302 results on '"Microbiology, Virology & Host Pathogen Interaction"'
Search Results
2. Structural basis for specific inhibition of the highly sensitive ShHTL7 receptor
- Author
-
Shahul Hameed, Umar, Haider, Imran, Jamil, Muhammad, Kountche, Boubacar A, Guo, Xianrong, Zarban, Randa A, Kim, Dongjin, Al‐Babili, Salim, and Arold, Stefan T
- Subjects
MAX2 ,Hydrolases ,Octoxynol ,Protein Conformation ,Weed Control ,Plant Biology ,Plant Weeds ,Germination ,Striga ,X‐ray crystallography ,Crystallography, X-Ray ,Biochemistry ,Article ,Structural Biology ,Genetics ,Molecular Biology ,Plant Proteins ,seed germination inhibitor ,Herbicides ,Articles ,Microbiology, Virology & Host Pathogen Interaction ,Striga hermonthica ,hydrolase ,Corrigendum ,Protein Binding ,Signal Transduction - Abstract
Striga hermonthica is a root parasitic plant that infests cereals, decimating yields, particularly in sub‐Saharan Africa. For germination, Striga seeds require host‐released strigolactones that are perceived by the family of HYPOSENSITIVE to LIGHT (ShHTL) receptors. Inhibiting seed germination would thus be a promising approach for combating Striga. However, there are currently no strigolactone antagonists that specifically block ShHTLs and do not bind to DWARF14, the homologous strigolactone receptor of the host. Here, we show that the octyl phenol ethoxylate Triton X‐100 inhibits S. hermonthica seed germination without affecting host plants. High‐resolution X‐ray structures reveal that Triton X‐100 specifically plugs the catalytic pocket of ShHTL7. ShHTL7‐specific inhibition by Triton X‐100 demonstrates the dominant role of this particular ShHTL receptor for Striga germination. Our structural analysis provides a rationale for the broad specificity and high sensitivity of ShHTL7, and reveals that strigolactones trigger structural changes in ShHTL7 that are required for downstream signaling. Our findings identify Triton and the related 2‐[4‐(2,4,4‐trimethylpentan‐2‐yl)phenoxy]acetic acid as promising lead compounds for the rational design of efficient Striga‐specific herbicides.
- Published
- 2022
- Full Text
- View/download PDF
3. TICAM-1/TRIF associates with Act1 and suppresses IL-17 receptor–mediated inflammatory responses
- Author
-
Kimitoshi Nakamura, Yusuke Miyashita, Masaaki Okamoto, Takahisa Kouwaki, Hirotake Tsukamoto, and Hiroyuki Oshiumi
- Subjects
Chemokine ,Health, Toxicology and Mutagenesis ,Immunology ,Autoimmunity ,macromolecular substances ,Plant Science ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Autoimmune Diseases ,Proinflammatory cytokine ,Mice ,medicine ,Animals ,Molecular Biology ,Research Articles ,Inflammation ,Ecology ,biology ,Chemistry ,Experimental autoimmune encephalomyelitis ,Receptors, Interleukin ,medicine.disease ,Peptide Fragments ,Microbiology, Virology & Host Pathogen Interaction ,Cell biology ,CXCL1 ,Adaptor Proteins, Vesicular Transport ,CXCL2 ,TRIF ,Connexin 43 ,Gene Knockdown Techniques ,TLR3 ,biology.protein ,Disease Susceptibility ,Cytokine receptor ,Biomarkers ,Signal Transduction ,Research Article - Abstract
TICAM-1/TRIF, a TLR3 adaptor molecule, associates with Act1 to inhibit the interaction between IL-17RA and Act1, resulting in attenuated IL-17-mediated inflammatory responses., TICAM-1 (also called TRIF) is the sole adaptor of TLR3 that recognizes double-stranded RNA. Here, we report that TICAM-1 is involved not only in TLR3 signaling but also in the cytokine receptor IL-17RA signaling. We found that TICAM-1 bound to IL-17R adaptor Act1 to inhibit the interaction between IL-17RA and Act1. Interestingly, TICAM-1 knockout promoted IL-17RA/Act1 interaction and increased IL-17A–mediated activation of NF-κB and MAP kinases, leading to enhanced expression of inflammatory cytokines and chemokines upon IL-17A stimulation. Moreover, Ticam-1 knockout augmented IL-17A–mediated CXCL1 and CXCL2 expression in vivo, resulting in accumulation of myeloid cells. Furthermore, Ticam-1 knockout enhanced delayed type hypersensitivity and exacerbated experimental autoimmune encephalomyelitis. Ticam-1 knockout promoted accumulation of myeloid and lymphoid cells in the spinal cord of EAE-induced mice. Collectively, these data indicate that TICAM-1 inhibits the interaction between IL-17RA and Act1 and functions as a negative regulator in IL-17A–mediated inflammatory responses.
- Published
- 2022
4. Glycolysis/gluconeogenesis specialization in microbes is driven by biochemical constraints of flux sensing
- Author
-
Schink, Severin Josef, Christodoulou, Dimitris, Mukherjee, Avik, Athaide, Edward, Brunner, Viktoria, Fuhrer, Tobias, Bradshaw, Gary Andrew, Sauer, Uwe, and Basan, Markus
- Subjects
Medicine (General) ,QH301-705.5 ,Microorganism ,Metabolite ,Metabolic network ,lag time ,Article ,General Biochemistry, Genetics and Molecular Biology ,chemistry.chemical_compound ,specialization ,R5-920 ,flux sensing ,Glycolysis ,trade‐off ,Biology (General) ,Organism ,trade-off ,biology ,General Immunology and Microbiology ,Pseudomonas putida ,Applied Mathematics ,Gluconeogenesis ,Metabolism ,Articles ,biology.organism_classification ,Carbon ,Microbiology, Virology & Host Pathogen Interaction ,Glucose ,chemistry ,Computational Theory and Mathematics ,metabolism ,Biophysics ,General Agricultural and Biological Sciences ,Information Systems - Abstract
Central carbon metabolism is highly conserved across microbial species, but can catalyze very different pathways depending on the organism and their ecological niche. Here, we study the dynamic reorganization of central metabolism after switches between the two major opposing pathway configurations of central carbon metabolism, glycolysis, and gluconeogenesis in Escherichia coli, Pseudomonas aeruginosa, and Pseudomonas putida. We combined growth dynamics and dynamic changes in intracellular metabolite levels with a coarse-grained model that integrates fluxes, regulation, protein synthesis, and growth and uncovered fundamental limitations of the regulatory network: After nutrient shifts, metabolite concentrations collapse to their equilibrium, rendering the cell unable to sense which direction the flux is supposed to flow through the metabolic network. The cell can partially alleviate this by picking a preferred direction of regulation at the expense of increasing lag times in the opposite direction. Moreover, decreasing both lag times simultaneously comes at the cost of reduced growth rate or higher futile cycling between metabolic enzymes. These three trade-offs can explain why microorganisms specialize for either glycolytic or gluconeogenic substrates and can help elucidate the complex growth patterns exhibited by different microbial species., Molecular Systems Biology, 18 (1), ISSN:1744-4292
- Published
- 2022
5. Dimensionless parameter predicts bacterial prodrug success
- Author
-
Brandon Alexander Holt, McKenzie Tuttle, Yilin Xu, Melanie Su, Joachim J Røise, Xioajian Wang, Niren Murthy, and Gabriel A Kwong
- Subjects
Medicine (General) ,antibiotic failure ,General Immunology and Microbiology ,Bacteria ,QH301-705.5 ,Applied Mathematics ,enzymes ,Articles ,Bacterial Infections ,minimum inhibitory concentration ,General Biochemistry, Genetics and Molecular Biology ,Article ,Microbiology, Virology & Host Pathogen Interaction ,Anti-Bacterial Agents ,R5-920 ,Computational Theory and Mathematics ,prodrugs ,Humans ,Pharmacology & Drug Discovery ,Biology (General) ,General Agricultural and Biological Sciences ,Antimicrobial Peptides ,Information Systems - Abstract
Understanding mechanisms of antibiotic failure is foundational to combating the growing threat of multidrug‐resistant bacteria. Prodrugs—which are converted into a pharmacologically active compound after administration—represent a growing class of therapeutics for treating bacterial infections but are understudied in the context of antibiotic failure. We hypothesize that strategies that rely on pathogen‐specific pathways for prodrug conversion are susceptible to competing rates of prodrug activation and bacterial replication, which could lead to treatment escape and failure. Here, we construct a mathematical model of prodrug kinetics to predict rate‐dependent conditions under which bacteria escape prodrug treatment. From this model, we derive a dimensionless parameter we call the Bacterial Advantage Heuristic (BAH) that predicts the transition between prodrug escape and successful treatment across a range of time scales (1–104 h), bacterial carrying capacities (5 × 104–105 CFU/µl), and Michaelis constants (KM = 0.747–7.47 mM). To verify these predictions in vitro, we use two models of bacteria‐prodrug competition: (i) an antimicrobial peptide hairpin that is enzymatically activated by bacterial surface proteases and (ii) a thiomaltose‐conjugated trimethoprim that is internalized by bacterial maltodextrin transporters and hydrolyzed by free thiols. We observe that prodrug failure occurs at BAH values above the same critical threshold predicted by the model. Furthermore, we demonstrate two examples of how failing prodrugs can be rescued by decreasing the BAH below the critical threshold via (i) substrate design and (ii) nutrient control. We envision such dimensionless parameters serving as supportive pharmacokinetic quantities that guide the design and administration of prodrug therapeutics., Analyses of potential prodrug failure mechanisms show that prodrugs that rely on pathogen‐specific pathways for conversion are susceptible to competing rates of prodrug activation and bacterial replication, which could lead to prodrug treatment escape and failure.
- Published
- 2022
6. Shiga toxin (Stx) type 2‐induced increase in O‐linked N‐acetyl glucosamine protein modification: a new therapeutic target?
- Author
-
Rebecca A Bova and Angela Melton‐Celsa
- Subjects
Medicine (General) ,QH426-470 ,Kidney ,Shiga Toxins ,Shiga Toxin 2 ,Article ,Shiga Toxin ,Mice ,R5-920 ,Post-translational Modifications & Proteolysis ,hemic and lymphatic diseases ,Genetics ,Animals ,News & Views ,Escherichia coli Infections ,Glucosamine ,Shiga-Toxigenic Escherichia coli ,apoptosis ,O‐GlcNAcylation ,Articles ,Endoplasmic Reticulum Stress ,Microbiology, Virology & Host Pathogen Interaction ,inflammation ,Hemolytic-Uremic Syndrome ,hemolytic uremic syndrome ,Molecular Medicine ,Digestive System - Abstract
Shiga toxins (Stxs) produced by enterohemorrhagic Escherichia coli (EHEC) are the major virulence factors responsible for hemorrhagic colitis, which can lead to life‐threatening systemic complications including acute renal failure (hemolytic uremic syndrome) and neuropathy. Here, we report that O‐GlcNAcylation, a type of post‐translational modification, was acutely increased upon induction of endoplasmic reticulum (ER) stress in host cells by Stxs. Suppression of the abnormal Stx‐mediated increase in O‐GlcNAcylation effectively inhibited apoptotic and inflammatory responses in Stx‐susceptible cells. The protective effect of O‐GlcNAc inhibition for Stx‐mediated pathogenic responses was also verified using three‐dimensional (3D)‐cultured spheroids or organoids mimicking the human kidney. Treatment with an O‐GlcNAcylation inhibitor remarkably improved the major disease symptoms and survival rate for mice intraperitoneally injected with a lethal dose of Stx. In conclusion, this study elucidates O‐GlcNAcylation‐dependent pathogenic mechanisms of Stxs and demonstrates that inhibition of aberrant O‐GlcNAcylation is a potential approach to treat Stx‐mediated diseases., Here we identify a previously unknown link between the O‐GlcNAcylation pathway and the pathogenesis of enterohemorrhagic E. coli Shiga toxin‐mediated diseases. Given the capacity of these bacterial toxins to subvert normal cellular regulation in the host, this study demonstrates that Shiga toxins alter O‐GlcNAcylation, a type of post‐translational modification, to exacerbate dysfunction in host cell signaling.
- Published
- 2021
7. Deciphering the physiological response of Escherichia coli under high ATP demand
- Author
-
Boecker, Simon, Slaviero, Giulia, Schramm, Thorben, Szymanski, Witold, Steuer, Ralf, Link, Hannes, and Klamt, Steffen
- Subjects
Medicine (General) ,QH301-705.5 ,General Biochemistry, Genetics and Molecular Biology ,Article ,ATP homeostasis ,03 medical and health sciences ,R5-920 ,Adenosine Triphosphate ,Escherichia coli ,Biology (General) ,030304 developmental biology ,0303 health sciences ,General Immunology and Microbiology ,030306 microbiology ,Applied Mathematics ,Articles ,kinetic model ,glycolysis ,Microbiology, Virology & Host Pathogen Interaction ,Metabolism ,Computational Theory and Mathematics ,central metabolism ,General Agricultural and Biological Sciences ,metabolic engineering ,Energy Metabolism ,Information Systems - Abstract
One long‐standing question in microbiology is how microbes buffer perturbations in energy metabolism. In this study, we systematically analyzed the impact of different levels of ATP demand in Escherichia coli under various conditions (aerobic and anaerobic, with and without cell growth). One key finding is that, under all conditions tested, the glucose uptake increases with rising ATP demand, but only to a critical level beyond which it drops markedly, even below wild‐type levels. Focusing on anaerobic growth and using metabolomics and proteomics data in combination with a kinetic model, we show that this biphasic behavior is induced by the dual dependency of the phosphofructokinase on ATP (substrate) and ADP (allosteric activator). This mechanism buffers increased ATP demands by a higher glycolytic flux but, as shown herein, it collapses under very low ATP concentrations. Model analysis also revealed two major rate‐controlling steps in the glycolysis under high ATP demand, which could be confirmed experimentally. Our results provide new insights on fundamental mechanisms of bacterial energy metabolism and guide the rational engineering of highly productive cell factories., A systematic analysis of the impact of different levels of ATP demand on E. coli under various conditions reveals a biphasic response curve of the glucose uptake rate with respect to increasing ATP demand.
- Published
- 2021
8. Suboptimal resource allocation in changing environments constrains response and growth in bacteria
- Author
-
Rohan Balakrishnan, Roshali T de Silva, Terence Hwa, and Jonas Cremer
- Subjects
Medicine (General) ,General Immunology and Microbiology ,Bacteria ,QH301-705.5 ,Applied Mathematics ,resource allocation ,diauxie ,Articles ,cellular response ,General Biochemistry, Genetics and Molecular Biology ,Article ,Microbiology, Virology & Host Pathogen Interaction ,R5-920 ,Phenotype ,Metabolism ,Computational Theory and Mathematics ,environmental changes ,Escherichia coli ,Biology (General) ,General Agricultural and Biological Sciences ,growth optimality ,Information Systems - Abstract
To respond to fluctuating conditions, microbes typically need to synthesize novel proteins. As this synthesis relies on sufficient biosynthetic precursors, microbes must devise effective response strategies to manage depleting precursors. To better understand these strategies, we investigate the active response of Escherichia coli to changes in nutrient conditions, connecting transient gene expression to growth phenotypes. By synthetically modifying gene expression during changing conditions, we show how the competition by genes for the limited protein synthesis capacity constrains cellular response. Despite this constraint cells substantially express genes that are not required, trapping them in states where precursor levels are low and the genes needed to replenish the precursors are outcompeted. Contrary to common modeling assumptions, our findings highlight that cells do not optimize growth under changing environments but rather exhibit hardwired response strategies that may have evolved to promote fitness in their native environment. The constraint and the suboptimality of the cellular response uncovered provide a conceptual framework relevant for many research applications, from the prediction of evolution to the improvement of gene circuits in biotechnology., Analyses of how allocation of cellular resources to different genes shapes Escherichia coli's response to changing nutrient conditions show that growth transitions are determined by the competition between genes that are directly required in the encountered conditions and those that are not.
- Published
- 2021
9. Inhibition of IRGM establishes a robust antiviral immune state to restrict pathogenic viruses
- Author
-
Kautilya Kumar Jena, Nishant Ranjan Chauhan, Soma Chattopadhyay, Ramyasingh Bal, Punit Prasad, Tapas Kumar Nayak, Krushna Chandra Murmu, Saikat De, Parej Nath, Srinivas Patnaik, Santosh Chauhan, Nilima Dinesh Kumar, Fulvio Reggiori, Swatismita Priyadarsini, Kshitish Rout, Ankita Datey, Manjula Kalia, Subhash Mehto, Center for Liver, Digestive and Metabolic Diseases (CLDM), and Microbes in Health and Disease (MHD)
- Subjects
IRGM1 ,viruses ,CHIKV ,Immunology ,Virus Replication ,Antiviral Agents ,Biochemistry ,SARS‐CoV‐2 ,Virus ,Mice ,GTP-Binding Proteins ,Interferon ,Report ,Genetics ,medicine ,Animals ,Humans ,Coronaviridae ,Molecular Biology ,ZIKV ,biology ,Rhabdoviridae ,IRGM ,biology.organism_classification ,Virology ,Microbiology, Virology & Host Pathogen Interaction ,Viral replication ,Virus Diseases ,Viperin ,Tetherin ,Reports ,Signal Transduction ,medicine.drug - Abstract
The type I interferon (IFN) response is the major host arsenal against invading viruses. IRGM is a negative regulator of IFN responses under basal conditions. However, the role of human IRGM during viral infection has remained unclear. In this study, we show that IRGM expression is increased upon viral infection. IFN responses induced by viral PAMPs are negatively regulated by IRGM. Conversely, IRGM depletion results in a robust induction of key viral restriction factors including IFITMs, APOBECs, SAMHD1, tetherin, viperin, and HERC5/6. Additionally, antiviral processes such as MHC‐I antigen presentation and stress granule signaling are enhanced in IRGM‐deficient cells, indicating a robust cell‐intrinsic antiviral immune state. Consistently, IRGM‐depleted cells are resistant to the infection with seven viruses from five different families, including Togaviridae, Herpesviridae, Flaviviverdae, Rhabdoviridae, and Coronaviridae. Moreover, we show that Irgm1 knockout mice are highly resistant to chikungunya virus (CHIKV) infection. Altogether, our work highlights IRGM as a broad therapeutic target to promote defense against a large number of human viruses, including SARS‐CoV‐2, CHIKV, and Zika virus., IRGM negatively regulates IFN responses and IRGM expression is increased upon viral infection. Its depletion triggers anti‐viral restriction factors and promotes resistance to a large number of human viruses, including SARS‐CoV2, CHIKV and ZIKV.
- Published
- 2021
- Full Text
- View/download PDF
10. Inhibiting parasite proliferation using a rationally designed anti‐tubulin agent
- Author
-
Carolyn A. Moores, Mamata Bangera, Valentin Ehrhard, Naomi S. Morrissette, Anthony J. Roberts, Natacha Gaillard, Izra Abbaali, Tianyang Liu, Fiona Shilliday, Ashwani Sharma, Michel O. Steinmetz, and Alexander D. Cook
- Subjects
Medicine (General) ,macromolecular substances ,Biology ,QH426-470 ,microtubules ,R5-920 ,Tubulin ,Structural Biology ,Microtubule ,Report ,parasitic diseases ,Genetics ,Animals ,Humans ,Parasite hosting ,Parasites ,anti‐parasite ,Biological sciences ,Cell Proliferation ,Binding Sites ,Antiparasitic Agents ,Drug discovery ,Rational design ,species‐specific tubulin inhibitor ,biology.organism_classification ,Tubulin Modulators ,Microbiology, Virology & Host Pathogen Interaction ,Cell biology ,rational structure‐based drug design ,biology.protein ,Molecular Medicine ,Protozoa - Abstract
Infectious diseases caused by apicomplexan parasites remain a global public health threat. The presence of multiple ligand‐binding sites in tubulin makes this protein an attractive target for anti‐parasite drug discovery. However, despite remarkable successes as anti‐cancer agents, the rational development of protozoan parasite‐specific tubulin drugs has been hindered by a lack of structural and biochemical information on protozoan tubulins. Here, we present atomic structures for a protozoan tubulin and microtubule and delineate the architectures of apicomplexan tubulin drug‐binding sites. Based on this information, we rationally designed the parasite‐specific tubulin inhibitor parabulin and show that it inhibits growth of parasites while displaying no effects on human cells. Our work presents for the first time the rational design of a species‐specific tubulin drug providing a framework to exploit structural differences between human and protozoa tubulin variants enabling the development of much‐needed, novel parasite inhibitors., In an effort to discover novel drug‐scaffolds targeting unique parasite proteins and pathways, specific inhibition of parasite tubulin was achieved using structure‐guided rational drug design.
- Published
- 2021
11. Protective antigenic sites identified in respiratory syncytial virus fusion protein reveals importance of p27 domain
- Author
-
Laura Klenow, Juanjie Tang, Youri Lee, Surender Khurana, Hana Golding, Supriya Ravichandran, Elizabeth M. Coyle, and Jeehyun Lee
- Subjects
Medicine (General) ,viruses ,Respiratory Syncytial Virus Infections ,Biology ,QH426-470 ,Antibodies, Viral ,Virus ,Epitope ,Article ,Mice ,Immune system ,R5-920 ,Antigen ,vaccine ,Genetics ,Respiratory Syncytial Virus Vaccines ,Animals ,Antibody-dependent cell-mediated cytotoxicity ,epitope ,Mice, Inbred BALB C ,F protein ,virus diseases ,RSV ,Articles ,respiratory system ,neutralization ,Virology ,Fusion protein ,Antibodies, Neutralizing ,Microbiology, Virology & Host Pathogen Interaction ,Respiratory Syncytial Virus, Human ,biology.protein ,Molecular Medicine ,Antibody ,Viral load ,Viral Fusion Proteins - Abstract
Respiratory syncytial virus (RSV) vaccines primarily focused on surface fusion (F) protein are under development. Therefore, to identify RSV‐F protective epitopes, we evaluated 14 antigenic sites recognized following primary human RSV infection. BALB/c mice were vaccinated with F peptides, F proteins, or RSV‐A2, followed by rA2‐Line19F challenge. F peptides generated binding antibodies with minimal in vitro neutralization titers. However, several F peptides (including Site II) reduced lung viral loads and lung pathology scores in animals, suggesting partial protection from RSV disease. Interestingly, animals vaccinated with peptides (aa 101–121 and 110–136) spanning the F‐p27 sequence, which is only present in unprocessed F0 protein, showed control of viral loads with significantly reduced pathology compared with mock‐vaccinated controls. Furthermore, we observed F‐p27 expression on the surface of RSV‐infected cells as well as lungs from RSV‐infected mice. The anti‐p27 antibodies demonstrated antibody‐dependent cellular cytotoxicity (ADCC) of RSV‐infected A549 cells. These findings suggest that p27‐mediated immune response may play a role in control of RSV disease in vivo, and F‐p27 should be considered for inclusion in an effective RSV vaccine., This study identifies possible protective linear antigenic sites on the RSV F protein in a mouse RSV challenge model for development of RSV vaccine. We show that F‐p27 peptide control viral loads and reduced RSV disease in vivo. Therefore, F‐p27 should be included in an effective RSV vaccine.
- Published
- 2021
12. SARS‐CoV‐2–host proteome interactions for antiviral drug discovery
- Author
-
Lisa Gawriyski, Salla Keskitalo, Ina Pöhner, Antti Hassinen, Tuomo Laitinen, Markku Varjosalo, Kari Salokas, Tiina Öhman, Taras A. Redchuk, Maria K. Vartiainen, Arun Kumar Tonduru, Sini Huuskonen, Xiaonan Liu, Antti Poso, Vilja Pietiäinen, Mariia S. Bogacheva, Institute of Biotechnology, Molecular Systems Biology, Helsinki Institute of Life Science HiLIFE, Department of Virology, Tissue engineering for drug research, Institute for Molecular Medicine Finland, Nuclear organization by actin, Precision Systems Medicine, and Biosciences
- Subjects
INVOLVEMENT ,Medicine (General) ,Proteome ,viruses ,Viral pathogenesis ,virus–host interactions ,SOFTWARE ,Virus Replication ,Proteomics ,SARS‐CoV‐2 ,0302 clinical medicine ,RESPIRATORY SYNDROME CORONAVIRUS ,Biology (General) ,mass spectrometry ,11832 Microbiology and virology ,0303 health sciences ,Drug discovery ,Applied Mathematics ,Articles ,Microbiology, Virology & Host Pathogen Interaction ,3. Good health ,Computational Theory and Mathematics ,030220 oncology & carcinogenesis ,Host-Pathogen Interactions ,VIRUS ,RIBAVIRIN ,General Agricultural and Biological Sciences ,Information Systems ,EXPRESSION ,QH301-705.5 ,PROTEINS ,medicine.drug_class ,INHIBITION ,Computational biology ,Biology ,Antiviral Agents ,Article ,General Biochemistry, Genetics and Molecular Biology ,drug discovery ,03 medical and health sciences ,R5-920 ,proteomics ,Human interactome ,medicine ,Humans ,Pharmacology & Drug Discovery ,virus-host interactions ,030304 developmental biology ,Virtual screening ,IDENTIFICATION ,General Immunology and Microbiology ,SARS-CoV-2 ,Drug Repositioning ,COVID-19 ,COVID-19 Drug Treatment ,Methotrexate ,Viral replication ,1182 Biochemistry, cell and molecular biology ,Antiviral drug - Abstract
Treatment options for COVID‐19, caused by SARS‐CoV‐2, remain limited. Understanding viral pathogenesis at the molecular level is critical to develop effective therapy. Some recent studies have explored SARS‐CoV‐2–host interactomes and provided great resources for understanding viral replication. However, host proteins that functionally associate with SARS‐CoV‐2 are localized in the corresponding subnetwork within the comprehensive human interactome. Therefore, constructing a downstream network including all potential viral receptors, host cell proteases, and cofactors is necessary and should be used as an additional criterion for the validation of critical host machineries used for viral processing. This study applied both affinity purification mass spectrometry (AP‐MS) and the complementary proximity‐based labeling MS method (BioID‐MS) on 29 viral ORFs and 18 host proteins with potential roles in viral replication to map the interactions relevant to viral processing. The analysis yields a list of 693 hub proteins sharing interactions with both viral baits and host baits and revealed their biological significance for SARS‐CoV‐2. Those hub proteins then served as a rational resource for drug repurposing via a virtual screening approach. The overall process resulted in the suggested repurposing of 59 compounds for 15 protein targets. Furthermore, antiviral effects of some candidate drugs were observed in vitro validation using image‐based drug screen with infectious SARS‐CoV‐2. In addition, our results suggest that the antiviral activity of methotrexate could be associated with its inhibitory effect on specific protein–protein interactions., A large‐scale proteomics study identifies critical host proteins for SARS‐CoV‐2 processing. Proteins from these core subnetworks are used for drug repurposing analyses, indicating drugs with antiviral effects.
- Published
- 2021
- Full Text
- View/download PDF
13. Ontogeny and function of the circadian clock in intestinal organoids
- Author
-
Suengwon Lee, James M. Wells, Jennifer Hawkins, Krithika R Subramanian, Andrew E. Rosselot, Gang Wu, John B. Hogenesch, Nathan Salomonis, Michael A. Helmrath, Nambirajan Sundaram, Danilo E. F. L. Flôres, Sean R. Moore, Christian I. Hong, Toru Matsu-ura, Miri Park, Heather A. McCauley, Kashish Chetal, Noah F. Shroyer, and Taylor Broda
- Subjects
human enteroids ,Circadian clock ,Motility ,Clostridium difficile toxin B ,RAC1 ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Ticks ,0302 clinical medicine ,Circadian Clocks ,Gene expression ,Organoid ,Animals ,Humans ,Small GTPase ,Circadian rhythm ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,General Immunology and Microbiology ,General Neuroscience ,Articles ,Microbiology, Virology & Host Pathogen Interaction ,3. Good health ,Cell biology ,Intestines ,Organoids ,circadian rhythms ,intestinal organoids ,Chromatin, Transcription & Genomics ,Digestive System ,Function (biology) ,Rac1 ,030217 neurology & neurosurgery - Abstract
Circadian rhythms regulate diverse aspects of gastrointestinal physiology ranging from the composition of microbiota to motility. However, development of the intestinal circadian clock and detailed mechanisms regulating circadian physiology of the intestine remain largely unknown. In this report, we show that both pluripotent stem cell‐derived human intestinal organoids engrafted into mice and patient‐derived human intestinal enteroids possess circadian rhythms and demonstrate circadian phase‐dependent necrotic cell death responses to Clostridium difficile toxin B (TcdB). Intriguingly, mouse and human enteroids demonstrate anti‐phasic necrotic cell death responses to TcdB. RNA‐Seq analysis shows that ~3–10% of the detectable transcripts are rhythmically expressed in mouse and human enteroids. Remarkably, we observe anti‐phasic gene expression of Rac1, a small GTPase directly inactivated by TcdB, between mouse and human enteroids, and disruption of Rac1 abolishes clock‐dependent necrotic cell death responses. Our findings uncover robust functions of circadian rhythms regulating clock‐controlled genes in both mouse and human enteroids governing organism‐specific, circadian phase‐dependent necrotic cell death responses, and lay a foundation for human organ‐ and disease‐specific investigation of clock functions using human organoids for translational applications., Mouse and human 3D in vitro models show tissue maturation to determine species‐specific circadian rhytms in the intestine.
- Published
- 2021
- Full Text
- View/download PDF
14. A versatile genetic toolbox for Prevotella copri enables studying polysaccharide utilization systems
- Author
-
Agata A. Bielecka, Eric J. C. Gálvez, Till Robin Lesker, Aida Iljazovic, Eva-Magdalena Schorr, Lena Amend, Jing Li, Till Strowig, Éva Almási, and HZI,Helmholtz-Zentrum für Infektionsforschung GmbH, Inhoffenstr. 7,38124 Braunschweig, Germany.
- Subjects
Prevotella ,hybrid two‐component system ,Computational biology ,Methods & Resources ,Biology ,human diet ,Polysaccharide ,General Biochemistry, Genetics and Molecular Biology ,Article ,chemistry.chemical_compound ,Feces ,Human gut ,Polysaccharides ,Humans ,Allele ,Molecular Biology ,chemistry.chemical_classification ,General Immunology and Microbiology ,General Neuroscience ,Diet, Vegetarian ,polysaccharide utilization locus ,Articles ,Galactan ,Health states ,Toolbox ,Microbiology, Virology & Host Pathogen Interaction ,genetic manipulation ,Gastrointestinal Microbiome ,Metabolism ,chemistry ,Genetic Loci ,Prevotella copri ,hybrid two-component system ,Genome, Bacterial - Abstract
Prevotella copri is a prevalent inhabitant of the human gut and has been associated with plant‐rich diet consumption and diverse health states. The underlying genetic basis of these associations remains enigmatic due to the lack of genetic tools. Here, we developed a novel versatile genetic toolbox for rapid and efficient genetic insertion and allelic exchange applicable to P. copri strains from multiple clades. Enabled by the genetic platform, we systematically investigated the specificity of polysaccharide utilization loci (PULs) and identified four highly conserved PULs for utilizing arabinan, pectic galactan, arabinoxylan, and inulin, respectively. Further genetic and functional analysis of arabinan utilization systems illustrate that P. copri has evolved two distinct types of arabinan‐processing PULs (PULAra) and that the type‐II PULAra is significantly enriched in individuals consuming a vegan diet compared to other diets. In summary, this genetic toolbox will enable functional genetic studies for P. copri in future., A novel conjugation‐based gene insertion/excision system allowing genetic manipulation of the previously intractable bacterium Prevotella copri provides insight into human diet effects on commensal bacterial metabolism.
- Published
- 2021
15. Flower lose, a cell fitness marker, predicts COVID‐19 prognosis
- Author
-
Christopher J Pelham, Esha Madan, Markus Tolnay, António M Palma, Emad A. Rakha, Stephanie M. McGregor, Maximilian Ackermann, Everlyne Nkadori, Alexandar Tzankov, Laura K. Muller, Linbu Liao, Mark S. Parker, Emily S Rice, Jasmin D. Haslbauer, Thomas Braun, Raquel Cruz-Duarte, Andrew K. Godwin, Max S. Wicha, Thomas Menter, Matthias S. Matter, Denise Camacho, Benjamin Tang, Kirsty R. Short, Kyoung-Jae Won, Paul B. Fisher, Michail Yekelchyk, Kristina A Matkwoskyj, Eduardo Moreno, Sahil Chaudhary, Rajan Gogna, Kornelia Galior, Inês Rolim, Masaki Nagane, Ronny Drapkin, Michael T. Winters, Kartik Gupta, Antonio Lopez-Beltran, Ivan Martinez, Arutha Kulasinghe, Clay B. Marsh, Jochen Wilhelm, Steven R Grossman, and Raghavendra Pillappa
- Subjects
Oncology ,Medicine (General) ,medicine.medical_specialty ,Flowers ,Disease ,QH426-470 ,Lung injury ,Severity of Illness Index ,cell fitness ,Article ,R5-920 ,COVID‐19 ,Internal medicine ,Severity of illness ,Genetics ,medicine ,Humans ,Pandemics ,Retrospective Studies ,Receiver operating characteristic ,SARS-CoV-2 ,business.industry ,COVID-19 ,Retrospective cohort study ,Articles ,Triage ,Microbiology, Virology & Host Pathogen Interaction ,flower ,ROC Curve ,biomarker ,Molecular Medicine ,Biomarker (medicine) ,Observational study ,prognosis ,business ,Biomarkers - Abstract
Risk stratification of COVID‐19 patients is essential for pandemic management. Changes in the cell fitness marker, hFwe‐Lose, can precede the host immune response to infection, potentially making such a biomarker an earlier triage tool. Here, we evaluate whether hFwe‐Lose gene expression can outperform conventional methods in predicting outcomes (e.g., death and hospitalization) in COVID‐19 patients. We performed a post‐mortem examination of infected lung tissue in deceased COVID‐19 patients to determine hFwe‐Lose’s biological role in acute lung injury. We then performed an observational study (n = 283) to evaluate whether hFwe‐Lose expression (in nasopharyngeal samples) could accurately predict hospitalization or death in COVID‐19 patients. In COVID‐19 patients with acute lung injury, hFwe‐Lose is highly expressed in the lower respiratory tract and is co‐localized to areas of cell death. In patients presenting in the early phase of COVID‐19 illness, hFwe‐Lose expression accurately predicts subsequent hospitalization or death with positive predictive values of 87.8–100% and a negative predictive value of 64.1–93.2%. hFwe‐Lose outperforms conventional inflammatory biomarkers and patient age and comorbidities, with an area under the receiver operating characteristic curve (AUROC) 0.93–0.97 in predicting hospitalization/death. Specifically, this is significantly higher than the prognostic value of combining biomarkers (serum ferritin, D‐dimer, C‐reactive protein, and neutrophil–lymphocyte ratio), patient age and comorbidities (AUROC of 0.67–0.92). The cell fitness marker, hFwe‐Lose, accurately predicts outcomes in COVID‐19 patients. This finding demonstrates how tissue fitness pathways dictate the response to infection and disease and their utility in managing the current COVID‐19 pandemic., A post‐mortem examination of COVID‐19 infected lung tissues and an observational study were performed in order to evaluate whether expression of cell fitness marker hFwe‐Lose in patient's nasopharyngeal swabs could predict hospitalization or death from COVID‐19.
- Published
- 2021
- Full Text
- View/download PDF
16. Brain Cross‐Protection against SARS‐CoV‐2 Variants by a Lentiviral Vaccine in New Transgenic Mice
- Author
-
Ku, Min-Wen, Authié, Pierre, Bourgine, Maryline, Anna, François, Noirat, Amandine, Moncoq, Fanny, Vesin, Benjamin, Nevo, Fabien, Lopez, Jodie, Souque, Philippe, Blanc, Catherine, Fert, Ingrid, Chardenoux, Sébastien, Lafosse, llta, Cussigh, Delphine, Hardy, David, Nemirov, Kirill, Guinet, Françoise, Vives, Francina Langa, Majlessi, Laleh, Charneau, Pierre, Laboratoire commun Pasteur-TheraVectys, Institut Pasteur [Paris]-TheraVectys, Centre d'Ingénierie génétique murine - Mouse Genetics Engineering Center (CIGM), Institut Pasteur [Paris], Neuropathologie expérimentale / Experimental neuropathology, Institut Pasteur [Paris]-Université de Paris (UP), Lymphocytes et Immunité - Lymphocytes and Immunity, Institut Pasteur [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), This work was supported by the «URGENCE COVID-19» fundraising campaign of Institut Pasteur, TheraVectys and Agence Nationale de la Recherche (ANR) HuMoCID. M.W. Ku is part of the Pasteur - Paris University (PPU) International PhD Program and received funding from the Institut Carnot Pasteur Microbes & Santé, and the European Union's Horizon 2020 research and innovation program under the Marie Sklodowska-Curie grant agreement No 665807., The authors are grateful to Pr S. van der Werf (National Reference Centre for Respiratory Viruses hosted by Institut Pasteur, Paris, France), F. Guivel-Benhassine and Pr O. Schwartz (Institut Pasteur) for providing the BetaCoV/France/IDF0372/2020 and Gamma (P.1) SARS-CoV-2 clinical isolates. The strain BetaCoV/France/IDF0372/2020 was supplied through the European Virus Archive goes Global (Evag) platform, a project that has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement No 653316. The authors thank Pr G. Milon and Dr L.A. Chakrabarti for fruitful advice and discussion, Dr H. Mouquet and Dr C. Planchais for providing recombinant homotrimeric S proteins, Dr N. Escriou and Dr M. Gransagne for providing a plasmid containing the prefusion SCoV-2 sequence,M. Tichit and N. Dominique, for excellent technical assistance, respectively, in preparing histological sections and in animal immunization., ANR-20-COVI-0028,HuMoCID,Développement de modèles murins de COVID-19(2020), European Project: 665807,H2020,H2020-MSCA-COFUND-2014,PASTEURDOC(2015), Institut Pasteur [Paris] (IP)-TheraVectys-Université Paris Cité (UPCité), Institut Pasteur [Paris] (IP)-Université Paris Cité (UPCité), Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), Hardy, David, Développement de modèles murins de COVID-19 - - HuMoCID2020 - ANR-20-COVI-0028 - COVID-19 - VALID, and Institut Pasteur International Docotal Program - PASTEURDOC - - H20202015-10-01 - 2020-10-01 - 665807 - VALID
- Subjects
Central Nervous System ,Medicine (General) ,COVID-19 Vaccines ,[SDV]Life Sciences [q-bio] ,Immunology ,Mice, Transgenic ,QH426-470 ,Antibodies, Viral ,Article ,Mice ,R5-920 ,Genetics ,Animals ,Humans ,SARS-CoV-2 ,Brain ,COVID-19 ,Articles ,hACE2 Transgenic Mice ,Antibodies, Neutralizing ,Olfactory Bulb ,SARS-CoV-2 Emerging Variants of Concern ,Microbiology, Virology & Host Pathogen Interaction ,[SDV] Life Sciences [q-bio] ,Intranasal Vaccination ,Spike Glycoprotein, Coronavirus ,SARS‐CoV‐2 emerging variants of concern - Abstract
COVID‐19 vaccines already in use or in clinical development may have reduced efficacy against emerging SARS‐CoV‐2 variants. In addition, although the neurotropism of SARS‐CoV‐2 is well established, the vaccine strategies currently developed have not taken into account protection of the central nervous system. Here, we generated a transgenic mouse strain expressing the human angiotensin‐converting enzyme 2, and displaying unprecedented brain permissiveness to SARS‐CoV‐2 replication, in addition to high permissiveness levels in the lung. Using this stringent transgenic model, we demonstrated that a non‐integrative lentiviral vector, encoding for the spike glycoprotein of the ancestral SARS‐CoV‐2, used in intramuscular prime and intranasal boost elicits sterilizing protection of lung and brain against both the ancestral virus, and the Gamma (P.1) variant of concern, which carries multiple vaccine escape mutations. Beyond induction of strong neutralizing antibodies, the mechanism underlying this broad protection spectrum involves a robust protective T‐cell immunity, unaffected by the recent mutations accumulated in the emerging SARS‐CoV‐2 variants., A lentiviral vector‐based vaccine, used in prime and intranasal boost, induces strong B‐ and T‐cell immunity. This vaccine candidate protects against both pulmonary and neurological COVID‐19 caused by either ancestral or emerging SARS‐CoV‐2.
- Published
- 2021
- Full Text
- View/download PDF
17. Uncovering a conserved vulnerability site in SARS-CoV-2 by a human antibody
- Author
-
Shiqi Xu, Yuhuan Gong, Yanfang Li, Bowen Duan, JianFeng Chen, Liu Daisy Liu, Zhong Huang, Yanyan Wang, Haisheng Zhou, Hebang Yao, Zhou Sun, Wenming Qin, Lu Lu, Hervé Raoul, Ligang Wu, Dimitri Lavillette, Chenqi Xu, Barry Rockx, Bingjie Zhou, Dianfan Li, Xuechao Zhang, Yapei Zhao, Ning Zhang, Yanling Lai, Tingting Li, Yuhai Bi, Audrey S Richard, Moumita Mondal, Jun Li, Hongmin Cai, Martje Fentener van Vlissingen, Junfang Chen, Corine H. GeurtsvanKessel, Thijs Kuiken, Fei-Long Meng, Erasmus MC other, and Virology
- Subjects
Medicine (General) ,receptor‐binding domain ,medicine.drug_class ,Mutant ,QH426-470 ,medicine.disease_cause ,Monoclonal antibody ,Antibodies, Viral ,variants of concern ,Neutralization ,Epitope ,Article ,cross‐active neutralizing antibody ,03 medical and health sciences ,R5-920 ,0302 clinical medicine ,COVID‐19 ,Structural Biology ,Genetics ,medicine ,Humans ,030304 developmental biology ,0303 health sciences ,Mutation ,biology ,SARS-CoV-2 ,destruction of spike ,COVID-19 ,Articles ,Virology ,In vitro ,Microbiology, Virology & Host Pathogen Interaction ,3. Good health ,Viral replication ,Spike Glycoprotein, Coronavirus ,biology.protein ,Molecular Medicine ,Antibody ,030217 neurology & neurosurgery - Abstract
An essential step for SARS‐CoV‐2 infection is the attachment to the host cell receptor by its Spike receptor‐binding domain (RBD). Most of the existing RBD‐targeting neutralizing antibodies block the receptor‐binding motif (RBM), a mutable region with the potential to generate neutralization escape mutants. Here, we isolated and structurally characterized a non‐RBM‐targeting monoclonal antibody (FD20) from convalescent patients. FD20 engages the RBD at an epitope distal to the RBM with a K D of 5.6 nM, neutralizes SARS‐CoV‐2 including the current Variants of Concern such as B.1.1.7, B.1.351, P.1, and B.1.617.2 (Delta), displays modest cross‐reactivity against SARS‐CoV, and reduces viral replication in hamsters. The epitope coincides with a predicted “ideal” vulnerability site with high functional and structural constraints. Mutation of the residues of the conserved epitope variably affects FD20‐binding but confers little or no resistance to neutralization. Finally, in vitro mode‐of‐action characterization and negative‐stain electron microscopy suggest a neutralization mechanism by which FD20 destructs the Spike. Our results reveal a conserved vulnerability site in the SARS‐CoV‐2 Spike for the development of potential antiviral drugs., A monoclonal antibody (FD20) from convalescent COVID‐19 patients has been isolated and structurally and biologically characterized. Various SARS‐CoV‐2 strains, including the Alpha, Beta, Gamma, and Delta variants, and naturally occurring epitope mutants, can be neutralized by FD20 with similar potency.
- Published
- 2021
18. A dual‐function RNA balances carbon uptake and central metabolism in Vibrio cholerae
- Author
-
Andreas Tholey, Beatrice Engelmann, Ulrike Rolle-Kampczyk, Martin von Bergen, Liam Cassidy, James R. J. Haycocks, David C. Grainger, Mona Hoyos, Kavyaa Venkat, and Kai Papenfort
- Subjects
Cholera Toxin ,Down-Regulation ,Repressor ,Citrate (si)-Synthase ,Biology ,medicine.disease_cause ,Article ,Hfq ,General Biochemistry, Genetics and Molecular Biology ,Open Reading Frames ,dual‐function RNA ,Bacterial Proteins ,Transcription (biology) ,Gene expression ,medicine ,Genetic Testing ,Vibrio cholerae ,Molecular Biology ,General Immunology and Microbiology ,General Neuroscience ,Cholera toxin ,RNA ,Biological Transport ,Gene Expression Regulation, Bacterial ,Articles ,RNA Biology ,Carbon ,Microbiology, Virology & Host Pathogen Interaction ,Citric acid cycle ,RNA, Bacterial ,Open reading frame ,Phenotype ,Biochemistry ,small protein ,citrate synthase - Abstract
Bacterial small RNAs (sRNAs) are well known to modulate gene expression by base pairing with trans‐encoded transcripts and are typically non‐coding. However, several sRNAs have been reported to also contain an open reading frame and thus are considered dual‐function RNAs. In this study, we discovered a dual‐function RNA from Vibrio cholerae, called VcdRP, harboring a 29 amino acid small protein (VcdP), as well as a base‐pairing sequence. Using a forward genetic screen, we identified VcdRP as a repressor of cholera toxin production and link this phenotype to the inhibition of carbon transport by the base‐pairing segment of the regulator. By contrast, we demonstrate that the VcdP small protein acts downstream of carbon transport by binding to citrate synthase (GltA), the first enzyme of the citric acid cycle. Interaction of VcdP with GltA results in increased enzyme activity and together VcdR and VcdP reroute carbon metabolism. We further show that transcription of vcdRP is repressed by CRP allowing us to provide a model in which VcdRP employs two different molecular mechanisms to synchronize central metabolism in V. cholerae., A screen for repressors of cholera toxin production identifies VcdPR as the first V. cholerae RNA that acts both as a non‐coding expression modulator and by encoding a small protein.
- Published
- 2021
- Full Text
- View/download PDF
19. Vaccines beyond antibodies
- Author
-
King, Anthony
- Subjects
COVID-19 Vaccines ,SARS-CoV-2 ,T-Lymphocytes ,fungi ,education ,COVID-19 ,Antibodies, Viral ,Microbiology, Virology & Host Pathogen Interaction ,body regions ,Immunogenicity, Vaccine ,Humans ,Pharmacology & Drug Discovery ,skin and connective tissue diseases ,Science & Society ,Pandemics - Abstract
Lessons learned from the vaccines against SARS-CoV-2 has encouraged research and vaccine development aimed at mustering strong T cell responses against the pathogen.
- Published
- 2021
20. Interaction between the inflammasome and commensal microorganisms in gastrointestinal health and disease
- Author
-
Nobuhiko Kamada, Daisuke Watanabe, and Yijie Guo
- Subjects
Medicine (General) ,interleukin 18 ,Inflammasomes ,Immunology ,Interleukin-1beta ,Reviews ,Inflammation ,Disease ,Review ,QH426-470 ,Gut flora ,digestive system ,Pathogenesis ,R5-920 ,inflammasome ,Genetics ,medicine ,Pyroptosis ,Humans ,Caspase ,biology ,gut microbiota ,Inflammasome ,biology.organism_classification ,Microbiology, Virology & Host Pathogen Interaction ,Cell biology ,Gastrointestinal Microbiome ,Gastrointestinal Tract ,biology.protein ,Molecular Medicine ,interleukin 1β ,medicine.symptom ,Inflammasome complex ,medicine.drug - Abstract
The inflammasome is a cytosolic multiprotein complex that plays a crucial role in inflammation and cell death. The sensor proteins in the inflammasome complex detect various microbial and endogenous stimuli, leading to subsequent caspase activation. The activation of caspases results in the maturation of pro‐inflammatory cytokines IL‐1β and IL‐18 or pyroptosis. Inflammasome dysfunction is associated with the pathogenesis of various diseases, including autoimmune disease and cancer. It appears that the interactions between the gut microbiota and the inflammasome play crucial roles in the gastrointestinal tract. The gut microbiota induces the expression and activation of inflammasome proteins, which contribute to both homeostasis and disease in the gut. Likewise, although controversial, mounting evidence suggests that inflammasome activation can modulate the composition of the gut microbiota, which, in turn, affects disease progression. In this review, we summarize the current concepts and recent insights linking the inflammasome and gut commensal microorganisms. We describe how the reciprocal interaction between the inflammasome and the commensal microbiota relates to physiological and pathophysiological consequences in the host., In this review, N. Kamada and colleagues discuss the effects of the crosstalk between the inflammasome and commensal microorganisms on the physiology and pathophysiology of the host.
- Published
- 2021
21. Negative interactions determine Clostridioides difficile growth in synthetic human gut communities
- Author
-
Nasia Safdar, Tyler B. Jacobson, Susan E. Hromada, Yili Qian, Ophelia S. Venturelli, Ryan L. Clark, Lauren Watson, and Daniel Amador-Noguez
- Subjects
computational modeling ,Medicine (General) ,QH301-705.5 ,media_common.quotation_subject ,Systems biology ,Computational biology ,Colonisation resistance ,pathogen invasion ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Competition (biology) ,R5-920 ,Human gut ,Clostridioides ,Abundance (ecology) ,Humans ,Microbiome ,Biology (General) ,media_common ,Bacteria ,General Immunology and Microbiology ,Resistance (ecology) ,Clostridioides difficile ,Applied Mathematics ,Biotechnology & Synthetic Biology ,systems biology ,Articles ,Microbiology, Virology & Host Pathogen Interaction ,Gastrointestinal Microbiome ,ecological interactions ,Computational Theory and Mathematics ,Clostridium Infections ,Species richness ,General Agricultural and Biological Sciences ,Information Systems - Abstract
Understanding the principles of colonization resistance of the gut microbiome to the pathogen Clostridioides difficile will enable the design of defined bacterial therapeutics. We investigate the ecological principles of community resistance to C. difficile using a synthetic human gut microbiome. Using a dynamic computational model, we demonstrate that C. difficile receives the largest number and magnitude of incoming negative interactions. Our results show that C. difficile is in a unique class of species that display a strong negative dependence between growth and species richness. We identify molecular mechanisms of inhibition including acidification of the environment and competition over resources. We demonstrate that Clostridium hiranonis strongly inhibits C. difficile partially via resource competition. Increasing the initial density of C. difficile can increase its abundance in the assembled community, but community context determines the maximum achievable C. difficile abundance. Our work suggests that the C. difficile inhibitory potential of defined bacterial therapeutics can be optimized by designing communities featuring a combination of mechanisms including species richness, environment acidification, and resource competition., A combination of bottom‐up community assembly and computational modeling reveals determinants of Clostridioides difficile growth in synthetic human gut communities.
- Published
- 2021
- Full Text
- View/download PDF
22. SARS-CoV-2 Alpha, Beta, and Delta variants display enhanced Spike-mediated syncytia formation
- Author
-
Mathieu Hubert, Jérémy Dufloo, Rémy Robinot, Hugo Mouquet, Olivier Schwartz, Ludivine Grzelak, Nell Saunders, Elodie Bishop, Françoise Porrot, Delphine Planas, Maaran Michael Rajah, Julian Buchrieser, Lisa A. Chakrabarti, Marija Zivaljic, Alice Bongers, Stacy Gellenoncourt, Florence Guivel-Benhassine, Cyril Planchais, Virus et Immunité - Virus and immunity, Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Université Paris Cité (UPCité), Vaccine Research Institute (VRI), Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12), Sorbonne Université (SU), Neurobiologie intégrative des Systèmes cholinergiques / Integrative Neurobiology of Cholinergic Systems (NISC), Institut Pasteur [Paris]-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Ecole doctorale Cerveau Cognition et Comportement [Paris] (ED 158 - 3C), Immunologie humorale - Humoral Immunology, Institut Pasteur [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Work in OS lab is funded by Institut Pasteur, Urgence COVID-19 Fundraising Campaign of Institut Pasteur, ANRS, the Vaccine Research Institute (ANR-10-LABX-77), Fondation Pour la Recherche Médicale (FRM), Labex IBEID (ANR-10-LABX62-IBEID), ANR/FRM Flash Covid PROTEO-SARS-CoV-2, ANR CoronaMito AAP RA-COVID-19 V14, and IDISCOVR. Work in UPBI is funded by grant ANR-10-INSB-04-01 and Région Ile-de-France program DIM1-Health. MMR and MZ are supported by the Pasteur-Paris University (PPU) International Doctoral Program. MMR is also supported by Institut Pasteur Department of Virology 'Bourse de Soudure' fellowship. DP is supported by the Vaccine Research Institute. LG is supported by the French Ministry of Higher Education, Research and Innovation. EB is supported by the Medecine-Sciences ENS-PSL Program. HM laboratory is funded by the Institut Pasteur, the Milieu Intérieur Program (ANR-10-LABX-69- 01), the INSERM, REACTing, EU (RECOVER), and Fondation de France (#00106077) grants., We thank members of the Virus and Immunity Unit for helpful discussions, Dr. Nicoletta Casartelli for her critical reading of the manuscript, and Nathalie Aulner and the UtechS Photonic BioImaging (UPBI) core facility (Institut Pasteur), a member of the France BioImaging network, for image acquisition and analysis support., ANR-10-LABX-0077,VRI,Initiative for the creation of a Vaccine Research Institute(2010), ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), ANR-20-COVI-0059,PROTEO-SARS-CoV-2,Protéomique du SARS-CoV-2(2020), ANR-21-CO14-0007,CoronaMito,Conséquences de l'infection par le SRAS-CoV-2 sur la fonction mitochondriale(2021), ANR-10-INBS-0004,France-BioImaging,Développment d'une infrastructure française distribuée coordonnée(2010), ANR-10-LABX-0069,MILIEU INTERIEUR,GENETIC & ENVIRONMENTAL CONTROL OF IMMUNE PHENOTYPE VARIANCE: ESTABLISHING A PATH TOWARDS PERSONALIZED MEDICINE(2010), Virus et Immunité - Virus and immunity (CNRS-UMR3569), Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Vaccine Research Institute [Créteil, France] (VRI), Institut Pasteur [Paris] (IP)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM), Ziani, Isma, Laboratoires d'excellence - Initiative for the creation of a Vaccine Research Institute - - VRI2010 - ANR-10-LABX-0077 - LABX - VALID, Integrative Biology of Emerging Infectious Diseases - - IBEID2010 - ANR-10-LABX-0062 - LABX - VALID, Protéomique du SARS-CoV-2 - - PROTEO-SARS-CoV-22020 - ANR-20-COVI-0059 - COVID-19 - VALID, Conséquences de l'infection par le SRAS-CoV-2 sur la fonction mitochondriale - - CoronaMito2021 - ANR-21-CO14-0007 - COVID-19 - VALID, Développment d'une infrastructure française distribuée coordonnée - - France-BioImaging2010 - ANR-10-INBS-0004 - INBS - VALID, and Laboratoires d'excellence - GENETIC & ENVIRONMENTAL CONTROL OF IMMUNE PHENOTYPE VARIANCE: ESTABLISHING A PATH TOWARDS PERSONALIZED MEDICINE - - MILIEU INTERIEUR2010 - ANR-10-LABX-0069 - LABX - VALID
- Subjects
fusion ,coronavirus ,MESH: Spike Glycoprotein, Coronavirus ,MESH: Angiotensin-Converting Enzyme 2 ,medicine.disease_cause ,Virus Replication ,Giant Cells ,SARS‐CoV‐2 ,MESH: Antibodies, Monoclonal ,MESH: Giant Cells ,MESH: Chlorocebus aethiops ,Chlorocebus aethiops ,MESH: Animals ,Receptor ,Coronavirus ,[SDV.MP.VIR] Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,Syncytium ,Strain (chemistry) ,General Neuroscience ,Antibodies, Monoclonal ,Articles ,Transmembrane protein ,Microbiology, Virology & Host Pathogen Interaction ,Cell biology ,MESH: HEK293 Cells ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,Spike Glycoprotein, Coronavirus ,MESH: Caco-2 Cells ,Angiotensin-Converting Enzyme 2 ,MESH: Mutation ,medicine.drug_class ,Immunology ,Alpha (ethology) ,MESH: Vero Cells ,Biology ,Monoclonal antibody ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,medicine ,Animals ,Humans ,MESH: SARS-CoV-2 ,Beta (finance) ,syncytia ,Molecular Biology ,Vero Cells ,MESH: Humans ,General Immunology and Microbiology ,SARS-CoV-2 ,MESH: Virus Replication ,spike ,MESH: Cell Line ,HEK293 Cells ,Mutation ,Caco-2 Cells - Abstract
Severe COVID‐19 is characterized by lung abnormalities, including the presence of syncytial pneumocytes. Syncytia form when SARS‐CoV‐2 spike protein expressed on the surface of infected cells interacts with the ACE2 receptor on neighboring cells. The syncytia forming potential of spike variant proteins remain poorly characterized. Here, we first assessed Alpha (B.1.1.7) and Beta (B.1.351) spread and fusion in cell cultures, compared with the ancestral D614G strain. Alpha and Beta replicated similarly to D614G strain in Vero, Caco‐2, Calu‐3, and primary airway cells. However, Alpha and Beta formed larger and more numerous syncytia. Variant spike proteins displayed higher ACE2 affinity compared with D614G. Alpha, Beta, and D614G fusion was similarly inhibited by interferon‐induced transmembrane proteins (IFITMs). Individual mutations present in Alpha and Beta spikes modified fusogenicity, binding to ACE2 or recognition by monoclonal antibodies. We further show that Delta spike also triggers faster fusion relative to D614G. Thus, SARS‐CoV‐2 emerging variants display enhanced syncytia formation., Spike protein mutations expressed by emerging SARS‐CoV‐2 variants‐of‐concern differentially affect host cell‐to‐cell fusion, ACE2 receptor binding, and antibody escape.
- Published
- 2021
- Full Text
- View/download PDF
23. The ubiquitin ligation machinery in the defense against bacterial pathogens
- Author
-
Arno F. Alpi, Ishita Tripathi-Giesgen, and Christian Behrends
- Subjects
Salmonella typhimurium ,Ubiquitin-Protein Ligases ,Review ,Biochemistry ,Post-translational Modifications & Proteolysis ,Ubiquitin ,Salmonella ,xenophagy ,Genetics ,Xenophagy ,innate immunity ,Molecular Biology ,E3 ligase ,Innate immune system ,biology ,Host (biology) ,Effector ,Ubiquitination ,Mycobacterium tuberculosis ,biology.organism_classification ,Microbiology, Virology & Host Pathogen Interaction ,Ubiquitin ligase ,Cell biology ,biology.protein ,Autophagy & Cell Death ,Ligation ,Bacteria - Abstract
The ubiquitin system is an important part of the host cellular defense program during bacterial infection. This is in particular evident for a number of bacteria including Salmonella Typhimurium and Mycobacterium tuberculosis which—inventively as part of their invasion strategy or accidentally upon rupture of seized host endomembranes—become exposed to the host cytosol. Ubiquitylation is involved in the detection and clearance of these bacteria as well as in the activation of innate immune and inflammatory signaling. Remarkably, all these defense responses seem to emanate from a dense layer of ubiquitin which coats the invading pathogens. In this review, we focus on the diverse group of host cell E3 ubiquitin ligases that help to tailor this ubiquitin coat. In particular, we address how the divergent ubiquitin conjugation mechanisms of these ligases contribute to the complexity of the anti‐bacterial coating and the recruitment of different ubiquitin‐binding effectors. We also discuss the activation and coordination of the different E3 ligases and which strategies bacteria evolved to evade the activities of the host ubiquitin system., The ubiquitin system is an important part of the cellular defense against invading bacterial pathogens. This review discusses the function and interplay of the different E3 ligases that act in this tug of war between bacteria and the host cell.
- Published
- 2021
- Full Text
- View/download PDF
24. Porin threading drives receptor disengagement and establishes active colicin transport through Escherichia coli OmpF
- Author
-
Renata Kaminska, Natalya Lukoyanova, Boonyaporn Chinthammit, Emma Elliston, Colin Kleanthous, Melissa N. Webby, Marie-Louise R. Francis, and Nicholas G. Housden
- Subjects
cryo‐electron microscopy ,Models, Molecular ,Protein Conformation, alpha-Helical ,medicine.disease_cause ,0303 health sciences ,biology ,Escherichia coli Proteins ,General Neuroscience ,Articles ,Translocon ,Microbiology, Virology & Host Pathogen Interaction ,Protein Transport ,Colicin ,Porin ,Thermodynamics ,lipids (amino acids, peptides, and proteins) ,Periplasmic Proteins ,Bacterial outer membrane ,Bacterial Outer Membrane Proteins ,Protein Binding ,Gram-negative bacteria ,Colicins ,Porins ,outer membrane ,Article ,bacteriocins ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Protein Domains ,Bacteriocin ,Escherichia coli ,medicine ,Protein Interaction Domains and Motifs ,Molecular Biology ,030304 developmental biology ,Binding Sites ,General Immunology and Microbiology ,030306 microbiology ,Cryoelectron Microscopy ,Membrane Proteins ,Membrane Transport Proteins ,Gene Expression Regulation, Bacterial ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Membranes & Trafficking ,Kinetics ,Bacterial Outer Membrane ,Gram‐negative bacteria ,Colicin transport ,Biophysics ,bacteria ,Protein Conformation, beta-Strand ,fluorescent microscopy - Abstract
Bacteria deploy weapons to kill their neighbours during competition for resources and to aid survival within microbiomes. Colicins were the first such antibacterial system identified, yet how these bacteriocins cross the outer membrane (OM) of Escherichia coli is unknown. Here, by solving the structures of translocation intermediates via cryo‐EM and by imaging toxin import, we uncover the mechanism by which the Tol‐dependent nuclease colicin E9 (ColE9) crosses the bacterial OM. We show that threading of ColE9’s disordered N‐terminal domain through two pores of the trimeric porin OmpF causes the colicin to disengage from its primary receptor, BtuB, and reorganises the translocon either side of the membrane. Subsequent import of ColE9 through the lumen of a single OmpF subunit is driven by the proton‐motive force, which is delivered by the TolQ‐TolR‐TolA‐TolB assembly. Our study answers longstanding questions, such as why OmpF is a better translocator than OmpC, and reconciles the mechanisms by which both Tol‐ and Ton‐dependent bacteriocins cross the bacterial outer membrane., Cryo‐EM structures and fluorescence imaging reveal how the Tol‐dependent bacteriocin ColE9 crosses the outer bacterial membrane to exert microbial killing.
- Published
- 2021
- Full Text
- View/download PDF
25. High-efficiency delivery of CRISPR-Cas9 by engineered probiotics enables precise microbiome editing
- Author
-
Kevin Neil, Sébastien Rodrigue, Alfredo Menendez, Vincent Burrus, Patricia Roy, Nancy Allard, and Frédéric Grenier
- Subjects
Medicine (General) ,antibiotic resistance ,QH301-705.5 ,Computational biology ,Gut flora ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Article ,antimicrobials ,03 medical and health sciences ,Synthetic biology ,Mice ,R5-920 ,Antibiotic resistance ,medicine ,Citrobacter rodentium ,CRISPR ,Animals ,Microbiome ,Biology (General) ,Escherichia coli ,030304 developmental biology ,Gene Editing ,0303 health sciences ,General Immunology and Microbiology ,biology ,030306 microbiology ,Applied Mathematics ,Bacterial conjugation ,Microbiota ,Probiotics ,Biotechnology & Synthetic Biology ,Articles ,biology.organism_classification ,Microbiology, Virology & Host Pathogen Interaction ,bacterial conjugation ,Computational Theory and Mathematics ,Conjugation, Genetic ,synthetic biology ,CRISPR-Cas Systems ,General Agricultural and Biological Sciences ,CRISPR‐Cas9 ,Information Systems - Abstract
Antibiotic resistance threatens our ability to treat infectious diseases, spurring interest in alternative antimicrobial technologies. The use of bacterial conjugation to deliver CRISPR‐cas systems programmed to precisely eliminate antibiotic‐resistant bacteria represents a promising approach but requires high in situ DNA transfer rates. We have optimized the transfer efficiency of conjugative plasmid TP114 using accelerated laboratory evolution. We hence generated a potent conjugative delivery vehicle for CRISPR‐cas9 that can eliminate > 99.9% of targeted antibiotic‐resistant Escherichia coli in the mouse gut microbiota using a single dose. We then applied this system to a Citrobacter rodentium infection model, achieving full clearance within four consecutive days of treatment., A conjugative plasmid with high transfer rates is leveraged to deliver the CRISPR system into targeted bacteria. The resulting conjugative system can clear a C. rodentium infection in mice.
- Published
- 2021
26. COVID‐19 and the gain of function debates
- Author
-
Gigi Kwik Gronvall, Kelsey Lane Warmbrod, and Michael Montague
- Subjects
Hot Temperature ,viruses ,Respiratory System ,Hemagglutinin Glycoproteins, Influenza Virus ,Biochemistry ,Biosafety ,Influenza A Virus, H1N1 Subtype ,Zoonoses ,Pandemic ,Medicine ,Gain of function mutation ,Science & Society ,Molecular Epidemiology ,Protein Stability ,Economics, Law & Politics ,virus diseases ,Containment of Biohazards ,Adaptation, Physiological ,Microbiology, Virology & Host Pathogen Interaction ,Body Fluids ,Risk analysis (engineering) ,Gain of Function Mutation ,Population Surveillance ,Receptors, Virus ,Female ,Reassortant Viruses ,2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Article ,Security Measures ,Cell Line ,Birds ,Evolution, Molecular ,Dogs ,Orthomyxoviridae Infections ,Influenza, Human ,Genetics ,Animals ,Humans ,Pandemics ,Molecular Biology ,Influenza A Virus, H5N1 Subtype ,SARS-CoV-2 ,business.industry ,Ferrets ,COVID-19 ,Science Policy & Publishing ,Bioterrorism ,HEK293 Cells ,Gain of function ,Influenza in Birds ,business ,HeLa Cells - Abstract
Highly pathogenic avian H5N1 influenza A viruses occasionally infect humans, but currently do not transmit efficiently among humans. The viral haemagglutinin (HA) protein is a known host-range determinant as it mediates virus binding to host-specific cellular receptors. Here we assess the molecular changes in HA that would allow a virus possessing subtype H5 HA to be transmissible among mammals. We identified a reassortant H5 HA/H1N1 virus-comprising H5 HA (from an H5N1 virus) with four mutations and the remaining seven gene segments from a 2009 pandemic H1N1 virus-that was capable of droplet transmission in a ferret model. The transmissible H5 reassortant virus preferentially recognized human-type receptors, replicated efficiently in ferrets, caused lung lesions and weight loss, but was not highly pathogenic and did not cause mortality. These results indicate that H5 HA can convert to an HA that supports efficient viral transmission in mammals; however, we do not know whether the four mutations in the H5 HA identified here would render a wholly avian H5N1 virus transmissible. The genetic origin of the remaining seven viral gene segments may also critically contribute to transmissibility in mammals. Nevertheless, as H5N1 viruses continue to evolve and infect humans, receptor-binding variants of H5N1 viruses with pandemic potential, including avian-human reassortant viruses as tested here, may emerge. Our findings emphasize the need to prepare for potential pandemics caused by influenza viruses possessing H5 HA, and will help individuals conducting surveillance in regions with circulating H5N1 viruses to recognize key residues that predict the pandemic potential of isolates, which will inform the development, production and distribution of effective countermeasures.
- Published
- 2021
- Full Text
- View/download PDF
27. Targeted delivery of a phosphoinositide 3‐kinase γ inhibitor to restore organ function in sepsis
- Author
-
Adrian T Press, Petra Babic, Bianca Hoffmann, Tina Müller, Wanling Foo, Walter Hauswald, Jovana Benecke, Martina Beretta, Zoltán Cseresnyés, Stephanie Hoeppener, Ivo Nischang, Sina M Coldewey, Markus H Gräler, Reinhard Bauer, Falk Gonnert, Nikolaus Gaßler, Reinhard Wetzker, Marc Thilo Figge, Ulrich S Schubert, and Michael Bauer
- Subjects
Medicine (General) ,Liver Diseases ,liver failure ,Articles ,QH426-470 ,PI3K ,Article ,Microbiology, Virology & Host Pathogen Interaction ,sepsis ,Mice ,Phosphatidylinositol 3-Kinases ,R5-920 ,Neutrophil Infiltration ,drug delivery ,Genetics ,Animals ,cholestasis ,Phosphoinositide-3 Kinase Inhibitors - Abstract
Jaundice, the clinical hallmark of infection‐associated liver dysfunction, reflects altered membrane organization of the canalicular pole of hepatocytes and portends poor outcomes. Mice lacking phosphoinositide 3‐kinase‐γ (PI3Kγ) are protected against membrane disintegration and hepatic excretory dysfunction. However, they exhibit a severe immune defect that hinders neutrophil recruitment to sites of infection. To exploit the therapeutic potential of PI3Kγ inhibition in sepsis, a targeted approach to deliver drugs to hepatic parenchymal cells without compromising other cells, in particular immune cells, seems warranted. Here, we demonstrate that nanocarriers functionalized through DY‐635, a fluorescent polymethine dye, and a ligand of organic anion transporters can selectively deliver therapeutics to hepatic parenchymal cells. Applying this strategy to a murine model of sepsis, we observed the PI3Kγ‐dependent restoration of biliary canalicular architecture, maintained excretory liver function, and improved survival without impairing host defense mechanisms. This strategy carries the potential to expand targeted nanomedicines to disease entities with systemic inflammation and concomitantly impaired barrier functionality., Dye‐functionalized liposomes allow delivery of a PI3K‐γ inhibitor to hepatocytes to resolve sepsis‐related liver failure without ‘off‐target’ effects on immunity.
- Published
- 2021
28. SARS‐CoV‐2 structural coverage map reveals viral protein assembly, mimicry, and hijacking mechanisms
- Author
-
James B. Procter, Sandeep Kaur, Christian Stolte, Seán I. O'Donoghue, Neblina Sikta, Matt Adcock, Bosco K. Ho, Christian Dallago, Burkhard Rost, Andrea Schafferhans, Stuart Anderson, and Nicola Bordin
- Subjects
Models, Molecular ,Protein Conformation, alpha-Helical ,Medicine (General) ,medicine.disease_cause ,Virus Replication ,Mitochondrial Membrane Transport Proteins ,SARS‐CoV‐2 ,Viroporin Proteins ,Mitochondrial Precursor Protein Import Complex Proteins ,Protein Interaction Mapping ,data visualization ,structural biology ,Biology (General) ,Coronavirus ,Applied Mathematics ,Translation (biology) ,Articles ,bioinformatics ,Microbiology, Virology & Host Pathogen Interaction ,Molecular mimicry ,Computational Theory and Mathematics ,Proteome ,Host-Pathogen Interactions ,Spike Glycoprotein, Coronavirus ,Angiotensin-Converting Enzyme 2 ,General Agricultural and Biological Sciences ,Information Systems ,Protein Binding ,Viral protein ,QH301-705.5 ,Computational biology ,Methods & Resources ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Viral Matrix Proteins ,Coronavirus Envelope Proteins ,R5-920 ,COVID‐19 ,medicine ,Coronavirus Nucleocapsid Proteins ,Humans ,Protein Interaction Domains and Motifs ,Viral matrix protein ,Binding Sites ,General Immunology and Microbiology ,SARS-CoV-2 ,Molecular Mimicry ,COVID-19 ,Computational Biology ,Phosphoproteins ,Neuropilin-1 ,Amino Acid Transport Systems, Neutral ,Viral replication ,Structural biology ,Protein Conformation, beta-Strand ,Protein Multimerization ,Protein Processing, Post-Translational - Abstract
We modeled 3D structures of all SARS‐CoV‐2 proteins, generating 2,060 models that span 69% of the viral proteome and provide details not available elsewhere. We found that ˜6% of the proteome mimicked human proteins, while ˜7% was implicated in hijacking mechanisms that reverse post‐translational modifications, block host translation, and disable host defenses; a further ˜29% self‐assembled into heteromeric states that provided insight into how the viral replication and translation complex forms. To make these 3D models more accessible, we devised a structural coverage map, a novel visualization method to show what is—and is not—known about the 3D structure of the viral proteome. We integrated the coverage map into an accompanying online resource (https://aquaria.ws/covid) that can be used to find and explore models corresponding to the 79 structural states identified in this work. The resulting Aquaria‐COVID resource helps scientists use emerging structural data to understand the mechanisms underlying coronavirus infection and draws attention to the 31% of the viral proteome that remains structurally unknown or dark., 2,060 structural models spanning 69% of the SARS‐CoV‐2 proteome are generated and presented in a novel visual layout that summarises current knowledge on viral protein structures, and provides insight into viral replication, mimicry, and hijacking.
- Published
- 2021
29. Machine learning identifies molecular regulators and therapeutics for targeting SARS‐CoV2‐induced cytokine release
- Author
-
Siddharth Vijay, John McNevin, M. Juliana McElrath, Taranjit S. Gujral, Marina Chan, and Eric C. Holland
- Subjects
Lipopolysaccharides ,Male ,Medicine (General) ,Neutrophils ,medicine.medical_treatment ,computer.software_genre ,SARS‐CoV‐2 ,Machine Learning ,chemistry.chemical_compound ,Mice ,Interleukin-1 Receptor-Associated Kinases ,Biology (General) ,Sulfonamides ,Janus kinase 1 ,Kinase ,Applied Mathematics ,Ponatinib ,Imidazoles ,Articles ,Microbiology, Virology & Host Pathogen Interaction ,Pyridazines ,Cytokine ,kinases ,Computational Theory and Mathematics ,Host-Pathogen Interactions ,Spike Glycoprotein, Coronavirus ,Cytokines ,Signal transduction ,medicine.symptom ,General Agricultural and Biological Sciences ,Information Systems ,QH301-705.5 ,Phenotypic screening ,Immunology ,Inflammation ,N‐terminal domain ,Biology ,Machine learning ,Antiviral Agents ,General Biochemistry, Genetics and Molecular Biology ,Article ,R5-920 ,Chemical Biology ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,General Immunology and Microbiology ,business.industry ,SARS-CoV-2 ,Janus Kinase 1 ,Mice, Inbred C57BL ,RAW 264.7 Cells ,chemistry ,Purines ,Azetidines ,Pyrazoles ,Artificial intelligence ,business ,computer - Abstract
Although 15–20% of COVID‐19 patients experience hyper‐inflammation induced by massive cytokine production, cellular triggers of this process and strategies to target them remain poorly understood. Here, we show that the N‐terminal domain (NTD) of the SARS‐CoV‐2 spike protein substantially induces multiple inflammatory molecules in myeloid cells and human PBMCs. Using a combination of phenotypic screening with machine learning‐based modeling, we identified and experimentally validated several protein kinases, including JAK1, EPHA7, IRAK1, MAPK12, and MAP3K8, as essential downstream mediators of NTD‐induced cytokine production, implicating the role of multiple signaling pathways in cytokine release. Further, we found several FDA‐approved drugs, including ponatinib, and cobimetinib as potent inhibitors of the NTD‐mediated cytokine release. Treatment with ponatinib outperforms other drugs, including dexamethasone and baricitinib, inhibiting all cytokines in response to the NTD from SARS‐CoV‐2 and emerging variants. Finally, ponatinib treatment inhibits lipopolysaccharide‐mediated cytokine release in myeloid cells in vitro and lung inflammation mouse model. Together, we propose that agents targeting multiple kinases required for SARS‐CoV‐2‐mediated cytokine release, such as ponatinib, may represent an attractive therapeutic option for treating moderate to severe COVID‐19., Massive cytokine production and hyper‐inflammation have been associated with fatal SARS‐CoV‐2 infection outcomes. Phenotypic screening combined with machine learning‐based modeling identifies regulators and therapeutics for targeting SARS‐CoV‐2‐induced cytokine release.
- Published
- 2021
30. Infection and transmission of SARS‐CoV‐2 depend on heparan sulfate proteoglycans
- Author
-
Ad C. van Nuenen, Philip J. M. Brouwer, Marcel Spaargaren, Tanja M. Kaptein, Bernadien M. Nijmeijer, Godelieve J. de Bree, Julia Eder, John L. van Hamme, Marta Bermejo-Jambrina, Marit J. van Gils, Leanne C. Helgers, Neeltje A. Kootstra, Killian E Vlaming, Rogier W. Sanders, Teunis B. H. Geijtenbeek, Experimental Immunology, Graduate School, AII - Infectious diseases, APH - Aging & Later Life, Pathology, Infectious diseases, APH - Global Health, and Medical Microbiology and Infection Prevention
- Subjects
Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,viruses ,Heparan Sulfate Proteoglycans ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Article ,SARS‐CoV‐2 ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,Chlorocebus aethiops ,Animals ,Humans ,Receptor ,skin and connective tissue diseases ,Molecular Biology ,Vero Cells ,030304 developmental biology ,0303 health sciences ,Mucous Membrane ,General Immunology and Microbiology ,biology ,Transmission (medicine) ,SARS-CoV-2 ,General Neuroscience ,fungi ,virus diseases ,COVID-19 ,Epithelial Cells ,Articles ,Dendritic Cells ,Heparin, Low-Molecular-Weight ,Antibodies, Neutralizing ,Microbiology, Virology & Host Pathogen Interaction ,3. Good health ,COVID-19 Drug Treatment ,body regions ,030220 oncology & carcinogenesis ,Host-Pathogen Interactions ,Vero cell ,biology.protein ,Syndecan-4 ,Angiotensin-Converting Enzyme 2 ,Syndecan-1 ,Antibody ,low molecular weight heparins ,Heparan sulfate proteoglycans - Abstract
The current pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) and outbreaks of new variants highlight the need for preventive treatments. Here, we identified heparan sulfate proteoglycans as attachment receptors for SARS‐CoV‐2. Notably, neutralizing antibodies against SARS‐CoV‐2 isolated from COVID‐19 patients interfered with SARS‐CoV‐2 binding to heparan sulfate proteoglycans, which might be an additional mechanism of antibodies to neutralize infection. SARS‐CoV‐2 binding to and infection of epithelial cells was blocked by low molecular weight heparins (LMWH). Although dendritic cells (DCs) and mucosal Langerhans cells (LCs) were not infected by SARS‐CoV‐2, both DC subsets efficiently captured SARS‐CoV‐2 via heparan sulfate proteoglycans and transmitted the virus to ACE2‐positive cells. Notably, human primary nasal cells were infected by SARS‐CoV‐2, and infection was blocked by pre‐treatment with LMWH. These data strongly suggest that heparan sulfate proteoglycans are important attachment receptors facilitating infection and transmission, and support the use of LMWH as prophylaxis against SARS‐CoV‐2 infection., In addition to the essential host cell receptor ACE2, extracellular matrix molecules also contribute to SARS‐CoV‐2 attachment to human epithelial cells and dendritic cells.
- Published
- 2021
31. A Brucella effector modulates the Arf6‐Rab8a GTPase cascade to promote intravacuolar replication
- Author
-
Kelsey A. Binder, Elizabeth Borghesan, Erin P. Smith, Jean Celli, Sebenzile K. Myeni, and Leigh A. Knodler
- Subjects
Brucella abortus ,Endosomes ,GTPase ,Vacuole ,Biology ,Models, Biological ,Brucellosis ,Article ,General Biochemistry, Genetics and Molecular Biology ,Type IV Secretion Systems ,Mice ,Bacterial Proteins ,Animals ,Humans ,Secretion ,Molecular Biology ,General Immunology and Microbiology ,Effector ,pathogenesis ,General Neuroscience ,Endoplasmic reticulum ,GTPase-Activating Proteins ,Articles ,retrograde membrane transport ,Membrane transport ,Membranes & Trafficking ,Brucella ,type IV secretion ,Microbiology, Virology & Host Pathogen Interaction ,Cell biology ,Vesicular transport protein ,Protein Transport ,ADP-Ribosylation Factor 6 ,rab GTP-Binding Proteins ,Host-Pathogen Interactions ,Vacuoles ,ACAP1 ,Intracellular ,HeLa Cells ,Protein Binding ,trans-Golgi Network - Abstract
Remodeling of host cellular membrane transport pathways is a common pathogenic trait of many intracellular microbes that is essential to their intravacuolar life cycle and proliferation. The bacterium Brucella abortus generates a host endoplasmic reticulum‐derived vacuole (rBCV) that supports its intracellular growth, via VirB Type IV secretion system‐mediated delivery of effector proteins, whose functions and mode of action are mostly unknown. Here, we show that the effector BspF specifically promotes Brucella replication within rBCVs by interfering with vesicular transport between the trans‐Golgi network (TGN) and recycling endocytic compartment. BspF targeted the recycling endosome, inhibited retrograde traffic to the TGN, and interacted with the Arf6 GTPase‐activating Protein (GAP) ACAP1 to dysregulate Arf6‐/Rab8a‐dependent transport within the recycling endosome, which resulted in accretion of TGN‐associated vesicles by rBCVs and enhanced bacterial growth. Altogether, these findings provide mechanistic insight into bacterial modulation of membrane transport used to promote their own proliferation within intracellular vacuoles., Brucella abortus effector protein BspF binding to host GTPase‐activating protein ACAP1 facilitates intracellular bacterial growth by decreasing retrograde vesicular transport to the trans‐Golgi network.
- Published
- 2021
- Full Text
- View/download PDF
32. PLK1-dependent phosphorylation restrains EBNA2 activity and lymphomagenesis in EBV-infected mice
- Author
-
Xiang Zhang, André Mourão, Sophie Beer, Anita Murer, Christian Münz, Ralf Küppers, Sybille Thumann, Stefanie M. Hauck, Monika Raab, Klaus Strebhardt, Patrick Schuhmachers, Piero Giansanti, Cornelia Kuklik-Roos, Bernhard Kuster, Wolfgang Hammerschmidt, Michael Sattler, and Bettina Kempkes
- Subjects
Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Medizin ,Cell Cycle Proteins ,Biology ,Protein Serine-Threonine Kinases ,Biochemistry ,PLK1 ,Virus ,Article ,03 medical and health sciences ,Transactivation ,Mice ,0302 clinical medicine ,Antigen ,EBV ,hemic and lymphatic diseases ,Proto-Oncogene Proteins ,Genetics ,Animals ,Phosphorylation ,Molecular Biology ,Transcription factor ,030304 developmental biology ,Cancer ,EBNA2 ,0303 health sciences ,B-lymphomagenesis ,Ebna2 ,Ebv ,Plk1 ,Humanized Mice ,Kinase ,Articles ,B‐lymphomagenesis ,Microbiology, Virology & Host Pathogen Interaction ,3. Good health ,Virus Latency ,humanized mice ,Epstein-Barr Virus Nuclear Antigens ,030220 oncology & carcinogenesis ,Monoclonal ,Cancer research ,Signal Transduction - Abstract
While Epstein–Barr virus (EBV) establishes a life‐long latent infection in apparently healthy human immunocompetent hosts, immunodeficient individuals are at particular risk to develop lymphoproliferative B‐cell malignancies caused by EBV. A key EBV protein is the transcription factor EBV nuclear antigen 2 (EBNA2), which initiates B‐cell proliferation. Here, we combine biochemical, cellular, and in vivo experiments demonstrating that the mitotic polo‐like kinase 1 (PLK1) binds to EBNA2, phosphorylates its transactivation domain, and thereby inhibits its biological activity. EBNA2 mutants that impair PLK1 binding or prevent EBNA2 phosphorylation are gain‐of‐function mutants. They exhibit enhanced transactivation capacities, accelerate the proliferation of infected B cells, and promote the development of monoclonal B‐cell lymphomas in infected mice. Thus, PLK1 coordinates the activity of EBNA2 to attenuate the risk of tumor incidences in favor of the establishment of latency in the infected but healthy host., EBNA2 is a key transactivator that initiates B cell immortalization. PLK1 phosphorylates the C‐ terminal transactivation domain of EBNA2 to attenuate its oncogenic activities and promote the establishment of long term latency in the infected host.
- Published
- 2021
33. Degradation of WTAP blocks antiviral responses by reducing the m
- Author
-
Yong, Ge, Tao, Ling, Yao, Wang, Xin, Jia, Xiongmei, Xie, Rong, Chen, Shangwu, Chen, Shaochun, Yuan, and Anlong, Xu
- Subjects
Proteasome Endopeptidase Complex ,Immunology ,Ubiquitination ,Receptor, Interferon alpha-beta ,Articles ,IRF3 ,Antiviral Agents ,RNA Biology ,Article ,Microbiology, Virology & Host Pathogen Interaction ,WTAP ,m6A modification ,antiviral responses ,Interferon Regulatory Factor-3 ,RNA, Messenger ,IFNAR1 - Abstract
N 6‐methyladenosine (m6A) is a chemical modification present in multiple RNA species and is most abundant in mRNAs. Studies on m6A reveal its comprehensive roles in almost every aspect of mRNA metabolism, as well as in a variety of physiological processes. Although some recent discoveries indicate that m6A can affect the life cycles of numerous viruses as well as the cellular antiviral immune response, the roles of m6A modification in type I interferon (IFN‐I) signaling are still largely unknown. Here, we reveal that WT1‐associated protein (WTAP), one of the m6A “writers”, is degraded via the ubiquitination‐proteasome pathway upon activation of IFN‐I signaling. With the degradation of WTAP, the m6A levels of IFN‐regulatory factor 3 (IRF3) and interferon alpha/beta receptor subunit 1 (IFNAR1) mRNAs are reduced, leading to translational suppression of IRF3 and instability of IFNAR1 mRNA. Thus, the WTAP‐IRF3/IFNAR1 axis may serve as negative feedback pathway to fine‐tune the activation of IFN‐I signaling, which highlights the roles of m6A in the antiviral response by dictating the fate of mRNAs associated with IFN‐I signaling., Proteasome mediated degradation of the m6A writer WTAP upon viral infection reduces m6A levels on IRF3 and IFNAR1 mRNAs and subsequently their protein output, thereby blocking IFN‐I‐mediated antiviral responses.
- Published
- 2021
34. Neutralization of SARS‐CoV‐2 by highly potent, hyperthermostable, and mutation‐tolerant nanobodies
- Author
-
Oleh Rymarenko, Kathrin Gregor, Volker C. Cordes, Jürgen Schünemann, Dirk Görlich, Christian Dienemann, Antje Dickmanns, Kim M. Stegmann, Uwe Groß, Bianka Mussil, Matthias Dobbelstein, Renate Rees, Waltraud Taxer, Jens Krull, Thomas Güttler, Philip Gunkel, M. Aksu, and Ulrike Teichmann
- Subjects
Coronaviridae ,Immunology ,Biology ,Antibodies, Viral ,medicine.disease_cause ,Article ,SARS‐CoV‐2 ,General Biochemistry, Genetics and Molecular Biology ,Neutralization ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,COVID‐19 ,Escherichia coli ,medicine ,Animals ,Humans ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Mutation ,General Immunology and Microbiology ,SARS-CoV-2 ,General Neuroscience ,COVID-19 ,Articles ,Single-Domain Antibodies ,biology.organism_classification ,Antibodies, Neutralizing ,Virology ,Microbiology, Virology & Host Pathogen Interaction ,3. Good health ,nanobody ,Cell culture ,Spike Glycoprotein, Coronavirus ,Monoclonal ,biology.protein ,VHH antibody ,Female ,Antibody ,Camelids, New World ,030217 neurology & neurosurgery - Abstract
Monoclonal anti‐SARS‐CoV‐2 immunoglobulins represent a treatment option for COVID‐19. However, their production in mammalian cells is not scalable to meet the global demand. Single‐domain (VHH) antibodies (also called nanobodies) provide an alternative suitable for microbial production. Using alpaca immune libraries against the receptor‐binding domain (RBD) of the SARS‐CoV‐2 Spike protein, we isolated 45 infection‐blocking VHH antibodies. These include nanobodies that can withstand 95°C. The most effective VHH antibody neutralizes SARS‐CoV‐2 at 17–50 pM concentration (0.2–0.7 µg per liter), binds the open and closed states of the Spike, and shows a tight RBD interaction in the X‐ray and cryo‐EM structures. The best VHH trimers neutralize even at 40 ng per liter. We constructed nanobody tandems and identified nanobody monomers that tolerate the K417N/T, E484K, N501Y, and L452R immune‐escape mutations found in the Alpha, Beta, Gamma, Epsilon, Iota, and Delta/Kappa lineages. We also demonstrate neutralization of the Beta strain at low‐picomolar VHH concentrations. We further discovered VHH antibodies that enforce native folding of the RBD in the E. coli cytosol, where its folding normally fails. Such “fold‐promoting” nanobodies may allow for simplified production of vaccines and their adaptation to viral escape‐mutations., Single‐domain camelid antibodies that neutralize a range of common and emerging immune‐escape mutant strains of SARS‐CoV‐2 may constitute an easily‐producible option for treatment of COVID‐19 patients.
- Published
- 2021
- Full Text
- View/download PDF
35. Adaptive laboratory evolution of microbial co-cultures for improved metabolite secretion
- Author
-
Konstantinidis, Dimitrios, Pereira, Filipa, Geissen, Eva‐Maria, Grkovska, Kristina, Kafkia, Eleni, Jouhten, Paula, Kim, Yongkyu, Devendran, Saravanan, Zimmermann, Michael, Patil, Kiran Raosaheb, Konstantinidis, Dimitrios [0000-0002-2134-6823], Pereira, Filipa [0000-0002-0557-8480], Geissen, Eva-Maria [0000-0002-0423-7019], Grkovska, Kristina [0000-0002-3784-494X], Kafkia, Eleni [0000-0001-9550-4487], Jouhten, Paula [0000-0003-1075-7448], Kim, Yongkyu [0000-0002-3336-6741], Devendran, Saravanan [0000-0002-1540-5241], Zimmermann, Michael [0000-0002-5797-3589], Patil, Kiran Raosaheb [0000-0002-6166-8640], Apollo - University of Cambridge Repository, and Geissen, Eva‐Maria [0000-0002-0423-7019]
- Subjects
Proteomics ,Medicine (General) ,GENE DISRUPTION ,Metabolite ,Microbial metabolism ,EMBO21 ,EMBO23 ,DISEASE ,chemistry.chemical_compound ,multi‐omics ,Biology (General) ,ADAPTATION ,2. Zero hunger ,0303 health sciences ,Experimental evolution ,biology ,Applied Mathematics ,Articles ,Microbiology, Virology & Host Pathogen Interaction ,Flux balance analysis ,Lactic acid ,Computational Theory and Mathematics ,Biochemistry ,coevolution ,General Agricultural and Biological Sciences ,Life Sciences & Biomedicine ,Information Systems ,Biochemistry & Molecular Biology ,QH301-705.5 ,GENOMES ,EMBO41 ,Saccharomyces cerevisiae ,metabolic cooperation ,vitamin secretion ,LACTIC-ACID BACTERIA ,METABOLOMICS ,RIBOFLAVIN ,TRAGEDY ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,R5-920 ,Metabolomics ,Secretion ,experimental evolution ,Symbiosis ,030304 developmental biology ,Science & Technology ,General Immunology and Microbiology ,IDENTIFICATION ,030306 microbiology ,multi-omics ,biology.organism_classification ,Coculture Techniques ,PROTEOME ,Metabolism ,chemistry ,Laboratories ,Synthetic Biology & Biotechnology ,Bacteria - Abstract
Adaptive laboratory evolution has proven highly effective for obtaining microorganisms with enhanced capabilities. Yet, this method is inherently restricted to the traits that are positively linked to cell fitness, such as nutrient utilization. Here, we introduce coevolution of obligatory mutualistic communities for improving secretion of fitness‐costly metabolites through natural selection. In this strategy, metabolic cross‐feeding connects secretion of the target metabolite, despite its cost to the secretor, to the survival and proliferation of the entire community. We thus co‐evolved wild‐type lactic acid bacteria and engineered auxotrophic Saccharomyces cerevisiae in a synthetic growth medium leading to bacterial isolates with enhanced secretion of two B‐group vitamins, viz., riboflavin and folate. The increased production was specific to the targeted vitamin, and evident also in milk, a more complex nutrient environment that naturally contains vitamins. Genomic, proteomic and metabolomic analyses of the evolved lactic acid bacteria, in combination with flux balance analysis, showed altered metabolic regulation towards increased supply of the vitamin precursors. Together, our findings demonstrate how microbial metabolism adapts to mutualistic lifestyle through enhanced metabolite exchange., Adaptive laboratory evolution of obligatory mutualistic communities shows how coevolution can be used for targeted improvement in fitness‐costly metabolite secretion.
- Published
- 2021
- Full Text
- View/download PDF
36. Rational design of West Nile virus vaccine through large replacement of 3′ UTR with internal poly(A)
- Author
-
Jing Liu, Zhiming Yuan, Ya-Nan Zhang, Yuan Shaopeng, Cheng-Lin Deng, Qiu-Yan Zhang, Xiao-Dan Li, Bo Zhang, Hong-Qing Zhang, Pei Yong Shi, Han-Qing Ye, Fang Yu, Zhan Shunli, Gao Lei, Na Li, and Xiang-Yue Zeng
- Subjects
Untranslated region ,Medicine (General) ,live‐attenuated vaccine ,viruses ,Immunology ,QH426-470 ,Biology ,Antibodies, Viral ,Article ,UTR ,Mice ,R5-920 ,Immune system ,flavivirus ,Chlorocebus aethiops ,Genetics ,West Nile Virus ,Animals ,West Nile Virus Vaccines ,3' Untranslated Regions ,Vero Cells ,Pathogen ,internal poly(A) ,Attenuated vaccine ,virus diseases ,Outbreak ,Articles ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Virology ,Microbiology, Virology & Host Pathogen Interaction ,Vaccination ,Flavivirus ,Vero cell ,Molecular Medicine ,Poly A ,West Nile Fever - Abstract
The genus Flavivirus comprises numerous emerging and re‐emerging arboviruses causing human illness. Vaccines are the best approach to prevent flavivirus diseases. But pathogen diversities are always one of the major hindrances for timely development of new vaccines when confronting unpredicted flavivirus outbreaks. We used West Nile virus (WNV) as a model to develop a new live‐attenuated vaccine (LAV), WNV‐poly(A), by replacing 5ʹ portion (corresponding to SL and DB domains in WNV) of 3ʹ‐UTR with internal poly(A) tract. WNV‐poly(A) not only propagated efficiently in Vero cells, but also was highly attenuated in mouse model. A single‐dose vaccination elicited robust and long‐lasting immune responses, conferring full protection against WNV challenge. Such “poly(A)” vaccine strategy may be promising for wide application in the development of flavivirus LAVs because of its general target regions in flaviviruses., West Nile virus (WNV) is one of the many members of the genus Flavivirus known to cause human diseases. Here we present a novel live attenuated WNV vaccine, WNV‐poly(A), that protects against WNV infections in a mouse model.
- Published
- 2021
- Full Text
- View/download PDF
37. Alterations in mitochondrial morphology as a key driver of immunity and host defence
- Author
-
Anne M. Curtis, Shannon L. Cox, and Mariana P. Cervantes-Silva
- Subjects
Chemokine ,Programmed cell death ,Immunology ,Cell ,Reviews ,Review ,virus ,Mitochondrion ,Mitochondrial Dynamics ,Biochemistry ,immune response ,Virus ,Immune system ,Immunity ,Organelle ,Genetics ,medicine ,Humans ,bacteria ,Molecular Biology ,Organelles ,therapy ,Cell Death ,biology ,Bacterial Infections ,Microbiology, Virology & Host Pathogen Interaction ,Mitochondria ,Cell biology ,medicine.anatomical_structure ,biology.protein ,Signal Transduction - Abstract
Mitochondria are dynamic organelles whose architecture changes depending on the cell’s energy requirements and other signalling events. These structural changes are collectively known as mitochondrial dynamics. Mitochondrial dynamics are crucial for cellular functions such as differentiation, energy production and cell death. Importantly, it has become clear in recent years that mitochondrial dynamics are a critical control point for immune cell function. Mitochondrial remodelling allows quiescent immune cells to rapidly change their metabolism and become activated, producing mediators, such as cytokines, chemokines and even metabolites to execute an effective immune response. The importance of mitochondrial dynamics in immunity is evident, as numerous pathogens have evolved mechanisms to manipulate host cell mitochondrial remodelling in order to promote their own survival. In this review, we comprehensively address the roles of mitochondrial dynamics in immune cell function, along with modulation of host cell mitochondrial morphology during viral and bacterial infections to facilitate either pathogen survival or host immunity. We also speculate on what the future may hold in terms of therapies targeting mitochondrial morphology for bacterial and viral control., Mitochondrial dynamics is crucial for several cellular processes including immune cell function. This review discusses how modulation of host cell mitochondrial morphology affects pathogen survival and host immunity and speculates on related therapeutic approaches.
- Published
- 2021
- Full Text
- View/download PDF
38. TIFA has dual functions in Helicobacter pylori-induced classical and alternative NF-κB pathways
- Author
-
Michelle Chin Chia Lim, Gunter Maubach, Olga Sokolova, Michael Naumann, Steffen Backert, and Thomas F. Meyer
- Subjects
TRAF2 ,Immunology ,ALPK1 ,Inflammation ,Inhibitor of apoptosis ,Biochemistry ,Helicobacter Infections ,chemistry.chemical_compound ,Report ,TRAF ,Genetics ,medicine ,Humans ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,TNF Receptor-Associated Factor 6 ,Helicobacter pylori ,NIK ,biology ,Kinase ,Chemistry ,NF-kappa B ,Cancer ,NF-κB ,medicine.disease ,biology.organism_classification ,Microbiology, Virology & Host Pathogen Interaction ,cIAP1 ,Cancer research ,Tumor necrosis factor alpha ,medicine.symptom ,Reports ,Signal Transduction - Abstract
Helicobacter pylori infection constitutes one of the major risk factors for the development of gastric diseases including gastric cancer. The activation of nuclear factor‐kappa‐light‐chain‐enhancer of activated B cells (NF‐κB) via classical and alternative pathways is a hallmark of H. pylori infection leading to inflammation in gastric epithelial cells. Tumor necrosis factor receptor‐associated factor (TRAF)‐interacting protein with forkhead‐associated domain (TIFA) was previously suggested to trigger classical NF‐κB activation, but its role in alternative NF‐κB activation remains unexplored. Here, we identify TRAF6 and TRAF2 as binding partners of TIFA, contributing to the formation of TIFAsomes upon H. pylori infection. Importantly, the TIFA/TRAF6 interaction enables binding of TGFβ‐activated kinase 1 (TAK1), leading to the activation of classical NF‐κB signaling, while the TIFA/TRAF2 interaction causes the transient displacement of cellular inhibitor of apoptosis 1 (cIAP1) from TRAF2, and proteasomal degradation of cIAP1, to facilitate the activation of the alternative NF‐κB pathway. Our findings therefore establish a dual function of TIFA in the activation of classical and alternative NF‐κB signaling in H. pylori‐infected gastric epithelial cells., Helicobacter pylori’s ADP‐heptose binds to the kinase ALPK1 in gastric epithelial cells and directs activation of classical and alternative NF‐κB pathways in a TIFA dependent manner, thereby promoting gastric inflammation.
- Published
- 2021
39. Interactomes of SARS‐CoV‐2 and human coronaviruses reveal host factors potentially affecting pathogenesis
- Author
-
Xu Feng, Mrinal Srivastava, Chao Wang, Zhen Chen, Samuel K Swisher, Mengfan Tang, Yun Xiong, Junjie Chen, Huimin Zhang, and Litong Nie
- Subjects
Resource ,Proteomics ,viruses ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,interactome ,host–virus protein–protein interaction ,Disease ,Computational biology ,Viral Nonstructural Proteins ,Biology ,Virus Replication ,Interactome ,SARS‐CoV‐2 ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Pathogenesis ,Coronavirus Nucleocapsid Proteins ,Humans ,Protein Interaction Maps ,Molecular Biology ,Tandem affinity purification ,NSP1 ,General Immunology and Microbiology ,SARS-CoV-2 ,Host (biology) ,General Neuroscience ,COVID-19 ,virus diseases ,BioID2 ,Resources ,Microbiology, Virology & Host Pathogen Interaction ,Host-Pathogen Interactions ,SFB‐TAP - Abstract
Host–virus protein–protein interactions play key roles in the life cycle of severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2). We conducted a comprehensive interactome study between the virus and host cells using tandem affinity purification and proximity‐labeling strategies and identified 437 human proteins as the high‐confidence interacting proteins. Further characterization of these interactions and comparison to other large‐scale study of cellular responses to SARS‐CoV‐2 infection elucidated how distinct SARS‐CoV‐2 viral proteins participate in its life cycle. With these data mining, we discovered potential drug targets for the treatment of COVID‐19. The interactomes of two key SARS‐CoV‐2‐encoded viral proteins, NSP1 and N, were compared with the interactomes of their counterparts in other human coronaviruses. These comparisons not only revealed common host pathways these viruses manipulate for their survival, but also showed divergent protein–protein interactions that may explain differences in disease pathology. This comprehensive interactome of SARS‐CoV‐2 provides valuable resources for the understanding and treating of this disease., Human proteins identified as copurifying with or proximal to SARS‐CoV‐2 proteins suggest cellular response pathways and potential drug targets, as well as divergence of NSP1 and N protein binding specificities compared to related human coronaviruses.
- Published
- 2021
- Full Text
- View/download PDF
40. Tunable phenotypic variability through an autoregulatory alternative sigma factor circuit
- Author
-
Sandra Cortijo, Vassili Kusmartsev, Casandra Villava, Teresa Saez, Christian Schwall, Bruno M. C. Martins, James C. W. Locke, Toby Livesey, Torkel E Loman, University of Cambridge [UK] (CAM), University of Warwick [Coventry], European Research Council under the European Union's Seventh Framework Programme (FP/2007-2013)/ERC Grant 338060, Gatsby Foundation GAT3272/GLC, European Project: 0721456(2007), Locke, James CW [0000-0003-0670-1943], Apollo - University of Cambridge Repository, Locke, James C W [0000-0003-0670-1943], and Cortijo, Sandra [0000-0003-3291-6729]
- Subjects
Medicine (General) ,Operon ,stochastic gene expression ,[SDV]Life Sciences [q-bio] ,single-cell time-lapse microscopy ,Bacillus subtilis ,EMBO23 ,Stress level ,Sigma factor ,Homeostasis ,Autoregulation ,Biology (General) ,0303 health sciences ,Applied Mathematics ,microbial systems biology ,Significant phenotypic variability ,Sigma ,Articles ,Response Variability ,Phenotype ,Microbiology, Virology & Host Pathogen Interaction ,Cell biology ,Computational Theory and Mathematics ,General Agricultural and Biological Sciences ,Information Systems ,stress priming Subject Category Microbiology ,QH301-705.5 ,Sigma Factor ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,R5-920 ,Bacterial Proteins ,Humans ,stress priming ,030304 developmental biology ,General Immunology and Microbiology ,030306 microbiology ,Mechanism (biology) ,single‐cell time‐lapse microscopy ,QH ,fungi ,QK ,Gene Expression Regulation, Bacterial ,biology.organism_classification ,stress primingphenoty ,QR ,Virology & Host Pathogen Interaction ,Biological Variation, Population ,Biophysics - Abstract
Funder: FP7 Ideas: European Research Council (FP7 Ideas); Id: http://dx.doi.org/10.13039/100011199; Grant(s): 338060, Genetically identical individuals in bacterial populations can display significant phenotypic variability. This variability can be functional, for example by allowing a fraction of stress prepared cells to survive an otherwise lethal stress. The optimal fraction of stress prepared cells depends on environmental conditions. However, how bacterial populations modulate their level of phenotypic variability remains unclear. Here we show that the alternative sigma factor σV circuit in Bacillus subtilis generates functional phenotypic variability that can be tuned by stress level, environmental history and genetic perturbations. Using single‐cell time‐lapse microscopy and microfluidics, we find the fraction of cells that immediately activate σV under lysozyme stress depends on stress level and on a transcriptional memory of previous stress. Iteration between model and experiment reveals that this tunability can be explained by the autoregulatory feedback structure of the sigV operon. As predicted by the model, genetic perturbations to the operon also modulate the response variability. The conserved sigma‐anti‐sigma autoregulation motif is thus a simple mechanism for bacterial populations to modulate their heterogeneity based on their environment.
- Published
- 2021
- Full Text
- View/download PDF
41. Bacterial lysis, autophagy and innate immune responses during adjunctive phage therapy in a child
- Author
-
Aleksandra Petrovic-Fabijan, Sulaiman Almuzam, Jonathan R. Iredell, Michael J Brownstein, Quang Dao, Ameneh Khatami, Ran Nir-Paz, Sivan Alkalay-Oren, Philip N Britton, Joe Fackler, Bri'Anna Horne, Ronen Hazan, and Ruby C.Y. Lin
- Subjects
Medicine (General) ,Phage therapy ,medicine.medical_treatment ,Immunology ,osteoarticular infection ,QH426-470 ,medicine.disease_cause ,immune response ,Bacteriophage ,Transcriptome ,R5-920 ,Immune system ,Downregulation and upregulation ,bacteriophage ,Report ,Genetics ,medicine ,Autophagy ,Humans ,Bacteriophages ,Phage Therapy ,child ,Innate immune system ,biology ,Pseudomonas aeruginosa ,business.industry ,biology.organism_classification ,Immunity, Innate ,Microbiology, Virology & Host Pathogen Interaction ,Molecular Medicine ,Autophagy & Cell Death ,business - Abstract
Adjunctive phage therapy was used in an attempt to avoid catastrophic outcomes from extensive chronic Pseudomonas aeruginosa osteoarticular infection in a 7‐year‐old child. Monitoring of phage and bacterial kinetics allowed real‐time phage dose adjustment, and along with markers of the human host response, indicated a significant therapeutic effect within two weeks of starting adjunctive phage therapy. These findings strongly suggested the release of bacterial cells or cell fragments into the bloodstream from deep bony infection sites early in treatment. This was associated with transient fever and local pain and with evidence of marked upregulation of innate immunity genes in the host transcriptome. Adaptive immune responses appeared to develop after a week of therapy and some immunomodulatory elements were also observed to be upregulated., Monitoring of phage and bacterial kinetics during adjunctive phage therapy for extensive chronic osteoarticular infection in a child demonstrated an initial flush of bacterial contents into the bloodstream associated with an inflammatory response marked by fever, local pain and upregulation of genes linked to autophagy and innate immunity.
- Published
- 2021
42. Individual COVID‐19 disease trajectories revealed by plasma proteomics
- Author
-
Inigo Barrio-Hernandez, Pedro Beltrao, and Danish Memon
- Subjects
Proteomics ,Medicine (General) ,medicine.medical_specialty ,Proteome ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Disease ,QH426-470 ,medicine.disease_cause ,R5-920 ,Pandemic ,Genetics ,medicine ,Humans ,News & Views ,Intensive care medicine ,Pandemics ,Coronavirus ,SARS-CoV-2 ,business.industry ,COVID-19 ,Human physiology ,Microbiology, Virology & Host Pathogen Interaction ,Time course ,Molecular Medicine ,Plasma proteomics ,business - Abstract
EMBO Molecular Medicine, 13 (8), ISSN:1757-4676, ISSN:1757-4684
- Published
- 2021
- Full Text
- View/download PDF
43. Fungal phytochrome chromophore biosynthesis at mitochondria
- Author
-
Kai Leister, Norbert Krauß, Franco Weth, Reinhard Fischer, Christian Streng, Martin Bastmeyer, Jana Hartmann, Nicole Frankenberg-Dinkel, Zhenzhong Yu, and Tilman Lamparter
- Subjects
Life sciences ,biology ,Mitochondrion ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,metabolon ,Fungal Proteins ,chemistry.chemical_compound ,Biosynthesis ,ddc:570 ,Organelle ,chromophore ,heme ,Molecular Biology ,Heme ,Homeodomain Proteins ,Organelles ,phytochrome ,General Immunology and Microbiology ,Phytochrome ,General Neuroscience ,Alternaria ,Articles ,heme oxygenase ,Tetrapyrrole ,Microbiology, Virology & Host Pathogen Interaction ,Mitochondria ,Cell biology ,Chloroplast ,Protein Transport ,Metabolism ,chemistry ,Metabolon - Abstract
Mitochondria are essential organelles because of their function in energy conservation. Here, we show an involvement of mitochondria in phytochrome‐dependent light sensing in fungi. Phytochrome photoreceptors are found in plants, bacteria, and fungi and contain a linear, heme‐derived tetrapyrrole as chromophore. Linearization of heme requires heme oxygenases (HOs) which reside inside chloroplasts in planta. Despite the poor degree of conservation of HOs, we identified two candidates in the fungus Alternaria alternata. Deletion of either one phenocopied phytochrome deletion. The two enzymes had a cooperative effect and physically interacted with phytochrome, suggesting metabolon formation. The metabolon was attached to the surface of mitochondria with a C‐terminal anchor (CTA) sequence in HoxA. The CTA was necessary and sufficient for mitochondrial targeting. The affinity of phytochrome apoprotein to HoxA was 57,000‐fold higher than the affinity of the holoprotein, suggesting a “kiss‐and‐go” mechanism for chromophore loading and a function of mitochondria as assembly platforms for functional phytochrome. Hence, two alternative approaches for chromophore biosynthesis and insertion into phytochrome evolved in plants and fungi., Characterization of fungal heme oxygenases suggests a role for the mitochondrial outer membrane in chromophore production for light‐sensing phytochromes.
- Published
- 2021
- Full Text
- View/download PDF
44. High‐resolution serum proteome trajectories in COVID‐19 reveal patient‐specific seroconversion
- Author
-
Marie Luise Louiset, Philipp E. Geyer, Johannes B. Müller-Reif, Sebastian Virreira Winter, Daniel Teupser, Mathias Bruegel, Peter Eichhorn, Furkan M. Torun, Michael Weigand, Florian M Arend, Maximilian T. Strauss, Sophia Doll, Lesca M. Holdt, and Matthias Mann
- Subjects
Proteomics ,Medicine (General) ,Proteome ,immunoglobulins ,Disease ,QH426-470 ,Antibodies, Viral ,Article ,SARS‐CoV‐2 ,R5-920 ,biobanking ,Genetics ,Humans ,Seroconversion ,Innate immune system ,individual‐specific ,biology ,Aryldialkylphosphatase ,SARS-CoV-2 ,COVID-19 ,Articles ,PON1 ,Microbiology, Virology & Host Pathogen Interaction ,Immunology ,biology.protein ,biomarker ,Molecular Medicine ,Biomarker (medicine) ,Antibody ,Biomarkers - Abstract
A deeper understanding of COVID‐19 on human molecular pathophysiology is urgently needed as a foundation for the discovery of new biomarkers and therapeutic targets. Here we applied mass spectrometry (MS)‐based proteomics to measure serum proteomes of COVID‐19 patients and symptomatic, but PCR‐negative controls, in a time‐resolved manner. In 262 controls and 458 longitudinal samples of 31 patients, hospitalized for COVID‐19, a remarkable 26% of proteins changed significantly. Bioinformatics analyses revealed co‐regulated groups and shared biological functions. Proteins of the innate immune system such as CRP, SAA1, CD14, LBP, and LGALS3BP decreased early in the time course. Regulators of coagulation (APOH, FN1, HRG, KNG1, PLG) and lipid homeostasis (APOA1, APOC1, APOC2, APOC3, PON1) increased over the course of the disease. A global correlation map provides a system‐wide functional association between proteins, biological processes, and clinical chemistry parameters. Importantly, five SARS‐CoV‐2 immunoassays against antibodies revealed excellent correlations with an extensive range of immunoglobulin regions, which were quantified by MS‐based proteomics. The high‐resolution profile of all immunoglobulin regions showed individual‐specific differences and commonalities of potential pathophysiological relevance., A total of 720 proteomes from 458 longitudinal serum samples of hospitalized COVID‐19 cases and 262 symptomatic controls were measured. In‐depth analysis revealed regulation of innate immune and coagulation systems and individual‐specific trajectories of immunoglobulin regions.
- Published
- 2021
- Full Text
- View/download PDF
45. SARS-CoV-2 nucleocapsid suppresses host pyroptosis by blocking Gasdermin D cleavage
- Author
-
Dong Jiang, Shuo Wang, Fei Shao, Fangrui Zhu, Dou Yu, Jiangwen Ma, Weitao Li, Juan Ma, Min Zhao, Yan Qian, Yan Zhang, Xusheng Zhang, and Pengyan Xia
- Subjects
Inflammasomes ,THP-1 Cells ,viruses ,Interleukin-1beta ,nucleocapsid ,Biology ,medicine.disease_cause ,Cleavage (embryo) ,General Biochemistry, Genetics and Molecular Biology ,Monocytes ,Article ,SARS‐CoV‐2 ,Mice ,Immune system ,inflammasome ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Pyroptosis ,Animals ,Humans ,Secretion ,Receptor ,skin and connective tissue diseases ,Molecular Biology ,Coronavirus ,General Immunology and Microbiology ,SARS-CoV-2 ,General Neuroscience ,fungi ,Caspase 1 ,Intracellular Signaling Peptides and Proteins ,virus diseases ,COVID-19 ,Inflammasome ,Articles ,Phosphate-Binding Proteins ,Microbiology, Virology & Host Pathogen Interaction ,Cell biology ,body regions ,HEK293 Cells ,Host-Pathogen Interactions ,Spike Glycoprotein, Coronavirus ,Autophagy & Cell Death ,Angiotensin-Converting Enzyme 2 ,GSDMD ,Linker ,medicine.drug - Abstract
SARS‐CoV‐2 is an emerging coronavirus that causes dysfunctions in multiple human cells and tissues. Studies have looked at the entry of SARS‐CoV‐2 into host cells mediated by the viral spike protein and human receptor ACE2. However, less is known about the cellular immune responses triggered by SARS‐CoV‐2 viral proteins. Here, we show that the nucleocapsid of SARS‐CoV‐2 inhibits host pyroptosis by blocking Gasdermin D (GSDMD) cleavage. SARS‐CoV‐2‐infected monocytes show enhanced cellular interleukin‐1β (IL‐1β) expression, but reduced IL‐1β secretion. While SARS‐CoV‐2 infection promotes activation of the NLRP3 inflammasome and caspase‐1, GSDMD cleavage and pyroptosis are inhibited in infected human monocytes. SARS‐CoV‐2 nucleocapsid protein associates with GSDMD in cells and inhibits GSDMD cleavage in vitro and in vivo. The nucleocapsid binds the GSDMD linker region and hinders GSDMD processing by caspase‐1. These insights into how SARS‐CoV‐2 antagonizes cellular inflammatory responses may open new avenues for treating COVID‐19 in the future., Despite caspase‐1 and NLRP3 inflammasome activation, pyroptotic cell death of SARS‐CoV2‐infected human monocytes is prevented by cleavage site masking in the key effector GSDMD by a viral protein.
- Published
- 2021
46. Single‐cell transcriptomics reveals immune response of intestinal cell types to viral infection
- Author
-
Diana Ordoñez-Rueda, Theodore Alexandrov, Vladimir Benes, Steeve Boulant, Clara Serger, Camila Metz-Zumaran, Markus Mukenhirn, Malte Paulsen, Mohammed Shahraz, Megan L. Stanifer, Carmon Kee, Ronald Koschny, and Sergio Triana
- Subjects
Cell type ,Medicine (General) ,QH301-705.5 ,Viral pathogenesis ,Immunology ,Chromatin, Epigenetics, Genomics & Functional Genomics ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Article ,immune response ,Astrovirus ,03 medical and health sciences ,astrovirus ,0302 clinical medicine ,Immune system ,R5-920 ,Interferon ,medicine ,Humans ,Intestinal Mucosa ,Biology (General) ,organoids ,030304 developmental biology ,0303 health sciences ,General Immunology and Microbiology ,biology ,Cell growth ,Applied Mathematics ,Immunity ,Articles ,biology.organism_classification ,Intestinal epithelium ,Microbiology, Virology & Host Pathogen Interaction ,3. Good health ,Cell biology ,Intestines ,Computational Theory and Mathematics ,Virus Diseases ,intestinal epithelial cells ,General Agricultural and Biological Sciences ,Transcriptome ,single‐cell transcriptomics ,030217 neurology & neurosurgery ,Information Systems ,medicine.drug - Abstract
Human intestinal epithelial cells form a primary barrier protecting us from pathogens, yet only limited knowledge is available about individual contribution of each cell type to mounting an immune response against infection. Here, we developed a framework combining single‐cell RNA‐Seq and highly multiplex RNA FISH and applied it to human intestinal organoids infected with human astrovirus, a model human enteric virus. We found that interferon controls the infection and that astrovirus infects all major cell types and lineages and induces expression of the cell proliferation marker MKI67. Intriguingly, each intestinal epithelial cell lineage exhibits a unique basal expression of interferon‐stimulated genes and, upon astrovirus infection, undergoes an antiviral transcriptional reprogramming by upregulating distinct sets of interferon‐stimulated genes. These findings suggest that in the human intestinal epithelium, each cell lineage plays a unique role in resolving virus infection. Our framework is applicable to other organoids and viruses, opening new avenues to unravel roles of individual cell types in viral pathogenesis., Single‐cell sequencing and multiplex single‐molecule RNA FISH analyses of human astrovirus 1 (HAstV1)‐infected human intestinal organoids characterize viral tropism and unravel the cell lineage‐specific immune response to viral infection.
- Published
- 2021
47. Model-guided development of an evolutionarily stable yeast chassis
- Author
-
Isabel Rocha, Justyna Nocon, Filipa Pereira, Kiran Raosaheb Patil, Peter Kötter, Miguel Rocha, Paulo Maia, Britta Meyer, Dimitrios Konstantinidis, Paula Jouhten, H. Lopes, Eleni Kafkia, Universidade do Minho, Pereira, Filipa [0000-0002-0557-8480], Lopes, Helder [0000-0001-9563-3844], Maia, Paulo [0000-0002-0848-8683], Konstantinidis, Dimitrios [0000-0002-2134-6823], Kafkia, Eleni [0000-0001-9550-4487], Rocha, Isabel [0000-0001-9494-3410], Patil, Kiran R [0000-0002-6166-8640], and Apollo - University of Cambridge Repository
- Subjects
0106 biological sciences ,Medicine (General) ,GENE KNOCKOUT ,Succinic Acid ,Chassis cell ,EMBO21 ,01 natural sciences ,EMBO23 ,SACCHAROMYCES-CEREVISIAE ,Biology (General) ,adaptive laboratory evolution ,multi‐objective optimization ,0303 health sciences ,biology ,Applied Mathematics ,systems biology ,Articles ,Microbiology, Virology & Host Pathogen Interaction ,Flux balance analysis ,Computational Theory and Mathematics ,ESCHERICHIA-COLI ,ACID ,General Agricultural and Biological Sciences ,metabolic engineering ,Adaptive laboratory evolution ,Systems biology ,Life Sciences & Biomedicine ,Information Systems ,Biochemistry & Molecular Biology ,STRAIN ,Chassis ,QH301-705.5 ,Citric Acid Cycle ,Saccharomyces cerevisiae ,Computational biology ,EMBO41 ,Malate dehydrogenase ,General Biochemistry, Genetics and Molecular Biology ,Article ,Metabolic engineering ,03 medical and health sciences ,R5-920 ,010608 biotechnology ,Metabolomics ,OPTIMIZATION ,030304 developmental biology ,chassis cell ,Science & Technology ,General Immunology and Microbiology ,Robustness (evolution) ,biology.organism_classification ,FRAMEWORK ,Yeast ,PROTEOME ,Multi-objective optimization ,Metabolism ,multi-objective optimization ,DISCOVERY ,Synthetic Biology & Biotechnology ,Flux (metabolism) ,GENERATION - Abstract
First-principle metabolic modelling holds potential for designing microbial chassis that are resilient against phenotype reversal due to adaptive mutations. Yet, the theory of model-based chassis design has rarely been put to rigorous experimental test. Here, we report the development of Saccharomyces cerevisiae chassis strains for dicarboxylic acid production using genome-scale metabolic modelling. The chassis strains, albeit geared for higher flux towards succinate, fumarate and malate, do not appreciably secrete these metabolites. As predicted by the model, introducing product-specific TCA cycle disruptions resulted in the secretion of the corresponding acid. Adaptive laboratory evolution further improved production of succinate and fumarate, demonstrating the evolutionary robustness of the engineered cells. In the case of malate, multi-omics analysis revealed a flux bypass at peroxisomal malate dehydrogenase that was missing in the yeast metabolic model. In all three cases, flux balance analysis integrating transcriptomics, proteomics and metabolomics data confirmed the flux re-routing predicted by the model. Taken together, our modelling and experimental results have implications for the computer-aided design of microbial cell factories., We would like to acknowledge the support of R. Mattel and F. Stein from the Proteomics Core Facility and the Genomics Core Facility at the European Molecular Biology Laboratory (EMBL Heidelberg, Germany). This study was supported by national funds through FCT/MCTES (Portugal, Ref. ERA-IB-2/0003/2013) and BMBF (Germany, Grant number: 031A343A, Ref. ERA-IB-2/0003/2013). The Portuguese Foundation for Science and Technology (FCT) supported HL through grant ref. PD/BD/52336/2013. FCT also supported this study under the scope of the strategic funding of UID/BIO/04469/2013 unit and COMPETE 2020 (POCI-01-0145-FEDER-006684) and through the Project RECI/BBB-EBI/0179/2012 (FCOMP-01-0124-FEDER-027462). Open Access funding enabled and organized by Projekt DEAL., info:eu-repo/semantics/publishedVersion
- Published
- 2021
48. TMPRSS2 expression dictates the entry route used by SARS‐CoV‐2 to infect host cells
- Author
-
Patricio Doldan, Zina M Uckeley, Steeve Boulant, Megan L. Stanifer, Pierre-Yves Lozach, Jana Koch, Infections Virales et Pathologie Comparée - UMR 754 (IVPC), École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Claude Bernard Lyon 1 (UCBL), and Université de Lyon-Université de Lyon-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)
- Subjects
viruses ,Cathepsin L ,[SDV.BC.BC]Life Sciences [q-bio]/Cellular Biology/Subcellular Processes [q-bio.SC] ,virus entry ,medicine.disease_cause ,urologic and male genital diseases ,SARS‐CoV‐2 ,Tissue culture ,Chlorocebus aethiops ,Membrane & Intracellular Transport ,skin and connective tissue diseases ,Coronavirus ,0303 health sciences ,biology ,General Neuroscience ,030302 biochemistry & molecular biology ,Serine Endopeptidases ,virus diseases ,Articles ,Hydrogen-Ion Concentration ,Microbiology, Virology & Host Pathogen Interaction ,Endocytosis ,3. Good health ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,Signal Transduction ,Lung injury ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Article ,03 medical and health sciences ,Viral entry ,COVID‐19 ,medicine ,Animals ,Humans ,Molecular Biology ,Vero Cells ,030304 developmental biology ,General Immunology and Microbiology ,Host Microbial Interactions ,SARS-CoV-2 ,fungi ,COVID-19 ,protease ,Virus Internalization ,Virology ,Proteolysis ,Tissue tropism ,Vero cell ,biology.protein ,Caco-2 Cells - Abstract
SARS‐CoV‐2 is a newly emerged coronavirus that caused the global COVID‐19 outbreak in early 2020. COVID‐19 is primarily associated with lung injury, but many other clinical symptoms such as loss of smell and taste demonstrated broad tissue tropism of the virus. Early SARS‐CoV‐2–host cell interactions and entry mechanisms remain poorly understood. Investigating SARS‐CoV‐2 infection in tissue culture, we found that the protease TMPRSS2 determines the entry pathway used by the virus. In the presence of TMPRSS2, the proteolytic process of SARS‐CoV‐2 was completed at the plasma membrane, and the virus rapidly entered the cells within 10 min in a pH‐independent manner. When target cells lacked TMPRSS2 expression, the virus was endocytosed and sorted into endolysosomes, from which SARS‐CoV‐2 entered the cytosol via acid‐activated cathepsin L protease 40–60 min post‐infection. Overexpression of TMPRSS2 in non‐TMPRSS2 expressing cells abolished the dependence of infection on the cathepsin L pathway and restored sensitivity to the TMPRSS2 inhibitors. Together, our results indicate that SARS‐CoV‐2 infects cells through distinct, mutually exclusive entry routes and highlight the importance of TMPRSS2 for SARS‐CoV‐2 sorting into either pathway., SARS‐CoV‐2 infects cells through mutually‐exclusive, pH‐independent versus pH‐dependent entry routes, whose distinction is governed by processing protease expression in target cells.
- Published
- 2021
- Full Text
- View/download PDF
49. A functional assay for serum detection of antibodies against SARS‐CoV‐2 nucleoprotein
- Author
-
Tyler Rhinesmith, Anna Albecka, Leo C. James, Marina Vaysburd, Dean Clift, David M Favara, Sarah L Caddy, Helen Baxendale, Albecka, Anna [0000-0002-3672-5498], Clift, Dean [0000-0001-8141-7817], Caddy, Sarah L [0000-0002-9790-7420], James, Leo C [0000-0003-2131-0334], and Apollo - University of Cambridge Repository
- Subjects
Resource ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Antigen presentation ,Fc receptor ,Antibodies, Viral ,EMBO23 ,General Biochemistry, Genetics and Molecular Biology ,Neutralization ,SARS‐CoV‐2 ,Immune system ,antibodies ,Humans ,Molecular Biology ,nucleoprotein ,General Immunology and Microbiology ,biology ,SARS-CoV-2 ,General Neuroscience ,COVID-19 ,neutralization ,Virology ,Antibodies, Neutralizing ,Resources ,Microbiology, Virology & Host Pathogen Interaction ,Nucleoprotein ,Cytosol ,Nucleoproteins ,biology.protein ,Antibody ,TRIM21 - Abstract
The humoral immune response to SARS‐CoV‐2 results in antibodies against spike (S) and nucleoprotein (N). However, whilst there are widely available neutralization assays for S antibodies, there is no assay for N‐antibody activity. Here, we present a simple in vitro method called EDNA (electroporated‐antibody‐dependent neutralization assay) that provides a quantitative measure of N‐antibody activity in unpurified serum from SARS‐CoV‐2 convalescents. We show that N antibodies neutralize SARS‐CoV‐2 intracellularly and cell‐autonomously but require the cytosolic Fc receptor TRIM21. Using EDNA, we show that low N‐antibody titres can be neutralizing, whilst some convalescents possess serum with high titres but weak activity. N‐antibody and N‐specific T‐cell activity correlates within individuals, suggesting N antibodies may protect against SARS‐CoV‐2 by promoting antigen presentation. This work highlights the potential benefits of N‐based vaccines and provides an in vitro assay to allow the antibodies they induce to be tested., A new in vitro assay, EDNA, measures neutralizing activities of patient antibodies against coronaviral N protein, complementing available methods for evaluating antiviral activities of anti‐spike (S) protein antibodies.
- Published
- 2021
50. Attenuation of clinical and immunological outcomes during SARS-CoV-2 infection by ivermectin
- Author
-
de Melo, Guilherme Dias, Lazarini, Françoise, Larrous, Florence, Feige, Lena, Kornobis, Etienne, Levallois, Sylvain, Marchio, Agnès, Kergoat, Lauriane, Hardy, David, Cokelaer, Thomas, Pineau, Pascal, Lecuit, Marc, Lledo, Pierre-Marie, Changeux, Jean‐pierre, Bourhy, Hervé, Lyssavirus, épidémiologie et neuropathologie - Lyssavirus Epidemiology and Neuropathology, Institut Pasteur [Paris], Perception et Mémoire / Perception and Memory, Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Biomics (plateforme technologique), Hub Bioinformatique et Biostatistique - Bioinformatics and Biostatistics HUB, Biologie des Infections - Biology of Infection, Institut Pasteur [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Organisation Nucléaire et Oncogenèse / Nuclear Organization and Oncogenesis, Neuropathologie expérimentale / Experimental neuropathology, Institut Pasteur [Paris]-Université de Paris (UP), Service des Maladies infectieuses et tropicales [CHU Necker], CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Collège de France (CdF (institution)), Département de Neuroscience - Department of Neuroscience, This work was supported by Institut Pasteur TASK FORCE SARS COV2 (NicoSARS, NeuroCovid and Cov-DROP projects) and received help from the European Union’s Horizon 2020 Framework Programme for Research and Innovation under Specific Grant Agreement No.945539 (Human Brain Project SGA3). We thank Elodie Turc and Laure Lemée, Biomics Platform, C2RT,Institut Pasteur, Paris, France, supported by France Génomique (ANR-10-INBS-09-09), IBISA and theIllumina COVID-19 Projects’offer., ANR-10-INBS-0009,France-Génomique,Organisation et montée en puissance d'une Infrastructure Nationale de Génomique(2010), European Project: 945539,HBP, Institut Pasteur [Paris] (IP), Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Pasteur [Paris] (IP)-Université Paris Cité (UPCité), European Project: 945539,H2020,H2020-SGA-FETFLAG-HBP-2019,HBP SGA3(2020), Cova Rodrigues, Ana, Organisation et montée en puissance d'une Infrastructure Nationale de Génomique - - France-Génomique2010 - ANR-10-INBS-0009 - INBS - VALID, and Human Brain Project Specific Grant Agreement 3 - HBP SGA3 - - H20202020-01-01 - 2023-09-30 - 945539 - VALID
- Subjects
[SDV.MP.VIR] Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,Ivermectin ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,SARS-CoV-2 ,[SDV]Life Sciences [q-bio] ,viral infections ,Immunology ,coronavirus ,COVID-19 ,Articles ,Article ,SARS‐CoV‐2 ,Microbiology, Virology & Host Pathogen Interaction ,[SDV] Life Sciences [q-bio] ,[SDV.MP]Life Sciences [q-bio]/Microbiology and Parasitology ,inflammation ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Animals ,Humans ,[SDV.MP] Life Sciences [q-bio]/Microbiology and Parasitology ,Lung ,Pandemics - Abstract
The devastating pandemic due to SARS‐CoV‐2 and the emergence of antigenic variants that jeopardize the efficacy of current vaccines create an urgent need for a comprehensive understanding of the pathophysiology of COVID‐19, including the contribution of inflammation to disease. It also warrants for the search of immunomodulatory drugs that could improve disease outcome. Here, we show that standard doses of ivermectin (IVM), an anti‐parasitic drug with potential immunomodulatory activities through the cholinergic anti‐inflammatory pathway, prevent clinical deterioration, reduce olfactory deficit, and limit the inflammation of the upper and lower respiratory tracts in SARS‐CoV‐2‐infected hamsters. Whereas it has no effect on viral load in the airways of infected animals, transcriptomic analyses of infected lungs reveal that IVM dampens type I interferon responses and modulates several other inflammatory pathways. In particular, IVM dramatically reduces the Il‐6/Il‐10 ratio in lung tissue and promotes macrophage M2 polarization, which might account for the more favorable clinical presentation of IVM‐treated animals. Altogether, this study supports the use of immunomodulatory drugs such as IVM, to improve the clinical condition of SARS‐CoV‐2‐infected patients., COVID‐19, caused by SARS‐CoV‐2, induces airways and pulmonary symptoms, and in severe cases can lead to respiratory distress and death. This study shows that the modulation of the host's inflammatory response using ivermectin as a repurposed drug, independently of the viral load, strongly diminished the clinical score and severity of the disease (including anosmia) observed in SARS‐CoV‐2‐infected golden hamsters. This study brings the proof‐of‐concept that a chemical therapy using ivermectin can preserve the clinical condition by modulating the inflammatory response, even without antiviral activity.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.