Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 18.571
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 35: 337-370, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28142321

ABSTRACT

Transcriptomics, the high-throughput characterization of RNAs, has been instrumental in defining pathogenic signatures in human autoimmunity and autoinflammation. It enabled the identification of new therapeutic targets in IFN-, IL-1- and IL-17-mediated diseases. Applied to immunomonitoring, transcriptomics is starting to unravel diagnostic and prognostic signatures that stratify patients, track molecular changes associated with disease activity, define personalized treatment strategies, and generally inform clinical practice. Herein, we review the use of transcriptomics to define mechanistic, diagnostic, and predictive signatures in human autoimmunity and autoinflammation. We discuss some of the analytical approaches applied to extract biological knowledge from high-dimensional data sets. Finally, we touch upon emerging applications of transcriptomics to study eQTLs, B and T cell repertoire diversity, and isoform usage.


Subject(s)
Autoimmune Diseases/diagnosis , Inflammation/diagnosis , Transcriptome , Autoimmune Diseases/immunology , Datasets as Topic , High-Throughput Nucleotide Sequencing , Humans , Inflammation/immunology , Information Storage and Retrieval , Molecular Targeted Therapy , Monitoring, Immunologic , Prognosis
2.
Cell ; 187(19): 5238-5252.e20, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39208796

ABSTRACT

Fanzor (Fz) is an ωRNA-guided endonuclease extensively found throughout the eukaryotic domain with unique gene editing potential. Here, we describe the structures of Fzs from three different organisms. We find that Fzs share a common ωRNA interaction interface, regardless of the length of the ωRNA, which varies considerably across species. The analysis also reveals Fz's mode of DNA recognition and unwinding capabilities as well as the presence of a non-canonical catalytic site. The structures demonstrate how protein conformations of Fz shift to allow the binding of double-stranded DNA to the active site within the R-loop. Mechanistically, examination of structures in different states shows that the conformation of the lid loop on the RuvC domain is controlled by the formation of the guide/DNA heteroduplex, regulating the activation of nuclease and DNA double-stranded displacement at the single cleavage site. Our findings clarify the mechanism of Fz, establishing a foundation for engineering efforts.


Subject(s)
DNA Cleavage , DNA , DNA/metabolism , DNA/chemistry , Catalytic Domain , Models, Molecular , RNA, Guide, CRISPR-Cas Systems/metabolism , RNA, Guide, CRISPR-Cas Systems/chemistry , Humans , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/chemistry , Gene Editing , CRISPR-Cas Systems
3.
Cell ; 187(15): 4113-4127.e13, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38876107

ABSTRACT

Vector-borne diseases are a leading cause of death worldwide and pose a substantial unmet medical need. Pathogens binding to host extracellular proteins (the "exoproteome") represents a crucial interface in the etiology of vector-borne disease. Here, we used bacterial selection to elucidate host-microbe interactions in high throughput (BASEHIT)-a technique enabling interrogation of microbial interactions with 3,324 human exoproteins-to profile the interactomes of 82 human-pathogen samples, including 30 strains of arthropod-borne pathogens and 8 strains of related non-vector-borne pathogens. The resulting atlas revealed 1,303 putative interactions, including hundreds of pairings with potential roles in pathogenesis, including cell invasion, tissue colonization, immune evasion, and host sensing. Subsequent functional investigations uncovered that Lyme disease spirochetes recognize epidermal growth factor as an environmental cue of transcriptional regulation and that conserved interactions between intracellular pathogens and thioredoxins facilitate cell invasion. In summary, this interactome atlas provides molecular-level insights into microbial pathogenesis and reveals potential host-directed targets for next-generation therapeutics.


Subject(s)
Host-Pathogen Interactions , Humans , Animals , Lyme Disease/microbiology , Vector Borne Diseases , Host Microbial Interactions , Borrelia burgdorferi/pathogenicity , Borrelia burgdorferi/metabolism
4.
Cell ; 186(14): 2959-2976.e22, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37339633

ABSTRACT

Snakes are a remarkable squamate lineage with unique morphological adaptations, especially those related to the evolution of vertebrate skeletons, organs, and sensory systems. To clarify the genetic underpinnings of snake phenotypes, we assembled and analyzed 14 de novo genomes from 12 snake families. We also investigated the genetic basis of the morphological characteristics of snakes using functional experiments. We identified genes, regulatory elements, and structural variations that have potentially contributed to the evolution of limb loss, an elongated body plan, asymmetrical lungs, sensory systems, and digestive adaptations in snakes. We identified some of the genes and regulatory elements that might have shaped the evolution of vision, the skeletal system and diet in blind snakes, and thermoreception in infrared-sensitive snakes. Our study provides insights into the evolution and development of snakes and vertebrates.


Subject(s)
Genome , Snakes , Animals , Snakes/genetics , Adaptation, Physiological , Acclimatization , Evolution, Molecular , Phylogeny , Biological Evolution
5.
Annu Rev Cell Dev Biol ; 40(1): 265-281, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39356809

ABSTRACT

Antibodies that gain specificity by a large insert encoding for an extra domain were described for the first time in 2016. In malaria-exposed individuals, an exon deriving from the leukocyte-associated immunoglobulin-like 1 (LAIR1) gene integrated via a copy-and-paste insertion into the immunoglobulin heavy chain encoding region. A few years later, a second example was identified, namely a dual exon integration from the leukocyte immunoglobulin-like receptor B1 (LILRB1) gene that is located in close proximity to LAIR1. A dedicated high-throughput characterization of chimeric immunoglobulin heavy chain transcripts unraveled, that insertions from distant genomic regions (including mitochondrial DNA) can contribute to human antibody diversity. This review describes the modalities of insert-containing antibodies. The role of known DNA mobility aspects, such as genomic translocation, gene conversion, and DNA fragility, is discussed in the context of insert-antibody generation. Finally, the review covers why insert antibodies were omitted from the past repertoire analyses and how insert antibodies can contribute to protective immunity or an autoreactive response.


Subject(s)
Exons , V(D)J Recombination , Humans , V(D)J Recombination/genetics , Exons/genetics , Animals , Antibodies/immunology , Antibodies/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Immunologic/immunology , Antibody Diversity/genetics
6.
Annu Rev Immunol ; 33: 201-25, 2015.
Article in English | MEDLINE | ID: mdl-25533702

ABSTRACT

Helminth parasites are a highly successful group of pathogens that challenge the immune system in a manner distinct from rapidly replicating infectious agents. Of this group, roundworms (nematodes) that dwell in the intestines of humans and other animals are prevalent worldwide. Currently, more than one billion people are infected by at least one species, often for extended periods of time. Thus, host-protective immunity is rarely complete. The reasons for this are complex, but laboratory investigation of tractable model systems in which protective immunity is effective has provided a mechanistic understanding of resistance that is characterized almost universally by a type 2/T helper 2 response. Greater understanding of the mechanisms of susceptibility has also provided the basis for defining host immunoregulation and parasite-evasion strategies, helping place in context the changing patterns of immunological disease observed worldwide.


Subject(s)
Helminthiasis/immunology , Helminthiasis/parasitology , Helminths/immunology , Host-Pathogen Interactions/immunology , Adaptive Immunity , Animals , Antigens, Helminth/immunology , Disease Resistance , Disease Susceptibility , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/microbiology , Gastrointestinal Diseases/parasitology , Gastrointestinal Microbiome , Humans , Immunity, Innate , Nematoda/immunology , Nematode Infections/immunology , Nematode Infections/microbiology , Nematode Infections/parasitology
7.
Annu Rev Biochem ; 89: 45-75, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32569524

ABSTRACT

Ribonucleotide reductases (RNRs) catalyze the de novo conversion of nucleotides to deoxynucleotides in all organisms, controlling their relative ratios and abundance. In doing so, they play an important role in fidelity of DNA replication and repair. RNRs' central role in nucleic acid metabolism has resulted in five therapeutics that inhibit human RNRs. In this review, we discuss the structural, dynamic, and mechanistic aspects of RNR activity and regulation, primarily for the human and Escherichia coli class Ia enzymes. The unusual radical-based organic chemistry of nucleotide reduction, the inorganic chemistry of the essential metallo-cofactor biosynthesis/maintenance, the transport of a radical over a long distance, and the dynamics of subunit interactions all present distinct entry points toward RNR inhibition that are relevant for drug discovery. We describe the current mechanistic understanding of small molecules that target different elements of RNR function, including downstream pathways that lead to cell cytotoxicity. We conclude by summarizing novel and emergent RNR targeting motifs for cancer and antibiotic therapeutics.


Subject(s)
Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemistry , Escherichia coli Infections/drug therapy , Neoplasms/drug therapy , Nucleotides/metabolism , Ribonucleotide Reductases/chemistry , Anti-Bacterial Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Biocatalysis , Drug Discovery/methods , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Infections/enzymology , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Humans , Molecular Docking Simulation , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Nucleotides/chemistry , Oxidation-Reduction , Protein Structure, Secondary , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Ribonucleotide Reductases/antagonists & inhibitors , Ribonucleotide Reductases/genetics , Ribonucleotide Reductases/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use , Structure-Activity Relationship
8.
Cell ; 183(4): 860-874, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33186528

ABSTRACT

Persistent cancer cells are the discrete and usually undetected cells that survive cancer drug treatment and constitute a major cause of treatment failure. These cells are characterized by their slow proliferation, highly flexible energy consumption, adaptation to their microenvironment, and phenotypic plasticity. Mechanisms that underlie their persistence offer highly coveted and sought-after therapeutic targets, and include diverse epigenetic, transcriptional, and translational regulatory processes, as well as complex cell-cell interactions. Although the successful clinical targeting of persistent cancer cells remains to be realized, immense progress has been made in understanding their persistence, yielding promising preclinical results.


Subject(s)
Neoplasms/pathology , Animals , Cell Survival , Energy Metabolism , Epithelial-Mesenchymal Transition , Humans , Mitochondria/metabolism , Neoplasms/therapy , Tumor Microenvironment
9.
Cell ; 182(2): 357-371.e13, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32610085

ABSTRACT

Excitatory neurotransmission meditated by glutamate receptors including N-methyl-D-aspartate receptors (NMDARs) is pivotal to brain development and function. NMDARs are heterotetramers composed of GluN1 and GluN2 subunits, which bind glycine and glutamate, respectively, to activate their ion channels. Despite importance in brain physiology, the precise mechanisms by which activation and inhibition occur via subunit-specific binding of agonists and antagonists remain largely unknown. Here, we show the detailed patterns of conformational changes and inter-subunit and -domain reorientation leading to agonist-gating and subunit-dependent competitive inhibition by providing multiple structures in distinct ligand states at 4 Å or better. The structures reveal that activation and competitive inhibition by both GluN1 and GluN2 antagonists occur by controlling the tension of the linker between the ligand-binding domain and the transmembrane ion channel of the GluN2 subunit. Our results provide detailed mechanistic insights into NMDAR pharmacology, activation, and inhibition, which are fundamental to the brain physiology.


Subject(s)
Receptors, N-Methyl-D-Aspartate/metabolism , Binding Sites , Binding, Competitive , Cryoelectron Microscopy , Crystallography, X-Ray , Dimerization , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Glycine/chemistry , Glycine/metabolism , Humans , Ligands , Molecular Dynamics Simulation , Protein Structure, Quaternary , Protein Subunits/agonists , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
10.
Cell ; 180(4): 645-654.e13, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32004460

ABSTRACT

Drugs selectively targeting CB2 hold promise for treating neurodegenerative disorders, inflammation, and pain while avoiding psychotropic side effects mediated by CB1. The mechanisms underlying CB2 activation and signaling are poorly understood but critical for drug design. Here we report the cryo-EM structure of the human CB2-Gi signaling complex bound to the agonist WIN 55,212-2. The 3D structure reveals the binding mode of WIN 55,212-2 and structural determinants for distinguishing CB2 agonists from antagonists, which are supported by a pair of rationally designed agonist and antagonist. Further structural analyses with computational docking results uncover the differences between CB2 and CB1 in receptor activation, ligand recognition, and Gi coupling. These findings are expected to facilitate rational structure-based discovery of drugs targeting the cannabinoid system.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Receptor, Cannabinoid, CB2/chemistry , Signal Transduction , Animals , Binding Sites , CHO Cells , Cannabinoid Receptor Agonists/chemical synthesis , Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Antagonists/chemical synthesis , Cannabinoid Receptor Antagonists/pharmacology , Cricetinae , Cricetulus , Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Molecular Docking Simulation , Protein Binding , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptor, Cannabinoid, CB2/metabolism , Sf9 Cells , Spodoptera
11.
Annu Rev Biochem ; 87: 451-478, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29570352

ABSTRACT

Genetic information is translated into proteins by the ribosome. Structural studies of the ribosome and of its complexes with factors and inhibitors have provided invaluable information on the mechanism of protein synthesis. Ribosome inhibitors are among the most successful antimicrobial drugs and constitute more than half of all medicines used to treat infections. However, bacterial infections are becoming increasingly difficult to treat because the microbes have developed resistance to the most effective antibiotics, creating a major public health care threat. This has spurred a renewed interest in structure-function studies of protein synthesis inhibitors, and in few cases, compounds have been developed into potent therapeutic agents against drug-resistant pathogens. In this review, we describe the modes of action of many ribosome-targeting antibiotics, highlight the major resistance mechanisms developed by pathogenic bacteria, and discuss recent advances in structure-assisted design of new molecules.


Subject(s)
Anti-Bacterial Agents/pharmacology , Ribosomes/drug effects , Animals , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Binding Sites , Drug Design , Drug Resistance, Microbial , Humans , Models, Biological , Models, Molecular , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , Ribosomes/chemistry , Ribosomes/metabolism , Structure-Activity Relationship
12.
Immunity ; 57(3): 541-558.e7, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38442708

ABSTRACT

Cancer patients often receive a combination of antibodies targeting programmed death-ligand 1 (PD-L1) and cytotoxic T lymphocyte antigen-4 (CTLA4). We conducted a window-of-opportunity study in head and neck squamous cell carcinoma (HNSCC) to examine the contribution of anti-CTLA4 to anti-PD-L1 therapy. Single-cell profiling of on- versus pre-treatment biopsies identified T cell expansion as an early response marker. In tumors, anti-PD-L1 triggered the expansion of mostly CD8+ T cells, whereas combination therapy expanded both CD4+ and CD8+ T cells. Such CD4+ T cells exhibited an activated T helper 1 (Th1) phenotype. CD4+ and CD8+ T cells co-localized with and were surrounded by dendritic cells expressing T cell homing factors or antibody-producing plasma cells. T cell receptor tracing suggests that anti-CTLA4, but not anti-PD-L1, triggers the trafficking of CD4+ naive/central-memory T cells from tumor-draining lymph nodes (tdLNs), via blood, to the tumor wherein T cells acquire a Th1 phenotype. Thus, CD4+ T cell activation and recruitment from tdLNs are hallmarks of early response to anti-PD-L1 plus anti-CTLA4 in HNSCC.


Subject(s)
CD8-Positive T-Lymphocytes , Head and Neck Neoplasms , Humans , Squamous Cell Carcinoma of Head and Neck , B7-H1 Antigen/genetics , CTLA-4 Antigen , Head and Neck Neoplasms/drug therapy , CD4-Positive T-Lymphocytes , Tumor Microenvironment
13.
Cell ; 172(3): 423-438.e25, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29249360

ABSTRACT

Stem cells are highly resistant to viral infection compared to their differentiated progeny; however, the mechanism is mysterious. Here, we analyzed gene expression in mammalian stem cells and cells at various stages of differentiation. We find that, conserved across species, stem cells express a subset of genes previously classified as interferon (IFN) stimulated genes (ISGs) but that expression is intrinsic, as stem cells are refractory to interferon. This intrinsic ISG expression varies in a cell-type-specific manner, and many ISGs decrease upon differentiation, at which time cells become IFN responsive, allowing induction of a broad spectrum of ISGs by IFN signaling. Importantly, we show that intrinsically expressed ISGs protect stem cells against viral infection. We demonstrate the in vivo importance of intrinsic ISG expression for protecting stem cells and their differentiation potential during viral infection. These findings have intriguing implications for understanding stem cell biology and the evolution of pathogen resistance.


Subject(s)
Immunity, Innate , Pluripotent Stem Cells/immunology , Virus Diseases/immunology , Animals , Cells, Cultured , Female , HEK293 Cells , Humans , Interferons/metabolism , Male , Mice , Mice, Inbred NOD , Pluripotent Stem Cells/virology , Species Specificity
14.
Annu Rev Cell Dev Biol ; 35: 433-452, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31340126

ABSTRACT

Cellular reprogramming experiments from somatic cell types have demonstrated the plasticity of terminally differentiated cell states. Recent efforts in understanding the mechanisms of cellular reprogramming have begun to elucidate the differentiation trajectories along the reprogramming processes. In this review, we focus mainly on direct reprogramming strategies by transcription factors and highlight the variables that contribute to cell fate conversion outcomes. We review key studies that shed light on the cellular and molecular mechanisms by investigating differentiation trajectories and alternative cell states as well as transcription factor regulatory activities during cell fate reprogramming. Finally, we highlight a few concepts that we believe require attention, particularly when measuring the success of cell reprogramming experiments.


Subject(s)
Cell Transdifferentiation/physiology , Cellular Reprogramming/genetics , Epigenesis, Genetic/genetics , Transcription Factors/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Transdifferentiation/genetics , Epigenesis, Genetic/physiology , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Humans , Signal Transduction/genetics , Transcription Factors/genetics
15.
Immunity ; 56(11): 2602-2620.e10, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37967532

ABSTRACT

Human cytomegalovirus (HCMV) can cause severe diseases in fetuses, newborns, and immunocompromised individuals. Currently, no vaccines are approved, and treatment options are limited. Here, we analyzed the human B cell response of four HCMV top neutralizers from a cohort of 9,000 individuals. By single-cell analyses of memory B cells targeting the pentameric and trimeric HCMV surface complexes, we identified vulnerable sites on the shared gH/gL subunits as well as complex-specific subunits UL128/130/131A and gO. Using high-resolution cryogenic electron microscopy, we revealed the structural basis of the neutralization mechanisms of antibodies targeting various binding sites. Moreover, we identified highly potent antibodies that neutralized a broad spectrum of HCMV strains, including primary clinical isolates, that outperform known antibodies used in clinical trials. Our study provides a deep understanding of the mechanisms of HCMV neutralization and identifies promising antibody candidates to prevent and treat HCMV infection.


Subject(s)
Cytomegalovirus , Viral Envelope Proteins , Infant, Newborn , Humans , Membrane Glycoproteins , Antibodies, Neutralizing , Memory B Cells , Antibodies, Viral , Single-Cell Analysis
16.
Cell ; 168(4): 707-723, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28187290

ABSTRACT

Cancer immunotherapy can induce long lasting responses in patients with metastatic cancers of a wide range of histologies. Broadening the clinical applicability of these treatments requires an improved understanding of the mechanisms limiting cancer immunotherapy. The interactions between the immune system and cancer cells are continuous, dynamic, and evolving from the initial establishment of a cancer cell to the development of metastatic disease, which is dependent on immune evasion. As the molecular mechanisms of resistance to immunotherapy are elucidated, actionable strategies to prevent or treat them may be derived to improve clinical outcomes for patients.


Subject(s)
Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Animals , Drug Therapy, Combination , Humans , Molecular Targeted Therapy , T-Lymphocytes/immunology
17.
Cell ; 171(3): 710-722.e12, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-28965761

ABSTRACT

To further our understanding of the genetic etiology of autism, we generated and analyzed genome sequence data from 516 idiopathic autism families (2,064 individuals). This resource includes >59 million single-nucleotide variants (SNVs) and 9,212 private copy number variants (CNVs), of which 133,992 and 88 are de novo mutations (DNMs), respectively. We estimate a mutation rate of ∼1.5 × 10-8 SNVs per site per generation with a significantly higher mutation rate in repetitive DNA. Comparing probands and unaffected siblings, we observe several DNM trends. Probands carry more gene-disruptive CNVs and SNVs, resulting in severe missense mutations and mapping to predicted fetal brain promoters and embryonic stem cell enhancers. These differences become more pronounced for autism genes (p = 1.8 × 10-3, OR = 2.2). Patients are more likely to carry multiple coding and noncoding DNMs in different genes, which are enriched for expression in striatal neurons (p = 3 × 10-3), suggesting a path forward for genetically characterizing more complex cases of autism.


Subject(s)
Autistic Disorder/genetics , DNA Copy Number Variations , Polymorphism, Single Nucleotide , Animals , DNA Mutational Analysis , Female , Genome-Wide Association Study , Humans , INDEL Mutation , Male , Mice
18.
Annu Rev Cell Dev Biol ; 34: 495-521, 2018 10 06.
Article in English | MEDLINE | ID: mdl-30044649

ABSTRACT

After an injury in the adult mammalian central nervous system (CNS), lesioned axons fail to regenerate. This failure to regenerate contrasts with axons' remarkable potential to grow during embryonic development and after an injury in the peripheral nervous system (PNS). Several intracellular mechanisms-including cytoskeletal dynamics, axonal transport and trafficking, signaling and transcription of regenerative programs, and epigenetic modifications-control axon regeneration. In this review, we describe how manipulation of intrinsic mechanisms elicits a regenerative response in different organisms and how strategies are implemented to form the basis of a future regenerative treatment after CNS injury.


Subject(s)
Axons/metabolism , Central Nervous System/growth & development , Nerve Regeneration/genetics , Peripheral Nervous System/growth & development , Animals , Axonal Transport/genetics , Axons/physiology , Humans , Mammals
19.
Mol Cell ; 84(11): 2185-2202.e12, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38788717

ABSTRACT

Retrons are toxin-antitoxin systems protecting bacteria against bacteriophages via abortive infection. The Retron-Eco1 antitoxin is formed by a reverse transcriptase (RT) and a non-coding RNA (ncRNA)/multi-copy single-stranded DNA (msDNA) hybrid that neutralizes an uncharacterized toxic effector. Yet, the molecular mechanisms underlying phage defense remain unknown. Here, we show that the N-glycosidase effector, which belongs to the STIR superfamily, hydrolyzes NAD+ during infection. Cryoelectron microscopy (cryo-EM) analysis shows that the msDNA stabilizes a filament that cages the effector in a low-activity state in which ADPr, a NAD+ hydrolysis product, is covalently linked to the catalytic E106 residue. Mutations shortening the msDNA induce filament disassembly and the effector's toxicity, underscoring the msDNA role in immunity. Furthermore, we discovered a phage-encoded Retron-Eco1 inhibitor (U56) that binds ADPr, highlighting the intricate interplay between retron systems and phage evolution. Our work outlines the structural basis of Retron-Eco1 defense, uncovering ADPr's pivotal role in immunity.


Subject(s)
Bacteriophages , Cryoelectron Microscopy , NAD , NAD/metabolism , Bacteriophages/genetics , Bacteriophages/metabolism , Bacteriophages/immunology , Hydrolysis , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/immunology , Toxin-Antitoxin Systems/genetics , Escherichia coli/virology , Escherichia coli/genetics , Escherichia coli/immunology , Escherichia coli/metabolism
20.
Annu Rev Biochem ; 85: 543-72, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27023848

ABSTRACT

The determination of the crystal structures of small-molecule transporters has shed light on the conformational changes that take place during structural isomerization from outward- to inward-facing states. Rather than using a simple rocking movement of two bundles around a central substrate-binding site, it has become clear that even the most simplistic transporters utilize rearrangements of nonrigid bodies. In the most dramatic cases, one bundle is fixed while the other, structurally divergent, bundle carries the substrate some 18 Å across the membrane, which in this review is termed an elevator alternating-access mechanism. Here, we compare and contrast rocker-switch, rocking-bundle, and elevator alternating-access mechanisms to highlight shared features and novel refinements to the basic alternating-access model.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Membrane Transport Proteins/chemistry , Molecular Dynamics Simulation , Biological Transport , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Evolution, Molecular , Gene Expression , Kinetics , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Protein Domains , Protein Structure, Secondary , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL