Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41.342
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 38: 23-48, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32340570

ABSTRACT

The gastrointestinal tract harbors numerous commensal bacteria, referred to as the microbiota, that benefit host health by digesting dietary components and eliminating pathogens. The intestinal microbiota maintains epithelial barrier integrity and shapes the mucosal immune system, balancing host defense and oral tolerance with microbial metabolites, components, and attachment to host cells. To avoid aberrant immune responses, epithelial cells segregate the intestinal microbiota from immune cells by constructing chemical and physical barriers, leading to the establishment of host-commensal mutualism. Furthermore, intestinal immune cells participate in the maintenance of a healthy microbiota community and reinforce epithelial barrier functions. Perturbations of the microbiota composition are commonly observed in patients with autoimmune diseases and chronic inflammatory disorders. An understanding of the intimate interactions between the intestinal microbiota, epithelial cells, and immune cells that are crucial for the maintenance of intestinal homeostasis might promote advances in diagnostic and therapeutic approaches for various diseases.


Subject(s)
Gastrointestinal Microbiome/immunology , Host-Pathogen Interactions/immunology , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Animals , Disease Susceptibility , Humans , Immune System/immunology , Immune System/metabolism , Peyer's Patches/immunology , Peyer's Patches/metabolism
2.
Annu Rev Immunol ; 38: 203-228, 2020 04 26.
Article in English | MEDLINE | ID: mdl-31986071

ABSTRACT

Mucosal-associated invariant T (MAIT) cells have been attracting increasing attention over the last few years as a potent unconventional T cell subset. Three factors largely account for this emerging interest. Firstly, these cells are abundant in humans, both in circulation and especially in some tissues such as the liver. Secondly is the discovery of a ligand that has uncovered their microbial targets, and also allowed for the development of tools to accurately track the cells in both humans and mice. Finally, it appears that the cells not only have a diverse range of functions but also are sensitive to a range of inflammatory triggers that can enhance or even bypass T cell receptor-mediated signals-substantially broadening their likely impact in health and disease. In this review we discuss how MAIT cells display antimicrobial, homeostatic, and amplifier roles in vivo, and how this may lead to protection and potentially pathology.


Subject(s)
Disease Susceptibility , Homeostasis , Mucosal-Associated Invariant T Cells/immunology , Mucosal-Associated Invariant T Cells/metabolism , Animals , Biomarkers , Host-Pathogen Interactions , Humans , Immunity, Mucosal , Mucous Membrane/immunology , Mucous Membrane/metabolism , Mucous Membrane/microbiology , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
3.
Cell ; 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39089253

ABSTRACT

The choroid plexus (ChP) is a vital brain barrier and source of cerebrospinal fluid (CSF). Here, we use longitudinal two-photon imaging in awake mice and single-cell transcriptomics to elucidate the mechanisms of ChP regulation of brain inflammation. We used intracerebroventricular injections of lipopolysaccharides (LPS) to model meningitis in mice and observed that neutrophils and monocytes accumulated in the ChP stroma and surged across the epithelial barrier into the CSF. Bi-directional recruitment of monocytes from the periphery and, unexpectedly, macrophages from the CSF to the ChP helped eliminate neutrophils and repair the barrier. Transcriptomic analyses detailed the molecular steps accompanying this process and revealed that ChP epithelial cells transiently specialize to nurture immune cells, coordinating their recruitment, survival, and differentiation as well as regulation of the tight junctions that control the permeability of the ChP brain barrier. Collectively, we provide a mechanistic understanding and a comprehensive roadmap of neuroinflammation at the ChP brain barrier.

4.
Cell ; 186(4): 877-891.e14, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36708705

ABSTRACT

We introduce BacDrop, a highly scalable technology for bacterial single-cell RNA sequencing that has overcome many challenges hindering the development of scRNA-seq in bacteria. BacDrop can be applied to thousands to millions of cells from both gram-negative and gram-positive species. It features universal ribosomal RNA depletion and combinatorial barcodes that enable multiplexing and massively parallel sequencing. We applied BacDrop to study Klebsiella pneumoniae clinical isolates and to elucidate their heterogeneous responses to antibiotic stress. In an unperturbed population presumed to be homogeneous, we found within-population heterogeneity largely driven by the expression of mobile genetic elements that promote the evolution of antibiotic resistance. Under antibiotic perturbation, BacDrop revealed transcriptionally distinct subpopulations associated with different phenotypic outcomes including antibiotic persistence. BacDrop thus can capture cellular states that cannot be detected by bulk RNA-seq, which will unlock new microbiological insights into bacterial responses to perturbations and larger bacterial communities such as the microbiome.


Subject(s)
Gene Expression Profiling , Single-Cell Gene Expression Analysis , Sequence Analysis, RNA , RNA-Seq , Bacteria/genetics , Single-Cell Analysis
5.
Cell ; 186(17): 3619-3631.e13, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37595565

ABSTRACT

During viral infection, cells can deploy immune strategies that deprive viruses of molecules essential for their replication. Here, we report a family of immune effectors in bacteria that, upon phage infection, degrade cellular adenosine triphosphate (ATP) and deoxyadenosine triphosphate (dATP) by cleaving the N-glycosidic bond between the adenine and sugar moieties. These ATP nucleosidase effectors are widely distributed within multiple bacterial defense systems, including cyclic oligonucleotide-based antiviral signaling systems (CBASS), prokaryotic argonautes, and nucleotide-binding leucine-rich repeat (NLR)-like proteins, and we show that ATP and dATP degradation during infection halts phage propagation. By analyzing homologs of the immune ATP nucleosidase domain, we discover and characterize Detocs, a family of bacterial defense systems with a two-component phosphotransfer-signaling architecture. The immune ATP nucleosidase domain is also encoded within diverse eukaryotic proteins with immune-like architectures, and we show biochemically that eukaryotic homologs preserve the ATP nucleosidase activity. Our findings suggest that ATP and dATP degradation is a cell-autonomous innate immune strategy conserved across the tree of life.


Subject(s)
Virus Diseases , Humans , Eukaryotic Cells , Prokaryotic Cells , Adenosine Triphosphate , N-Glycosyl Hydrolases
6.
Cell ; 186(9): 1863-1876.e16, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37030292

ABSTRACT

Over the past few years, numerous anti-phage defense systems have been discovered in bacteria. Although the mechanism of defense for some of these systems is understood, a major unanswered question is how these systems sense phage infection. To systematically address this question, we isolated 177 phage mutants that escape 15 different defense systems. In many cases, these escaper phages were mutated in the gene sensed by the defense system, enabling us to map the phage determinants that confer sensitivity to bacterial immunity. Our data identify specificity determinants of diverse retron systems and reveal phage-encoded triggers for multiple abortive infection systems. We find general themes in phage sensing and demonstrate that mechanistically diverse systems have converged to sense either the core replication machinery of the phage, phage structural components, or host takeover mechanisms. Combining our data with previous findings, we formulate key principles on how bacterial immune systems sense phage invaders.


Subject(s)
Bacteria , Bacteriophages , Bacteria/genetics , Bacteria/virology , Bacteriophages/genetics , CRISPR-Cas Systems , Viral Proteins/metabolism , Mutation , Bacterial Physiological Phenomena
7.
Cell ; 185(13): 2338-2353.e18, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35662409

ABSTRACT

Hijacking the cellular protein degradation system offers unique opportunities for drug discovery, as exemplified by proteolysis-targeting chimeras. Despite their great promise for medical chemistry, so far, it has not been possible to reprogram the bacterial degradation machinery to interfere with microbial infections. Here, we develop small-molecule degraders, so-called BacPROTACs, that bind to the substrate receptor of the ClpC:ClpP protease, priming neo-substrates for degradation. In addition to their targeting function, BacPROTACs activate ClpC, transforming the resting unfoldase into its functional state. The induced higher-order oligomer was visualized by cryo-EM analysis, providing a structural snapshot of activated ClpC unfolding a protein substrate. Finally, drug susceptibility and degradation assays performed in mycobacteria demonstrate in vivo activity of BacPROTACs, allowing selective targeting of endogenous proteins via fusion to an established degron. In addition to guiding antibiotic discovery, the BacPROTAC technology presents a versatile research tool enabling the inducible degradation of bacterial proteins.


Subject(s)
Bacterial Proteins , Molecular Chaperones , Bacteria/metabolism , Bacterial Proteins/metabolism , Molecular Chaperones/metabolism , Proteolysis
8.
Cell ; 185(7): 1157-1171.e22, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35259335

ABSTRACT

Enterococci are a part of human microbiota and a leading cause of multidrug resistant infections. Here, we identify a family of Enterococcus pore-forming toxins (Epxs) in E. faecalis, E. faecium, and E. hirae strains isolated across the globe. Structural studies reveal that Epxs form a branch of ß-barrel pore-forming toxins with a ß-barrel protrusion (designated the top domain) sitting atop the cap domain. Through a genome-wide CRISPR-Cas9 screen, we identify human leukocyte antigen class I (HLA-I) complex as a receptor for two members (Epx2 and Epx3), which preferentially recognize human HLA-I and homologous MHC-I of equine, bovine, and porcine, but not murine, origin. Interferon exposure, which stimulates MHC-I expression, sensitizes human cells and intestinal organoids to Epx2 and Epx3 toxicity. Co-culture with Epx2-harboring E. faecium damages human peripheral blood mononuclear cells and intestinal organoids, and this toxicity is neutralized by an Epx2 antibody, demonstrating the toxin-mediated virulence of Epx-carrying Enterococcus.


Subject(s)
Bacterial Toxins/metabolism , Enterococcus , Leukocytes, Mononuclear , Virulence Factors/metabolism , Animals , Cattle , Enterococcus/metabolism , Enterococcus/pathogenicity , Horses , Mice , Microbial Sensitivity Tests , Swine
9.
Annu Rev Biochem ; 90: 817-846, 2021 06 20.
Article in English | MEDLINE | ID: mdl-33823652

ABSTRACT

Sulfonates include diverse natural products and anthropogenic chemicals and are widespread in the environment. Many bacteria can degrade sulfonates and obtain sulfur, carbon, and energy for growth, playing important roles in the biogeochemical sulfur cycle. Cleavage of the inert sulfonate C-S bond involves a variety of enzymes, cofactors, and oxygen-dependent and oxygen-independent catalytic mechanisms. Sulfonate degradation by strictly anaerobic bacteria was recently found to involve C-S bond cleavage through O2-sensitive free radical chemistry, catalyzed by glycyl radical enzymes (GREs). The associated discoveries of new enzymes and metabolic pathways for sulfonate metabolism in diverse anaerobic bacteria have enriched our understanding of sulfonate chemistry in the anaerobic biosphere. An anaerobic environment of particular interest is the human gut microbiome, where sulfonate degradation by sulfate- and sulfite-reducing bacteria (SSRB) produces H2S, a process linked to certain chronic diseases and conditions.


Subject(s)
Carbon-Carbon Lyases/metabolism , Gastrointestinal Microbiome/physiology , Sulfonic Acids/metabolism , Acetyltransferases/chemistry , Acetyltransferases/metabolism , Alkanesulfonates/metabolism , Anaerobiosis , Bacteria/metabolism , Carbon-Carbon Lyases/chemistry , Glycine/metabolism , Humans , Hydrogen Sulfide/metabolism , Isethionic Acid/metabolism , Microbiota/physiology , Taurine/metabolism
10.
Cell ; 184(10): 2665-2679.e19, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33882274

ABSTRACT

The bacterial flagellar motor is a supramolecular protein machine that drives rotation of the flagellum for motility, which is essential for bacterial survival in different environments and a key determinant of pathogenicity. The detailed structure of the flagellar motor remains unknown. Here we present an atomic-resolution cryoelectron microscopy (cryo-EM) structure of the bacterial flagellar motor complexed with the hook, consisting of 175 subunits with a molecular mass of approximately 6.3 MDa. The structure reveals that 10 peptides protruding from the MS ring with the FlgB and FliE subunits mediate torque transmission from the MS ring to the rod and overcome the symmetry mismatch between the rotational and helical structures in the motor. The LP ring contacts the distal rod and applies electrostatic forces to support its rotation and torque transmission to the hook. This work provides detailed molecular insights into the structure, assembly, and torque transmission mechanisms of the flagellar motor.


Subject(s)
Flagella/physiology , Flagella/ultrastructure , Salmonella typhimurium/physiology , Cryoelectron Microscopy , Protein Conformation , Torque
11.
Cell ; 184(3): 675-688.e19, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33421369

ABSTRACT

CRISPR-Cas systems provide prokaryotes with acquired immunity against viruses and plasmids, but how these systems are regulated to prevent autoimmunity is poorly understood. Here, we show that in the S. pyogenes CRISPR-Cas system, a long-form transactivating CRISPR RNA (tracr-L) folds into a natural single guide that directs Cas9 to transcriptionally repress its own promoter (Pcas). Further, we demonstrate that Pcas serves as a critical regulatory node. De-repression causes a dramatic 3,000-fold increase in immunization rates against viruses; however, heightened immunity comes at the cost of increased autoimmune toxicity. Using bioinformatic analyses, we provide evidence that tracrRNA-mediated autoregulation is widespread in type II-A CRISPR-Cas systems. Collectively, we unveil a new paradigm for the intrinsic regulation of CRISPR-Cas systems by natural single guides, which may facilitate the frequent horizontal transfer of these systems into new hosts that have not yet evolved their own regulatory strategies.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Gene Expression , Homeostasis/genetics , RNA, Guide, Kinetoplastida/genetics , Autoimmunity/genetics , Base Sequence , Conserved Sequence , Down-Regulation/genetics , Models, Genetic , Mutation/genetics , Operon/genetics , Promoter Regions, Genetic/genetics , Streptococcus pyogenes/genetics , Stress, Physiological/genetics , Transcription, Genetic , Transcriptional Activation/genetics
12.
Cell ; 184(8): 2053-2067.e18, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33794144

ABSTRACT

Industrialization has impacted the human gut ecosystem, resulting in altered microbiome composition and diversity. Whether bacterial genomes may also adapt to the industrialization of their host populations remains largely unexplored. Here, we investigate the extent to which the rates and targets of horizontal gene transfer (HGT) vary across thousands of bacterial strains from 15 human populations spanning a range of industrialization. We show that HGTs have accumulated in the microbiome over recent host generations and that HGT occurs at high frequency within individuals. Comparison across human populations reveals that industrialized lifestyles are associated with higher HGT rates and that the functions of HGTs are related to the level of host industrialization. Our results suggest that gut bacteria continuously acquire new functionality based on host lifestyle and that high rates of HGT may be a recent development in human history linked to industrialization.


Subject(s)
Bacteria/genetics , Gastrointestinal Microbiome , Gene Transfer, Horizontal , Bacteria/classification , Bacteria/isolation & purification , DNA, Bacterial/chemistry , DNA, Bacterial/isolation & purification , DNA, Bacterial/metabolism , Feces/microbiology , Genome, Bacterial , Humans , Phylogeny , Rural Population , Sequence Analysis, DNA , Urban Population , Whole Genome Sequencing
13.
Cell ; 184(9): 2302-2315.e12, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33838112

ABSTRACT

By following up the gut microbiome, 51 human phenotypes and plasma levels of 1,183 metabolites in 338 individuals after 4 years, we characterize microbial stability and variation in relation to host physiology. Using these individual-specific and temporally stable microbial profiles, including bacterial SNPs and structural variations, we develop a microbial fingerprinting method that shows up to 85% accuracy in classifying metagenomic samples taken 4 years apart. Application of our fingerprinting method to the independent HMP cohort results in 95% accuracy for samples taken 1 year apart. We further observe temporal changes in the abundance of multiple bacterial species, metabolic pathways, and structural variation, as well as strain replacement. We report 190 longitudinal microbial associations with host phenotypes and 519 associations with plasma metabolites. These associations are enriched for cardiometabolic traits, vitamin B, and uremic toxins. Finally, mediation analysis suggests that the gut microbiome may influence cardiometabolic health through its metabolites.


Subject(s)
Bacteria/genetics , Bacterial Proteins/metabolism , Gastrointestinal Microbiome , Metabolome , Metagenome , Microbiota , Adult , Aged , Aged, 80 and over , Bacteria/classification , Bacteria/isolation & purification , Bacteria/metabolism , Bacterial Proteins/genetics , Drug Resistance, Microbial , Feces/microbiology , Female , Genomic Instability , Humans , Longitudinal Studies , Male , Middle Aged , Phenotype , Polymorphism, Single Nucleotide , Virulence Factors/genetics , Virulence Factors/metabolism , Young Adult
14.
Cell ; 181(3): 653-664.e19, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32359438

ABSTRACT

Gram-negative bacteria are surrounded by an outer membrane composed of phospholipids and lipopolysaccharide, which acts as a barrier and contributes to antibiotic resistance. The systems that mediate phospholipid trafficking across the periplasm, such as MCE (Mammalian Cell Entry) transporters, have not been well characterized. Our ~3.5 Å cryo-EM structure of the E. coli MCE protein LetB reveals an ~0.6 megadalton complex that consists of seven stacked rings, with a central hydrophobic tunnel sufficiently long to span the periplasm. Lipids bind inside the tunnel, suggesting that it functions as a pathway for lipid transport. Cryo-EM structures in the open and closed states reveal a dynamic tunnel lining, with implications for gating or substrate translocation. Our results support a model in which LetB establishes a physical link between the two membranes and creates a hydrophobic pathway for the translocation of lipids across the periplasm.


Subject(s)
Bacterial Proteins/metabolism , Lipopolysaccharides/metabolism , Membrane Transport Proteins/metabolism , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/physiology , Biological Transport , Cell Membrane/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Membrane Proteins/metabolism , Phospholipids/metabolism , Protein Transport/physiology
15.
Cell ; 183(1): 244-257.e16, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32931735

ABSTRACT

Many bacteria use the flagellum for locomotion and chemotaxis. Its bidirectional rotation is driven by a membrane-embedded motor, which uses energy from the transmembrane ion gradient to generate torque at the interface between stator units and rotor. The structural organization of the stator unit (MotAB), its conformational changes upon ion transport, and how these changes power rotation of the flagellum remain unknown. Here, we present ~3 Å-resolution cryoelectron microscopy reconstructions of the stator unit in different functional states. We show that the stator unit consists of a dimer of MotB surrounded by a pentamer of MotA. Combining structural data with mutagenesis and functional studies, we identify key residues involved in torque generation and present a detailed mechanistic model for motor function and switching of rotational direction.


Subject(s)
Bacterial Proteins/ultrastructure , Flagella/ultrastructure , Bacteria/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cryoelectron Microscopy/methods , Flagella/metabolism , Protein Conformation , Torque
16.
Cell ; 182(4): 919-932.e19, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32763156

ABSTRACT

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


Subject(s)
Biofilms/growth & development , DNA/chemistry , Pseudomonas aeruginosa/physiology , Pyocyanine/chemistry , DNA/metabolism , Electrochemical Techniques , Electrodes , Electron Transport/drug effects , Fluorescent Dyes/chemistry , Hydrogen-Ion Concentration , Oxidation-Reduction , Phenazines/chemistry , Phenazines/metabolism , Phenazines/pharmacology , Pyocyanine/metabolism
17.
Cell ; 182(2): 345-356.e16, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32589945

ABSTRACT

Pathogenic clostridial species secrete potent toxins that induce severe host tissue damage. Paeniclostridium sordellii lethal toxin (TcsL) causes an almost invariably lethal toxic shock syndrome associated with gynecological infections. TcsL is 87% similar to C. difficile TcdB, which enters host cells via Frizzled receptors in colon epithelium. However, P. sordellii infections target vascular endothelium, suggesting that TcsL exploits another receptor. Here, using CRISPR/Cas9 screening, we establish semaphorins SEMA6A and SEMA6B as TcsL receptors. We demonstrate that recombinant SEMA6A can protect mice from TcsL-induced edema. A 3.3 Å cryo-EM structure shows that TcsL binds SEMA6A with the same region that in TcdB binds structurally unrelated Frizzled. Remarkably, 15 mutations in this evolutionarily divergent surface are sufficient to switch binding specificity of TcsL to that of TcdB. Our findings establish semaphorins as physiologically relevant receptors for TcsL and reveal the molecular basis for the difference in tissue targeting and disease pathogenesis between highly related toxins.


Subject(s)
Bacterial Toxins/metabolism , Clostridium sordellii/metabolism , Semaphorins/metabolism , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/toxicity , Binding Sites , CRISPR-Cas Systems/genetics , Cell Line , Cryoelectron Microscopy , Edema/pathology , Edema/prevention & control , Female , Humans , Lung/drug effects , Lung/pathology , Mice , Mice, Inbred C57BL , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/therapeutic use , Semaphorins/chemistry , Semaphorins/genetics
18.
Annu Rev Cell Dev Biol ; 37: 1-21, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34186006

ABSTRACT

One of the most common bacterial shapes is a rod, yet we have a limited understanding of how this simple shape is constructed. While only six proteins are required for rod shape, we are just beginning to understand how they self-organize to build the micron-sized enveloping structures that define bacterial shape out of nanometer-sized glycan strains. Here, we detail and summarize the insights gained over the last 20 years into this complex problem that have been achieved with a wide variety of different approaches. We also explain and compare both current and past models of rod shape formation and maintenance and then highlight recent insights into how the Rod complex might be regulated.


Subject(s)
Bacteria , Bacterial Proteins , Bacteria/genetics , Bacteria/metabolism , Bacterial Proteins/genetics
19.
Annu Rev Biochem ; 88: 811-837, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30388027

ABSTRACT

Botulinum neurotoxins (BoNTs) and tetanus neurotoxin (TeNT) are the most potent toxins known and cause botulism and tetanus, respectively. BoNTs are also widely utilized as therapeutic toxins. They contain three functional domains responsible for receptor-binding, membrane translocation, and proteolytic cleavage of host proteins required for synaptic vesicle exocytosis. These toxins also have distinct features: BoNTs exist within a progenitor toxin complex (PTC), which protects the toxin and facilitates its absorption in the gastrointestinal tract, whereas TeNT is uniquely transported retrogradely within motor neurons. Our increasing knowledge of these toxins has allowed the development of engineered toxins for medical uses. The discovery of new BoNTs and BoNT-like proteins provides additional tools to understand the evolution of the toxins and to engineer toxin-based therapeutics. This review summarizes the progress on our understanding of BoNTs and TeNT, focusing on the PTC, receptor recognition, new BoNT-like toxins, and therapeutic toxin engineering.


Subject(s)
Botulinum Toxins/therapeutic use , Metalloendopeptidases/therapeutic use , Tetanus Toxin/therapeutic use , Animals , Botulinum Toxins/metabolism , Botulinum Toxins/toxicity , Humans , Metalloendopeptidases/metabolism , Metalloendopeptidases/toxicity , Protein Conformation , Protein Engineering , Tetanus Toxin/metabolism , Tetanus Toxin/toxicity
20.
Cell ; 179(7): 1512-1524.e15, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31835030

ABSTRACT

During cell division, newly replicated DNA is actively segregated to the daughter cells. In most bacteria, this process involves the DNA-binding protein ParB, which condenses the centromeric regions of sister DNA molecules into kinetochore-like structures that recruit the DNA partition ATPase ParA and the prokaroytic SMC/condensin complex. Here, we report the crystal structure of a ParB-like protein (PadC) that emerges to tightly bind the ribonucleotide CTP. The CTP-binding pocket of PadC is conserved in ParB and composed of signature motifs known to be essential for ParB function. We find that ParB indeed interacts with CTP and requires nucleotide binding for DNA condensation in vivo. We further show that CTP-binding modulates the affinity of ParB for centromeric parS sites, whereas parS recognition stimulates its CTPase activity. ParB proteins thus emerge as a new class of CTP-dependent molecular switches that act in concert with ATPases and GTPases to control fundamental cellular functions.


Subject(s)
Bacterial Proteins/chemistry , Cytidine Triphosphate/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Myxococcus xanthus/genetics , Myxococcus xanthus/metabolism , Nucleotide Motifs , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL