic bile acid levels in patients with AD or aMCI, prior/in combination with other treatments.
Collapse
Affiliation(s)
- Touraj Ehtezazi
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Khalid Rahman
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Rhys Davies
- The Walton Centre, NHS Foundation Trust, Liverpool, UK
| | - Andrew G Leach
- School of Pharmacy, University of Manchester, Manchester, UK
| |
Collapse
|
9
|
Pan X, Dutta D, Lu S, Bellen HJ. Sphingolipids in neurodegenerative diseases. Front Neurosci 2023; 17:1137893. [PMID: 36875645 PMCID: PMC9978793 DOI: 10.3389/fnins.2023.1137893] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/27/2023] [Indexed: 02/18/2023] Open
Abstract
Neurodegenerative Diseases (NDDs) are a group of disorders that cause progressive deficits of neuronal function. Recent evidence argues that sphingolipid metabolism is affected in a surprisingly broad set of NDDs. These include some lysosomal storage diseases (LSDs), hereditary sensory and autonomous neuropathy (HSAN), hereditary spastic paraplegia (HSP), infantile neuroaxonal dystrophy (INAD), Friedreich's ataxia (FRDA), as well as some forms of amyotrophic lateral sclerosis (ALS) and Parkinson's disease (PD). Many of these diseases have been modeled in Drosophila melanogaster and are associated with elevated levels of ceramides. Similar changes have also been reported in vertebrate cells and mouse models. Here, we summarize studies using fly models and/or patient samples which demonstrate the nature of the defects in sphingolipid metabolism, the organelles that are implicated, the cell types that are initially affected, and potential therapeutics for these diseases.
Collapse
Affiliation(s)
- Xueyang Pan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Debdeep Dutta
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Shenzhao Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Hugo J. Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
10
|
Sailer SA, Burkhalter MD, Philipp M. Cholesterol and Phosphoinositides in Cilia Biology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:121-142. [PMID: 36988879 DOI: 10.1007/978-3-031-21547-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Cilia are evolutionarily conserved organelles that can be found on virtually every cell. They appear as hair-like structures emanating from the cellular surface either as single or as bundles of cilia. There, they sense external stimuli and translate them into intracellular signals. Motile cilia beat for the generation of locomotion of unicellular organisms or fluid flow in certain body cavities of vertebrate organisms. Defects in cilia are detrimental and account for the development of ciliopathies, one of the fastest-growing family of afflictions. In the past decade, membrane lipids, such as cholesterol and phosphoinositides, have emerged as essential elements in both the signal transduction via cilia and the building of cilia itself. Here, we summarize the current knowledge on the impact of cholesterol and phosphoinositides on cilium biology.
Collapse
Affiliation(s)
- Steffen-Alexander Sailer
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Division of Pharmacogenomics, University Hospital Tübingen, Tübingen, Germany
| | - Martin D Burkhalter
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Division of Pharmacogenomics, University Hospital Tübingen, Tübingen, Germany
| | - Melanie Philipp
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Division of Pharmacogenomics, University Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
11
|
Kunc M, Dobeš P, Ward R, Lee S, Čegan R, Dostálková S, Holušová K, Hurychová J, Eliáš S, Pinďáková E, Čukanová E, Prodělalová J, Petřivalský M, Danihlík J, Havlík J, Hobza R, Kavanagh K, Hyršl P. Omics-based analysis of honey bee (Apis mellifera) response to Varroa sp. parasitisation and associated factors reveals changes impairing winter bee generation. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 152:103877. [PMID: 36403678 DOI: 10.1016/j.ibmb.2022.103877] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/24/2022] [Accepted: 11/13/2022] [Indexed: 06/16/2023]
Abstract
The extensive annual loss of honey bees (Apis mellifera L.) represents a global problem affecting agriculture and biodiversity. The parasitic mite Varroa destructor, associated with viral co-infections, plays a key role in this loss. Despite years of intensive research, the complex mechanisms of Varroa - honey bee interaction are still not fully defined. Therefore, this study employed a unique combination of transcriptomic, proteomic, metabolomic, and functional analyses to reveal new details about the effect of Varroa mites and naturally associated factors, including viruses, on honey bees. We focused on the differences between Varroa parasitised and unparasitised ten-day-old worker bees collected before overwintering from the same set of colonies reared without anti-mite treatment. Supplementary comparison to honey bees collected from colonies with standard anti-Varroa treatment can provide further insights into the effect of a pyrethroid flumethrin. Analysis of the honey bees exposed to mite parasitisation revealed alterations in the transcriptome and proteome related to immunity, oxidative stress, olfactory recognition, metabolism of sphingolipids, and RNA regulatory mechanisms. The immune response and sphingolipid metabolism were strongly activated, whereas olfactory recognition and oxidative stress pathways were inhibited in Varroa parasitised honey bees compared to unparasitised ones. Moreover, metabolomic analysis confirmed the depletion of nutrients and energy stores, resulting in a generally disrupted metabolism in the parasitised workers. The combined omics-based analysis conducted on strictly parasitised bees revealed the key molecular components and mechanisms underlying the detrimental effects of Varroa sp. and its associated pathogens. This study provides the theoretical basis and interlinked datasets for further research on honey bee response to biological threats and the development of efficient control strategies against Varroa mites.
Collapse
Affiliation(s)
- Martin Kunc
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Pavel Dobeš
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.
| | - Rachel Ward
- Department of Biology, Maynooth University, W23 F2K8 Maynooth, Co. Kildare, Ireland
| | - Saetbyeol Lee
- Department of Food Science, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 00, Prague, Czech Republic
| | - Radim Čegan
- Department of Plant Developmental Genetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 00, Brno, Czech Republic
| | - Silvie Dostálková
- Department of Biochemistry, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 783 71, Olomouc, Czech Republic
| | - Kateřina Holušová
- Institute of Experimental Botany of the Czech Academy of Sciences, Centre of the Region Haná for Biotechnological and Agricultural Research, Šlechtitelů 31, 779 00, Olomouc, Czech Republic
| | - Jana Hurychová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Sara Eliáš
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Eliška Pinďáková
- Department of Biochemistry, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 783 71, Olomouc, Czech Republic
| | - Eliška Čukanová
- Department of Infectious Disease and Preventive Medicine, Veterinary Research Institute, Hudcova 296/70, 621 00, Brno, Czech Republic
| | - Jana Prodělalová
- Department of Infectious Disease and Preventive Medicine, Veterinary Research Institute, Hudcova 296/70, 621 00, Brno, Czech Republic
| | - Marek Petřivalský
- Department of Biochemistry, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 783 71, Olomouc, Czech Republic
| | - Jiří Danihlík
- Department of Biochemistry, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 783 71, Olomouc, Czech Republic
| | - Jaroslav Havlík
- Department of Food Science, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 00, Prague, Czech Republic
| | - Roman Hobza
- Department of Plant Developmental Genetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 00, Brno, Czech Republic
| | - Kevin Kavanagh
- Department of Biology, Maynooth University, W23 F2K8 Maynooth, Co. Kildare, Ireland
| | - Pavel Hyršl
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| |
Collapse
|
12
|
Linnemannstöns K, Karuna M P, Witte L, Choezom D, Honemann‐Capito M, Lagurin AS, Schmidt CV, Shrikhande S, Steinmetz L, Wiebke M, Lenz C, Gross JC. Microscopic and biochemical monitoring of endosomal trafficking and extracellular vesicle secretion in an endogenous in vivo model. J Extracell Vesicles 2022; 11:e12263. [PMID: 36103151 PMCID: PMC9473323 DOI: 10.1002/jev2.12263] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/17/2022] [Accepted: 05/22/2022] [Indexed: 11/10/2022] Open
Abstract
Extracellular vesicle (EV) secretion enables cell-cell communication in multicellular organisms. During development, EV secretion and the specific loading of signalling factors in EVs contributes to organ development and tissue differentiation. Here, we present an in vivo model to study EV secretion using the fat body and the haemolymph of the fruit fly, Drosophila melanogaster. The system makes use of tissue-specific EV labelling and is amenable to genetic modification by RNAi. This allows the unique combination of microscopic visualisation of EVs in different organs and quantitative biochemical purification to study how EVs are generated within the cells and which factors regulate their secretion in vivo. Characterisation of the system revealed that secretion of EVs from the fat body is mainly regulated by Rab11 and Rab35, highlighting the importance of recycling Rab GTPase family members for EV secretion. We furthermore discovered a so far unknown function of Rab14 along with the kinesin Klp98A in EV biogenesis and secretion.
Collapse
Affiliation(s)
- Karen Linnemannstöns
- Developmental BiochemistryUniversity Medical Center GoettingenGoettingenGermany
- Hematology and OncologyUniversity Medical Center GoettingenGoettingenGermany
- Molecular OncologyUniversity Medical Center GoettingenGoettingenGermany
| | - Pradhipa Karuna M
- Developmental BiochemistryUniversity Medical Center GoettingenGoettingenGermany
| | - Leonie Witte
- Developmental BiochemistryUniversity Medical Center GoettingenGoettingenGermany
| | - Dolma Choezom
- Developmental BiochemistryUniversity Medical Center GoettingenGoettingenGermany
| | | | - Alex Simon Lagurin
- Developmental BiochemistryUniversity Medical Center GoettingenGoettingenGermany
| | | | - Shreya Shrikhande
- Developmental BiochemistryUniversity Medical Center GoettingenGoettingenGermany
| | | | - Möbius Wiebke
- Electron Microscopy Core Unit, Department of NeurogeneticsMax Planck Institute of Experimental MedicineGöttingenGermany
| | - Christof Lenz
- Institute of Clinical ChemistryUniversity Medical Center GöttingenGöttingenGermany
- Bioanalytical Mass Spectrometry GroupMax Planck Institute for Biophysical ChemistryGöttingenGermany
| | - Julia Christina Gross
- Developmental BiochemistryUniversity Medical Center GoettingenGoettingenGermany
- Hematology and OncologyUniversity Medical Center GoettingenGoettingenGermany
- Department of MedicineHealth and Medical UniversityPotsdamGermany
| |
Collapse
|
13
|
Kinnebrew M, Woolley RE, Ansell TB, Byrne EFX, Frigui S, Luchetti G, Sircar R, Nachtergaele S, Mydock-McGrane L, Krishnan K, Newstead S, Sansom MSP, Covey DF, Siebold C, Rohatgi R. Patched 1 regulates Smoothened by controlling sterol binding to its extracellular cysteine-rich domain. SCIENCE ADVANCES 2022; 8:eabm5563. [PMID: 35658032 PMCID: PMC9166294 DOI: 10.1126/sciadv.abm5563] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 04/15/2022] [Indexed: 05/06/2023]
Abstract
Smoothened (SMO) transduces the Hedgehog (Hh) signal across the plasma membrane in response to accessible cholesterol. Cholesterol binds SMO at two sites: one in the extracellular cysteine-rich domain (CRD) and a second in the transmembrane domain (TMD). How these two sterol-binding sites mediate SMO activation in response to the ligand Sonic Hedgehog (SHH) remains unknown. We find that mutations in the CRD (but not the TMD) reduce the fold increase in SMO activity triggered by SHH. SHH also promotes the photocrosslinking of a sterol analog to the CRD in intact cells. In contrast, sterol binding to the TMD site boosts SMO activity regardless of SHH exposure. Mutational and computational analyses show that these sites are in allosteric communication despite being 45 angstroms apart. Hence, sterols function as both SHH-regulated orthosteric ligands at the CRD and allosteric ligands at the TMD to regulate SMO activity and Hh signaling.
Collapse
Affiliation(s)
- Maia Kinnebrew
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rachel E. Woolley
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | - Eamon F. X. Byrne
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Sara Frigui
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Giovanni Luchetti
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ria Sircar
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sigrid Nachtergaele
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Laurel Mydock-McGrane
- Department of Developmental Biology, Washington School of Medicine, St. Louis, MO, USA
| | - Kathiresan Krishnan
- Department of Developmental Biology, Washington School of Medicine, St. Louis, MO, USA
| | - Simon Newstead
- Department of Biochemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | | | - Douglas F. Covey
- Department of Developmental Biology, Washington School of Medicine, St. Louis, MO, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Rajat Rohatgi
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
14
|
The Role of Membrane Lipids in Light-Activation of Drosophila TRP Channels. Biomolecules 2022; 12:biom12030382. [PMID: 35327573 PMCID: PMC8945425 DOI: 10.3390/biom12030382] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 12/28/2022] Open
Abstract
Transient Receptor Potential (TRP) channels constitute a large superfamily of polymodal channel proteins with diverse roles in many physiological and sensory systems that function both as ionotropic and metabotropic receptors. From the early days of TRP channel discovery, membrane lipids were suggested to play a fundamental role in channel activation and regulation. A prominent example is the Drosophila TRP and TRP-like (TRPL) channels, which are predominantly expressed in the visual system of Drosophila. Light activation of the TRP and TRPL channels, the founding members of the TRP channel superfamily, requires activation of phospholipase Cβ (PLC), which hydrolyzes phosphatidylinositol 4,5-bisphosphate (PIP2) into Diacylglycerol (DAG) and Inositol 1, 4,5-trisphosphate (IP3). However, the events required for channel gating downstream of PLC activation are still under debate and led to several hypotheses regarding the mechanisms by which lipids gate the channels. Despite many efforts, compelling evidence of the involvement of DAG accumulation, PIP2 depletion or IP3-mediated Ca2+ release in light activation of the TRP/TRPL channels are still lacking. Exogeneous application of poly unsaturated fatty acids (PUFAs), a product of DAG hydrolysis was demonstrated as an efficient way to activate the Drosophila TRP/TRPL channels. However, compelling evidence for the involvement of PUFAs in physiological light-activation of the TRP/TRPL channels is still lacking. Light-induced mechanical force generation was measured in photoreceptor cells prior to channel opening. This mechanical force depends on PLC activity, suggesting that the enzymatic activity of PLC converting PIP2 into DAG generates membrane tension, leading to mechanical gating of the channels. In this review, we will present the roles of membrane lipids in light activation of Drosophila TRP channels and present the many advantages of this model system in the exploration of TRP channel activation under physiological conditions.
Collapse
|
15
|
Eickelberg V, Lüersen K, Staats S, Rimbach G. Phenotyping of Drosophila Melanogaster-A Nutritional Perspective. Biomolecules 2022; 12:221. [PMID: 35204721 PMCID: PMC8961528 DOI: 10.3390/biom12020221] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/15/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
The model organism Drosophila melanogaster was increasingly applied in nutrition research in recent years. A range of methods are available for the phenotyping of D. melanogaster, which are outlined in the first part of this review. The methods include determinations of body weight, body composition, food intake, lifespan, locomotor activity, reproductive capacity and stress tolerance. In the second part, the practical application of the phenotyping of flies is demonstrated via a discussion of obese phenotypes in response to high-sugar diet (HSD) and high-fat diet (HFD) feeding. HSD feeding and HFD feeding are dietary interventions that lead to an increase in fat storage and affect carbohydrate-insulin homeostasis, lifespan, locomotor activity, reproductive capacity and stress tolerance. Furthermore, studies regarding the impacts of HSD and HFD on the transcriptome and metabolome of D. melanogaster are important for relating phenotypic changes to underlying molecular mechanisms. Overall, D. melanogaster was demonstrated to be a valuable model organism with which to examine the pathogeneses and underlying molecular mechanisms of common chronic metabolic diseases in a nutritional context.
Collapse
Affiliation(s)
- Virginia Eickelberg
- Department of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6-8, D-24118 Kiel, Germany; (K.L.); (S.S.); (G.R.)
| | | | | | | |
Collapse
|
16
|
Dispatching plasma membrane cholesterol and Sonic Hedgehog dispatch: two sides of the same coin? Biochem Soc Trans 2021; 49:2455-2463. [PMID: 34515747 PMCID: PMC8589413 DOI: 10.1042/bst20210918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 11/19/2022]
Abstract
Vertebrate and invertebrate Hedgehog (Hh) morphogens signal over short and long distances to direct cell fate decisions during development and to maintain tissue homeostasis after birth. One of the most important questions in Hh biology is how such Hh signaling to distant target cells is achieved, because all Hh proteins are secreted as dually lipidated proteins that firmly tether to the outer plasma membrane leaflet of their producing cells. There, Hhs multimerize into light microscopically visible storage platforms that recruit factors required for their regulated release. One such recruited release factor is the soluble glycoprotein Scube2 (Signal sequence, cubulin domain, epidermal-growth-factor-like protein 2), and maximal Scube2 function requires concomitant activity of the resistance-nodulation-division (RND) transporter Dispatched (Disp) at the plasma membrane of Hh-producing cells. Although recently published cryo-electron microscopy-derived structures suggest possible direct modes of Scube2/Disp-regulated Hh release, the mechanism of Disp-mediated Hh deployment is still not fully understood. In this review, we discuss suggested direct modes of Disp-dependent Hh deployment and relate them to the structural similarities between Disp and the related RND transporters Patched (Ptc) and Niemann-Pick type C protein 1. We then discuss open questions and perspectives that derive from these structural similarities, with particular focus on new findings that suggest shared small molecule transporter functions of Disp to deplete the plasma membrane of cholesterol and to modulate Hh release in an indirect manner.
Collapse
|
17
|
Manikowski D, Ehring K, Gude F, Jakobs P, Froese J, Grobe K. Hedgehog lipids: Promotors of alternative morphogen release and signaling?: Conflicting findings on lipidated Hedgehog transport and signaling can be explained by alternative regulated mechanisms to release the morphogen. Bioessays 2021; 43:e2100133. [PMID: 34611914 DOI: 10.1002/bies.202100133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/27/2021] [Accepted: 09/06/2021] [Indexed: 12/19/2022]
Abstract
Two posttranslational lipid modifications present on all Hedgehog (Hh) morphogens-an N-terminal palmitate and a C-terminal cholesterol-are established and essential regulators of Hh biofunction. Yet, for several decades, the question of exactly how both lipids contribute to Hh signaling remained obscure. Recently, cryogenic electron microscopy revealed different modes by which one or both lipids may contribute directly to Hh binding and signaling to its receptor Patched1 (Ptc). Some of these modes demand that the established release factor Dispatched1 (Disp) extracts dual-lipidated Hh from the cell surface, and that another known upstream signaling modulator called Scube2 chaperones the dual-lipidated morphogen to Ptc. By mechanistically and biochemically aligning this concept with established in vivo and recent in vitro findings, this reflection identifies remaining questions in lipidated Hh transport and evaluates additional mechanisms of Disp- and Scube2-regulated release of a second bioactive Hh fraction that has one or both lipids removed.
Collapse
Affiliation(s)
- Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Kristina Ehring
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Fabian Gude
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Petra Jakobs
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Jurij Froese
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| |
Collapse
|
18
|
Wang X, Liu H, Liu Y, Han G, Wang Y, Chen H, He L, Ma G. Highly Conserved C-Terminal Region of Indian Hedgehog N-Fragment Contributes to Its Auto-Processing and Multimer Formation. Biomolecules 2021; 11:biom11060792. [PMID: 34070546 PMCID: PMC8227148 DOI: 10.3390/biom11060792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/15/2021] [Accepted: 05/18/2021] [Indexed: 11/30/2022] Open
Abstract
Hedgehog (HH) is a highly conserved secretory signalling protein family mainly involved in embryonic development, homeostasis, and tumorigenesis. HH is generally synthesised as a precursor, which subsequently undergoes autoproteolytic cleavage to generate an amino-terminal fragment (HH-N), mediating signalling, and a carboxyl-terminal fragment (HH-C), catalysing the auto-processing reaction. The N-terminal region of HH-N is required for HH multimer formation to promote signal transduction, whilst the functions of the C-terminal region of HH-N remain ambiguous. This study focused on Indian Hedgehog (IHH), a member of the HH family, to explore the functions of the C-terminal region of the amino-terminal fragment of IHH (IHH-N) via protein truncation, cell-based assays, and 3D structure prediction. The results revealed that three amino acids, including S195, A196, and A197, were crucial for the multimer formation by inserting the mutual binding of IHH-N proteins. K191, S192, E193, and H194 had an extremely remarkable effect on IHH self-cleavage. In addition, A198, K199, and T200 evidently affected the stability of IHH-N. This work suggested that the C-terminus of IHH-N played an important role in the physiological function of IHH at multiple levels, thus deepening the understanding of HH biochemical properties.
Collapse
Affiliation(s)
- Xiaoqing Wang
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China;
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.L.); (G.H.); (Y.W.)
| | - Hao Liu
- State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China;
| | - Yanfang Liu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.L.); (G.H.); (Y.W.)
| | - Gefei Han
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.L.); (G.H.); (Y.W.)
| | - Yushu Wang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.L.); (G.H.); (Y.W.)
| | - Haifeng Chen
- State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China;
- Correspondence: (H.C.); (L.H.); (G.M.)
| | - Lin He
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.L.); (G.H.); (Y.W.)
- Correspondence: (H.C.); (L.H.); (G.M.)
| | - Gang Ma
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China;
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.L.); (G.H.); (Y.W.)
- Correspondence: (H.C.); (L.H.); (G.M.)
| |
Collapse
|
19
|
Hall ET, Dillard ME, Stewart DP, Zhang Y, Wagner B, Levine RM, Pruett-Miller SM, Sykes A, Temirov J, Cheney RE, Mori M, Robinson CG, Ogden SK. Cytoneme delivery of Sonic Hedgehog from ligand-producing cells requires Myosin 10 and a Dispatched-BOC/CDON co-receptor complex. eLife 2021; 10:61432. [PMID: 33570491 PMCID: PMC7968926 DOI: 10.7554/elife.61432] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
Morphogens function in concentration-dependent manners to instruct cell fate during tissue patterning. The cytoneme morphogen transport model posits that specialized filopodia extend between morphogen-sending and responding cells to ensure that appropriate signaling thresholds are achieved. How morphogens are transported along and deployed from cytonemes, how quickly a cytoneme-delivered, receptor-dependent signal is initiated, and whether these processes are conserved across phyla are not known. Herein, we reveal that the actin motor Myosin 10 promotes vesicular transport of Sonic Hedgehog (SHH) morphogen in mouse cell cytonemes, and that SHH morphogen gradient organization is altered in neural tubes of Myo10-/- mice. We demonstrate that cytoneme-mediated deposition of SHH onto receiving cells induces a rapid, receptor-dependent signal response that occurs within seconds of ligand delivery. This activity is dependent upon a novel Dispatched (DISP)-BOC/CDON co-receptor complex that functions in ligand-producing cells to promote cytoneme occurrence and facilitate ligand delivery for signal activation.
Collapse
Affiliation(s)
- Eric T Hall
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Miriam E Dillard
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Daniel P Stewart
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Yan Zhang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Ben Wagner
- Cell and Tissue Imaging Center, St. Jude Children's Research Hospital, Memphis, United States
| | - Rachel M Levine
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States.,Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, United States
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States.,Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, United States
| | - April Sykes
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, United States
| | - Jamshid Temirov
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Richard E Cheney
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, United States
| | - Motomi Mori
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, United States
| | - Camenzind G Robinson
- Cell and Tissue Imaging Center, St. Jude Children's Research Hospital, Memphis, United States
| | - Stacey K Ogden
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| |
Collapse
|
20
|
Abstract
Lipids exert diverse functions in living organisms. They form cellular membranes, store and transport energy and play signalling roles. Some lipid species function in all of these processes, making them ideal candidates to coordinate metabolism with cellular homeostasis and animal development. This theme was central to Suzanne Eaton's research in the fruit fly, Drosophila Here, we discuss her work on membrane lipid homeostasis in changing environments and on functions for lipids in the Hedgehog signalling pathway. We further highlight lipoproteins as inter-organ carriers of lipids and lipid-linked morphogens, which communicate dietary and developmental signals throughout the organism.
Collapse
Affiliation(s)
- Wilhelm Palm
- Cell and Tumor Biology Program, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jonathan Rodenfels
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| |
Collapse
|
21
|
Fabri JHTM, de Sá NP, Malavazi I, Del Poeta M. The dynamics and role of sphingolipids in eukaryotic organisms upon thermal adaptation. Prog Lipid Res 2020; 80:101063. [PMID: 32888959 DOI: 10.1016/j.plipres.2020.101063] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/18/2020] [Accepted: 08/27/2020] [Indexed: 01/09/2023]
Abstract
All living beings have an optimal temperature for growth and survival. With the advancement of global warming, the search for understanding adaptive processes to climate changes has gained prominence. In this context, all living beings monitor the external temperature and develop adaptive responses to thermal variations. These responses ultimately change the functioning of the cell and affect the most diverse structures and processes. One of the first structures to detect thermal variations is the plasma membrane, whose constitution allows triggering of intracellular signals that assist in the response to temperature stress. Although studies on this topic have been conducted, the underlying mechanisms of recognizing thermal changes and modifying cellular functioning to adapt to this condition are not fully understood. Recently, many reports have indicated the participation of sphingolipids (SLs), major components of the plasma membrane, in the regulation of the thermal stress response. SLs can structurally reinforce the membrane or/and send signals intracellularly to control numerous cellular processes, such as apoptosis, cytoskeleton polarization, cell cycle arresting and fungal virulence. In this review, we discuss how SLs synthesis changes during both heat and cold stresses, focusing on fungi, plants, animals and human cells. The role of lysophospholipids is also discussed.
Collapse
Affiliation(s)
- João Henrique Tadini Marilhano Fabri
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA; Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | - Nivea Pereira de Sá
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Iran Malavazi
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA; Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, New York, USA; Veterans Administration Medical Center, Northport, New York, USA.
| |
Collapse
|
22
|
Molecular evolution of a collage of cholesterol interaction motifs in transmembrane helix V of the serotonin 1A receptor. Chem Phys Lipids 2020; 232:104955. [PMID: 32846149 DOI: 10.1016/j.chemphyslip.2020.104955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/08/2020] [Accepted: 08/16/2020] [Indexed: 12/20/2022]
Abstract
The human serotonin1A receptor is a representative member of the superfamily of G protein-coupled receptors (GPCRs) and an important drug target for neurological disorders. Using a combination of biochemical, biophysical and molecular dynamics simulation approaches, we and others have shown that membrane cholesterol modulates the organization, dynamics and function of vertebrate serotonin1A receptors. Previous studies have shown that the cytoplasmic portion of transmembrane helix V (TM V) and the extramembraneous intracellular loop 3 are critical for G-protein coupling, phosphorylation and desensitization of the receptor. We have recently resolved a collage of putative cholesterol interaction motifs from the amino acid sequence overlapping this region. In this paper, we explore the sequence plasticity of this fragment that may have adapted to altered membrane lipidome, after vertebrates evolved from primordial invertebrates. Since invertebrates have lower levels of membrane cholesterol relative to vertebrates, we compared TM V sequence fragments from invertebrate serotonin1 receptors with vertebrate orthologs to infer the sequence plasticity in TM V. We report that the average number of cholesterol interaction motifs in TM V for diverse phyla represents an increasing trend that could mirror vertebrate evolution from primordial invertebrates. By statistical modeling, we propose that the collage of cholesterol interaction motifs in TM V of the human serotonin1A receptor may have evolved from rudimentary collages, reminiscent of primordial invertebrate orthologs. Taken together, we propose that a repertoire of cholesterol-philic nonsynonymous substitutions may have enhanced collage complexity in TM V during vertebrate evolution.
Collapse
|
23
|
Gao L, Cazenave-Gassiot A, Burla B, Wenk MR, Torta F. Dual mass spectrometry as a tool to improve annotation and quantification in targeted plasma lipidomics. Metabolomics 2020; 16:53. [PMID: 32303853 DOI: 10.1007/s11306-020-01677-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022]
Abstract
INTRODUCTION High quality data, based on reliable quantification and clear identification of the reported lipid species, are required for the clinical translation of human plasma lipidomic studies. OBJECTIVE Lipid quantification can be efficiently performed on triple quadrupole (QqQ) mass spectrometers in targeted multiple reaction monitoring (MRM) mode. However, a series of issues can be encountered when aiming at unambiguous identification and accurate quantification, including (i) resolving peaks of polyunsaturated species, (ii) discriminating between plasmanyl-, plasmenyl- and odd chain species and (iii) resolving the isotopic overlap between co-eluting lipid species. METHODS As a practical tool to improve the quality of targeted lipidomics studies, we applied a Dual MS platform by simultaneously coupling a reversed-phase liquid chromatography separation to a QqQ and a quadrupole-time of flight (Q-ToF) mass spectrometers. In one single experiment, this platform allows to correctly identify, by high-resolution MS and MS/MS, the peaks that are quantified by MRM. RESULTS As proof of concept, we applied the platform on glycerophosphocholines (GPCs) and sphingomyelins (SMs), which are highly abundant in human plasma and play crucial roles in various physiological functions. Our results demonstrated that Dual MS could provide a higher level of confidence in the identification and quantification of GPCs and SMs in human plasma. The same approach can also be applied to improve the study of other lipid classes and expanded for the identification of novel lipid molecular species. CONCLUSIONS This methodology might have a great potential to achieve a better specificity in the quantification of lipids by targeted lipidomics in high-throughput studies.
Collapse
Affiliation(s)
- Liang Gao
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Amaury Cazenave-Gassiot
- Singapore Lipidomics Incubator (SLING), Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Bo Burla
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Markus R Wenk
- Singapore Lipidomics Incubator (SLING), Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Federico Torta
- Singapore Lipidomics Incubator (SLING), Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
24
|
Kinnebrew M, Iverson EJ, Patel BB, Pusapati GV, Kong JH, Johnson KA, Luchetti G, Eckert KM, McDonald JG, Covey DF, Siebold C, Radhakrishnan A, Rohatgi R. Cholesterol accessibility at the ciliary membrane controls hedgehog signaling. eLife 2019; 8:e50051. [PMID: 31657721 PMCID: PMC6850779 DOI: 10.7554/elife.50051] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 10/25/2019] [Indexed: 12/21/2022] Open
Abstract
Previously we proposed that transmission of the hedgehog signal across the plasma membrane by Smoothened is triggered by its interaction with cholesterol (Luchetti et al., 2016). But how is cholesterol, an abundant lipid, regulated tightly enough to control a signaling system that can cause birth defects and cancer? Using toxin-based sensors that distinguish between distinct pools of cholesterol, we find that Smoothened activation and Hedgehog signaling are driven by a biochemically-defined, small fraction of membrane cholesterol, termed accessible cholesterol. Increasing cholesterol accessibility by depletion of sphingomyelin, which sequesters cholesterol in complexes, amplifies Hedgehog signaling. Hedgehog ligands increase cholesterol accessibility in the membrane of the primary cilium by inactivating the transporter-like protein Patched 1. Trapping this accessible cholesterol blocks Hedgehog signal transmission across the membrane. Our work shows that the organization of cholesterol in the ciliary membrane can be modified by extracellular ligands to control the activity of cilia-localized signaling proteins.
Collapse
Affiliation(s)
- Maia Kinnebrew
- Department of BiochemistryStanford University School of MedicineStanfordUnited States
| | - Ellen J Iverson
- Department of BiochemistryStanford University School of MedicineStanfordUnited States
| | - Bhaven B Patel
- Department of BiochemistryStanford University School of MedicineStanfordUnited States
| | - Ganesh V Pusapati
- Department of BiochemistryStanford University School of MedicineStanfordUnited States
| | - Jennifer H Kong
- Department of BiochemistryStanford University School of MedicineStanfordUnited States
| | - Kristen A Johnson
- Department of Molecular GeneticsUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Giovanni Luchetti
- Department of BiochemistryStanford University School of MedicineStanfordUnited States
| | - Kaitlyn M Eckert
- Center for Human NutritionUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Jeffrey G McDonald
- Department of Molecular GeneticsUniversity of Texas Southwestern Medical CenterDallasUnited States
- Center for Human NutritionUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Douglas F Covey
- Taylor Family Institute for Innovative Psychiatric ResearchWashington University School of MedicineSt. LouisUnited States
- Department of Developmental BiologyWashington University School of MedicineSt. LouisUnited States
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUnited Kingdom
| | - Arun Radhakrishnan
- Department of Molecular GeneticsUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Rajat Rohatgi
- Department of BiochemistryStanford University School of MedicineStanfordUnited States
- Department of MedicineStanford University School of MedicineStanfordUnited States
| |
Collapse
|
25
|
Sato M, Arita M, Kawashima T. Uncovering Ecdysozoa-specific Sphingomyelin Synthase by Phylogenetic Analysis of Metazoan Sequences. Zoolog Sci 2019; 36:316-321. [PMID: 34664902 DOI: 10.2108/zs180168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/01/2019] [Indexed: 11/17/2022]
Abstract
Sphingomyelin (SM) is a membrane phospholipid that is widely distributed in Metazoa; it is the major constituent of myelin sheaths in vertebrates. In mammals, two genes (SMS1 and SMS2) are responsible for its synthesis. No SM-producing genes have been clearly identified in insects and crustaceans (Ecdysozoa) despite the presence of a myelin sheath-like structure in shrimps. Since the rapid transmission of electrical signals requires the use of an insulating material in the nerve, it is possible that the convergent evolution of enzymes to synthesize the insulating compounds for the nervous system also occurred in animals other than vertebrates. Our exhaustive phylogenetic search for metazoan SM synthase identified an Ecdysozoa-specific SM synthase candidate, SMSe, which is absent in Drosophila and Lophotrochozoa. All Ecdysozoa lack the homolog of myelin basic- and proteolipid proteins present in mammals. We propose an evolutionary path of SM synthase and discuss the origin of the myelin structure in Metazoa.
Collapse
Affiliation(s)
- Mitsuharu Sato
- National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan,
| | - Masanori Arita
- National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan.,RIKEN Center for Sustainable Resource Science, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | | |
Collapse
|
26
|
Ceramide phosphoethanolamine, an enigmatic cellular membrane sphingolipid. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:1284-1292. [DOI: 10.1016/j.bbamem.2019.05.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 12/14/2022]
|
27
|
Guo W, Roelink H. Loss of the Heparan Sulfate Proteoglycan Glypican5 Facilitates Long-Range Sonic Hedgehog Signaling. Stem Cells 2019; 37:899-909. [PMID: 30977233 PMCID: PMC8491322 DOI: 10.1002/stem.3018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/31/2019] [Indexed: 01/01/2023]
Abstract
As a morphogen, Sonic Hedgehog (Shh) mediates signaling at a distance from its sites of synthesis. After secretion, Shh must traverse a distance through the extracellular matrix (ECM) to reach the target cells and activate the Hh response. ECM proteins, in particular, the heparan sulfate proteoglycans (HSPGs) of the glypican family, have both negative and positive effects on Shh signaling, all attributed to their ability to bind Shh. Using mouse embryonic stem cell-derived mosaic tissues with compartments that lack the glycosyltransferases Exostosin1 and Exostosin2, or the HSPG core protein Glypican5, we show that Shh accumulates around its source cells when they are surrounded by cells that have a mutated ECM. This accumulation of Shh is correlated with an increased noncell autonomous Shh response. Our results support a model in which Shh presented on the cell surface accumulates at or near ECM that lacks HSPGs, possibly due to the absence of these Shh sequestering molecules. Stem Cells 2019;37:899-909.
Collapse
Affiliation(s)
- Wei Guo
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, California, USA
| | - Henk Roelink
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, California, USA
| |
Collapse
|
28
|
Sokolov SS, Trushina NI, Severin FF, Knorre DA. Ergosterol Turnover in Yeast: An Interplay between Biosynthesis and Transport. BIOCHEMISTRY (MOSCOW) 2019; 84:346-357. [DOI: 10.1134/s0006297919040023] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
29
|
Abramyan J. Hedgehog Signaling and Embryonic Craniofacial Disorders. J Dev Biol 2019; 7:E9. [PMID: 31022843 PMCID: PMC6631594 DOI: 10.3390/jdb7020009] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
Since its initial discovery in a Drosophila mutagenesis screen, the Hedgehog pathway has been revealed to be instrumental in the proper development of the vertebrate face. Vertebrates possess three hedgehog paralogs: Sonic hedgehog (Shh), Indian hedgehog (Ihh), and Desert hedgehog (Dhh). Of the three, Shh has the broadest range of functions both in the face and elsewhere in the embryo, while Ihh and Dhh play more limited roles. The Hedgehog pathway is instrumental from the period of prechordal plate formation early in the embryo, until the fusion of the lip and secondary palate, which complete the major patterning events of the face. Disruption of Hedgehog signaling results in an array of developmental disorders in the face, ranging from minor alterations in the distance between the eyes to more serious conditions such as severe clefting of the lip and palate. Despite its critical role, Hedgehog signaling seems to be disrupted through a number of mechanisms that may either be direct, as in mutation of a downstream target of the Hedgehog ligand, or indirect, such as mutation in a ciliary protein that is otherwise seemingly unrelated to the Hedgehog pathway. A number of teratogens such as alcohol, statins and steroidal alkaloids also disrupt key aspects of Hedgehog signal transduction, leading to developmental defects that are similar, if not identical, to those of Hedgehog pathway mutations. The aim of this review is to highlight the variety of roles that Hedgehog signaling plays in developmental disorders of the vertebrate face.
Collapse
Affiliation(s)
- John Abramyan
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, MI 48128, USA.
| |
Collapse
|
30
|
Specification of positional identity in forebrain organoids. Nat Biotechnol 2019; 37:436-444. [PMID: 30936566 PMCID: PMC6447454 DOI: 10.1038/s41587-019-0085-3] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 02/22/2019] [Indexed: 01/28/2023]
Abstract
Human brain organoids generated with current technologies recapitulate histological features of the human brain, but they lack a reproducible topographic organization. During development, spatial topography is determined by gradients of signaling molecules released from discrete signaling centers. We hypothesized that introduction of a signaling center into forebrain organoids would specify the positional identity of neural tissue in a distance-dependent manner. Here, we present a system to trigger a sonic hedgehog (SHH) protein gradient in developing forebrain organoids that enables ordered self-organization along dorso-ventral and antero-posterior positional axes. SHH-patterned forebrain organoids establish major forebrain subdivisions that are positioned with in vivo-like topography. Consistent with its behavior in vivo, SHH exhibits long-range signaling activity in organoids. Finally, we use SHH-patterned cerebral organoids as a tool to study the role of cholesterol metabolism in SHH signaling. Together, this work identifies inductive signaling as an effective organizing strategy to recapitulate in vivo-like topography in human brain organoids.
Collapse
|
31
|
Huang Y, Huang S, Di Scala C, Wang Q, Wandall HH, Fantini J, Zhang YQ. The glycosphingolipid MacCer promotes synaptic bouton formation in Drosophila by interacting with Wnt. eLife 2018; 7:38183. [PMID: 30355446 PMCID: PMC6202054 DOI: 10.7554/elife.38183] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 09/09/2018] [Indexed: 01/05/2023] Open
Abstract
Lipids are structural components of cellular membranes and signaling molecules that are widely involved in development and diseases, but the underlying molecular mechanisms are poorly understood, partly because of the vast variety of lipid species and complexity of synthetic and turnover pathways. From a genetic screen, we identify that mannosyl glucosylceramide (MacCer), a species of glycosphingolipid (GSL), promotes synaptic bouton formation at the Drosophila neuromuscular junction (NMJ). Pharmacological and genetic analysis shows that the NMJ growth-promoting effect of MacCer depends on normal lipid rafts, which are known to be composed of sphingolipids, sterols and select proteins. MacCer positively regulates the synaptic level of Wnt1/Wingless (Wg) and facilitates presynaptic Wg signaling, whose activity is raft-dependent. Furthermore, a functional GSL-binding motif in Wg exhibiting a high affinity for MacCer is required for normal NMJ growth. These findings reveal a novel mechanism whereby the GSL MacCer promotes synaptic bouton formation via Wg signaling.
Collapse
Affiliation(s)
- Yan Huang
- Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Beijing, China
| | - Sheng Huang
- Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Beijing, China.,Sino-Danish College, Sino-Danish Center for Education and Research, Chinese Academy of Sciences, Beijing, China
| | | | - Qifu Wang
- Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Beijing, China
| | - Hans H Wandall
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jacques Fantini
- UNIS UMR_S 1072, INSERM, Aix-Marseille Université, Marseille, France
| | - Yong Q Zhang
- Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Beijing, China
| |
Collapse
|
32
|
Kunduri G, Turner-Evans D, Konya Y, Izumi Y, Nagashima K, Lockett S, Holthuis J, Bamba T, Acharya U, Acharya JK. Defective cortex glia plasma membrane structure underlies light-induced epilepsy in cpes mutants. Proc Natl Acad Sci U S A 2018; 115:E8919-E8928. [PMID: 30185559 PMCID: PMC6156639 DOI: 10.1073/pnas.1808463115] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Seizures induced by visual stimulation (photosensitive epilepsy; PSE) represent a common type of epilepsy in humans, but the molecular mechanisms and genetic drivers underlying PSE remain unknown, and no good genetic animal models have been identified as yet. Here, we show an animal model of PSE, in Drosophila, owing to defective cortex glia. The cortex glial membranes are severely compromised in ceramide phosphoethanolamine synthase (cpes)-null mutants and fail to encapsulate the neuronal cell bodies in the Drosophila neuronal cortex. Expression of human sphingomyelin synthase 1, which synthesizes the closely related ceramide phosphocholine (sphingomyelin), rescues the cortex glial abnormalities and PSE, underscoring the evolutionarily conserved role of these lipids in glial membranes. Further, we show the compromise in plasma membrane structure that underlies the glial cell membrane collapse in cpes mutants and leads to the PSE phenotype.
Collapse
Affiliation(s)
- Govind Kunduri
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD 21702
| | | | - Yutaka Konya
- Department of Metabolomics, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshihiro Izumi
- Department of Metabolomics, Kyushu University, Fukuoka 812-8582, Japan
| | - Kunio Nagashima
- Electron Microscopy Laboratory, National Cancer Institute, Frederick, MD 21702
| | - Stephen Lockett
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Joost Holthuis
- Molecular Cell Biology Division, University of Osnabrück, 49074 Osnabrück, Germany
| | - Takeshi Bamba
- Department of Metabolomics, Kyushu University, Fukuoka 812-8582, Japan
| | - Usha Acharya
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Jairaj K Acharya
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD 21702;
| |
Collapse
|
33
|
Manikowski D, Kastl P, Grobe K. Taking the Occam's Razor Approach to Hedgehog Lipidation and Its Role in Development. J Dev Biol 2018; 6:jdb6010003. [PMID: 29615552 PMCID: PMC5875562 DOI: 10.3390/jdb6010003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 01/24/2018] [Accepted: 01/25/2018] [Indexed: 01/01/2023] Open
Abstract
All Hedgehog (Hh) proteins signal from producing cells to distant receiving cells despite being synthesized as N-and C-terminally lipidated, membrane-tethered molecules. To explain this paradoxical situation, over the past 15 years, several hypotheses have been postulated that tie directly into this property, such as Hh transport on cellular extensions called cytonemes or on secreted vesicles called lipophorins and exosomes. The alternative situation that tight membrane association merely serves to prevent unregulated Hh solubilization has been addressed by biochemical and structural studies suggesting Hh extraction from the membrane or proteolytic Hh release. While some of these models may act in different organisms, tissues or developmental programs, others may act together to specify Hh short- and long-range signaling in the same tissues. To test and rank these possibilities, we here review major models of Hh release and transport and hypothesize that the (bio)chemical and physical properties of firmly established, homologous, and functionally essential biochemical Hh modifications are adapted to specify and determine interdependent steps of Hh release, transport and signaling, while ruling out other steps. This is also described by the term “congruence”, meaning that the logical combination of biochemical Hh modifications can reveal their true functional implications. This combined approach reveals potential links between models of Hh release and transport that were previously regarded as unrelated, thereby expanding our view of how Hhs can steer development in a simple, yet extremely versatile, manner.
Collapse
Affiliation(s)
- Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Münster, D-48149 Münster, Germany.
| | - Philipp Kastl
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Münster, D-48149 Münster, Germany.
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Münster, D-48149 Münster, Germany.
| |
Collapse
|
34
|
Hentschel A, Zahedi RP, Ahrends R. Protein lipid modifications--More than just a greasy ballast. Proteomics 2016; 16:759-82. [PMID: 26683279 DOI: 10.1002/pmic.201500353] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 10/24/2015] [Accepted: 12/14/2015] [Indexed: 12/21/2022]
Abstract
Covalent lipid modifications of proteins are crucial for regulation of cellular plasticity, since they affect the chemical and physical properties and therefore protein activity, localization, and stability. Most recently, lipid modifications on proteins are increasingly attracting important regulatory entities in diverse signaling events and diseases. In all cases, the lipid moiety of modified proteins is essential to allow water-soluble proteins to strongly interact with membranes or to induce structural changes in proteins that are critical for elemental processes such as respiration, transport, signal transduction, and motility. Until now, roughly about ten lipid modifications on different amino acid residues are described at the UniProtKB database and even well-known modifications are underrepresented. Thus, it is of fundamental importance to develop a better understanding of this emerging and so far under-investigated type of protein modification. Therefore, this review aims to give a comprehensive and detailed overview about enzymatic and nonenzymatic lipidation events, will report their role in cellular biology, discuss their relevancy for diseases, and describe so far available bioanalytical strategies to analyze this highly challenging type of modification.
Collapse
Affiliation(s)
- Andreas Hentschel
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V, Dortmund, Germany
| | - René P Zahedi
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V, Dortmund, Germany
| | - Robert Ahrends
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V, Dortmund, Germany
| |
Collapse
|
35
|
Simon E, Aguirre-Tamaral A, Aguilar G, Guerrero I. Perspectives on Intra- and Intercellular Trafficking of Hedgehog for Tissue Patterning. J Dev Biol 2016; 4:jdb4040034. [PMID: 29615597 PMCID: PMC5831803 DOI: 10.3390/jdb4040034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/07/2016] [Accepted: 11/08/2016] [Indexed: 12/14/2022] Open
Abstract
Intercellular communication is a fundamental process for correct tissue development. The mechanism of this process involves, among other things, the production and secretion of signaling molecules by specialized cell types and the capability of these signals to reach the target cells in order to trigger specific responses. Hedgehog (Hh) is one of the best-studied signaling pathways because of its importance during morphogenesis in many organisms. The Hh protein acts as a morphogen, activating its targets at a distance in a concentration-dependent manner. Post-translational modifications of Hh lead to a molecule covalently bond to two lipid moieties. These lipid modifications confer Hh high affinity to lipidic membranes, and intense studies have been carried out to explain its release into the extracellular matrix. This work reviews Hh molecule maturation, the intracellular recycling needed for its secretion and the proposed carriers to explain Hh transportation to the receiving cells. Special focus is placed on the role of specialized filopodia, also named cytonemes, in morphogen transport and gradient formation.
Collapse
Affiliation(s)
- Eléanor Simon
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, CSIC-UAM, Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain.
| | - Adrián Aguirre-Tamaral
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, CSIC-UAM, Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain.
| | - Gustavo Aguilar
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, CSIC-UAM, Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain.
| | - Isabel Guerrero
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, CSIC-UAM, Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain.
| |
Collapse
|
36
|
Vigoder FM, Parker DJ, Cook N, Tournière O, Sneddon T, Ritchie MG. Inducing Cold-Sensitivity in the Frigophilic Fly Drosophila montana by RNAi. PLoS One 2016; 11:e0165724. [PMID: 27832122 PMCID: PMC5104470 DOI: 10.1371/journal.pone.0165724] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/17/2016] [Indexed: 12/31/2022] Open
Abstract
Cold acclimation is a critical physiological adaptation for coping with seasonal cold. By increasing their cold tolerance individuals can remain active for longer at the onset of winter and can recover more quickly from a cold shock. In insects, despite many physiological studies, little is known about the genetic basis of cold acclimation. Recently, transcriptomic analyses in Drosophila virilis and D. montana revealed candidate genes for cold acclimation by identifying genes upregulated during exposure to cold. Here, we test the role of myo-inositol-1-phosphate synthase (Inos), in cold tolerance in D. montana using an RNAi approach. D. montana has a circumpolar distribution and overwinters as an adult in northern latitudes with extreme cold. We assessed cold tolerance of dsRNA knock-down flies using two metrics: chill-coma recovery time (CCRT) and mortality rate after cold acclimation. Injection of dsRNAInos did not alter CCRT, either overall or in interaction with the cold treatment, however it did induced cold-specific mortality, with high levels of mortality observed in injected flies acclimated at 5°C but not at 19°C. Overall, injection with dsRNAInos induced a temperature-sensitive mortality rate of over 60% in this normally cold-tolerant species. qPCR analysis confirmed that dsRNA injection successfully reduced gene expression of Inos. Thus, our results demonstrate the involvement of Inos in increasing cold tolerance in D. montana. The potential mechanisms involved by which Inos increases cold tolerance are also discussed.
Collapse
Affiliation(s)
- Felipe M. Vigoder
- Centre for Biological Diversity, School of Biology, University of St Andrews, Fife, United Kingdom
- Departamento de Genética, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Darren J. Parker
- Centre for Biological Diversity, School of Biology, University of St Andrews, Fife, United Kingdom
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| | - Nicola Cook
- Centre for Biological Diversity, School of Biology, University of St Andrews, Fife, United Kingdom
| | - Océane Tournière
- Centre for Biological Diversity, School of Biology, University of St Andrews, Fife, United Kingdom
- Sars International Centre for Marine Molecular Biology, Thormøhlensgt, Bergen, Norway
| | - Tanya Sneddon
- Centre for Biological Diversity, School of Biology, University of St Andrews, Fife, United Kingdom
| | - Michael G. Ritchie
- Centre for Biological Diversity, School of Biology, University of St Andrews, Fife, United Kingdom
| |
Collapse
|
37
|
Huang Y, Huang S, Lam SM, Liu Z, Shui G, Zhang YQ. Acsl, the Drosophila ortholog of intellectual-disability-related ACSL4, inhibits synaptic growth by altered lipids. J Cell Sci 2016; 129:4034-4045. [PMID: 27656110 DOI: 10.1242/jcs.195032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/16/2016] [Indexed: 12/17/2022] Open
Abstract
Nervous system development and function are tightly regulated by metabolic processes, including the metabolism of lipids such as fatty acids. Mutations in long-chain acyl-CoA synthetase 4 (ACSL4) are associated with non-syndromic intellectual disabilities. We previously reported that Acsl, the Drosophila ortholog of mammalian ACSL3 and ACSL4, inhibits neuromuscular synapse growth by suppressing bone morphogenetic protein (BMP) signaling. Here, we report that Acsl regulates the composition of fatty acids and membrane lipids, which in turn affects neuromuscular junction (NMJ) synapse development. Acsl mutant brains had a decreased abundance of C16:1 fatty acyls; restoration of Acsl expression abrogated NMJ overgrowth and the increase in BMP signaling. A lipidomic analysis revealed that Acsl suppressed the levels of three lipid raft components in the brain, including mannosyl glucosylceramide (MacCer), phosphoethanolamine ceramide and ergosterol. The MacCer level was elevated in Acsl mutant NMJs and, along with sterol, promoted NMJ overgrowth, but was not associated with the increase in BMP signaling in the mutants. These findings suggest that Acsl inhibits NMJ growth by stimulating C16:1 fatty acyl production and concomitantly suppressing raft-associated lipid levels.
Collapse
Affiliation(s)
- Yan Huang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Sheng Huang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.,Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Sin Man Lam
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhihua Liu
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Guanghou Shui
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yong Q Zhang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
38
|
Wolbachia Modulates Lipid Metabolism in Aedes albopictus Mosquito Cells. Appl Environ Microbiol 2016; 82:3109-3120. [PMID: 26994075 PMCID: PMC4959074 DOI: 10.1128/aem.00275-16] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/10/2016] [Indexed: 12/11/2022] Open
Abstract
Certain strains of the intracellular endosymbiont Wolbachia can strongly inhibit or block the transmission of viruses such as dengue virus (DENV) by Aedes mosquitoes, and the mechanisms responsible are still not well understood. Direct infusion and liquid chromatography-Fourier transform ion cyclotron resonance (FT-ICR) mass spectrometry-based lipidomics analyses were conducted using Aedes albopictus Aa23 cells that were infected with the wMel and wMelPop strains of Wolbachia in comparison to uninfected Aa23-T cells. Substantial shifts in the cellular lipid profile were apparent in the presence of Wolbachia. Most significantly, almost all sphingolipid classes were depleted, and some reductions in diacylglycerols and phosphatidylcholines were also observed. These lipid classes have previously been shown to be selectively enriched in DENV-infected mosquito cells, suggesting that Wolbachia may produce a cellular lipid environment that is antagonistic to viral replication. The data improve our understanding of the intracellular interactions between Wolbachia and mosquitoes. IMPORTANCE Mosquitoes transmit a variety of important viruses to humans, such as dengue virus and Zika virus. Certain strains of the intracellular bacterial genus called Wolbachia found in or introduced into mosquitoes can block the transmission of viruses, including dengue virus, but the mechanisms responsible are not well understood. We found substantial shifts in the cellular lipid profiles in the presence of these bacteria. Some lipid classes previously shown to be enriched in dengue virus-infected mosquito cells were depleted in the presence of Wolbachia, suggesting that Wolbachia may produce a cellular lipid environment that inhibits mosquito-borne viruses.
Collapse
|
39
|
Stalinski R, Laporte F, Després L, Tetreau G. Alkaline phosphatases are involved in the response ofAedes aegyptilarvae to intoxication withBacillus thuringiensissubsp.israelensis Cry toxins. Environ Microbiol 2016; 18:1022-36. [DOI: 10.1111/1462-2920.13186] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 12/08/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Renaud Stalinski
- Laboratoire d'Ecologie Alpine LECA UMR5553; Université Grenoble Alpes; F-38000 Grenoble France
- Laboratoire d'Ecologie Alpine LECA UMR5553; Centre National de la Recherche Scientifique; F-38000 Grenoble France
| | - Frédéric Laporte
- Laboratoire d'Ecologie Alpine LECA UMR5553; Université Grenoble Alpes; F-38000 Grenoble France
- Laboratoire d'Ecologie Alpine LECA UMR5553; Centre National de la Recherche Scientifique; F-38000 Grenoble France
| | - Laurence Després
- Laboratoire d'Ecologie Alpine LECA UMR5553; Université Grenoble Alpes; F-38000 Grenoble France
- Laboratoire d'Ecologie Alpine LECA UMR5553; Centre National de la Recherche Scientifique; F-38000 Grenoble France
| | - Guillaume Tetreau
- Laboratoire d'Ecologie Alpine LECA UMR5553; Université Grenoble Alpes; F-38000 Grenoble France
- Laboratoire d'Ecologie Alpine LECA UMR5553; Centre National de la Recherche Scientifique; F-38000 Grenoble France
| |
Collapse
|
40
|
Function of Membrane-Associated Proteoglycans in the Regulation of Satellite Cell Growth. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 900:61-95. [DOI: 10.1007/978-3-319-27511-6_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
41
|
The many lives of SHH in limb development and evolution. Semin Cell Dev Biol 2016; 49:116-24. [DOI: 10.1016/j.semcdb.2015.12.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/21/2015] [Accepted: 12/23/2015] [Indexed: 01/17/2023]
|
42
|
Koleva MV, Rothery S, Spitaler M, Neil MAA, Magee AI. Sonic hedgehog multimerization: a self-organizing event driven by post-translational modifications? Mol Membr Biol 2015; 32:65-74. [PMID: 26312641 DOI: 10.3109/09687688.2015.1066895] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 06/02/2015] [Indexed: 01/05/2023]
Abstract
Sonic hedgehog (Shh) is a morphogen active during vertebrate development and tissue homeostasis in adulthood. Dysregulation of the Shh signalling pathway is known to incite carcinogenesis. Due to the highly lipophilic nature of this protein imparted by two post-translational modifications, Shh's method of transit through the aqueous extracellular milieu has been a long-standing conundrum, prompting the proposition of numerous hypotheses to explain the manner of its displacement from the surface of the producing cell. Detection of high molecular-weight complexes of Shh in the intercellular environment has indicated that the protein achieves this by accumulating into multimeric structures prior to release from producing cells. The mechanism of assembly of the multimers, however, has hitherto remained mysterious and contentious. Here, with the aid of high-resolution optical imaging and post-translational modification mutants of Shh, we show that the C-terminal cholesterol and the N-terminal palmitate adducts contribute to the assembly of large multimers and regulate their shape. Moreover, we show that small Shh multimers are produced in the absence of any lipid modifications. Based on an assessment of the distribution of various dimensional characteristics of individual Shh clusters, in parallel with deductions about the kinetics of release of the protein from the producing cells, we conclude that multimerization is driven by self-assembly underpinned by the law of mass action. We speculate that the lipid modifications augment the size of the multimolecular complexes through prolonging their association with the exoplasmic membrane.
Collapse
Affiliation(s)
- Mirella V Koleva
- a Department of Chemistry
- b Institute of Chemical Biology
- c Photonics Group, Department of Physics
- d Molecular Medicine Section, National Heart & Lung Institute , and
| | - Stephen Rothery
- e Facility for Imaging by Light Microscopy, Imperial College London , London , UK
| | - Martin Spitaler
- e Facility for Imaging by Light Microscopy, Imperial College London , London , UK
| | - Mark A A Neil
- b Institute of Chemical Biology
- c Photonics Group, Department of Physics
| | - Anthony I Magee
- b Institute of Chemical Biology
- d Molecular Medicine Section, National Heart & Lung Institute , and
| |
Collapse
|
43
|
Lorent JH, Levental I. Structural determinants of protein partitioning into ordered membrane domains and lipid rafts. Chem Phys Lipids 2015; 192:23-32. [PMID: 26241883 DOI: 10.1016/j.chemphyslip.2015.07.022] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 07/27/2015] [Accepted: 07/29/2015] [Indexed: 11/16/2022]
Abstract
Increasing evidence supports the existence of lateral nanoscopic lipid domains in plasma membranes, known as lipid rafts. These domains preferentially recruit membrane proteins and lipids to facilitate their interactions and thereby regulate transmembrane signaling and cellular homeostasis. The functionality of raft domains is intrinsically dependent on their selectivity for specific membrane components; however, while the physicochemical determinants of raft association for lipids are known, very few systematic studies have focused on the structural aspects that guide raft partitioning of proteins. In this review, we describe biophysical and thermodynamic aspects of raft-mimetic liquid ordered phases, focusing on those most relevant for protein partitioning. Further, we detail the variety of experimental models used to study protein-raft interactions. Finally, we review the existing literature on mechanisms for raft targeting, including lipid post-translational modifications, lipid binding, and transmembrane domain features. We conclude that while protein palmitoylation is a clear raft-targeting signal, few other general structural determinants for raft partitioning have been revealed, suggesting that many discoveries lie ahead in this burgeoning field.
Collapse
Affiliation(s)
- Joseph Helmuth Lorent
- Department for Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, USA
| | - Ilya Levental
- Department for Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, USA.
| |
Collapse
|
44
|
Lin YC, Roffler SR, Yan YT, Yang RB. Disruption of Scube2 Impairs Endochondral Bone Formation. J Bone Miner Res 2015; 30:1255-67. [PMID: 25639508 DOI: 10.1002/jbmr.2451] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 12/17/2014] [Accepted: 01/05/2015] [Indexed: 11/11/2022]
Abstract
Signal peptide-CUB-EGF domain-containing protein 2 (SCUBE2) belongs to a secreted and membrane-tethered multidomain SCUBE protein family composed of three members found in vertebrates and mammals. Recent reports suggested that zebrafish scube2 could facilitate sonic hedgehog (Shh) signaling for proper development of slow muscle. However, whether SCUBE2 can regulate the signaling activity of two other hedgehog ligands (Ihh and Dhh), and the developmental relevance of the SCUBE2-induced hedgehog signaling in mammals remain poorly understood. In this study, we first showed that as compared with SCUBE1 or SCUBE3, SCUBE2 is the most potent modulator of IHH signaling in vitro. In addition, gain and loss-of-function studies demonstrated that SCUBE2 exerted an osteogenic function by enhancing Ihh-stimulated osteoblast differentiation in the mouse mesenchymal progenitor cells. Consistent with these in vitro studies and the prominent roles of Ihh in coordinating skeletogenesis, genetic ablation of Scube2 (-/-) caused defective endochondral bone formation and impaired Ihh-mediated chondrocyte differentiation and proliferation as well as osteoblast differentiation of -/- bone-marrow mesenchymal stromal-cell cultures. Our data demonstrate that Scube2 plays a key regulatory role in Ihh-dependent endochondral bone formation.
Collapse
Affiliation(s)
- Yuh-Charn Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yu-Ting Yan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ruey-Bing Yang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
45
|
Ortmann C, Pickhinke U, Exner S, Ohlig S, Lawrence R, Jboor H, Dreier R, Grobe K. Sonic hedgehog processing and release are regulated by glypican heparan sulfate proteoglycans. J Cell Sci 2015; 128:2374-85. [PMID: 25967551 DOI: 10.1242/jcs.170670] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/05/2015] [Indexed: 12/21/2022] Open
Abstract
All Hedgehog morphogens are released from producing cells, despite being synthesized as N- and C-terminally lipidated molecules, a modification that firmly tethers them to the cell membrane. We have previously shown that proteolytic removal of both lipidated peptides, called shedding, releases bioactive Sonic hedgehog (Shh) morphogens from the surface of transfected Bosc23 cells. Using in vivo knockdown together with in vitro cell culture studies, we now show that glypican heparan sulfate proteoglycans regulate this process, through their heparan sulfate chains, in a cell autonomous manner. Heparan sulfate specifically modifies Shh processing at the cell surface, and purified glycosaminoglycans enhance the proteolytic removal of N- and C-terminal Shh peptides under cell-free conditions. The most likely explanation for these observations is direct Shh processing in the extracellular compartment, suggesting that heparan sulfate acts as a scaffold or activator for Shh ligands and the factors required for their turnover. We also show that purified heparan sulfate isolated from specific cell types and tissues mediates the release of bioactive Shh from pancreatic cancer cells, revealing a previously unknown regulatory role for these versatile molecules in a pathological context.
Collapse
Affiliation(s)
- Corinna Ortmann
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Ute Pickhinke
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Sebastian Exner
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Stefanie Ohlig
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Roger Lawrence
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Hamodah Jboor
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Rita Dreier
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany
| | - Kay Grobe
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany
| |
Collapse
|
46
|
Abstract
Cholesterylation is a post-translational attachment of sterol to proteins. This modification has been a characteristic of a single family of hedgehog proteins (Hh). Hh is a well-established morphogenic molecule important in embryonic development. It was also found to be involved in the progression of many cancer types. Herein, we describe the mechanism of biosynthesis of cholesterylated Hh, the role of this unusual modification on protein functions and novel chemical probes, which could be used to specifically target this modification, both in vitro and in vivo.
Collapse
|
47
|
Long J, Tokhunts R, Old WM, Houel S, Rodgriguez-Blanco J, Singh S, Schilling N, J Capobianco A, Ahn NG, Robbins DJ. Identification of a family of fatty-acid-speciated sonic hedgehog proteins, whose members display differential biological properties. Cell Rep 2015; 10:1280-1287. [PMID: 25732819 DOI: 10.1016/j.celrep.2015.01.058] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 01/14/2015] [Accepted: 01/26/2015] [Indexed: 01/25/2023] Open
Abstract
Hedgehog (HH) proteins are proteolytically processed into a biologically active form that is covalently modified by cholesterol and palmitate. However, most studies of HH biogenesis have characterized protein from cells in which HH is overexpressed. We purified Sonic Hedgehog (SHH) from cells expressing physiologically relevant levels and showed that it was more potent than SHH isolated from overexpressing cells. Furthermore, the SHH in our preparations was modified with a diverse spectrum of fatty acids on its amino termini, and this spectrum of fatty acids varied dramatically depending on the growth conditions of the cells. The fatty acid composition of SHH affected its trafficking to lipid rafts as well as its potency. Our results suggest that HH proteins exist as a family of diverse lipid-speciated proteins that might be altered in different physiological and pathological contexts in order to regulate distinct properties of HH proteins.
Collapse
Affiliation(s)
- Jun Long
- Molecular Oncology Program, The DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136.,The Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida 33136
| | - Robert Tokhunts
- Molecular Oncology Program, The DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136.,Program in Experimental and Molecular Medicine, Dartmouth Medical School, Hanover, New Hampshire 03755
| | - William M Old
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado 80309
| | - Stephane Houel
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, Colorado 80309
| | - Jezabel Rodgriguez-Blanco
- Molecular Oncology Program, The DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136
| | - Samer Singh
- Molecular Oncology Program, The DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136
| | - Neal Schilling
- Molecular Oncology Program, The DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136.,Program in Experimental and Molecular Medicine, Dartmouth Medical School, Hanover, New Hampshire 03755
| | - Anthony J Capobianco
- Molecular Oncology Program, The DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136.,Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida 33136.,Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136
| | - Natalie G Ahn
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, Colorado 80309.,Howard Hughes Medical Institute, University of Colorado, Boulder, Colorado 80309
| | - David J Robbins
- Molecular Oncology Program, The DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136.,Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida 33136.,Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136
| |
Collapse
|
48
|
Després L, Stalinski R, Tetreau G, Paris M, Bonin A, Navratil V, Reynaud S, David JP. Gene expression patterns and sequence polymorphisms associated with mosquito resistance to Bacillus thuringiensis israelensis toxins. BMC Genomics 2014; 15:926. [PMID: 25341495 PMCID: PMC4223840 DOI: 10.1186/1471-2164-15-926] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 10/16/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Despite the intensive use of Bacillus thuringiensis israelensis (Bti) toxins for mosquito control, little is known about the long term effect of exposure to this cocktail of toxins on target mosquito populations. In contrast to the many cases of resistance to Bacillus thuringiensis Cry toxins observed in other insects, there is no evidence so far for Bti resistance evolution in field mosquito populations. High fitness costs measured in a Bti selected mosquito laboratory strain suggest that evolving resistance to Bti is costly. The aim of the present study was to identify transcription level and polymorphism variations associated with resistance to Bti toxins in the dengue vector Aedes aegypti. We used RNA sequencing (RNA-seq) for comparing a laboratory-selected strain showing elevated resistance to Bti toxins and its parental non-selected susceptible strain. As the resistant strain displayed two marked larval development phenotypes (slow and normal), each phenotype was analyzed separately in order to evidence potential links between resistance mechanisms and mosquito life-history traits. RESULTS A total of 12,458 genes were detected of which 844 were differentially transcribed between the resistant and susceptible strains. Polymorphism analysis revealed a total of 68,541 SNPs of which 12,571 SNPs exhibited more than 40% frequency difference between the resistant and susceptible strains, affecting 2,953 genes. Bti resistance is associated with changes in the transcription level of enzymes involved in detoxification and chitin metabolism. Among previously described Bti-toxin receptors, four alkaline phosphatases (ALPs) were differentially transcribed between resistant and susceptible larvae, and non-synonymous changes affected the protein sequence of one cadherin, six aminopeptidases (APNs) and four α-amylases. Other putative Cry receptors located in lipid rafts, such as flotillin and glycoside hydrolases, were under-transcribed and/or contained non-synonymous substitutions. Finally, immunity-related genes showed contrasted transcription and polymorphisms patterns between the two developmental resistant phenotypes, suggesting the existence of trade-offs between Bti-resistance, life-history traits and immunity. CONCLUSIONS The present study is the first to analyze the whole transcriptome of Bti-resistant mosquitoes by RNA-seq, shedding light on the importance of studying both transcription levels and sequence polymorphism variations to get a comprehensive view of insecticide resistance.
Collapse
Affiliation(s)
- Laurence Després
- Université Grenoble Alpes, Laboratoire d'Ecologie Alpine UMR5553, Grenoble, France.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Kornberg TB. The contrasting roles of primary cilia and cytonemes in Hh signaling. Dev Biol 2014; 394:1-5. [PMID: 25072627 DOI: 10.1016/j.ydbio.2014.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 07/11/2014] [Accepted: 07/19/2014] [Indexed: 12/16/2022]
Abstract
Hedgehog (Hh) is a paracrine signaling protein with major roles in development and disease. In vertebrates and invertebrates, Hh signal transduction is carried out almost entirely by evolutionarily conserved components, and in both, intercellular movement of Hh is mediated by cytonemes - specialized filopodia that serve as bridges that bring distant cells into contact. A significant difference is the role of the primary cilium, a slender, tubulin-based protuberance of many vertebrate cells. Although the primary cilium is essential for Hh signaling in cells that have one, most Drosophila cells lack a primary cilium. This perspective addresses the roles of primary cilia and cytonemes, and proposes that for Hh signaling, the role of primary cilia is to provide a specialized hydrophobic environment that hosts lipid-modified Hh and other components of Hh signal transduction after Hh has traveled from elsewhere in the cell. Implicit in this model is the idea that initial binding and uptake of Hh is independent of and segregated from the processes of signal transduction and activation.
Collapse
Affiliation(s)
- Thomas B Kornberg
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, United States.
| |
Collapse
|
50
|
Guerrero I, Kornberg TB. Hedgehog and its circuitous journey from producing to target cells. Semin Cell Dev Biol 2014; 33:52-62. [PMID: 24994598 DOI: 10.1016/j.semcdb.2014.06.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 06/23/2014] [Indexed: 12/12/2022]
Abstract
The hedgehog (Hh) signaling protein has essential roles in the growth, development and regulation of many vertebrate and invertebrate organs. The processes that make Hh and prepare it for release from producing cells and that move it to target cells are both diverse and complex. This article reviews the essential features of these processes and highlights recent work that provides a novel framework to understand how these processes contribute to an integrated pathway.
Collapse
Affiliation(s)
- Isabel Guerrero
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Cantoblanco, E-28049 Madrid, Spain.
| | - Thomas B Kornberg
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|