Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.949
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 89: 741-768, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32569526

ABSTRACT

Complex carbohydrates are essential for many biological processes, from protein quality control to cell recognition, energy storage, and cell wall formation. Many of these processes are performed in topologically extracellular compartments or on the cell surface; hence, diverse secretion systems evolved to transport the hydrophilic molecules to their sites of action. Polyprenyl lipids serve as ubiquitous anchors and facilitators of these transport processes. Here, we summarize and compare bacterial biosynthesis pathways relying on the recognition and transport of lipid-linked complex carbohydrates. In particular, we compare transporters implicated in O antigen and capsular polysaccharide biosyntheses with those facilitating teichoic acid and N-linked glycan transport. Further, we discuss recent insights into the generation, recognition, and recycling of polyprenyl lipids.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Glycolipids/biosynthesis , O Antigens/biosynthesis , Polyprenols/metabolism , Transferases (Other Substituted Phosphate Groups)/chemistry , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Transport , Carbon-Oxygen Ligases/chemistry , Carbon-Oxygen Ligases/genetics , Carbon-Oxygen Ligases/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Glycosyltransferases/chemistry , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Models, Molecular , Protein Structure, Secondary , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Teichoic Acids/metabolism , Transferases (Other Substituted Phosphate Groups)/genetics , Transferases (Other Substituted Phosphate Groups)/metabolism
2.
Cell ; 181(2): 293-305.e11, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32142653

ABSTRACT

Pulmonary tuberculosis, a disease caused by Mycobacterium tuberculosis (Mtb), manifests with a persistent cough as both a primary symptom and mechanism of transmission. The cough reflex can be triggered by nociceptive neurons innervating the lungs, and some bacteria produce neuron-targeting molecules. However, how pulmonary Mtb infection causes cough remains undefined, and whether Mtb produces a neuron-activating, cough-inducing molecule is unknown. Here, we show that an Mtb organic extract activates nociceptive neurons in vitro and identify the Mtb glycolipid sulfolipid-1 (SL-1) as the nociceptive molecule. Mtb organic extracts from mutants lacking SL-1 synthesis cannot activate neurons in vitro or induce cough in a guinea pig model. Finally, Mtb-infected guinea pigs cough in a manner dependent on SL-1 synthesis. Thus, we demonstrate a heretofore unknown molecular mechanism for cough induction by a virulent human pathogen via its production of a complex lipid.


Subject(s)
Cough/physiopathology , Glycolipids/metabolism , Nociceptors/physiology , Virulence Factors/metabolism , Adult , Animals , Cell Line , Cough/etiology , Cough/microbiology , Female , Glycolipids/physiology , Guinea Pigs , Host-Pathogen Interactions , Humans , Lipids/physiology , Lung/microbiology , Macrophages/microbiology , Male , Mice , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Primary Cell Culture , Tuberculosis/microbiology , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/physiopathology , Virulence Factors/physiology
3.
Cell ; 183(5): 1312-1324.e10, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33212011

ABSTRACT

Interferon (IFN)-Is are crucial mediators of antiviral immunity and homeostatic immune system regulation. However, the source of IFN-I signaling under homeostatic conditions is unclear. We discovered that commensal microbes regulate the IFN-I response through induction of IFN-ß by colonic DCs. Moreover, the mechanism by which a specific commensal microbe induces IFN-ß was identified. Outer membrane (OM)-associated glycolipids of gut commensal microbes belonging to the Bacteroidetes phylum induce expression of IFN-ß. Using Bacteroides fragilis and its OM-associated polysaccharide A, we determined that IFN-ß expression was induced via TLR4-TRIF signaling. Antiviral activity of this purified microbial molecule against infection with either vesicular stomatitis virus (VSV) or influenza was demonstrated to be dependent on the induction of IFN-ß. In a murine VSV infection model, commensal-induced IFN-ß regulated natural resistance to virus infection. Due to the physiological importance of IFN-Is, discovery of an IFN-ß-inducing microbial molecule represents a potential approach for the treatment of some human diseases.


Subject(s)
Immunity, Innate , Microbiota , Virus Diseases/microbiology , Animals , Bacteroides fragilis/physiology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Colon/pathology , Colon/virology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Female , Gene Expression Regulation/drug effects , Glycolipids/metabolism , Immunity, Innate/drug effects , Interferon-beta/blood , Interferon-beta/metabolism , Male , Mice, Inbred C57BL , Microbiota/drug effects , Polysaccharides, Bacterial/pharmacology , Toll-Like Receptor 4/metabolism , Vesiculovirus/physiology , Virus Diseases/genetics
4.
Annu Rev Biochem ; 88: 85-111, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30901263

ABSTRACT

Membrane proteins that exist in lipid bilayers are not isolated molecular entities. The lipid molecules that surround them play crucial roles in maintaining their full structural and functional integrity. Research directed at investigating these critical lipid-protein interactions is developing rapidly. Advancements in both instrumentation and software, as well as in key biophysical and biochemical techniques, are accelerating the field. In this review, we provide a brief outline of structural techniques used to probe protein-lipid interactions and focus on the molecular aspects of these interactions obtained from native mass spectrometry (native MS). We highlight examples in which lipids have been shown to modulate membrane protein structure and show how native MS has emerged as a complementary technique to X-ray crystallography and cryo-electron microscopy. We conclude with a short perspective on future developments that aim to better understand protein-lipid interactions in the native environment.


Subject(s)
Glycerophospholipids/metabolism , Glycolipids/metabolism , Mass Spectrometry/methods , Membrane Proteins/metabolism , Sphingolipids/metabolism , Sterols/metabolism , Bacteria/chemistry , Bacteria/metabolism , Binding Sites , Cell Membrane/chemistry , Cell Membrane/metabolism , Cryoelectron Microscopy/instrumentation , Cryoelectron Microscopy/methods , Fungi/chemistry , Fungi/metabolism , Glycerophospholipids/chemistry , Glycolipids/chemistry , Magnetic Resonance Spectroscopy/instrumentation , Magnetic Resonance Spectroscopy/methods , Mass Spectrometry/instrumentation , Membrane Proteins/chemistry , Membrane Proteins/ultrastructure , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Sphingolipids/chemistry , Sterols/chemistry
5.
Cell ; 170(5): 973-985.e10, 2017 Aug 24.
Article in English | MEDLINE | ID: mdl-28841420

ABSTRACT

Mycobacterium leprae causes leprosy and is unique among mycobacterial diseases in producing peripheral neuropathy. This debilitating morbidity is attributed to axon demyelination resulting from direct interaction of the M. leprae-specific phenolic glycolipid 1 (PGL-1) with myelinating glia and their subsequent infection. Here, we use transparent zebrafish larvae to visualize the earliest events of M. leprae-induced nerve damage. We find that demyelination and axonal damage are not directly initiated by M. leprae but by infected macrophages that patrol axons; demyelination occurs in areas of intimate contact. PGL-1 confers this neurotoxic response on macrophages: macrophages infected with M. marinum-expressing PGL-1 also damage axons. PGL-1 induces nitric oxide synthase in infected macrophages, and the resultant increase in reactive nitrogen species damages axons by injuring their mitochondria and inducing demyelination. Our findings implicate the response of innate macrophages to M. leprae PGL-1 in initiating nerve damage in leprosy.


Subject(s)
Antigens, Bacterial/metabolism , Disease Models, Animal , Glycolipids/metabolism , Leprosy/microbiology , Leprosy/pathology , Macrophages/immunology , Mycobacterium leprae/physiology , Animals , Axons/metabolism , Axons/pathology , Demyelinating Diseases , Larva/growth & development , Leprosy/immunology , Mycobacterium marinum/metabolism , Myelin Sheath/chemistry , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Neuroglia/metabolism , Neuroglia/pathology , Nitric Oxide/metabolism , Zebrafish
6.
Annu Rev Biochem ; 85: 599-630, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27145845

ABSTRACT

Glycoscience research has been significantly impeded by the complex compositions of the glycans present in biological molecules and the lack of convenient tools suitable for studying the glycosylation process and its function. Polysaccharides and glycoconjugates are not encoded directly by genes; instead, their biosynthesis relies on the differential expression of carbohydrate enzymes, resulting in heterogeneous mixtures of glycoforms, each with a distinct physiological activity. Access to well-defined structures is required for functional study, and this has been provided by chemical and enzymatic synthesis and by the engineering of glycosylation pathways. This review covers general methods for preparing glycans commonly found in mammalian systems and applying them to the synthesis of therapeutically significant glycoconjugates (glycosaminoglycans, glycoproteins, glycolipids, glycosylphosphatidylinositol-anchored proteins) and the development of carbohydrate-based vaccines.


Subject(s)
Glycoconjugates/chemical synthesis , Glycoproteins/chemical synthesis , Glycosaminoglycans/chemical synthesis , Haemophilus Infections/prevention & control , Haemophilus Vaccines/administration & dosage , Polysaccharides/chemical synthesis , Amino Acid Sequence , Carbohydrate Conformation , Carbohydrate Sequence , Glycoconjugates/immunology , Glycolipids/chemical synthesis , Glycolipids/immunology , Glycoproteins/immunology , Glycosaminoglycans/immunology , Glycosylation , Glycosylphosphatidylinositols/chemical synthesis , Glycosylphosphatidylinositols/immunology , Haemophilus Infections/immunology , Haemophilus Infections/microbiology , Haemophilus Vaccines/chemical synthesis , Haemophilus influenzae type b/drug effects , Haemophilus influenzae type b/growth & development , Haemophilus influenzae type b/pathogenicity , Humans , Polysaccharides/immunology
7.
Nature ; 628(8009): 901-909, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38570679

ABSTRACT

Capsular polysaccharides (CPSs) fortify the cell boundaries of many commensal and pathogenic bacteria1. Through the ABC-transporter-dependent biosynthesis pathway, CPSs are synthesized intracellularly on a lipid anchor and secreted across the cell envelope by the KpsMT ABC transporter associated with the KpsE and KpsD subunits1,2. Here we use structural and functional studies to uncover crucial steps of CPS secretion in Gram-negative bacteria. We show that KpsMT has broad substrate specificity and is sufficient for the translocation of CPSs across the inner bacterial membrane, and we determine the cell surface organization and localization of CPSs using super-resolution fluorescence microscopy. Cryo-electron microscopy analyses of the KpsMT-KpsE complex in six different states reveal a KpsE-encaged ABC transporter, rigid-body conformational rearrangements of KpsMT during ATP hydrolysis and recognition of a glycolipid inside a membrane-exposed electropositive canyon. In vivo CPS secretion assays underscore the functional importance of canyon-lining basic residues. Combined, our analyses suggest a molecular model of CPS secretion by ABC transporters.


Subject(s)
Bacterial Capsules , Escherichia coli Proteins , Escherichia coli , Polysaccharides, Bacterial , Adenosine Triphosphate/metabolism , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/metabolism , ATP-Binding Cassette Transporters/ultrastructure , Bacterial Capsules/metabolism , Bacterial Capsules/chemistry , Bacterial Capsules/ultrastructure , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cryoelectron Microscopy , Escherichia coli/chemistry , Escherichia coli/metabolism , Escherichia coli/ultrastructure , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/ultrastructure , Glycolipids/chemistry , Glycolipids/metabolism , Hydrolysis , Microscopy, Fluorescence , Models, Molecular , Polysaccharides, Bacterial/metabolism , Polysaccharides, Bacterial/chemistry , Substrate Specificity
8.
Annu Rev Biochem ; 83: 99-128, 2014.
Article in English | MEDLINE | ID: mdl-24580642

ABSTRACT

Lipopolysaccharide molecules represent a unique family of glycolipids based on a highly conserved lipid moiety known as lipid A. These molecules are produced by most gram-negative bacteria, in which they play important roles in the integrity of the outer-membrane permeability barrier and participate extensively in host-pathogen interplay. Few bacteria contain lipopolysaccharide molecules composed only of lipid A. In most forms, lipid A is glycosylated by addition of the core oligosaccharide that, in some bacteria, provides an attachment site for a long-chain O-antigenic polysaccharide. The complexity of lipopolysaccharide structures is reflected in the processes used for their biosynthesis and export. Rapid growth and cell division depend on the bacterial cell's capacity to synthesize and export lipopolysaccharide efficiently and in large amounts. We review recent advances in those processes, emphasizing the reactions that are essential for viability.


Subject(s)
Lipopolysaccharides/biosynthesis , Lipopolysaccharides/metabolism , Adenosine Triphosphate/metabolism , Bacteria , Bacterial Physiological Phenomena , Bacterial Proteins/metabolism , Biological Transport , Cell Membrane/metabolism , Glycolipids/metabolism , Glycosylation , Gram-Negative Bacteria/metabolism , O Antigens/metabolism , Permeability , Polysaccharides/metabolism
9.
Nature ; 613(7943): 324-331, 2023 01.
Article in English | MEDLINE | ID: mdl-36599989

ABSTRACT

Pathogens generate ubiquitous selective pressures and host-pathogen interactions alter social behaviours in many animals1-4. However, very little is known about the neuronal mechanisms underlying pathogen-induced changes in social behaviour. Here we show that in adult Caenorhabditis elegans hermaphrodites, exposure to a bacterial pathogen (Pseudomonas aeruginosa) modulates sensory responses to pheromones by inducing the expression of the chemoreceptor STR-44 to promote mating. Under standard conditions, C. elegans hermaphrodites avoid a mixture of ascaroside pheromones to facilitate dispersal5-13. We find that exposure to the pathogenic Pseudomonas bacteria enables pheromone responses in AWA sensory neurons, which mediate attractive chemotaxis, to suppress the avoidance. Pathogen exposure induces str-44 expression in AWA neurons, a process regulated by a transcription factor zip-5 that also displays a pathogen-induced increase in expression in AWA. STR-44 acts as a pheromone receptor and its function in AWA neurons is required for pathogen-induced AWA pheromone response and suppression of pheromone avoidance. Furthermore, we show that C. elegans hermaphrodites, which reproduce mainly through self-fertilization, increase the rate of mating with males after pathogen exposure and that this increase requires str-44 in AWA neurons. Thus, our results uncover a causal mechanism for pathogen-induced social behaviour plasticity, which can promote genetic diversity and facilitate adaptation of the host animals.


Subject(s)
Caenorhabditis elegans , Pheromones , Pseudomonas aeruginosa , Reproduction , Sexual Behavior, Animal , Animals , Female , Male , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/metabolism , Glycolipids/metabolism , Hermaphroditic Organisms/physiology , Pheromones/metabolism , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/physiology , Receptors, Pheromone/metabolism , Reproduction/physiology , Sensory Receptor Cells/metabolism
10.
Mol Cell ; 81(21): 4540-4551.e6, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34433091

ABSTRACT

Within the extensive range of self-propagating pathologic protein aggregates of mammals, prions are the most clearly infectious (e.g., ∼109 lethal doses per milligram). The structures of such lethal assemblies of PrP molecules have been poorly understood. Here we report a near-atomic core structure of a brain-derived, fully infectious prion (263K strain). Cryo-electron microscopy showed amyloid fibrils assembled with parallel in-register intermolecular ß sheets. Each monomer provides one rung of the ordered fibril core, with N-linked glycans and glycolipid anchors projecting outward. Thus, single monomers form the templating surface for incoming monomers at fibril ends, where prion growth occurs. Comparison to another prion strain (aRML) revealed major differences in fibril morphology but, like 263K, an asymmetric fibril cross-section without paired protofilaments. These findings provide structural insights into prion propagation, strains, species barriers, and membrane pathogenesis. This structure also helps frame considerations of factors influencing the relative transmissibility of other pathologic amyloids.


Subject(s)
Brain/metabolism , Cryoelectron Microscopy/methods , Polysaccharides/chemistry , Prions/chemistry , Prions/ultrastructure , Amyloid/chemistry , Animals , Glycolipids/chemistry , Humans , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Mice , Phenotype , Prion Proteins/chemistry , Protein Binding , Protein Structure, Secondary , Thermodynamics
11.
Immunity ; 47(3): 395-397, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28930653

ABSTRACT

Two recent studies (Cambier et al., 2017; Madigan et al., 2017) reveal in vivo functions for specific phenolic glycolipids (PGLs) in the mycobacteria that cause tuberculosis or leprosy. M. tuberculosis (and M. marinum) PGL promotes bacterial spread to growth-permissive macrophages, while M. leprae PGL-1 induces macrophages to cause nerve demyelination characteristic of human leprosy.


Subject(s)
Antigens, Bacterial , Mycobacterium leprae , Glycolipids , Humans , Leprosy/microbiology , Mycobacterium tuberculosis
12.
Immunity ; 47(3): 552-565.e4, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28844797

ABSTRACT

Mycobacterium tuberculosis (Mtb) enters the host in aerosol droplets deposited in lung alveoli, where the bacteria first encounter lung-resident alveolar macrophages. We studied the earliest mycobacterium-macrophage interactions in the optically transparent zebrafish. First-responding resident macrophages phagocytosed and eradicated infecting mycobacteria, suggesting that to establish a successful infection, mycobacteria must escape out of the initially infected resident macrophage into growth-permissive monocytes. We defined a critical role for mycobacterial membrane phenolic glycolipid (PGL) in engineering this transition. PGL activated the STING cytosolic sensing pathway in resident macrophages, inducing the production of the chemokine CCL2, which in turn recruited circulating CCR2+ monocytes toward infection. Transient fusion of infected macrophages with CCR2+ monocytes enabled bacterial transfer and subsequent dissemination, and interrupting this transfer so as to prolong mycobacterial sojourn in resident macrophages promoted clearing of infection. Human alveolar macrophages produced CCL2 in a PGL-dependent fashion following infection, arguing for the potential of PGL-blocking interventions or PGL-targeting vaccine strategies in the prevention of tuberculosis. VIDEO ABSTRACT.


Subject(s)
Glycolipids/immunology , Macrophages/microbiology , Macrophages/physiology , Mycobacterium tuberculosis/immunology , Animals , Chemokine CCL2/metabolism , Chemotaxis/immunology , Cytokines/metabolism , Disease Models, Animal , Gene Knockout Techniques , Humans , Inflammation Mediators/metabolism , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/physiology , Membrane Proteins/metabolism , Monocytes/immunology , Monocytes/metabolism , Monocytes/microbiology , Mutation , Mycobacterium tuberculosis/genetics , Organ Specificity/immunology , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/microbiology , Zebrafish
13.
Annu Rev Biochem ; 79: 619-53, 2010.
Article in English | MEDLINE | ID: mdl-20380561

ABSTRACT

Glycans are ubiquitous components of all organisms. Efforts to elucidate glycan function and to understand how they are assembled and disassembled can reap benefits in fields ranging from bioenergy to human medicine. Significant advances in our knowledge of glycan biosynthesis and function are emerging, and chemical biology approaches are accelerating the pace of discovery. Novel strategies for assembling oligosaccharides, glycoproteins, and other glycoconjugates are providing access to critical materials for interrogating glycan function. Chemoselective reactions that facilitate the synthesis of glycan-substituted imaging agents, arrays, and materials are yielding compounds to interrogate and perturb glycan function and dysfunction. To complement these advances, small molecules are being generated that inhibit key glycan-binding proteins or biosynthetic enzymes. These examples illustrate how chemical glycobiology is providing new insight into the functional roles of glycans and new opportunities to interfere with or exploit these roles.


Subject(s)
Glycolipids/metabolism , Glycoproteins/chemistry , Polysaccharides/chemistry , Animals , Glycomics/methods , Glycopeptides/chemistry , Glycosyltransferases/metabolism , Humans
14.
Hum Mol Genet ; 32(15): 2464-2472, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37145097

ABSTRACT

Fabry disease stems from a deficiency of alpha-galactosidase and results in the accumulation of globotriaosylceramide (Gb3). However, the production of its deacylated form globotriaosylsphingosine (lyso-Gb3) is also observed and its plasma levels have closer association with disease severity. Studies have shown that lyso-Gb3 directly affects podocytes and causes sensitisation of peripheral nociceptive neurons. However, little is understood of the mechanisms of this cytotoxicity. To study the effect on neuronal cells, we incubated SH-Sy5y cells with lyso-Gb3 at low (20 ng/mL) and high (200 ng/mL) levels, to mimic mild and classical FD serum levels. We used glucosylsphingosine as a positive control to determine specific effects of lyso-Gb3. Proteomic analyses revealed that cellular systems affected by lyso-Gb3 included cell signalling particularly protein ubiquitination and protein translation. To confirm ER/proteasome perturbations, we performed an immune enrichment of ubiquitinated proteins and demonstrated specific increased protein ubiquitination at both doses. The most ubiquitinated proteins observed included the chaperone/heat shock proteins, cytoskeletal proteins and synthesis/translation proteins. To detect proteins that interact directly with lyso-Gb3, we immobilised lyso-lipids, then incubated them with neuronal cellular extracts and identified bound proteins using mass spectrometry. Proteins that specifically bound were chaperones and included HSP90, HSP60 and the TRiC complex. In conclusion, lyso-Gb3 exposure affects pathways involved in protein translation and folding. This response is observed as increased ubiquitination and changes in signalling proteins which may explain the multiple biological processes, particularly cellular remodelling, often associated with FD.


Subject(s)
Fabry Disease , Neuroblastoma , Humans , Fabry Disease/genetics , Ubiquitinated Proteins , Proteomics , alpha-Galactosidase/genetics , Sphingolipids/metabolism , Glycolipids/metabolism , Glycolipids/pharmacology
15.
Genes Cells ; 29(4): 347-355, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38351722

ABSTRACT

CdsA is a CDP-diacylglycerol synthase essential for phospholipid and glycolipid MPIase biosynthesis, and therefore for growth. The initiation codon of CdsA has been assigned as "TTG," while methionine at the 37th codon was reported to be an initiation codon in the original report. Since a vector containing the open reading frame starting with "TTG" under a controllable promoter complemented the cdsA knockout, "TTG" could function as an initiation codon. However, no evidence supporting that this "TTG" is the sole initiation codon has been reported. We determined the initiation codon by examining the ability of mutants around the N-terminal region to complement cdsA mutants. Even if the "TTG" was substituted with a stop codon, the clear complementation was observed. Moreover, the clones with multiple mutations of stop codons complemented the cdsA mutant up to the 37th codon, indicating that cdsA possesses multiple codons that can function as initiation codons. We constructed an experimental system in which the chromosomal expression of cdsA can be analyzed. By means of this system, we found that the cdsA mutant with substitution of "TTG" with a stop codon is fully functional. Thus, we concluded that CdsA contains multiple initiation codons.


Subject(s)
Diacylglycerol Cholinephosphotransferase , Glycolipids , Phospholipids , Diacylglycerol Cholinephosphotransferase/metabolism , Codon, Initiator/genetics , Codon, Terminator/genetics , Protein Biosynthesis
16.
Plant Physiol ; 196(1): 397-408, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38850059

ABSTRACT

Alka(e)nes are produced by many living organisms and exhibit diverse physiological roles, reflecting a high functional versatility. Alka(e)nes serve as waterproof wax in plants, communicating pheromones for insects, and microbial signaling molecules in some bacteria. Although alka(e)nes have been found in cyanobacteria and algal chloroplasts, their importance for photosynthetic membranes has remained elusive. In this study, we investigated the consequences of the absence of alka(e)nes on membrane lipid composition and photosynthesis using the cyanobacterium Synechocystis PCC6803 as a model organism. By following the dynamics of membrane lipids and the photosynthetic performance in strains defected and altered in alka(e)ne biosynthesis, we show that drastic changes in the glycerolipid contents occur in the absence of alka(e)nes, including a decrease in the membrane carotenoid content, a decrease in some digalactosyldiacylglycerol (DGDG) species and a parallel increase in monogalactosyldiacylglycerol (MGDG) species. These changes are associated with a higher susceptibility of photosynthesis and growth to high light in alka(e)ne-deficient strains. All these phenotypes are reversed by expressing an algal photoenzyme producing alka(e)nes from fatty acids. Therefore, alkenes, despite their low abundance, are an essential component of the lipid composition of membranes. The profound remodeling of lipid composition that results from their absence suggests that they play an important role in one or more membrane properties in cyanobacteria. Moreover, the lipid compensatory mechanism observed is not sufficient to restore normal functioning of the photosynthetic membranes, particularly under high-light intensity. We conclude that alka(e)nes play a crucial role in maintaining the lipid homeostasis of thylakoid membranes, thereby contributing to the proper functioning of photosynthesis, particularly under elevated light intensities.


Subject(s)
Carotenoids , Glycolipids , Membrane Lipids , Photosynthesis , Synechocystis , Synechocystis/metabolism , Synechocystis/growth & development , Carotenoids/metabolism , Glycolipids/metabolism , Membrane Lipids/metabolism , Cell Membrane/metabolism , Galactolipids/metabolism , Waxes/metabolism
17.
PLoS Biol ; 20(2): e3001555, 2022 02.
Article in English | MEDLINE | ID: mdl-35180210

ABSTRACT

Bacterial membrane lipids are critical for membrane bilayer formation, cell division, protein localization, stress responses, and pathogenesis. Despite their critical roles, membrane lipids have not been fully elucidated for many pathogens. Here, we report the discovery of a novel cationic glycolipid, lysyl-glucosyl-diacylglycerol (Lys-Glc-DAG), which is synthesized in high abundance by the bacterium Streptococcus agalactiae (Group B Streptococcus, GBS). To our knowledge, Lys-Glc-DAG is more positively charged than any other known lipids. Lys-Glc-DAG carries 2 positive net charges per molecule, distinct from the widely described lysylated phospholipid lysyl-phosphatidylglycerol (Lys-PG) that carries one positive net charge due to the presence of a negatively charged phosphate moiety. We use normal phase liquid chromatography (NPLC) coupled with electrospray ionization (ESI) high-resolution tandem mass spectrometry (HRMS/MS) and genetic approaches to determine that Lys-Glc-DAG is synthesized by the enzyme MprF in GBS, which covalently modifies the neutral glycolipid Glc-DAG with the cationic amino acid lysine. GBS is a leading cause of neonatal meningitis, which requires traversal of the endothelial blood-brain barrier (BBB). We demonstrate that GBS strains lacking mprF exhibit a significant decrease in the ability to invade BBB endothelial cells. Further, mice challenged with a GBSΔmprF mutant developed bacteremia comparably to wild-type (WT) infected mice yet had less recovered bacteria from brain tissue and a lower incidence of meningitis. Thus, our data suggest that Lys-Glc-DAG may contribute to bacterial uptake into host cells and disease progression. Importantly, our discovery provides a platform for further study of cationic lipids at the host-pathogen interface.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Glycolipids/metabolism , Meningitis/metabolism , Streptococcus agalactiae/metabolism , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Transport/genetics , Cations/chemistry , Chromatography, Liquid/methods , Glycolipids/chemistry , Humans , Male , Mice , Mutation , Spectrometry, Mass, Electrospray Ionization/methods , Streptococcus agalactiae/genetics , Tandem Mass Spectrometry/methods
18.
J Immunol ; 211(9): 1385-1396, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37695687

ABSTRACT

Mycobacterium tuberculosis cell-wall glycolipids such as mannosylated lipoarabinomannan (ManLAM) can inhibit murine CD4+ T cells by blocking TCR signaling. This results in suppression of IL-2 production, reduced T cell proliferation, and induction of CD4+ T cell anergy. This study extended these findings to the interaction between primary human CD4+ T cells and macrophages infected by mycobacteria. Exposure of human CD4+ T cells to ManLAM before activation resulted in loss of polyfunctionality, as measured by IL-2, IFN-γ, and TNF-α expression, and reduced CD25 expression. This was not associated with upregulation of inhibitory receptors CTLA-4, PD-1, TIM-3, and Lag-3. By confocal microscopy and imaging flow cytometry, ManLAM exposure reduced conjugate formation between macrophages and CD4+ T cells. ManLAM colocalized to the immunological synapse (IS) and reduced translocation of lymphocyte-specific protein tyrosine kinase (LCK) to the IS. When CD4+ T cells and Mycobacterium bovis BCG-infected monocytes were cocultured, ManLAM colocalized to CD4+ T cells, which formed fewer conjugates with infected monocytes. These results demonstrate that mycobacterial cell-wall glycolipids such as ManLAM can traffic from infected macrophages to disrupt productive IS formation and inhibit CD4+ T cell activation, contributing to immune evasion by M. tuberculosis.


Subject(s)
Mycobacterium tuberculosis , Humans , CD4-Positive T-Lymphocytes , Glycolipids/metabolism , Immunological Synapses , Interleukin-2/metabolism , Macrophages/microbiology
19.
Cell ; 143(5): 672-6, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21111227

ABSTRACT

Cells run on carbohydrates. Glycans, sequences of carbohydrates conjugated to proteins and lipids, are arguably the most abundant and structurally diverse class of molecules in nature. Recent advances in glycomics reveal the scope and scale of their functional roles and their impact on human disease.


Subject(s)
Glycomics , Animals , Biomarkers, Tumor/analysis , Glycolipids/analysis , Glycosylation , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Humans , Neoplasms/diagnosis , Polysaccharides/analysis , Polysaccharides/metabolism , Protein Processing, Post-Translational , Proteoglycans/analysis
20.
Cell Mol Life Sci ; 81(1): 265, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38880863

ABSTRACT

Heterotopic ossification (HO) occurs as a common complication after injury, while its risk factor and mechanism remain unclear, which restricts the development of pharmacological treatment. Clinical research suggests that diabetes mellitus (DM) patients are prone to developing HO in the tendon, but solid evidence and mechanical research are still needed. Here, we combined the clinical samples and the DM mice model to identify that disordered glycolipid metabolism aggravates the senescence of tendon-derived stem cells (TSCs) and promotes osteogenic differentiation. Then, combining the RNA-seq results of the aging tendon, we detected the abnormally activated autocrine CXCL13-CXCR5 axis in TSCs cultured in a high fat, high glucose (HFHG) environment and also in the aged tendon. Genetic inhibition of CXCL13 successfully alleviated HO formation in DM mice, providing a potential therapeutic target for suppressing HO formation in DM patients after trauma or surgery.


Subject(s)
Chemokine CXCL13 , Glycolipids , Ossification, Heterotopic , Osteogenesis , Receptors, CXCR5 , Animals , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/pathology , Ossification, Heterotopic/genetics , Mice , Humans , Chemokine CXCL13/metabolism , Chemokine CXCL13/genetics , Glycolipids/metabolism , Receptors, CXCR5/metabolism , Receptors, CXCR5/genetics , Stem Cells/metabolism , Tendons/metabolism , Tendons/pathology , Male , Mice, Inbred C57BL , Cell Differentiation , Cellular Senescence , Signal Transduction , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology
SELECTION OF CITATIONS
SEARCH DETAIL