1
|
Modi M, Chauhan D, Gilmore MC, Cava F, Priyadarshini R. Deficiency in peptidoglycan recycling promotes β-lactam sensitivity in Caulobacter crescentus. mBio 2025; 16:e0297524. [PMID: 40066998 PMCID: PMC11980594 DOI: 10.1128/mbio.02975-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/07/2025] [Indexed: 04/10/2025] Open
Abstract
Peptidoglycan (PG)-modifying enzymes play a crucial role in cell wall remodeling, essential for growth and division. Cell wall degradation products are transported to the cytoplasm and recycled back in most gram-negative bacteria, and PG recycling is also linked to β-lactam resistance in many bacteria. Caulobacter crescentus is intrinsically resistant to β-lactams. Recently, it was shown that a soluble lytic transglycosylase, SdpA, is essential for β-lactam resistance. However, the precise role of SdpA in β-lactam resistance is unknown. This study investigated the PG recycling pathway and its role in antibiotic resistance in C. crescentus. Anhydromuropeptides generated by the action of lytic transglycosylases (LTs) are transported to the cytoplasm by the permease AmpG. C. crescentus encodes an ampG homolog, and deletion mutants of sdpA and ampG are sensitive to β-lactams. The ampG deletion mutant displays a significant accumulation of anhydromuropeptides in the periplasm of C. crescentus, demonstrating its essential role in PG recycling. While single knockout mutants of sdpA and ampG exhibit no growth defects, double-deletion mutants (∆sdpA∆ampG) exhibit severe growth and morphological defects. These double mutants also show enhanced sensitivity to β-lactams. Analysis of soluble muropeptides in wild-type (WT), ∆sdpA, and ∆ampG mutants revealed reduced levels of PG precursors (UDP-GlcNAc, UDP-MurNAc, and UDP-MurNAc-P5), suggesting that PG recycling products contribute toward de novo PG biosynthesis. Furthermore, supplementing the growth media with GlcNAc sugar enhanced the fitness of ∆sdpA and ∆ampG mutants under β-lactam stress. In conclusion, our study indicates that defects in PG recycling compromise cell wall biogenesis, leading to antibiotic sensitivity in C. crescentus.IMPORTANCEβ-lactam antibiotics target the peptidoglycan cell wall biosynthetic pathway in bacteria. In response to antibiotic pressures, bacteria have developed various resistance mechanisms. In many gram-negative species, cell wall degradation products are transported into the cytoplasm and induce the expression of β-lactamase enzymes. In this study, we investigated the cell wall recycling pathway and its role in antibiotic resistance in Caulobacter crescentus. Based on our data and prior studies, we propose that cell wall degradation products are utilized for the synthesis of peptidoglycan precursors in the cytoplasm. A deficiency in cell wall recycling leads to cell wall defects and increased antibiotic sensitivity in C. crescentus. These findings are crucial for understanding antibiotic resistance mechanisms in bacteria.
Collapse
Affiliation(s)
- Malvika Modi
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, Uttar Pradesh, India
| | - Deepika Chauhan
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, Uttar Pradesh, India
| | - Michael C. Gilmore
- Laboratory for Molecular Infection Medicine Sweden, Department of Molecular Biology, Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Felipe Cava
- Laboratory for Molecular Infection Medicine Sweden, Department of Molecular Biology, Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Richa Priyadarshini
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, Uttar Pradesh, India
| |
Collapse
|
2
|
Lakshminarasimhan K, Buck J, Kellendonk C, Horga G. A corticostriatal learning mechanism linking excess striatal dopamine and auditory hallucinations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643990. [PMID: 40166304 PMCID: PMC11956939 DOI: 10.1101/2025.03.18.643990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Auditory hallucinations are linked to elevated striatal dopamine, but their underlying computational mechanisms have been obscured by regional heterogeneity in striatal dopamine signaling. To address this, we developed a normative circuit model in which corticostriatal plasticity in the ventral striatum is modulated by reward prediction errors to drive reinforcement learning while that in the sensory-dorsal striatum is modulated by sensory prediction errors derived from internal belief to drive self-supervised learning. We then validate the key predictions of this model using dopamine recordings across striatal regions in mice, as well as human behavior in a hybrid learning task. Finally, we find that changes in learning resulting from optogenetic stimulation of the sensory striatum in mice and individual variability in hallucination proneness in humans are best explained by selectively enhancing dopamine levels in the model sensory striatum. These findings identify plasticity mechanisms underlying biased learning of sensory expectations as a biologically plausible link between excess dopamine and hallucinations.
Collapse
Affiliation(s)
- Kaushik Lakshminarasimhan
- Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY, USA
| | - Justin Buck
- Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - Christoph Kellendonk
- Department of Psychiatry, Columbia University, New York, NY, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, USA
| | - Guillermo Horga
- Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
| |
Collapse
|
3
|
Yaeger LN, Sychantha D, Luong P, Shekarriz S, Goncalves O, Dobrin A, Ranieri MR, Lamers RP, Harvey H, diCenzo GC, Surette M, Côté JP, Magolan J, Burrows LL. Perturbation of Pseudomonas aeruginosa peptidoglycan recycling by anti-folates and design of a dual-action inhibitor. mBio 2025; 16:e0298424. [PMID: 39878530 PMCID: PMC11898565 DOI: 10.1128/mbio.02984-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/17/2024] [Indexed: 01/31/2025] Open
Abstract
Peptidoglycan (PG) is an important bacterial macromolecule that confers cell shape and structural integrity, and is a key antibiotic target. Its synthesis and turnover are carefully coordinated with other cellular processes and pathways. Despite established connections between the biosynthesis of PG and the outer membrane, or PG and DNA replication, links between PG and folate metabolism remain comparatively unexplored. Folate is an essential cofactor for bacterial growth and is required for the synthesis of many important metabolites. Here we show that inhibition of folate synthesis in the important Gram-negative pathogen Pseudomonas aeruginosa has downstream effects on PG metabolism and integrity that can manifest as the formation of a subpopulation of round cells that can undergo explosive lysis. Folate inhibitors potentiated β-lactams by perturbation of PG recycling, reducing expression of the AmpC β-lactamase. Supporting this mechanism, folate inhibitors also synergized with fosfomycin, an inhibitor of MurA, the first committed step in PG synthesis that can be bypassed by PG recycling. These insights led to the design of a dual-active inhibitor that overcomes NDM-1 metallo-β lactamase-mediated meropenem resistance and synergizes with the folate inhibitor, trimethoprim. We show that folate and PG metabolism are intimately connected, and targeting this connection can overcome antibiotic resistance in Gram-negative pathogens. IMPORTANCE To combat the alarming global increase in superbugs amid the simultaneous scarcity of new drugs, we can create synergistic combinations of currently available antibiotics or chimeric molecules with dual activities, to minimize resistance. Here we show that older anti-folate drugs synergize with specific cell wall biosynthesis inhibitors to kill the priority pathogen, Pseudomonas aeruginosa. Anti-folate drugs caused a dose-dependent loss of rod cell shape followed by explosive lysis, and synergized with β-lactams that target D,D-carboxypeptidases required to tailor the cell wall. Anti-folates impaired cell wall recycling and subsequent downstream expression of the chromosomally encoded β-lactamase, AmpC, which normally destroys β-lactam antibiotics. Building on the anti-folate-like scaffold of a metallo-β-lactamase inhibitor, we created a new molecule, MLLB-2201, that potentiates β-lactams and anti-folates and restores meropenem activity against metallo-β-lactamase-expressing Escherichia coli. These strategies are useful ways to tackle the ongoing rise in dangerous bacterial pathogens.
Collapse
Affiliation(s)
- Luke N. Yaeger
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - David Sychantha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Princeton Luong
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Shahrokh Shekarriz
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Océane Goncalves
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Annamaria Dobrin
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Michael R. Ranieri
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Ryan P. Lamers
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Hanjeong Harvey
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - George C. diCenzo
- Department of Biology, Queen’s University, Kingston, Ontario, Canada
| | - Michael Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Jean-Phiippe Côté
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Jakob Magolan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Lori L. Burrows
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
4
|
Dombrovski M, Zang Y, Frighetto G, Vaccari A, Jang H, Mirshahidi PS, Xie F, Sanfilippo P, Hina BW, Rehan A, Hussein RH, Mirshahidi PS, Lee C, Morris A, Frye MA, von Reyn CR, Kurmangaliyev YZ, Card GM, Zipursky SL. Gradients of Cell Recognition Molecules Wire Visuomotor Transformation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.04.610846. [PMID: 39974884 PMCID: PMC11838220 DOI: 10.1101/2024.09.04.610846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Converting sensory information into motor commands is fundamental to most of our actions 1,2 . In Drosophila , visuomotor transformations are mediated by Visual Projection Neurons (VPNs) 3,4 . These neurons encode object location and motion to drive directional behaviors through a synaptic gradient mechanism 5 . However, the molecular origins of such graded connectivity remain unknown. We addressed this question in a VPN cell type called LPLC2 6 , which integrates looming motion and transforms it into an escape response through two separate dorsoventral synaptic gradients at its inputs and outputs. We identified two corresponding dorsoventral expression gradients of cell recognition molecules within the LPLC2 population that regulate this synaptic connectivity. Dpr13 determines synaptic outputs of LPLC2 axons by interacting with its binding partner, DIP-ε, expressed in the Giant Fiber - a neuron that mediates escape 7 . Similarly, Beat-VI regulates synaptic inputs onto LPLC2 dendrites by interacting with Side-II expressed in upstream motion-detecting neurons. Behavioral, physiological, and molecular experiments demonstrate that these coordinated molecular gradients regulate synaptic connectivity, enabling the accurate transformation of visual features into motor commands. As continuous variation in gene expression within a neuronal type is also observed in the mammalian brain 8 , graded expression of cell recognition molecules may represent a common mechanism underlying synaptic specificity.
Collapse
|
5
|
Raghavan AR, May K, Subramanian VV, Blitzblau HG, Patel NJ, Houseley J, Hochwagen A. Distinct chromatin regulators downmodulate meiotic axis formation and DNA break induction at chromosome ends. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640173. [PMID: 40093131 PMCID: PMC11908166 DOI: 10.1101/2025.02.27.640173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
In many organisms, meiotic crossover recombination is suppressed near the extreme ends of chromosomes. Here, we identified two chromatin modifiers, the histone methyltransferase Dot1 and the Sir silencing complex, as regulators of this process in Saccharomyces cerevisiae. We show that the recombination-promoting axis proteins Red1 and Hop1, but not the axis-associated cohesin Rec8, are significantly reduced within 20 kb of telomeres compared to the chromosome interior. Dot1, which preferentially methylates histones in the chromosome interior, is required for this pattern by directing Red1 binding toward the chromosome interior. In parallel, the Sir complex suppresses the induction of meiotic DNA double-strand breaks (DSBs) at chromosome ends. Sir-dependent DSB suppression is independent of axis deposition and occurs in a chromosome end-specific manner that mirrors the spreading and transcriptional silencing activity of the complex, suggesting that the Sir complex suppresses DSB formation by limiting the openness of promoters, the preferred sites of meiotic DSB formation. We conclude that multiple chromatin-based mechanisms collaborate to achieve a robust reduction of meiotic recombination near chromosome ends.
Collapse
Affiliation(s)
| | - Kieron May
- Epigenetics Programme, Babraham Institute, Cambridge, UK
| | - Vijayalakshmi V Subramanian
- Department of Biology, New York University, New York, USA
- Department of Biology, IISER Tirupati, Tirupati, India
| | | | - Neem J Patel
- Department of Biology, New York University, New York, USA
| | | | | |
Collapse
|
6
|
Kimura H, Lahouel K, Tomasetti C, Roberts NJ. Functional characterization of all CDKN2A missense variants and comparison to in silico models of pathogenicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.28.573507. [PMID: 38234851 PMCID: PMC10793438 DOI: 10.1101/2023.12.28.573507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Interpretation of variants identified during genetic testing is a significant clinical challenge. In this study, we developed a high-throughput CDKN2A functional assay and characterized all possible CDKN2A missense variants. We found that 17.7% of all missense variants were functionally deleterious. We also used our functional classifications to assess the performance of in silico models that predict the effect of variants, including recently reported models based on machine learning. Notably, we found that all in silico models performed similarly when compared to our functional classifications with accuracies of 39.5-85.4%. Furthermore, while we found that functionally deleterious variants were enriched within ankyrin repeats, we did not identify any residues where all missense variants were functionally deleterious. Our functional classifications are a resource to aid the interpretation of CDKN2A variants and have important implications for the application of variant interpretation guidelines, particularly the use of in silico models for clinical variant interpretation.
Collapse
Affiliation(s)
- Hirokazu Kimura
- Department of Pathology, the Johns Hopkins University School of Medicine; Baltimore, 21287, USA
| | - Kamel Lahouel
- Division of Integrated Genomics, Translational Genomics Research Institute; Phoenix, 85004, USA
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope; Duarte, 91010, USA
| | - Cristian Tomasetti
- Division of Integrated Genomics, Translational Genomics Research Institute; Phoenix, 85004, USA
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope; Duarte, 91010, USA
| | - Nicholas J. Roberts
- Department of Pathology, the Johns Hopkins University School of Medicine; Baltimore, 21287, USA
- Department of Oncology, the Johns Hopkins University School of Medicine; Baltimore, 21287, USA
| |
Collapse
|
7
|
El-Araby AM, Fisher JF, Mobashery S. Bacterial peptidoglycan as a living polymer. Curr Opin Chem Biol 2025; 84:102562. [PMID: 39700530 PMCID: PMC11788026 DOI: 10.1016/j.cbpa.2024.102562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024]
Abstract
The peptidoglycan manifests as a multifaceted component of the bacterial cell wall. Throughout the lifecycle of the bacterium, the peptidoglycan is deconstructed, rebuilt, and remodeled for bacterial cell growth and replication. Degradation products of the peptidoglycan serve as precursors for cell-wall building blocks via recycling processes and as signaling molecules. Cell-wall recycling and de novo cell-wall synthesis converge biochemically at the cytoplasmic compartment. Peptidoglycan biochemistry is finely tuned to maintain the polymer's functions and is intimately connected to antibiotic-resistance mechanisms. Cell-wall-modifying enzymes present a unique opportunity for the discovery of antibiotics and antibiotic adjuvants. The unique chemical template of the peptidoglycan has been a target of numerous chemical biology approaches for investigating its functions and modulation. In this review, we highlight the current perspective on peptidoglycan research. We present recent efforts to understand the peptidoglycan as a functional component of antibiotic resistance, and as a target for antimicrobial therapy.
Collapse
Affiliation(s)
- Amr M El-Araby
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Jed F Fisher
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA.
| |
Collapse
|
8
|
Mejia-Santana A, Collins R, Doud EH, Landeta C. Disulfide bonds are required for cell division, cell envelope biogenesis and antibiotic resistance proteins in mycobacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635063. [PMID: 39975046 PMCID: PMC11838256 DOI: 10.1101/2025.01.27.635063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Mycobacteria, including Mycobacterium tuberculosis-the etiological agent of tuberculosis-have a unique cell envelope critical for their survival and resistance. The cell envelope's assembly and maintenance influence permeability, making it a key target against multidrug-resistant strains. Disulfide bond (DSB) formation is crucial for the folding of cell envelope proteins. The DSB pathway in mycobacteria includes two enzymes, DsbA and VKOR, required for survival. Using bioinformatics and cysteine profiling proteomics, we identified cell envelope proteins dependent on DSBs. We validated via in vivo alkylation that key proteins like LamA (MmpS3), PstP, LpqW, and EmbB rely on DSBs for stability. Furthermore, chemical inhibition of VKOR results in phenotypes similar to those of Δvkor. Thus, targeting DsbA-VKOR systems could compromise both cell division and mycomembrane integrity. These findings emphasize the potential of DSB inhibition as a novel strategy to combat mycobacterial infections.
Collapse
Affiliation(s)
| | - Rebecca Collins
- Department of Biology. Indiana University. Bloomington, IN. U.S.A
| | - Emma H. Doud
- Biochemistry and Molecular Biology. Indiana University School of Medicine. Indianapolis, IN. U.S.A
- Center for Proteome Analysis; Indiana University School of Medicine. Indianapolis, IN. U.S.A
| | - Cristina Landeta
- Department of Biology. Indiana University. Bloomington, IN. U.S.A
| |
Collapse
|
9
|
McCausland JW, Kloos ZA, Irnov I, Sonnert ND, Zhou J, Putnick R, Mueller EA, Steere AC, Palm NW, Grimes CL, Jacobs-Wagner C. Bacterial and host enzymes modulate the inflammatory response produced by the peptidoglycan of the Lyme disease agent. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631998. [PMID: 39829805 PMCID: PMC11741416 DOI: 10.1101/2025.01.08.631998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The spirochete Borrelia burgdorferi causes Lyme disease. In some patients, an excessive, dysregulated proinflammatory immune response can develop in joints leading to persistent arthritis. In such patients, persistence of antigenic B. burgdorferi peptidoglycan (PGBb) fragments within joint tissues may contribute to the immunopathogenesis, even after appropriate antibiotic treatment. In live B. burgdorferi cells, the outer membrane shields the polymeric PGBb sacculus from exposure to the immune system. However, unlike most diderm bacteria, B. burgdorferi releases PGBb turnover products into its environment due to the absence of recycling activity. In this study, we identified the released PGBb fragments using a mass spectrometry-based approach. By characterizing the l,d-carboxypeptidase activity of B. burgdorferi protein BB0605 (renamed DacA), we found that PGBb turnover largely occurs at sites of PGBb synthesis. In parallel, we demonstrated that the lytic transglycosylase activity associated with BB0259 (renamed MltS) releases PGBb fragments with 1,6-anhydro bond on their N-acetylmuramyl residues. Stimulation of human cell lines with various synthetic PGBb fragments revealed that 1,6-anhydromuramyl-containing PGBb fragments are poor inducers of a NOD2-dependent immune response relative to their hydrated counterparts. We also showed that the activity of the human N-acetylmuramyl-l-alanine amidase PGLYRP2, which reduces the immunogenicity of PGBb material, is low in joint (synovial) fluids relative to serum. Altogether, our findings suggest that MltS activity helps B. burgdorferi evade PG-based immune detection by NOD2 during growth despite shedding PGBb fragments and that PGBb-induced immunopathology likely results from host sensing of PGBb material from dead (lysed) spirochetes. Additionally, our results suggest the possibility that natural variation in PGLYRP2 activity may contribute to differences in susceptibility to PG-induced inflammation across tissues and individuals.
Collapse
Affiliation(s)
- Joshua W McCausland
- Sarafan ChEM-H Institute, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Zachary A Kloos
- Microbiology Program, Yale University, West Haven, Connecticut, USA
| | - Irnov Irnov
- Sarafan ChEM-H Institute, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Nicole D Sonnert
- Microbiology Program, Yale University, West Haven, Connecticut, USA
- Department of Immunology, Yale University School of Medicine, New Haven, CT, USA
| | - Junhui Zhou
- Department of Chemistry and Biochemistry, University of Delaware, Neward, DE
| | - Rachel Putnick
- Department of Chemistry and Biochemistry, University of Delaware, Neward, DE
| | - Elizabeth A Mueller
- Sarafan ChEM-H Institute, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Alan C Steere
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Noah W Palm
- Department of Immunology, Yale University School of Medicine, New Haven, CT, USA
| | - Catherine L Grimes
- Department of Chemistry and Biochemistry, University of Delaware, Neward, DE
| | - Christine Jacobs-Wagner
- Sarafan ChEM-H Institute, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
10
|
Fei Y, Luh M, Ontiri A, Ghauri D, Hu W, Liang L. Coordination of distinct sources of excitatory inputs enhances motion selectivity in the mouse visual thalamus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631826. [PMID: 39829841 PMCID: PMC11741327 DOI: 10.1101/2025.01.08.631826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Multiple sources innervate the visual thalamus to influence image-forming vision prior to the cortex, yet it remains unclear how non-retinal and retinal input coordinate to shape thalamic visual selectivity. Using dual-color two-photon calcium imaging in the thalamus of awake mice, we observed similar coarse-scale retinotopic organization between axons of superior colliculus neurons and retinal ganglion cells, both providing strong converging excitatory input to thalamic neurons. At a fine scale of ∼10 µm, collicular boutons often shared visual feature preferences with nearby retinal boutons. Inhibiting collicular input significantly suppressed visual responses in thalamic neurons and specifically reduced motion selectivity in neurons preferring nasal-to-temporal motion. The reduction in motion selectivity could be the result of silencing sharply tuned direction-selective colliculogeniculate input. These findings suggest that the thalamus is not merely a relay but selectively integrates inputs from multiple regions to build stimulus selectivity and shape the information transmitted to the cortex. HIGHLIGHTS Chronic dual-color calcium imaging reveals diverse visual tuning of collicular axonal boutons.Nearby collicular and retinal boutons often share feature preferences at ∼10 µm scaleSilencing of collicular input suppresses visual responses in the majority of thalamic neurons.Silencing of collicular input reduces motion selectivity in thalamic neurons.
Collapse
|
11
|
van der Molen T, Spaeth A, Chini M, Hernandez S, Kaurala GA, Schweiger HE, Duncan C, McKenna S, Geng J, Lim M, Bartram J, Dendukuri A, Zhang Z, Gonzalez-Ferrer J, Bhaskaran-Nair K, Blauvelt LJ, Harder CR, Petzold LR, Alam El Din DM, Laird J, Schenke M, Smirnova L, Colquitt BM, Mostajo-Radji MA, Hansma PK, Teodorescu M, Hierlemann A, Hengen KB, Hanganu-Opatz IL, Kosik KS, Sharf T. Protosequences in brain organoids model intrinsic brain states Authors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.29.573646. [PMID: 38234832 PMCID: PMC10793448 DOI: 10.1101/2023.12.29.573646] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Neuronal firing sequences are thought to be the basic building blocks of neural coding and information broadcasting within the brain. However, when sequences emerge during neurodevelopment remains unknown. We demonstrate that structured firing sequences are present in spontaneous activity of human and murine brain organoids and ex vivo neonatal brain slices from the murine somatosensory cortex. We observed a balance between temporally rigid and flexible firing patterns that are emergent phenomena in human and murine brain organoids and early postnatal murine somatosensory cortex, but not in primary dissociated cortical cultures. Our findings suggest that temporal sequences do not arise in an experience-dependent manner, but are rather constrained by an innate preconfigured architecture established during neurogenesis. These findings highlight the potential for brain organoids to further explore how exogenous inputs can be used to refine neuronal circuits and enable new studies into the genetic mechanisms that govern assembly of functional circuitry during early human brain development.
Collapse
Affiliation(s)
- Tjitse van der Molen
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106, USA
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Alex Spaeth
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Mattia Chini
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Sebastian Hernandez
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Gregory A. Kaurala
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Hunter E. Schweiger
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Cole Duncan
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Sawyer McKenna
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jinghui Geng
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Max Lim
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106, USA
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Julian Bartram
- Department of Biosystems Science and Engineering, ETH Zürich, Klingelbergstrasse 48, 4056 Basel, Switzerland
| | - Aditya Dendukuri
- Department of Computer Science, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Zongren Zhang
- Department of Physics, University of California Santa Barbara, Santa Barbara, CA 93106
| | - Jesus Gonzalez-Ferrer
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Kiran Bhaskaran-Nair
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Lon J. Blauvelt
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
| | - Cole R.K. Harder
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Linda R. Petzold
- Department of Computer Science, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Dowlette-Mary Alam El Din
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jason Laird
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health Johns Hopkins University, Baltimore, MD 21205, USA
| | - Maren Schenke
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health Johns Hopkins University, Baltimore, MD 21205, USA
| | - Lena Smirnova
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health Johns Hopkins University, Baltimore, MD 21205, USA
| | - Bradley M. Colquitt
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
- Institute for the Biology of Stem Cells, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | | | - Paul K. Hansma
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106, USA
- Department of Physics, University of California Santa Barbara, Santa Barbara, CA 93106
| | - Mircea Teodorescu
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Andreas Hierlemann
- Department of Biosystems Science and Engineering, ETH Zürich, Klingelbergstrasse 48, 4056 Basel, Switzerland
| | - Keith B. Hengen
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Ileana L. Hanganu-Opatz
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Kenneth S. Kosik
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106, USA
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Tal Sharf
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Institute for the Biology of Stem Cells, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
12
|
Zeidler Z, Gomez MF, Gupta TA, Shari M, Wilke SA, DeNardo LA. Prefrontal dopamine activity is critical for rapid threat avoidance learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.02.592069. [PMID: 39803535 PMCID: PMC11722269 DOI: 10.1101/2024.05.02.592069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
The medial prefrontal cortex (mPFC) is required for learning associations that determine whether animals approach or avoid potential threats in the environment. Dopaminergic (DA) projections from the ventral tegmental area (VTA) to the mPFC carry information, particularly about aversive outcomes, that may inform prefrontal computations. But the role of prefrontal DA in learning based on aversive outcomes remains poorly understood. Here, we used platform mediated avoidance (PMA) to study the role of mPFC DA in threat avoidance learning in mice. We show that activity in VTA-mPFC dopaminergic terminals is required for avoidance learning, but not for escape, conditioned fear, or to recall a previously learned avoidance strategy. mPFC DA is most dynamic in the early stages of learning, and encodes aversive outcomes, their omissions, and threat-induced behaviors. Computational models of PMA behavior and DA activity revealed that mPFC DA influences learning rates and encodes the predictive relationships between cues and adaptive behaviors. Taken together, these data indicate that mPFC DA is necessary to rapidly learn behaviors required to avoid signaled threats, but not for learning cue-threat associations.
Collapse
Affiliation(s)
- Zachary Zeidler
- Department of Physiology; David Geffen School of Medicine, University of California, Los Angeles, California
| | - Marta Fernandez Gomez
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Tanya A. Gupta
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California
| | | | - Scott A. Wilke
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Laura A. DeNardo
- Department of Physiology; David Geffen School of Medicine, University of California, Los Angeles, California
- Lead contact
| |
Collapse
|
13
|
Waldern JM, Taylor C, Giannetti CA, Irving PS, Allen SR, Zhu M, Backofen R, Mathews D, Weeks KM, Laederach A. Structural determinants of inverted Alu-mediated backsplicing revealed by -MaP and -JuMP. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.13.628372. [PMID: 39713457 PMCID: PMC11661277 DOI: 10.1101/2024.12.13.628372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Biogenesis of circular RNA usually involves a backsplicing reaction where the downstream donor site is ligated to the upstream acceptor site by the spliceosome. For this reaction to occur, it is hypothesized that these sites must be in proximity. Inverted repeat sequences, such as Alu elements, in the upstream and downstream introns are predicted to base-pair and represent one mechanism for inducing proximity. Here, we investigate the pre-mRNA structure of the human HIPK3 gene at exon 2, which forms a circular RNA via backsplicing. We leverage multiple chemical probing techniques, including the recently developed SHAPE- JuMP strategy, to characterize secondary and tertiary interactions in the pre- mRNA that govern backsplicing. Our data confirm that the antisense Alu elements, AluSz(-) and AluSq2(+) in the upstream and downstream introns, form a highly- paired interaction. Circularization requires formation of long-range Alu-mediated base pairs but does not require the full-length AluSq2(+). In addition to confirming long-range base pairs, our SHAPE-JuMP data identified multiple long-range interactions between non-pairing nucleotides. Genome-wide analysis of inverted repeats flanking circular RNAs confirm that their presence favors circularization, but the overall effect is modest. Together these results suggest that secondary structure considerations alone cannot fully explain backsplicing and additional interactions are key.
Collapse
|
14
|
Murase S, Severin D, Dye L, Mesik L, Moreno C, Kirkwood A, Quinlan EM. Adult visual deprivation engages associative, presynaptic plasticity of thalamic input to cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626829. [PMID: 39677752 PMCID: PMC11643054 DOI: 10.1101/2024.12.04.626829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Associative plasticity at thalamocortical synapses is thought to be constrained by age in the mammalian cortex. However, here we show for the first time that prolonged visual deprivation induces robust and reversible plasticity at synapses between first order visual thalamus and cortical layer 4 pyramidal neurons. The plasticity is associative and expressed by changes in presynaptic function, thereby amplifying and relaying the change in efferent drive to the visual cortex.
Collapse
|
15
|
Vogler NW, Chen R, Virkler A, Tu VY, Gottfried JA, Geffen MN. Direct Piriform-to-Auditory Cortical Projections Shape Auditory-Olfactory Integration. J Neurosci 2024; 44:e1140242024. [PMID: 39510831 PMCID: PMC11622214 DOI: 10.1523/jneurosci.1140-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/12/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024] Open
Abstract
In a real-world environment, the brain must integrate information from multiple sensory modalities, including the auditory and olfactory systems. However, little is known about the neuronal circuits governing how odors influence and modulate sound processing. Here, we investigated the mechanisms underlying auditory-olfactory integration using anatomical, electrophysiological, and optogenetic approaches, focusing on the auditory cortex as a key locus for cross-modal integration. First, retrograde and anterograde viral tracing strategies revealed a direct projection from the piriform cortex to the auditory cortex. Next, using in vivo electrophysiological recordings of neuronal activity in the auditory cortex of awake male or female mice, we found that odors modulate auditory cortical responses to sound. Finally, we used in vivo optogenetic manipulations during electrophysiology to demonstrate that olfactory modulation in the auditory cortex, specifically, odor-driven enhancement of sound responses, depends on direct input from the piriform cortex. Together, our results identify a novel role of piriform-to-auditory cortical circuitry in shaping olfactory modulation in the auditory cortex, shedding new light on the neuronal mechanisms underlying auditory-olfactory integration.
Collapse
Affiliation(s)
- Nathan W Vogler
- Departments of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
| | - Ruoyi Chen
- Departments of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
| | - Alister Virkler
- Neurology, Perelman School of Medicine, University of Pennsylvania
| | - Violet Y Tu
- Departments of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
| | - Jay A Gottfried
- Neurology, Perelman School of Medicine, University of Pennsylvania
| | - Maria N Geffen
- Departments of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
- Neurology, Perelman School of Medicine, University of Pennsylvania
- Neuroscience, Perelman School of Medicine, University of Pennsylvania
| |
Collapse
|
16
|
Pan D, Martin CA, Nazareth J, Sze S, Al-Oraibi A, Gogoi M, Grolmusova N, Divall P, Decker J, Fletcher E, Williams C, Hay J, Baggaley RF, Wyllie AL, Stephenson I, Gaillard E, Nellums LB, Clark TW, Van-Tam JN, Cowling BJ, Hollingsworth TD, Gray L, Barer M, Pareek M. Defining within-host SARS-CoV-2 RNA viral load kinetics during acute COVID-19 infection within different respiratory compartments and their respective associations with host infectiousness: a protocol for a systematic review and meta-analysis. BMJ Open 2024; 14:e085127. [PMID: 39622563 PMCID: PMC11624818 DOI: 10.1136/bmjopen-2024-085127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024] Open
Abstract
INTRODUCTION Understanding how RNA viral load changes (viral load kinetics) during acute infection in SARS-CoV-2 can help to identify when and which patients are most infectious. We seek to summarise existing data on the longitudinal RNA viral load kinetics of SARS-CoV-2 sampled from different parts of the respiratory tract (nose, nasopharynx, oropharynx, saliva and exhaled breath) and how this may vary with age, sex, ethnicity, immune status, disease severity, vaccination, treatment and virus variant. METHODS AND ANALYSIS We will conduct a systematic review and meta-analysis, using studies identified through MEDLINE and EMBASE (via Ovid). All research studies reporting primary data on longitudinal RNA viral load kinetics of infected patients with SARS-CoV-2 will be included. Methodological quality will be assessed using a validated checklist for longitudinal studies as well as predefined quality criteria for assessment of individual-level RNA viral load. Should the data allow, we will aim to perform individual patient-level meta-analysis. Our primary outcomes are duration to, and quantity of peak RNA viral load, and total duration of viral load shedding within different respiratory compartments. Secondary outcomes include duration of lateral flow antigen and virus culture positivity and variation of RNA viral load by age, immune status, disease severity, vaccination, treatment, lateral flow tests, viral culture positivity and SARS-CoV-2 variant. Study-level effects affecting observations, but not related to properties of the patient, such as the PCR platform and gene target will also be recorded. Random-effects models will estimate the population mean and individual-level variation in viral shedding conditional on the aforementioned variables. Finally, we will summarise the key mechanistic models used in the literature to reconstruct individual-level viral kinetics and estimate how different factors shape viral dynamics over time. ETHICS AND DISSEMINATION Ethical approval is not needed as data will be obtained from published articles or studies with data that have already received and ethical review for analysis. Manuscript(s) will be prepared for publication. SYSTEMATIC REVIEW PROTOCOL REGISTRATION PROSPERO ID: CRD42023385315.
Collapse
Affiliation(s)
- Daniel Pan
- Development Centre for Population Health, University of Leicester, Leicester, UK
- Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
- Oxford Big Data Institute, University of Oxford, Oxford, UK
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, Hong Kong School of Public Health, Hong Kong, Hong Kong
| | - Christopher A Martin
- Development Centre for Population Health, University of Leicester, Leicester, UK
- Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
| | - Joshua Nazareth
- Development Centre for Population Health, University of Leicester, Leicester, UK
- Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
| | - Shirley Sze
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Amani Al-Oraibi
- Development Centre for Population Health, University of Leicester, Leicester, UK
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
- Lifespan and Population Health, University of Nottingham, Nottingham, UK
| | - Mayuri Gogoi
- Development Centre for Population Health, University of Leicester, Leicester, UK
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Natalia Grolmusova
- Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
| | - Pip Divall
- Education Centre Library, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Jonathan Decker
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Eve Fletcher
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
| | - Caroline Williams
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
- Department of Clinical Microbiology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - James Hay
- Oxford Big Data Institute, University of Oxford, Oxford, UK
| | - Rebecca F. Baggaley
- Development Centre for Population Health, University of Leicester, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
| | - Anne L Wyllie
- Department of Epidemiology of Microbial Diseases, Yale University, New Haven, Connecticut, USA
| | - Iain Stephenson
- Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Erol Gaillard
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
- Department of Paediatric Respiratory Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Laura B Nellums
- College of Population Health, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Tristan William Clark
- Clinical and Experimental Sciences, University of Southampton, Southampton, UK
- Department of Infection, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- NIHR Southampton Biomedical Research Centre, Southampton, UK
| | | | - Benjamin J Cowling
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, Hong Kong School of Public Health, Hong Kong, Hong Kong
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Parks Corporation, Hong Kong, Hong Kong
| | - T. Déirdre Hollingsworth
- Oxford Big Data Institute, University of Oxford, Oxford, UK
- NDM Centre for Global Health Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Laura Gray
- Leicester NIHR Biomedical Research Centre, Leicester, UK
- Department of Population Health Sciences, University of Leicester, Leicester, UK
- NIHR Applied Research Collaborative East Midlands, Leicester, UK
| | - Michael Barer
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
- Department of Clinical Microbiology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Manish Pareek
- Development Centre for Population Health, University of Leicester, Leicester, UK
- Department of Infectious Diseases and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
- NIHR Applied Research Collaborative East Midlands, Leicester, UK
| |
Collapse
|
17
|
Jin B, Gongwer MW, Kearney BP, Ohanian L, Holden-Wingate L, Le B, Darmawan A, Nakayama Y, Mora SAR, DeNardo LA. A developmental brain-wide screen identifies retrosplenial cortex as a key player in the emergence of persistent memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.07.574554. [PMID: 38260633 PMCID: PMC10802387 DOI: 10.1101/2024.01.07.574554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Memories formed early in life are short-lived while those formed later persist. Recent work revealed that infant memories are stored in a latent state. But why they fail to be retrieved is poorly understood. Here we investigated brain-wide circuit mechanisms underlying infantile amnesia. We performed a screen that combined contextual fear conditioning, activity-dependent neuronal tagging at different postnatal ages, tissue clearing and light sheet microscopy. We observed striking developmental changes in regional activity patterns between infant, juvenile, and adult mice, including changes in the retrosplenial cortex (RSP) that aligned with the emergence of persistent memory. We then performed a series of targeted investigations of RSP structure and function across development. Chronic chemogenetic reactivation of tagged RSP ensembles during the week after learning enhanced memory in adults and juveniles, but not in infants. However, after 33 days, reactivating infant-tagged RSP ensembles recovered forgotten memories. Changes in the developmental functions of RSP memory ensembles were accompanied by changes in dendritic spine density and the likelihood that those ensembles could be reactivated by contextual cues. These studies show that RSP ensembles store latent infant memories, reveal the time course of RSP functional maturation, and suggest that immature RSP functional networks contribute to infantile amnesia.
Collapse
|
18
|
Petty GH, Bruno RM. Attentional modulation of secondary somatosensory and visual thalamus of mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586242. [PMID: 38585833 PMCID: PMC10996504 DOI: 10.1101/2024.03.22.586242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Each sensory modality has its own primary and secondary thalamic nuclei. While the primary thalamic nuclei are well understood to relay sensory information from the periphery to the cortex, the role of secondary sensory nuclei is elusive. We trained head-fixed mice to ateend to one sensory modality while ignoring a second modality, namely to ateend to touch and ignore vision, or vice versa. Arrays were used to record simultaneously from secondary somatosensory thalamus (POm) and secondary visual thalamus (LP). In mice trained to respond to tactile stimuli and ignore visual stimuli, POm was robustly activated by touch and largely unresponsive to visual stimuli. A different pateern was observed when mice were trained to respond to visual stimuli and ignore touch, with POm now more robustly activated during visual trials. This POm activity was not explained by differences in movements (i.e., whisking, licking, pupil dilation) resulting from the two tasks. Post hoc histological reconstruction of array tracks through POm revealed that subregions varied in their degree of plasticity. LP exhibited similar phenomena. We conclude that behavioral training reshapes activity in secondary thalamic nuclei. Secondary nuclei respond to the same behaviorally relevant, reward-predicting stimuli regardless of stimulus modality.
Collapse
Affiliation(s)
- Gordon H Petty
- Department of Neuroscience, Columbia University, New York, NY 10027 USA
- Department of Physiology, Anatomy, & Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom
| | - Randy M Bruno
- Department of Neuroscience, Columbia University, New York, NY 10027 USA
- Department of Physiology, Anatomy, & Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom
| |
Collapse
|
19
|
Karampelias C, Băloiu B, Rathkolb B, da Silva-Buttkus P, Bachar-Wikström E, Marschall S, Fuchs H, Gailus-Durner V, Chu L, Hrabě de Angelis M, Andersson O. Examining the liver-pancreas crosstalk reveals a role for the molybdenum cofactor in β-cell regeneration. Life Sci Alliance 2024; 7:e202402771. [PMID: 39159974 PMCID: PMC11333758 DOI: 10.26508/lsa.202402771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/21/2024] Open
Abstract
Regeneration of insulin-producing β-cells is an alternative avenue to manage diabetes, and it is crucial to unravel this process in vivo during physiological responses to the lack of β-cells. Here, we aimed to characterize how hepatocytes can contribute to β-cell regeneration, either directly or indirectly via secreted proteins or metabolites, in a zebrafish model of β-cell loss. Using lineage tracing, we show that hepatocytes do not directly convert into β-cells even under extreme β-cell ablation conditions. A transcriptomic analysis of isolated hepatocytes after β-cell ablation displayed altered lipid- and glucose-related processes. Based on the transcriptomics, we performed a genetic screen that uncovers a potential role of the molybdenum cofactor (Moco) biosynthetic pathway in β-cell regeneration and glucose metabolism in zebrafish. Consistently, molybdenum cofactor synthesis 2 (Mocs2) haploinsufficiency in mice indicated dysregulated glucose metabolism and liver function. Together, our study sheds light on the liver-pancreas crosstalk and suggests that the molybdenum cofactor biosynthesis pathway should be further studied in relation to glucose metabolism and diabetes.
Collapse
Affiliation(s)
- Christos Karampelias
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
| | - Bianca Băloiu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Birgit Rathkolb
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Patricia da Silva-Buttkus
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Etty Bachar-Wikström
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Susan Marschall
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Valerie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Lianhe Chu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Freising, Germany
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
20
|
Chen C, Song S. Distinct Neuron Types Contribute to Hybrid Auditory Spatial Coding. J Neurosci 2024; 44:e0159242024. [PMID: 39261006 PMCID: PMC11502229 DOI: 10.1523/jneurosci.0159-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 07/20/2024] [Accepted: 07/28/2024] [Indexed: 09/13/2024] Open
Abstract
Neural decoding is a tool for understanding how activities from a population of neurons inside the brain relate to the outside world and for engineering applications such as brain-machine interfaces. However, neural decoding studies mainly focused on different decoding algorithms rather than different neuron types which could use different coding strategies. In this study, we used two-photon calcium imaging to assess three auditory spatial decoders (space map, opponent channel, and population pattern) in excitatory and inhibitory neurons in the dorsal inferior colliculus of male and female mice. Our findings revealed a clustering of excitatory neurons that prefer similar interaural level difference (ILD), the primary spatial cues in mice, while inhibitory neurons showed random local ILD organization. We found that inhibitory neurons displayed lower decoding variability under the opponent channel decoder, while excitatory neurons achieved higher decoding accuracy under the space map and population pattern decoders. Further analysis revealed that the inhibitory neurons' preference for ILD off the midline and the excitatory neurons' heterogeneous ILD tuning account for their decoding differences. Additionally, we discovered a sharper ILD tuning in the inhibitory neurons. Our computational model, linking this to increased presynaptic inhibitory inputs, was corroborated using monaural and binaural stimuli. Overall, this study provides experimental and computational insight into how excitatory and inhibitory neurons uniquely contribute to the coding of sound locations.
Collapse
Affiliation(s)
- Chenggang Chen
- Tsinghua Laboratory of Brain and Intelligence and School of Biomedical Engineering, McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Sen Song
- Tsinghua Laboratory of Brain and Intelligence and School of Biomedical Engineering, McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| |
Collapse
|
21
|
Lakey B, Alberge F, Donohue TJ. Insights into Alphaproteobacterial regulators of cell envelope remodeling. Curr Opin Microbiol 2024; 81:102538. [PMID: 39232444 DOI: 10.1016/j.mib.2024.102538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/06/2024]
Abstract
The cell envelope is at the center of many processes essential for bacterial lifestyles. In addition to giving bacteria shape and delineating it from the environment, it contains macromolecules important for energy transduction, cell division, protection against toxins, biofilm formation, or virulence. Hence, many systems coordinate different processes within the cell envelope to ensure function and integrity. Two-component systems have been identified as crucial regulators of cell envelope functions over the last few years. In this review, we summarize the new information obtained on the regulation of cell envelope biosynthesis and homeostasis in α-proteobacteria, as well as newly identified targets that coordinate the processes in the cell envelope.
Collapse
Affiliation(s)
- Bryan Lakey
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - François Alberge
- CEA, CNRS, Aix-Marseille Université, Institut de Biosciences et Biotechnologies d'Aix-Marseille, UMR 7265, CEA Cadarache, Saint Paul-lez Durance, France
| | - Timothy J Donohue
- Department of Bacteriology, Wisconsin Energy Institute, University of Wisconsin Madison, Madison, WI, USA.
| |
Collapse
|
22
|
Ramirez Carbo CA, Faromiki OG, Nan B. A lytic transglycosylase connects bacterial focal adhesion complexes to the peptidoglycan cell wall. eLife 2024; 13:RP99273. [PMID: 39352247 PMCID: PMC11444678 DOI: 10.7554/elife.99273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024] Open
Abstract
The Gram-negative bacterium Myxococcus xanthus glides on solid surfaces. Dynamic bacterial focal adhesion complexes (bFACs) convert proton motive force from the inner membrane into mechanical propulsion on the cell surface. It is unclear how the mechanical force transmits across the rigid peptidoglycan (PG) cell wall. Here, we show that AgmT, a highly abundant lytic PG transglycosylase homologous to Escherichia coli MltG, couples bFACs to PG. Coprecipitation assay and single-particle microscopy reveal that the gliding motors fail to connect to PG and thus are unable to assemble into bFACs in the absence of an active AgmT. Heterologous expression of E. coli MltG restores the connection between PG and bFACs and thus rescues gliding motility in the M. xanthus cells that lack AgmT. Our results indicate that bFACs anchor to AgmT-modified PG to transmit mechanical force across the PG cell wall.
Collapse
Affiliation(s)
- Carlos A Ramirez Carbo
- Department of Biology, Texas A&M University, College Station, United States
- The Genetics and Genomics Interdisciplinary Program, Texas A&M University, College Station, United States
| | | | - Beiyan Nan
- Department of Biology, Texas A&M University, College Station, United States
| |
Collapse
|
23
|
Korotkevich E, Conrad DN, Gartner ZJ, O’Farrell PH. Selection promotes age-dependent degeneration of the mitochondrial genome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615276. [PMID: 39386732 PMCID: PMC11463671 DOI: 10.1101/2024.09.27.615276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Somatic mutations in mitochondrial genomes (mtDNA) accumulate exponentially during aging. Using single cell sequencing, we characterize the spectrum of age-accumulated mtDNA mutations in mouse and human liver and identify directional forces that accelerate the accumulation of mutations beyond the rate predicted by a neutral model. "Driver" mutations that give genomes a replicative advantage rose to high cellular abundance and carried along "passenger" mutations, some of which are deleterious. In addition, alleles that alter mtDNA-encoded proteins selectively increased in abundance overtime, strongly supporting the idea of a "destructive" selection that favors genomes lacking function. Overall, this combination of selective forces acting in hepatocytes promotes somatic accumulation of mutations in coding regions of mtDNA that are otherwise conserved in evolution. We propose that these selective processes could contribute to the population prevalence of mtDNA mutations, accelerate the course of heteroplasmic mitochondrial diseases and promote age-associated erosion of the mitochondrial genome.
Collapse
Affiliation(s)
- Ekaterina Korotkevich
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, California, USA
| | - Daniel N. Conrad
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Patrick H. O’Farrell
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
24
|
Vogler NW, Chen R, Virkler A, Tu VY, Gottfried JA, Geffen MN. Direct piriform-to-auditory cortical projections shape auditory-olfactory integration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.602976. [PMID: 39071445 PMCID: PMC11275881 DOI: 10.1101/2024.07.11.602976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
In a real-world environment, the brain must integrate information from multiple sensory modalities, including the auditory and olfactory systems. However, little is known about the neuronal circuits governing how odors influence and modulate sound processing. Here, we investigated the mechanisms underlying auditory-olfactory integration using anatomical, electrophysiological, and optogenetic approaches, focusing on the auditory cortex as a key locus for cross-modal integration. First, retrograde and anterograde viral tracing strategies revealed a direct projection from the piriform cortex to the auditory cortex. Next, using in vivo electrophysiological recordings of neuronal activity in the auditory cortex of awake male or female mice, we found that odors modulate auditory cortical responses to sound. Finally, we used in vivo optogenetic manipulations during electrophysiology to demonstrate that olfactory modulation in auditory cortex, specifically, odor-driven enhancement of sound responses, depends on direct input from the piriform cortex. Together, our results identify a novel role of piriform-to-auditory cortical circuitry in shaping olfactory modulation in the auditory cortex, shedding new light on the neuronal mechanisms underlying auditory-olfactory integration.
Collapse
Affiliation(s)
- Nathan W. Vogler
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
| | - Ruoyi Chen
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
| | - Alister Virkler
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania
| | - Violet Y. Tu
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
| | - Jay A. Gottfried
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania
| | - Maria N. Geffen
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania
| |
Collapse
|
25
|
Alvarez L, Hernandez SB, Torrens G, Weaver AI, Dörr T, Cava F. Control of bacterial cell wall autolysins by peptidoglycan crosslinking mode. Nat Commun 2024; 15:7937. [PMID: 39261529 PMCID: PMC11390936 DOI: 10.1038/s41467-024-52325-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024] Open
Abstract
To withstand their internal turgor pressure and external threats, most bacteria have a protective peptidoglycan (PG) cell wall. The growth of this PG polymer relies on autolysins, enzymes that create space within the structure. Despite extensive research, the regulatory mechanisms governing these PG-degrading enzymes remain poorly understood. Here, we unveil a novel and widespread control mechanism of lytic transglycosylases (LTs), a type of autolysin responsible for breaking down PG glycan chains. Specifically, we show that LD-crosslinks within the PG sacculus act as an inhibitor of LT activity. Moreover, we demonstrate that this regulation controls the release of immunogenic PG fragments and provides resistance against predatory LTs of both bacterial and viral origin. Our findings address a critical gap in understanding the physiological role of the LD-crosslinking mode in PG homeostasis, highlighting how bacteria can enhance their resilience against environmental threats, including phage attacks, through a single structural PG modification.
Collapse
Affiliation(s)
- Laura Alvarez
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Sara B Hernandez
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Instituto de Bioquímica Vegetal y Fotosíntesis, Consejo Superior de Investigaciones Científicas and Universidad de Sevilla, Seville, Spain
| | - Gabriel Torrens
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Anna I Weaver
- Department of Microbiology, Cornell University, Ithaca, New York, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Tobias Dörr
- Department of Microbiology, Cornell University, Ithaca, New York, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, USA
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, USA
| | - Felipe Cava
- Department of Molecular Biology, Umeå University, Umeå, Sweden.
- Umeå Center for Microbial Research (UCMR), Umeå University, Umeå, Sweden.
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå, Sweden.
- Science for Life Laboratory (SciLifeLab), Umeå University, Umeå, Sweden.
| |
Collapse
|
26
|
Chaverra M, Cheney AM, Scheel A, Miller A, George L, Schultz A, Henningsen K, Kominsky D, Walk H, Kennedy WR, Kaufmann H, Walk S, Copié V, Lefcort F. ELP1, the Gene Mutated in Familial Dysautonomia, Is Required for Normal Enteric Nervous System Development and Maintenance and for Gut Epithelium Homeostasis. J Neurosci 2024; 44:e2253232024. [PMID: 39138000 PMCID: PMC11391678 DOI: 10.1523/jneurosci.2253-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 07/31/2024] [Accepted: 08/04/2024] [Indexed: 08/15/2024] Open
Abstract
Familial dysautonomia (FD) is a rare sensory and autonomic neuropathy that results from a mutation in the ELP1 gene. Virtually all patients report gastrointestinal (GI) dysfunction and we have recently shown that FD patients have a dysbiotic gut microbiome and altered metabolome. These findings were recapitulated in an FD mouse model and moreover, the FD mice had reduced intestinal motility, as did patients. To understand the cellular basis for impaired GI function in FD, the enteric nervous system (ENS; both female and male mice) from FD mouse models was analyzed during embryonic development and adulthood. We show here that not only is Elp1 required for the normal formation of the ENS, but it is also required in adulthood for the regulation of both neuronal and non-neuronal cells and for target innervation in both the mucosa and in intestinal smooth muscle. In particular, CGRP innervation was significantly reduced as was the number of dopaminergic neurons. Examination of an FD patient's gastric biopsy also revealed reduced and disoriented axons in the mucosa. Finally, using an FD mouse model in which Elp1 was deleted exclusively from neurons, we found significant changes to the colon epithelium including reduced E-cadherin expression, perturbed mucus layer organization, and infiltration of bacteria into the mucosa. The fact that deletion of Elp1 exclusively in neurons is sufficient to alter the intestinal epithelium and perturb the intestinal epithelial barrier highlights a critical role for neurons in regulating GI epithelium homeostasis.
Collapse
Affiliation(s)
- Marta Chaverra
- Departments of Microbiology and Cell Biology, Montana State University, Bozeman, Montana 59717
| | - Alexandra M Cheney
- Biochemistry and Chemistry, Montana State University, Bozeman, Montana 59717
| | - Alpha Scheel
- Departments of Microbiology and Cell Biology, Montana State University, Bozeman, Montana 59717
| | - Alessa Miller
- Departments of Microbiology and Cell Biology, Montana State University, Bozeman, Montana 59717
| | - Lynn George
- Department of Biological and Physical Sciences, Montana State University, Billings, Montana 59101
| | - Anastasia Schultz
- Departments of Microbiology and Cell Biology, Montana State University, Bozeman, Montana 59717
| | - Katelyn Henningsen
- Departments of Microbiology and Cell Biology, Montana State University, Bozeman, Montana 59717
| | - Douglas Kominsky
- Departments of Microbiology and Cell Biology, Montana State University, Bozeman, Montana 59717
| | - Heather Walk
- Departments of Microbiology and Cell Biology, Montana State University, Bozeman, Montana 59717
| | - William R Kennedy
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota 55455
| | - Horacio Kaufmann
- Department of Neurology, New York University School of Medicine, New York, New York 10016
| | - Seth Walk
- Departments of Microbiology and Cell Biology, Montana State University, Bozeman, Montana 59717
| | - Valérie Copié
- Biochemistry and Chemistry, Montana State University, Bozeman, Montana 59717
| | - Frances Lefcort
- Departments of Microbiology and Cell Biology, Montana State University, Bozeman, Montana 59717
| |
Collapse
|
27
|
Ramteke P, Watson B, Toci M, Tran VA, Johnston S, Tsingas M, Barve RA, Mitra R, Loeser RF, Collins JA, Risbud MV. SIRT6 loss causes intervertebral disc degeneration in mice by promoting senescence and SASP status. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612072. [PMID: 39314282 PMCID: PMC11419082 DOI: 10.1101/2024.09.09.612072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Intervertebral disc degeneration is a major risk factor contributing to chronic low back and neck pain. While the etiological factors for disc degeneration vary, age is still one of the most important risk factors. Recent studies have shown the promising role of SIRT6 in mammalian aging and skeletal tissue health, however its role in the intervertebral disc health remains unexplored. We investigated the contribution of SIRT6 to disc health by studying the age-dependent spinal phenotype of mice with conditional deletion of Sirt6 in the disc (Acan CreERT2 ; Sirt6 fl/fl ). Histological studies showed a degenerative phenotype in knockout mice compared to Sirt6 fl/fl control mice at 12 months which became pronounced at 24 months. RNA-Seq analysis of NP and AF tissues, quantitative histone analysis, and in vitro multiomics employing RNA-seq with ATAC-seq revealed that SIRT6-loss resulted in changes in acetylation and methylation status of specific Histone 3 lysine residues, thereby affecting DNA accessibility and transcriptomic landscape. A decrease in autophagy and an increase in DNA damage were also noted in Sirt6-deficient cells. Further mechanistic insights revealed that loss of SIRT6 increased senescence and SASP burden in the disc characterized by increased p21, γH2AX, IL-6, and TGF-β abundance. Taken together our study highlights the contribution of SIRT6 in modulating DNA damage, autophagy and cell senescence, and its importance in maintaining disc health during aging thereby underscoring it as a potential therapeutic target to treat intervertebral disc degeneration.
Collapse
Affiliation(s)
- Pranay Ramteke
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Bahiyah Watson
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Mallory Toci
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Victoria A Tran
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Shira Johnston
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Maria Tsingas
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Ruteja A. Barve
- Department of Genetics, Genome Technology Access Centre at the McDonnell Genome Institute, Washington University, School of Medicine, St. Louis, MO, 63110, USA
| | - Ramkrishna Mitra
- Department of Pharmacology and Biostatistics, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Richard F. Loeser
- Thurston Arthritis Research Center and the Division of Rheumatology, Allergy, and Immunology, 3300 Thurston Building, Campus Box 7280, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599-7280, USA
| | - John A. Collins
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Makarand V. Risbud
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| |
Collapse
|
28
|
Zutshi I, Apostolelli A, Yang W, Zheng ZS, Dohi T, Balzani E, Williams AH, Savin C, Buzsáki G. Hippocampal neuronal activity is aligned with action plans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611533. [PMID: 39282373 PMCID: PMC11398474 DOI: 10.1101/2024.09.05.611533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Neurons in the hippocampus are correlated with different variables, including space, time, sensory cues, rewards, and actions, where the extent of tuning depends on ongoing task demands. However, it remains uncertain whether such diverse tuning corresponds to distinct functions within the hippocampal network or if a more generic computation can account for these observations. To disentangle the contribution of externally driven cues versus internal computation, we developed a task in mice where space, auditory tones, rewards, and context were juxtaposed with changing relevance. High-density electrophysiological recordings revealed that neurons were tuned to each of these modalities. By comparing movement paths and action sequences, we observed that external variables had limited direct influence on hippocampal firing. Instead, spiking was influenced by online action plans modulated by goal uncertainty. Our results suggest that internally generated cell assembly sequences are selected and updated by action plans toward deliberate goals. The apparent tuning of hippocampal neuronal spiking to different sensory modalities might emerge due to alignment to the afforded action progression within a task rather than representation of external cues.
Collapse
|
29
|
Solano A, Lerner G, Griffa G, Deleglise A, Caffaro P, Riquelme L, Perez-Chada D, Della-Maggiore V. Sleep Consolidation Potentiates Sensorimotor Adaptation. J Neurosci 2024; 44:e0325242024. [PMID: 39074983 PMCID: PMC11376339 DOI: 10.1523/jneurosci.0325-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/23/2024] [Accepted: 06/12/2024] [Indexed: 07/31/2024] Open
Abstract
Contrary to its well-established role in declarative learning, the impact of sleep on motor memory consolidation remains a subject of debate. Current literature suggests that while motor skill learning benefits from sleep, consolidation of sensorimotor adaptation (SMA) depends solely on the passage of time. This has led to the proposal that SMA may be an exception to other types of memories. Here, we addressed this ongoing controversy in humans through three comprehensive experiments using the visuomotor adaptation paradigm (N = 290, 150 females). In Experiment 1, we investigated the impact of sleep on memory retention when the temporal gap between training and sleep was not controlled. In line with the previous literature, we found that memory consolidates with the passage of time. In Experiment 2, we used an anterograde interference protocol to determine the time window during which SMA memory is most fragile and, thus, potentially most sensitive to sleep intervention. Our results show that memory is most vulnerable during the initial hour post-training. Building on this insight, in Experiment 3, we investigated the impact of sleep when it coincided with the critical first hour of memory consolidation. This manipulation unveiled a benefit of sleep (30% memory enhancement) alongside an increase in spindle density and spindle-SO coupling during NREM sleep, two well-established neural markers of sleep consolidation. Our findings reconcile seemingly conflicting perspectives on the active role of sleep in motor learning and point to common mechanisms at the basis of memory formation.
Collapse
Affiliation(s)
- Agustin Solano
- Universidad de Buenos Aires-CONICET. Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Facultad de Medicina, Departamento de Ciencias Fisiológicas, Ciudad de Buenos Aires C1121ABG, Argentina
| | - Gonzalo Lerner
- Universidad de Buenos Aires-CONICET. Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Facultad de Medicina, Departamento de Ciencias Fisiológicas, Ciudad de Buenos Aires C1121ABG, Argentina
| | - Guillermina Griffa
- Universidad de Buenos Aires-CONICET. Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Facultad de Medicina, Departamento de Ciencias Fisiológicas, Ciudad de Buenos Aires C1121ABG, Argentina
| | - Alvaro Deleglise
- Universidad de Buenos Aires-CONICET. Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Facultad de Medicina, Departamento de Ciencias Fisiológicas, Ciudad de Buenos Aires C1121ABG, Argentina
| | - Pedro Caffaro
- Universidad de Buenos Aires-CONICET. Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Facultad de Medicina, Departamento de Ciencias Fisiológicas, Ciudad de Buenos Aires C1121ABG, Argentina
| | - Luis Riquelme
- Universidad de Buenos Aires-CONICET. Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Facultad de Medicina, Departamento de Ciencias Fisiológicas, Ciudad de Buenos Aires C1121ABG, Argentina
| | - Daniel Perez-Chada
- Departamento de Medicina Interna, Servicio de Medicina Pulmonar y Sueño, Hospital Universitario Austral, Pilar, Buenos Aires B1629AHJ, Argentina
| | - Valeria Della-Maggiore
- Universidad de Buenos Aires-CONICET. Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Facultad de Medicina, Departamento de Ciencias Fisiológicas, Ciudad de Buenos Aires C1121ABG, Argentina
- Department of Neurology and Neurosurgery, McGill University Montreal, Quebec H3A2B4, Canada
- Escuela de Ciencia y Tecnología (ECyT), Universidad Nacional de San Martin, San Martin, Buenos Aires, CP 1650, Argentina
| |
Collapse
|
30
|
Schlecht NJ, Lanier ER, Andersen TB, Brose J, Holmes D, Hamberger BR. CYP76BK1 orthologs catalyze furan and lactone ring formation in clerodane diterpenoids across the mint family. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.28.609960. [PMID: 39257772 PMCID: PMC11383695 DOI: 10.1101/2024.08.28.609960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The Lamiaceae (mint family) is the largest known source of furanoclerodanes, a subset of clerodane diterpenoids with broad bioactivities including insect antifeedant properties. The Ajugoideae subfamily, in particular, accumulates significant numbers of structurally related furanoclerodanes. The biosynthetic capacity for formation of these diterpenoids is retained across most Lamiaceae subfamilies, including the early-diverging Callicarpoideae which forms a sister clade to the rest of Lamiaceae. VacCYP76BK1, a cytochrome P450 monooxygenase from Vitex agnus-castus, was previously found to catalyze the formation of the proposed precursor to furan and lactone-containing labdane diterpenoids. Through transcriptome-guided pathway exploration, we identified orthologs of VacCYP76BK1 in Ajuga reptans and Callicarpa americana. Functional characterization demonstrated that both could catalyze the oxidative cyclization of clerodane backbones to yield a furan ring. Subsequent investigation revealed a total of ten CYP76BK1 orthologs across six Lamiaceae subfamilies. Through analysis of available chromosome-scale genomes, we identified four CYP76BK1 members as syntelogs within a conserved syntenic block across divergent subfamilies. This suggests an evolutionary lineage that predates the speciation of the Lamiaceae. Functional characterization of the CYP76BK1 orthologs affirmed conservation of function, as all catalyzed furan ring formation. Additionally, some orthologs yielded two novel lactone ring moieties. The presence of the CYP76BK1 orthologs across Lamiaceae subfamilies closely overlaps with the distribution of reported furanoclerodanes. Together, the activities and distribution of the CYP76BK1 orthologs identified here support their central role in furanoclerodane biosynthesis within the Lamiaceae family. Our findings lay the groundwork for biotechnological applications to harness the economic potential of this promising class of compounds.
Collapse
Affiliation(s)
- Nicholas J. Schlecht
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- DOE Great Lakes Bioenergy Research Center, Michigan State University, East Lansing, MI, USA
| | - Emily R. Lanier
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Trine B. Andersen
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- DOE Great Lakes Bioenergy Research Center, Michigan State University, East Lansing, MI, USA
| | - Julia Brose
- Department of Plant Biology, Michigan State University, East Lansing, MI, USA
| | - Daniel Holmes
- Department of Chemistry, Michigan State University, East Lansing MI, USA
| | - Björn R. Hamberger
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- DOE Great Lakes Bioenergy Research Center, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
31
|
LeBlang CJ, Pazyra-Murphy MF, Silagi ES, Dasgupta S, Tsolias M, Miller T, Petrova V, Zhen S, Jovanovic V, Castellano D, Gerrish K, Ormanoglu P, Tristan C, Singeç I, Woolf CJ, Tasdemir-Yilmaz O, Segal RA. Satellite glial contact enhances differentiation and maturation of human iPSC-derived sensory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604966. [PMID: 39211268 PMCID: PMC11361066 DOI: 10.1101/2024.07.24.604966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Sensory neurons generated from induced pluripotent stem cells (iSNs) are used to model human peripheral neuropathies, however current differentiation protocols produce sensory neurons with an embryonic phenotype. Peripheral glial cells contact sensory neurons early in development and contribute to formation of the canonical pseudounipolar morphology, but these signals are not encompassed in current iSN differentiation protocols. Here, we show that terminal differentiation of iSNs in co-culture with rodent Dorsal Root Ganglion satellite glia (rSG) advances their differentiation and maturation. Co-cultured iSNs develop a pseudounipolar morphology through contact with rSGs. This transition depends on semaphorin-plexin guidance cues and on glial gap junction signaling. In addition to morphological changes, iSNs terminally differentiated in co-culture exhibit enhanced spontaneous action potential firing, more mature gene expression, and increased susceptibility to paclitaxel induced axonal degeneration. Thus, iSNs differentiated in coculture with rSGs provide a better model for investigating human peripheral neuropathies.
Collapse
|
32
|
Nigro M, Tortorelli LS, Yang H. Distinct roles of prefrontal cortex neurons in set shifting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.608808. [PMID: 39229035 PMCID: PMC11370324 DOI: 10.1101/2024.08.20.608808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Cognitive flexibility, the ability to adjust behavioral strategies in response to changing environmental contingencies, requires adaptive processing of internal states and contextual cues to guide goal-oriented behavior, and is dependent on prefrontal cortex (PFC) functions. However, the neurophysiological underpinning of how the PFC supports cognitive flexibility is not well understood and has been under active investigation. We recorded spiking activity from single PFC neurons in mice performing the attentional set-shifting task, where mice learned to associate different contextually relevant sensory stimuli to reward. We identified subgroups of PFC neurons encoding task context, choice and trial outcome. Putative fast-spiking neurons were more involved in representing outcome and choice than putative regular-spiking neurons. Regression model further revealed that task context and trial outcome modulated the activity of choice-encoding neurons in rule-dependent and cell type-dependent manners. Together, our data provide new evidence to elucidate PFC's role in cognitive flexibility, suggesting differential cell type-specific engagement during set shifting, and that both contextual rule representation and trial outcome monitoring underlie PFC's unique capacity to support flexible behavioral switching.
Collapse
Affiliation(s)
- Marco Nigro
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA 92521, USA
| | - Lucas Silva Tortorelli
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA 92521, USA
| | - Hongdian Yang
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA 92521, USA
- Neuroscience Graduate Program, University of California, Riverside, CA 92521, USA
| |
Collapse
|
33
|
Ramírez Carbó CA, Faromiki OG, Nan B. A lytic transglycosylase connects bacterial focal adhesion complexes to the peptidoglycan cell wall. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588103. [PMID: 38617213 PMCID: PMC11014575 DOI: 10.1101/2024.04.04.588103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The Gram-negative bacterium Myxococcus xanthus glides on solid surfaces. Dynamic bacterial focal adhesion complexes (bFACs) convert proton motive force from the inner membrane into mechanical propulsion on the cell surface. It is unclear how the mechanical force transmits across the rigid peptidoglycan (PG) cell wall. Here we show that AgmT, a highly abundant lytic PG transglycosylase homologous to Escherichia coli MltG, couples bFACs to PG. Coprecipitation assay and single-particle microscopy reveal that the gliding motors fail to connect to PG and thus are unable to assemble into bFACs in the absence of an active AgmT. Heterologous expression of E. coli MltG restores the connection between PG and bFACs and thus rescues gliding motility in the M. xanthus cells that lack AgmT. Our results indicate that bFACs anchor to AgmT-modified PG to transmit mechanical force across the PG cell wall.
Collapse
Affiliation(s)
- Carlos A. Ramírez Carbó
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
- The Genetics and Genomics Interdisciplinary Program, Texas A&M University, College Station, TX 77843, USA
- C. A. R. C. and O. G. F. contribute equally to this work
| | - Olalekan G. Faromiki
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
- C. A. R. C. and O. G. F. contribute equally to this work
| | - Beiyan Nan
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
34
|
Gyger J, Torrens G, Cava F, Bernhardt TG, Fumeaux C. A potential space-making role in cell wall biogenesis for SltB1and DacB revealed by a beta-lactamase induction phenotype in Pseudomonas aeruginosa. mBio 2024; 15:e0141924. [PMID: 38920394 PMCID: PMC11253642 DOI: 10.1128/mbio.01419-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 06/27/2024] Open
Abstract
Pseudomonas aeruginosa encodes the beta-lactamase AmpC, which promotes resistance to beta-lactam antibiotics. Expression of ampC is induced by anhydro-muropeptides (AMPs) released from the peptidoglycan (PG) cell wall upon beta-lactam treatment. AmpC can also be induced via genetic inactivation of PG biogenesis factors such as the endopeptidase DacB that cleaves PG crosslinks. Mutants in dacB occur in beta-lactam-resistant clinical isolates of P. aeruginosa, but it has remained unclear why DacB inactivation promotes ampC induction. Similarly, the inactivation of lytic transglycosylase (LT) enzymes such as SltB1 that cut PG glycans has also been associated with ampC induction and beta-lactam resistance. Given that LT enzymes are capable of producing AMP products that serve as ampC inducers, this latter observation has been especially difficult to explain. Here, we show that ampC induction in sltB1 or dacB mutants requires another LT enzyme called MltG. In Escherichia coli, MltG has been implicated in the degradation of nascent PG strands produced upon beta-lactam treatment. Accordingly, in P. aeruginosa sltB1 and dacB mutants, we detected the MltG-dependent production of pentapeptide-containing AMP products that are signatures of nascent PG degradation. Our results therefore support a model in which SltB1 and DacB use their PG-cleaving activity to open space in the PG matrix for the insertion of new material. Thus, their inactivation mimics low-level beta-lactam treatment by reducing the efficiency of new PG insertion into the wall, causing the degradation of some nascent PG material by MltG to produce the ampC-inducing signal. IMPORTANCE Inducible beta-lactamases like the ampC system of Pseudomonas aeruginosa are a common determinant of beta-lactam resistance among gram-negative bacteria. The regulation of ampC is elegantly tuned to detect defects in cell wall synthesis caused by beta-lactam drugs. Studies of mutations causing ampC induction in the absence of drug therefore promise to reveal new insights into the process of cell wall biogenesis in addition to aiding our understanding of how resistance to beta-lactam antibiotics arises in the clinic. In this study, the ampC induction phenotype for mutants lacking a glycan-cleaving enzyme or an enzyme that cuts cell wall crosslinks was used to uncover a potential role for these enzymes in making space in the wall matrix for the insertion of new material during cell growth.
Collapse
Affiliation(s)
- Joël Gyger
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Gabriel Torrens
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Center for Microbial Research (UCMR), Umea, Sweden
- Department of Molecular Biology, Science for Life Laboratory (SciLifeLab), Umeå University, Umeå, Sweden
| | - Felipe Cava
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Center for Microbial Research (UCMR), Umea, Sweden
- Department of Molecular Biology, Science for Life Laboratory (SciLifeLab), Umeå University, Umeå, Sweden
| | - Thomas G. Bernhardt
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Coralie Fumeaux
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Son JE, Park SH, Choi U, Lee CR. Lytic transglycosylase repertoire diversity enables intrinsic antibiotic resistance and daughter cell separation in Escherichia coli under acidic stress. Antimicrob Agents Chemother 2024; 68:e0037224. [PMID: 38884456 PMCID: PMC11232391 DOI: 10.1128/aac.00372-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024] Open
Abstract
Peptidoglycan (PG) is an important architectural element that imparts physical toughness and rigidity to the bacterial envelope. It is also a dynamic structure that undergoes continuous turnover or autolysis. Escherichia coli possesses redundant PG degradation enzymes responsible for PG turnover; however, the advantage afforded by the existence of numerous PG degradation enzymes remains incompletely understood. In this study, we elucidated the physiological roles of MltE and MltC, members of the lytic transglycosylase (LTG) family that catalyze the cleavage of glycosidic bonds between disaccharide subunits within PG strands. MltE and MltC are acidic LTGs that exhibit increased enzymatic activity and protein levels under acidic pH conditions, respectively, and deletion of these two LTGs results in a pronounced growth defect at acidic pH. Furthermore, inactivation of these two LTGs induces increased susceptibility at acidic pH against various antibiotics, particularly vancomycin, which seems to be partially caused by elevated membrane permeability. Intriguingly, inactivation of these LTGs induces a chaining morphology, indicative of daughter cell separation defects, only under acidic pH conditions. Simultaneous deletion of PG amidases, known contributors to daughter cell separation, exacerbates the chaining phenotype at acidic pH. This suggests that the two LTGs may participate in the cleavage of glycan strands between daughter cells under acidic pH conditions. Collectively, our findings highlight the role of LTG repertoire diversity in facilitating bacterial survival and antibiotic resistance under stressful conditions.
Collapse
Affiliation(s)
- Ji Eun Son
- Department of Biological Sciences, Myongji University, Yongin, Gyeonggido, Republic of Korea
| | - Si Hyoung Park
- Department of Biological Sciences, Myongji University, Yongin, Gyeonggido, Republic of Korea
| | - Umji Choi
- Department of Biological Sciences, Myongji University, Yongin, Gyeonggido, Republic of Korea
| | - Chang-Ro Lee
- Department of Biological Sciences, Myongji University, Yongin, Gyeonggido, Republic of Korea
| |
Collapse
|
36
|
Avila‐Cobian LF, De Benedetti S, Hoshino H, Nguyen VT, El‐Araby AM, Sader S, Hu DD, Cole SL, Kim C, Fisher JF, Champion MM, Mobashery S. Lytic transglycosylase Slt of Pseudomonas aeruginosa as a periplasmic hub protein. Protein Sci 2024; 33:e5038. [PMID: 38864725 PMCID: PMC11168074 DOI: 10.1002/pro.5038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 05/10/2024] [Accepted: 05/12/2024] [Indexed: 06/13/2024]
Abstract
Peptidoglycan is a major constituent of the bacterial cell wall. Its integrity as a polymeric edifice is critical for bacterial survival and, as such, it is a preeminent target for antibiotics. The peptidoglycan is a dynamic crosslinked polymer that undergoes constant biosynthesis and turnover. The soluble lytic transglycosylase (Slt) of Pseudomonas aeruginosa is a periplasmic enzyme involved in this dynamic turnover. Using amber-codon-suppression methodology in live bacteria, we incorporated a fluorescent chromophore into the structure of Slt. Fluorescent microscopy shows that Slt populates the length of the periplasmic space and concentrates at the sites of septation in daughter cells. This concentration persists after separation of the cells. Amber-codon-suppression methodology was also used to incorporate a photoaffinity amino acid for the capture of partner proteins. Mass-spectrometry-based proteomics identified 12 partners for Slt in vivo. These proteomics experiments were complemented with in vitro pulldown analyses. Twenty additional partners were identified. We cloned the genes and purified to homogeneity 22 identified partners. Biophysical characterization confirmed all as bona fide Slt binders. The identities of the protein partners of Slt span disparate periplasmic protein families, inclusive of several proteins known to be present in the divisome. Notable periplasmic partners (KD < 0.5 μM) include PBPs (PBP1a, KD = 0.07 μM; PBP5 = 0.4 μM); other lytic transglycosylases (SltB2, KD = 0.09 μM; RlpA, KD = 0.4 μM); a type VI secretion system effector (Tse5, KD = 0.3 μM); and a regulatory protease for alginate biosynthesis (AlgO, KD < 0.4 μM). In light of the functional breadth of its interactome, Slt is conceptualized as a hub protein within the periplasm.
Collapse
Affiliation(s)
- Luis F. Avila‐Cobian
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| | - Stefania De Benedetti
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| | - Hidekazu Hoshino
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| | - Van T. Nguyen
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| | - Amr M. El‐Araby
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| | - Safaa Sader
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| | - Daniel D. Hu
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| | - Sara L. Cole
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| | - Choon Kim
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| | - Jed F. Fisher
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| | - Matthew M. Champion
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| | - Shahriar Mobashery
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| |
Collapse
|
37
|
Meylakh N, Crawford LS, Mills EP, Macefield VG, Vickers ER, Macey PM, Keay KA, Henderson LA. Altered Corticobrainstem Connectivity during Spontaneous Fluctuations in Pain Intensity in Painful Trigeminal Neuropathy. eNeuro 2024; 11:ENEURO.0522-23.2024. [PMID: 38997145 PMCID: PMC11277291 DOI: 10.1523/eneuro.0522-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 06/02/2024] [Accepted: 06/06/2024] [Indexed: 07/14/2024] Open
Abstract
Chronic neuropathic pain can result from nervous system injury and can persist in the absence of external stimuli. Although ongoing pain characterizes the disorder, in many individuals, the intensity of this ongoing pain fluctuates dramatically. Previously, it was identified that functional magnetic resonance imaging signal covariations between the midbrain periaqueductal gray (PAG) matter, rostral ventromedial medulla (RVM), and spinal trigeminal nucleus are associated with moment-to-moment fluctuations in pain intensity in individuals with painful trigeminal neuropathy (PTN). Since this brainstem circuit is modulated by higher brain input, we sought to determine which cortical sites might be influencing this brainstem network during spontaneous fluctuations in pain intensity. Over 12 min, we recorded the ongoing pain intensity in 24 PTN participants and classified them as fluctuating (n = 13) or stable (n = 11). Using a PAG seed, we identified connections between the PAG and emotional-affective sites such as the hippocampal and posterior cingulate cortices, the sensory-discriminative posterior insula, and cognitive-affective sites such as the dorsolateral prefrontal (dlPFC) and subgenual anterior cingulate cortices that were altered dependent on spontaneous high and low pain intensity. Additionally, sliding-window functional connectivity analysis revealed that the dlPFC-PAG connection anticorrelated with perceived pain intensity over the entire 12 min period. These findings reveal cortical systems underlying moment-to-moment changes in perceived pain in PTN, which likely cause dysregulation in the brainstem circuits previously identified, and consequently alter the appraisal of pain across time.
Collapse
Affiliation(s)
- Noemi Meylakh
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, Sydney, New South Wales 2050, Australia
| | - Lewis S Crawford
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, Sydney, New South Wales 2050, Australia
| | - Emily P Mills
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, Sydney, New South Wales 2050, Australia
| | - Vaughan G Macefield
- Department of Neuroscience, Monash University, Melbourne, Victoria 3800, Australia
| | - E Russell Vickers
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, Sydney, New South Wales 2050, Australia
| | - Paul M Macey
- UCLA School of Nursing and Brain Research Institute, University of California, Los Angeles, California 90095
| | - Kevin A Keay
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, Sydney, New South Wales 2050, Australia
| | - Luke A Henderson
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, Sydney, New South Wales 2050, Australia
| |
Collapse
|
38
|
Sladky VC, Strong MA, Tapias-Gomez D, Jewett CE, Drown CG, Scott PM, Holland AJ. The AID2 system offers a potent tool for rapid, reversible, or sustained degradation of essential proteins in live mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597287. [PMID: 38895390 PMCID: PMC11185741 DOI: 10.1101/2024.06.04.597287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Studying essential genes required for dynamic processes in live mice is challenging as genetic perturbations are irreversible and limited by slow protein depletion kinetics. The first-generation auxin-inducible-degron (AID) system is a powerful tool for analyzing inducible protein loss in cultured cells. However, auxin administration is toxic to mice, preventing its long-term use in animals. Here, we use an optimized second-generation AID system to achieve the conditional and reversible loss of the essential centrosomal protein CEP192 in live mice. We show that the auxin derivative 5-Ph-IAA is well tolerated over two weeks and drives near-complete CEP192-mAID degradation in less than one hour in vivo. Prolonged CEP192 loss led to cell division failure and cell death in proliferative tissues. Thus, the second-generation AID system is well suited for rapid and/or sustained protein depletion in live mice, offering a valuable new tool for interrogating protein function in vivo.
Collapse
Affiliation(s)
- Valentina C Sladky
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, 21205, MD, Baltimore, USA
| | - Margaret A Strong
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, 21205, MD, Baltimore, USA
| | - Daniel Tapias-Gomez
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, 21205, MD, Baltimore, USA
| | - Cayla E Jewett
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, 21205, MD, Baltimore, USA
| | - Chelsea G Drown
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, 21205, MD, Baltimore, USA
| | - Phillip M Scott
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, 21205, MD, Baltimore, USA
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, 21205, MD, Baltimore, USA
| |
Collapse
|
39
|
Fonda BD, Kato M, Li Y, Murray DT. Cryo-EM and Solid State NMR Together Provide a More Comprehensive Structural Investigation of Protein Fibrils. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596698. [PMID: 38853912 PMCID: PMC11160737 DOI: 10.1101/2024.05.30.596698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The Tropomyosin 1 isoform I/C C-terminal domain (Tm1-LC) fibril structure is studied jointly with cryogenic electron microscopy (cryo-EM) and solid state nuclear magnetic resonance (NMR). This study demonstrates the complementary nature of these two structural biology techniques. Chemical shift assignments from solid state NMR are used to determine the secondary structure at the level of individual amino acids, which is faithfully seen in cryo-EM reconstructions. Additionally, solid state NMR demonstrates that the region not observed in the reconstructed cryo-EM density is primarily in a highly mobile random coil conformation rather than adopting multiple rigid conformations. Overall, this study illustrates the benefit of investigations combining cryo-EM and solid state NMR to investigate protein fibril structure.
Collapse
Affiliation(s)
- Blake D. Fonda
- Department of Chemistry, University of California, Davis, California, 95616, United States of America
| | - Masato Kato
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
| | - Yang Li
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
| | - Dylan T. Murray
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, 06269, United States of America
| |
Collapse
|
40
|
Leuthner TC, Zhang S, Kohrn BF, Stapleton HM, Baugh LR. Structure-specific variation in per- and polyfluoroalkyl substances toxicity among genetically diverse Caenorhabditis elegans strains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596269. [PMID: 38854041 PMCID: PMC11160736 DOI: 10.1101/2024.05.29.596269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background There are >14,500 structurally diverse per- and polyfluoroalkyl substances (PFAS). Despite knowledge that these "forever chemicals" are in 99% of humans, mechanisms of toxicity and adverse health effects are incompletely known. Furthermore, the contribution of genetic variation to PFAS susceptibility and health consequences is unknown. Objectives We determined the toxicity of a structurally distinct set of PFAS in twelve genetically diverse strains of the genetic model system Caenorhabditis elegans. Methods Dose-response curves for four perfluoroalkyl carboxylic acids (PFNA, PFOA, PFPeA, and PFBA), two perfluoroalkyl sulfonic acids (PFOS and PFBS), two perfluoroalkyl sulfonamides (PFOSA and PFBSA), two fluoroether carboxylic acids (GenX and PFMOAA), one fluoroether sulfonic acid (PFEESA), and two fluorotelomers (6:2 FCA and 6:2 FTS) were determined in the C. elegans laboratory reference strain, N2, and eleven genetically diverse wild strains. Body length was quantified by image analysis at each dose after 48 hr of developmental exposure of L1 arrest-synchronized larvae to estimate effective concentration values (EC50). Results There was a significant range in toxicity among PFAS: PFOSA > PFBSA ≈ PFOS ≈ PFNA > PFOA > GenX ≈ PFEESA > PFBS ≈ PFPeA ≈ PFBA. Long-chain PFAS had greater toxicity than short-chain, and fluorosulfonamides were more toxic than carboxylic and sulfonic acids. Genetic variation explained variation in susceptibility to PFBSA, PFOS, PFBA, PFOA, GenX, PFEESA, PFPeA, and PFBA. There was significant variation in toxicity among C. elegans strains due to chain length, functional group, and between legacy and emerging PFAS. Conclusion C. elegans respond to legacy and emerging PFAS of diverse structures, and this depends on specific structures and genetic variation. Harnessing the natural genetic diversity of C. elegans and the structural complexity of PFAS is a powerful New Approach Methodology (NAM) to investigate structure-activity relationships and mechanisms of toxicity which may inform regulation of other PFAS to improve human and environmental health.
Collapse
Affiliation(s)
- Tess C. Leuthner
- Department of Biology, Duke University, Durham, North Carolina, USA
| | - Sharon Zhang
- Nicholas School of the Environment, Duke University, Durham, North Carolina, USA
| | - Brendan F Kohrn
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Heather M. Stapleton
- Nicholas School of the Environment, Duke University, Durham, North Carolina, USA
| | - L. Ryan Baugh
- Department of Biology, Duke University, Durham, North Carolina, USA
- Center for Genomic and Computational Biology, Duke University, North Carolina, USA
| |
Collapse
|
41
|
Clayton KK, McGill M, Awwad B, Stecyk KS, Kremer C, Skerleva D, Narayanan DP, Zhu J, Hancock KE, Kujawa SG, Kozin ED, Polley DB. Cortical determinants of loudness perception and auditory hypersensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596691. [PMID: 38853938 PMCID: PMC11160727 DOI: 10.1101/2024.05.30.596691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Parvalbumin-expressing inhibitory neurons (PVNs) stabilize cortical network activity, generate gamma rhythms, and regulate experience-dependent plasticity. Here, we observed that activation or inactivation of PVNs functioned like a volume knob in the mouse auditory cortex (ACtx), turning neural and behavioral classification of sound level up or down over a 20dB range. PVN loudness adjustments were "sticky", such that a single bout of 40Hz PVN stimulation sustainably suppressed ACtx sound responsiveness, potentiated feedforward inhibition, and behaviorally desensitized mice to loudness. Sensory sensitivity is a cardinal feature of autism, aging, and peripheral neuropathy, prompting us to ask whether PVN stimulation can persistently desensitize mice with ACtx hyperactivity, PVN hypofunction, and loudness hypersensitivity triggered by cochlear sensorineural damage. We found that a single 16-minute bout of 40Hz PVN stimulation session restored normal loudness perception for one week, showing that perceptual deficits triggered by irreversible peripheral injuries can be reversed through targeted cortical circuit interventions.
Collapse
Affiliation(s)
- Kameron K Clayton
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston MA 02114
| | - Matthew McGill
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston MA 02114
| | - Bshara Awwad
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston MA 02114
| | - Kamryn S Stecyk
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston MA 02114
| | - Caroline Kremer
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston MA 02114
| | | | - Divya P Narayanan
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston MA 02114
| | - Jennifer Zhu
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston MA 02114
| | - Kenneth E Hancock
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston MA 02114
| | - Sharon G Kujawa
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston MA 02114
| | - Elliott D Kozin
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston MA 02114
| | - Daniel B Polley
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston MA 02114
| |
Collapse
|
42
|
Hagan CE, Snyder AG, Headley M, Oberst A. Apoptotic cells promote circulating tumor cell survival and metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595217. [PMID: 38826267 PMCID: PMC11142129 DOI: 10.1101/2024.05.21.595217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
During tumor progression and especially following cytotoxic therapy, cell death of both tumor and stromal cells is widespread. Despite clinical observations that high levels of apoptotic cells correlate with poorer patient outcomes, the physiological effects of dying cells on tumor progression remain incompletely understood. Here, we report that circulating apoptotic cells robustly enhance tumor cell metastasis to the lungs. Using intravenous metastasis models, we observed that the presence of apoptotic cells, but not cells dying by other mechanisms, supports circulating tumor cell (CTC) survival following arrest in the lung vasculature. Apoptotic cells promote CTC survival by recruiting platelets to the forming metastatic niche. Apoptotic cells externalize the phospholipid phosphatidylserine to the outer leaflet of the plasma membrane, which we found increased the activity of the coagulation initiator Tissue Factor, thereby triggering the formation of platelet clots that protect proximal CTCs. Inhibiting the ability of apoptotic cells to induce coagulation by knocking out Tissue Factor, blocking phosphatidylserine, or administering the anticoagulant heparin abrogated the pro-metastatic effect of apoptotic cells. This work demonstrates a previously unappreciated role for apoptotic cells in facilitating metastasis by establishing CTC-supportive emboli, and suggests points of intervention that may reduce the pro-metastatic effect of apoptotic cells. GRAPHICAL ABSTRACT
Collapse
|
43
|
Rossi Sebastiano A, Poles K, Gualtiero S, Romeo M, Galigani M, Bruno V, Fossataro C, Garbarini F. Balancing the Senses: Electrophysiological Responses Reveal the Interplay between Somatosensory and Visual Processing During Body-Related Multisensory Conflict. J Neurosci 2024; 44:e1397232024. [PMID: 38508711 PMCID: PMC11079966 DOI: 10.1523/jneurosci.1397-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/27/2023] [Accepted: 01/03/2024] [Indexed: 03/22/2024] Open
Abstract
In the study of bodily awareness, the predictive coding theory has revealed that our brain continuously modulates sensory experiences to integrate them into a unitary body representation. Indeed, during multisensory illusions (e.g., the rubber hand illusion, RHI), the synchronous stroking of the participant's concealed hand and a fake visible one creates a visuotactile conflict, generating a prediction error. Within the predictive coding framework, through sensory processing modulation, prediction errors are solved, inducing participants to feel as if touches originated from the fake hand, thus ascribing the fake hand to their own body. Here, we aimed to address sensory processing modulation under multisensory conflict, by disentangling somatosensory and visual stimuli processing that are intrinsically associated during the illusion induction. To this aim, we designed two EEG experiments, in which somatosensory- (SEPs; Experiment 1; N = 18; F = 10) and visual-evoked potentials (VEPs; Experiment 2; N = 18; F = 9) were recorded in human males and females following the RHI. Our results show that, in both experiments, ERP amplitude is significantly modulated in the illusion as compared with both control and baseline conditions, with a modality-dependent diametrical pattern showing decreased SEP amplitude and increased VEP amplitude. Importantly, both somatosensory and visual modulations occur in long-latency time windows previously associated with tactile and visual awareness, thus explaining the illusion of perceiving touch at the sight location. In conclusion, we describe a diametrical modulation of somatosensory and visual processing as the neural mechanism that allows maintaining a stable body representation, by restoring visuotactile congruency under the occurrence of multisensory conflicts.
Collapse
Affiliation(s)
| | - Karol Poles
- MANIBUS Lab, Psychology Department, University of Turin, Turin 10124, Italy
| | - Stefano Gualtiero
- MANIBUS Lab, Psychology Department, University of Turin, Turin 10124, Italy
| | - Marcella Romeo
- MANIBUS Lab, Psychology Department, University of Turin, Turin 10124, Italy
- IMT School for Advanced Studies Lucca, Lucca 55100, Italy
| | - Mattia Galigani
- MANIBUS Lab, Psychology Department, University of Turin, Turin 10124, Italy
| | - Valentina Bruno
- MANIBUS Lab, Psychology Department, University of Turin, Turin 10124, Italy
| | - Carlotta Fossataro
- MANIBUS Lab, Psychology Department, University of Turin, Turin 10124, Italy
| | - Francesca Garbarini
- MANIBUS Lab, Psychology Department, University of Turin, Turin 10124, Italy
- Neuroscience Institute of Turin (NIT), Turin 10124, Italy
| |
Collapse
|
44
|
Gizzio J, Thakur A, Haldane A, Post CB, Levy RM. Evolutionary sequence and structural basis for the distinct conformational landscapes of Tyr and Ser/Thr kinases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584161. [PMID: 38559238 PMCID: PMC10979876 DOI: 10.1101/2024.03.08.584161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Protein kinases are molecular machines with rich sequence variation that distinguishes the two main evolutionary branches - tyrosine kinases (TKs) from serine/threonine kinases (STKs). Using a sequence co-variation Potts statistical energy model we previously concluded that TK catalytic domains are more likely than STKs to adopt an inactive conformation with the activation loop in an autoinhibitory "folded" conformation, due to intrinsic sequence effects. Here we investigated the structural basis for this phenomenon by integrating the sequence-based model with structure-based molecular dynamics (MD) to determine the effects of mutations on the free energy difference between active and inactive conformations, using a novel thermodynamic cycle involving many (n=108) protein-mutation free energy perturbation (FEP) simulations in the active and inactive conformations. The sequence and structure-based results are consistent and support the hypothesis that the inactive conformation "DFG-out Activation Loop Folded", is a functional regulatory state that has been stabilized in TKs relative to STKs over the course of their evolution via the accumulation of residue substitutions in the activation loop and catalytic loop that facilitate distinct substrate binding modes in trans and additional modes of regulation in cis for TKs.
Collapse
Affiliation(s)
- Joan Gizzio
- Center for Biophysics and Computational Biology, Temple University, Philadelphia, Pennsylvania 19122
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122
| | - Abhishek Thakur
- Center for Biophysics and Computational Biology, Temple University, Philadelphia, Pennsylvania 19122
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122
| | - Allan Haldane
- Center for Biophysics and Computational Biology, Temple University, Philadelphia, Pennsylvania 19122
- Department of Physics, Temple University, Philadelphia, Pennsylvania 19122
| | - Carol Beth Post
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907
| | - Ronald M. Levy
- Center for Biophysics and Computational Biology, Temple University, Philadelphia, Pennsylvania 19122
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122
| |
Collapse
|
45
|
Hogan AM, Motnenko A, Rahman ASMZ, Cardona ST. Cell envelope structural and functional contributions to antibiotic resistance in Burkholderia cenocepacia. J Bacteriol 2024; 206:e0044123. [PMID: 38501654 PMCID: PMC11025338 DOI: 10.1128/jb.00441-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/05/2024] [Indexed: 03/20/2024] Open
Abstract
Antibiotic activity is limited by the physical construction of the Gram-negative cell envelope. Species of the Burkholderia cepacia complex (Bcc) are known as intrinsically multidrug-resistant opportunistic pathogens with low permeability cell envelopes. Here, we re-examined a previously performed chemical-genetic screen of barcoded transposon mutants in B. cenocepacia K56-2, focusing on cell envelope structural and functional processes. We identified structures mechanistically important for resistance to singular and multiple antibiotic classes. For example, susceptibility to novobiocin, avibactam, and the LpxC inhibitor, PF-04753299, was linked to the BpeAB-OprB efflux pump, suggesting these drugs are substrates for this pump in B. cenocepacia. Defects in peptidoglycan precursor synthesis specifically increased susceptibility to cycloserine and revealed a new putative amino acid racemase, while defects in divisome accessory proteins increased susceptibility to multiple β-lactams. Additionally, disruption of the periplasmic disulfide bond formation system caused pleiotropic defects on outer membrane integrity and β-lactamase activity. Our findings highlight the layering of resistance mechanisms in the structure and function of the cell envelope. Consequently, we point out processes that can be targeted for developing antibiotic potentiators.IMPORTANCEThe Gram-negative cell envelope is a double-layered physical barrier that protects cells from extracellular stressors, such as antibiotics. The Burkholderia cell envelope is known to contain additional modifications that reduce permeability. We investigated Burkholderia cell envelope factors contributing to antibiotic resistance from a genome-wide view by re-examining data from a transposon mutant library exposed to an antibiotic panel. We identified susceptible phenotypes for defects in structures and functions in the outer membrane, periplasm, and cytoplasm. Overall, we show that resistance linked to the cell envelope is multifaceted and provides new targets for the development of antibiotic potentiators.
Collapse
Affiliation(s)
- Andrew M. Hogan
- Department of Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Anna Motnenko
- Department of Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Silvia T. Cardona
- Department of Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
46
|
Reddy NA, Clements RG, Brooks JCW, Bright MG. Simultaneous cortical, subcortical, and brainstem mapping of sensory activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.11.589099. [PMID: 38659741 PMCID: PMC11042175 DOI: 10.1101/2024.04.11.589099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Non-painful tactile sensory stimuli are processed in the cortex, subcortex, and brainstem. Recent functional magnetic resonance imaging (fMRI) studies have highlighted the value of whole-brain, systems-level investigation for examining pain processing. However, whole-brain fMRI studies are uncommon, in part due to challenges with signal to noise when studying the brainstem. Furthermore, the differentiation of small sensory brainstem structures such as the cuneate and gracile nuclei necessitates high resolution imaging. To address this gap in systems-level sensory investigation, we employed a whole-brain, multi-echo fMRI acquisition at 3T with multi-echo independent component analysis (ME-ICA) denoising and brainstem-specific modeling to enable detection of activation across the entire sensory system. In healthy participants, we examined patterns of activity in response to non-painful brushing of the right hand, left hand, and right foot, and found the expected lateralization, with distinct cortical and subcortical responses for upper and lower limb stimulation. At the brainstem level, we were able to differentiate the small, adjacent cuneate and gracile nuclei, corresponding to hand and foot stimulation respectively. Our findings demonstrate that simultaneous cortical, subcortical, and brainstem mapping at 3T could be a key tool to understand the sensory system in both healthy individuals and clinical cohorts with sensory deficits.
Collapse
Affiliation(s)
- Neha A. Reddy
- Department of Physical Therapy and Human Movement Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Department of Biomedical Engineering, McCormick School of Engineering and Applied Sciences, Northwestern University, Evanston, IL, United States
| | - Rebecca G. Clements
- Department of Physical Therapy and Human Movement Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Department of Biomedical Engineering, McCormick School of Engineering and Applied Sciences, Northwestern University, Evanston, IL, United States
| | | | - Molly G. Bright
- Department of Physical Therapy and Human Movement Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Department of Biomedical Engineering, McCormick School of Engineering and Applied Sciences, Northwestern University, Evanston, IL, United States
| |
Collapse
|
47
|
Singer A, Ramos A, Keating AE. Elaboration of the Homer1 Recognition Landscape Reveals Incomplete Divergence of Paralogous EVH1 Domains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.23.576863. [PMID: 38645240 PMCID: PMC11030225 DOI: 10.1101/2024.01.23.576863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Short sequences that mediate interactions with modular binding domains are ubiquitous throughout eukaryotic proteomes. Networks of Short Linear Motifs (SLiMs) and their corresponding binding domains orchestrate many cellular processes, and the low mutational barrier to evolving novel interactions provides a way for biological systems to rapidly sample selectable phenotypes. Mapping SLiM binding specificity and the rules that govern SLiM evolution is fundamental to uncovering the pathways regulated by these networks and developing the tools to manipulate them. We used high-throughput screening of the human proteome to identify sequences that bind to the Enabled/VASP homology 1 (EVH1) domain of the postsynaptic density scaffolding protein Homer1. In doing so, we expanded current understanding of the determinants of Homer EVH1 binding preferences and defined a new motif that can facilitate the discovery of additional Homer-mediated interactions. Interestingly, the Homer1 EVH1 domain preferentially binds to sequences containing an N-terminally overlapping motif that is bound by the paralogous family of Ena/VASP actin polymerases, and many of these sequences can bind to EVH1 domains from both protein families. We provide evidence from orthologous EVH1 domains in pre-metazoan organisms that the overlap in human Ena/VASP and Homer binding preferences corresponds to an incomplete divergence from a common Ena/VASP ancestor. Given this overlap in binding profiles, promiscuous sequences that can be recognized by both families either achieve specificity through extrinsic regulatory strategies or may provide functional benefits via multi-specificity. This may explain why these paralogs incompletely diverged despite the accessibility of further diverged isoforms.
Collapse
Affiliation(s)
- Avinoam Singer
- MIT Department of Biology, Cambridge, Massachusetts, USA
| | | | - Amy E. Keating
- MIT Department of Biology, Cambridge, Massachusetts, USA
- MIT Department of Biological Engineering, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Cambridge, Massachusetts, USA
| |
Collapse
|
48
|
Razew A, Herail Q, Miyachiro M, Anoyatis-Pelé C, Bougault C, Dessen A, Arthur M, Simorre JP. Monitoring Drug-Protein Interactions in the Bacterial Periplasm by Solution Nuclear Magnetic Resonance Spectroscopy. J Am Chem Soc 2024; 146:9252-9260. [PMID: 38500259 DOI: 10.1021/jacs.4c00604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
The rapid spread of antimicrobial resistance across bacterial pathogens poses a serious risk to the efficacy and sustainability of available treatments. This puts pressure on research concerning the development of new drugs. Here, we present an in-cell NMR-based research strategy to monitor the activity of the enzymes located in the periplasmic space delineated by the inner and outer membranes of Gram-negative bacteria. We demonstrate its unprecedented analytical power in monitoring in situ and in real time (i) the hydrolysis of β-lactams by β-lactamases, (ii) the interaction of drugs belonging to the β-lactam family with their essential targets, and (iii) the binding of inhibitors to these enzymes. We show that in-cell NMR provides a powerful analytical tool for investigating new drugs targeting the molecular components of the bacterial periplasm.
Collapse
Affiliation(s)
- Alicja Razew
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, 38044, France
| | - Quentin Herail
- INSERM, Sorbonne Université, Université Paris Cité, Paris, 75006, France
| | - Mayara Miyachiro
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, 38044, France
| | | | - Catherine Bougault
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, 38044, France
| | - Andrea Dessen
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, 38044, France
| | - Michel Arthur
- INSERM, Sorbonne Université, Université Paris Cité, Paris, 75006, France
| | - Jean-Pierre Simorre
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, 38044, France
| |
Collapse
|
49
|
Gharat V, Peter F, de Quervain DJF, Papassotiropoulos A, Stetak A. Role of GLR-1 in Age-Dependent Short-Term Memory Decline. eNeuro 2024; 11:ENEURO.0420-23.2024. [PMID: 38519128 PMCID: PMC11005081 DOI: 10.1523/eneuro.0420-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/10/2024] [Accepted: 03/06/2024] [Indexed: 03/24/2024] Open
Abstract
As the global elderly population grows, age-related cognitive decline is becoming an increasingly significant healthcare issue, often leading to various neuropsychiatric disorders. Among the many molecular players involved in memory, AMPA-type glutamate receptors are known to regulate learning and memory, but how their dynamics change with age and affect memory decline is not well understood. Here, we examined the in vivo properties of the AMPA-type glutamate receptor GLR-1 in the AVA interneuron of the Caenorhabditis elegans nervous system during physiological aging. We found that both total and membrane-bound GLR-1 receptor levels decrease with age in wild-type worms, regardless of their location along the axon. Using fluorescence recovery after photobleaching, we also demonstrated that a reduction in GLR-1 abundance correlates with decreased local, synaptic GLR-1 receptor dynamics. Importantly, we found that reduced GLR-1 levels strongly correlate with the age-related decline in short-term associative memory. Genetic manipulation of GLR-1 stability, by either deleting msi-1 or expressing a ubiquitination-defective GLR-1 (4KR) variant, prevented this age-related reduction in receptor abundance and improved the short-term memory performance in older animals, which reached performance levels similar to those of young animals. Overall, our data indicate that AMPA-type glutamate receptor abundance and dynamics are key factors in maintaining memory function and that changes in these parameters are linked to age-dependent short-term memory decline.
Collapse
Affiliation(s)
- Vaibhav Gharat
- Division of Molecular Neuroscience, Department of Biomedicine, University of Basel, Basel 4055, Switzerland
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel 4055, Switzerland
| | - Fabian Peter
- Division of Molecular Neuroscience, Department of Biomedicine, University of Basel, Basel 4055, Switzerland
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel 4055, Switzerland
| | - Dominique J-F de Quervain
- Division of Molecular Neuroscience, Department of Biomedicine, University of Basel, Basel 4055, Switzerland
- Division of Cognitive Neuroscience, Department of Biomedicine, University of Basel, Basel 4055, Switzerland
- University Psychiatric Clinics, University of Basel, Basel 4002, Switzerland
| | - Andreas Papassotiropoulos
- Division of Molecular Neuroscience, Department of Biomedicine, University of Basel, Basel 4055, Switzerland
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel 4055, Switzerland
- University Psychiatric Clinics, University of Basel, Basel 4002, Switzerland
| | - Attila Stetak
- Division of Molecular Neuroscience, Department of Biomedicine, University of Basel, Basel 4055, Switzerland
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel 4055, Switzerland
- University Psychiatric Clinics, University of Basel, Basel 4002, Switzerland
| |
Collapse
|
50
|
Alvarado Obando M, Rey-Varela D, Cava F, Dörr T. Genetic interaction mapping reveals functional relationships between peptidoglycan endopeptidases and carboxypeptidases. PLoS Genet 2024; 20:e1011234. [PMID: 38598601 PMCID: PMC11034669 DOI: 10.1371/journal.pgen.1011234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 04/22/2024] [Accepted: 03/25/2024] [Indexed: 04/12/2024] Open
Abstract
Peptidoglycan (PG) is the main component of the bacterial cell wall; it maintains cell shape while protecting the cell from internal osmotic pressure and external environmental challenges. PG synthesis is essential for bacterial growth and survival, and a series of PG modifications are required to allow expansion of the sacculus. Endopeptidases (EPs), for example, cleave the crosslinks between adjacent PG strands to allow the incorporation of newly synthesized PG. EPs are collectively essential for bacterial growth and must likely be carefully regulated to prevent sacculus degradation and cell death. However, EP regulation mechanisms are poorly understood. Here, we used TnSeq to uncover novel EP regulators in Vibrio cholerae. This screen revealed that the carboxypeptidase DacA1 (PBP5) alleviates EP toxicity. dacA1 is essential for viability on LB medium, and this essentiality was suppressed by EP overexpression, revealing that EP toxicity both mitigates, and is mitigated by, a defect in dacA1. A subsequent suppressor screen to restore viability of ΔdacA1 in LB medium identified hypomorphic mutants in the PG synthesis pathway, as well as mutations that promote EP activation. Our data thus reveal a more complex role of DacA1 in maintaining PG homeostasis than previously assumed.
Collapse
Affiliation(s)
- Manuela Alvarado Obando
- Department of Microbiology, Cornell University, Ithaca, New York, United States of America
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, United States of America
| | - Diego Rey-Varela
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Center for Microbial Research (UCMR), Science for Life Laboratory (SciLifeLab), Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Felipe Cava
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Center for Microbial Research (UCMR), Science for Life Laboratory (SciLifeLab), Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Tobias Dörr
- Department of Microbiology, Cornell University, Ithaca, New York, United States of America
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, United States of America
- Cornell Institute for Host-Microbe Interactions and Disease (CIHMID), Ithaca, New York, United States of America
| |
Collapse
|