51
|
Chatterjee R, Chaudhuri D, Setty SRG, Chakravortty D. Deceiving the big eaters: Salmonella Typhimurium SopB subverts host cell xenophagy in macrophages via dual mechanisms. Microbes Infect 2023; 25:105128. [PMID: 37019426 DOI: 10.1016/j.micinf.2023.105128] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023]
Abstract
Salmonella, a stealthy facultative intracellular pathogen, utilises an array of host immune evasion strategies. This facilitates successful survival via replicative niche establishment in otherwise hostile environments such as macrophages. Salmonella survives in and utilises macrophages for effective dissemination, ultimately leading to systemic infection. Bacterial xenophagy or macro-autophagy is an important host defense mechanism in macrophages. Here, we report for the first time that the Salmonella pathogenicity island-1 (SPI-1) effector SopB is involved in subverting host autophagy via dual mechanisms. SopB is a phosphoinositide phosphatase capable of altering the phosphoinositide dynamics of the host cell. Here, we demonstrate that SopB mediates escape from autophagy by inhibiting the terminal fusion of Salmonella-containing vacuoles (SCVs) with lysosomes and/or autophagosomes. We also report that SopB downregulates overall lysosomal biogenesis by modulating the Akt-transcription factor EB (TFEB) axis via restricting the latter's nuclear localisation. TFEB is a master regulator of lysosomal biogenesis and autophagy. This reduces the overall lysosome content inside host macrophages, further facilitating the survival of Salmonella in macrophages and systemic dissemination of Salmonella.
Collapse
Affiliation(s)
- Ritika Chatterjee
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India
| | - Debalina Chaudhuri
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India
| | - Subba Rao Gangi Setty
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India; Indian Institute of Science Research and Education, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
52
|
Xu D, Jiang W, Wu L, Gaudet RG, Park ES, Su M, Cheppali SK, Cheemarla NR, Kumar P, Uchil PD, Grover JR, Foxman EF, Brown CM, Stansfeld PJ, Bewersdorf J, Mothes W, Karatekin E, Wilen CB, MacMicking JD. PLSCR1 is a cell-autonomous defence factor against SARS-CoV-2 infection. Nature 2023; 619:819-827. [PMID: 37438530 PMCID: PMC10371867 DOI: 10.1038/s41586-023-06322-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 06/14/2023] [Indexed: 07/14/2023]
Abstract
Understanding protective immunity to COVID-19 facilitates preparedness for future pandemics and combats new SARS-CoV-2 variants emerging in the human population. Neutralizing antibodies have been widely studied; however, on the basis of large-scale exome sequencing of protected versus severely ill patients with COVID-19, local cell-autonomous defence is also crucial1-4. Here we identify phospholipid scramblase 1 (PLSCR1) as a potent cell-autonomous restriction factor against live SARS-CoV-2 infection in parallel genome-wide CRISPR-Cas9 screens of human lung epithelia and hepatocytes before and after stimulation with interferon-γ (IFNγ). IFNγ-induced PLSCR1 not only restricted SARS-CoV-2 USA-WA1/2020, but was also effective against the Delta B.1.617.2 and Omicron BA.1 lineages. Its robust activity extended to other highly pathogenic coronaviruses, was functionally conserved in bats and mice, and interfered with the uptake of SARS-CoV-2 in both the endocytic and the TMPRSS2-dependent fusion routes. Whole-cell 4Pi single-molecule switching nanoscopy together with bipartite nano-reporter assays found that PLSCR1 directly targeted SARS-CoV-2-containing vesicles to prevent spike-mediated fusion and viral escape. A PLSCR1 C-terminal β-barrel domain-but not lipid scramblase activity-was essential for this fusogenic blockade. Our mechanistic studies, together with reports that COVID-associated PLSCR1 mutations are found in some susceptible people3,4, identify an anti-coronavirus protein that interferes at a late entry step before viral RNA is released into the host-cell cytosol.
Collapse
Affiliation(s)
- Dijin Xu
- Howard Hughes Medical Institute, New Haven, CT, USA
- Yale Systems Biology Institute, West Haven, CT, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Weiqian Jiang
- Howard Hughes Medical Institute, New Haven, CT, USA
- Yale Systems Biology Institute, West Haven, CT, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Lizhen Wu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ryan G Gaudet
- Howard Hughes Medical Institute, New Haven, CT, USA
- Yale Systems Biology Institute, West Haven, CT, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Eui-Soon Park
- Howard Hughes Medical Institute, New Haven, CT, USA
- Yale Systems Biology Institute, West Haven, CT, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Maohan Su
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Sudheer Kumar Cheppali
- Yale Nanobiology Institute, West Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Nagarjuna R Cheemarla
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Pradeep Kumar
- Howard Hughes Medical Institute, New Haven, CT, USA
- Yale Systems Biology Institute, West Haven, CT, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Pradeep D Uchil
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Jonathan R Grover
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Ellen F Foxman
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Chelsea M Brown
- School of Life Sciences and Department of Chemistry, University of Warwick, Coventry, UK
| | - Phillip J Stansfeld
- School of Life Sciences and Department of Chemistry, University of Warwick, Coventry, UK
| | - Joerg Bewersdorf
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Walther Mothes
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Erdem Karatekin
- Yale Nanobiology Institute, West Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Saints-Pères Paris Institute for the Neurosciences, Université de Paris, Centre National de la Recherche Scientifique UMR 8003, Paris, France
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Craig B Wilen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - John D MacMicking
- Howard Hughes Medical Institute, New Haven, CT, USA.
- Yale Systems Biology Institute, West Haven, CT, USA.
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
53
|
Roy S, Wang B, Tian Y, Yin Q. Crystal structures reveal nucleotide-induced conformational changes in G motifs and distal regions in guanylate-binding protein 2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.28.546747. [PMID: 37425906 PMCID: PMC10327160 DOI: 10.1101/2023.06.28.546747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Guanylate-binding proteins (GBPs) are interferon-inducible GTPases that confer protective immunity against a variety of intracellular pathogens including bacteria, viruses, and protozoan parasites. GBP2 is one of the two highly inducible GBPs, yet the precise mechanisms underlying the activation and regulation of GBP2, in particular the nucleotide-induced conformational changes in GBP2, remain poorly understood. In this study, we elucidate the structural dynamics of GBP2 upon nucleotide binding through crystallographic analysis. GBP2 dimerizes upon GTP hydrolysis and returns to monomer state once GTP is hydrolyzed to GDP. By determining the crystal structures of GBP2 G domain (GBP2GD) in complex with GDP and nucleotide-free full-length GBP2, we unveil distinct conformational states adopted by the nucleotide-binding pocket and distal regions of the protein. Our findings demonstrate that the binding of GDP induces a distinct closed conformation both in the G motifs and the distal regions in the G domain. The conformational changes in the G domain are further transmitted to the C-terminal helical domain, leading to large-scale conformational rearrangements. Through comparative analysis, we identify subtle but critical differences in the nucleotide-bound states of GBP2, providing insights into the molecular basis of its dimer-monomer transition and enzymatic activity. Overall, our study expands the understanding of the nucleotide-induced conformational changes in GBP2, shedding light on the structural dynamics governing its functional versatility. These findings pave the way for future investigations aimed at elucidating the precise molecular mechanisms underlying GBP2's role in the immune response and may facilitate the development of targeted therapeutic strategies against intracellular pathogens.
Collapse
Affiliation(s)
- Sayantan Roy
- Department of Biological Science, Florida State University
| | - Bing Wang
- Department of Biological Science, Florida State University
| | - Yuan Tian
- Department of Biological Science, Florida State University
| | - Qian Yin
- Department of Biological Science, Florida State University
- Institute of Molecular Biophysics, Florida State University
| |
Collapse
|
54
|
Graham ML, Li M, Gong AY, Deng S, Jin K, Wang S, Chen XM. Cryptosporidium parvum hijacks a host's long noncoding RNA U90926 to evade intestinal epithelial cell-autonomous antiparasitic defense. Front Immunol 2023; 14:1205468. [PMID: 37346046 PMCID: PMC10280636 DOI: 10.3389/fimmu.2023.1205468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
Cryptosporidium is a zoonotic apicomplexan parasite that infects the gastrointestinal epithelium and other mucosal surfaces in humans. It is an important opportunistic pathogen in AIDS patients and a leading cause of infectious diarrhea and diarrheal-related death in children worldwide. The intestinal epithelial cells provide the first line of defense against Cryptosporidium infection and play a central role in activating and regulating the host's antiparasitic response. Increasing evidence suggests that long noncoding RNAs (lncRNAs) participate in host-pathogen interactions and play a regulatory role in the pathogenesis of diseases but the underlying molecular mechanisms are not fully understood. We previously identified a panel of host lncRNAs that are upregulated in murine intestinal epithelial cells following Cryptosporidium infection, including U90926. We demonstrate here that U90926 is acting in a pro-parasitic manner in regulating intestinal epithelial cell-autonomous antiparasitic defense. Inhibition of U90926 resulted in a decreased infection burden of the parasite while overexpression of U90926 showed an increase in infection burden in cultured murine intestinal epithelial cells. Induction of U90926 suppressed transcription of epithelial defense genes involved in controlling Cryptosporidium infection through epigenetic mechanisms. Specifically, transcription of Aebp1, which encodes the Aebp1 protein, a potent modulator of inflammation and NF-κB signaling, was suppressed by U90926. Gain- or loss-of-function of Aebp1 in the host's epithelial cells caused reciprocal alterations in the infection burden of the parasite. Interestingly, Cryptosporidium carries the Cryptosporidium virus 1 (CSpV1), a double-stranded (ds) RNA virus coding two dsRNA fragments, CSpV1-dsRdRp and CSpV1-dsCA. Both CSpV1-dsRdRp and CSpV1-dsCA can be delivered into infected cells as previously reported. We found that cells transfected with in vitro transcribed CSpV1-dsCA or CSpV1-dsRdRp displayed an increased level of U90926, suggesting that CSpV1 is involved in the upregulation of U90926 during Cryptosporidium infection. Our study highlights a new strategy by Cryptosporidium to hijack a host lncRNA to suppress epithelial cell-autonomous antiparasitic defense and allow for a robust infection.
Collapse
Affiliation(s)
- Marion L. Graham
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, United States
| | - Min Li
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, NE, United States
| | - Ai-Yu Gong
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, United States
| | - Silu Deng
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, United States
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, NE, United States
| | - Kehua Jin
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, United States
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Shuhong Wang
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, United States
| | - Xian-Ming Chen
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
55
|
Abstract
Immunity to infection has been extensively studied in humans and mice bearing naturally occurring or experimentally introduced germline mutations. Mouse studies are sometimes neglected by human immunologists, on the basis that mice are not humans and the infections studied are experimental and not natural. Conversely, human studies are sometimes neglected by mouse immunologists, on the basis of the uncontrolled conditions of study and small numbers of patients. However, both sides would agree that the infectious phenotypes of patients with inborn errors of immunity often differ from those of the corresponding mutant mice. Why is that? We argue that this important question is best addressed by revisiting and reinterpreting the findings of both mouse and human studies from a genetic perspective. Greater caution is required for reverse-genetics studies than for forward-genetics studies, but genetic analysis is sufficiently strong to define the studies likely to stand the test of time. Genetically robust mouse and human studies can provide invaluable complementary insights into the mechanisms of immunity to infection common and specific to these two species.
Collapse
Affiliation(s)
- Philippe Gros
- McGill University Research Center on Complex Traits, Department of Biochemistry, and Department of Human Genetics, McGill University, Montréal, Québec, Canada;
| | - Jean-Laurent Casanova
- Howard Hughes Medical Institute and St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA;
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, and University of Paris Cité, Imagine Institute and Necker Hospital for Sick Children, Paris, France
| |
Collapse
|
56
|
Dickinson M, Kutsch M, Sistemich L, Hernandez D, Piro A, Needham D, Lesser C, Herrmann C, Coers J. LPS-aggregating proteins GBP1 and GBP2 are each sufficient to enhance caspase-4 activation both in cellulo and in vitro. Proc Natl Acad Sci U S A 2023; 120:e2216028120. [PMID: 37023136 PMCID: PMC10104521 DOI: 10.1073/pnas.2216028120] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 02/26/2023] [Indexed: 04/07/2023] Open
Abstract
The gamma-interferon (IFNγ)-inducible guanylate-binding proteins (GBPs) promote host defense against gram-negative cytosolic bacteria in part through the induction of an inflammatory cell death pathway called pyroptosis. To activate pyroptosis, GBPs facilitate sensing of the gram-negative bacterial outer membrane component lipopolysaccharide (LPS) by the noncanonical caspase-4 inflammasome. There are seven human GBP paralogs, and it is unclear how each GBP contributes to LPS sensing and pyroptosis induction. GBP1 forms a multimeric microcapsule on the surface of cytosolic bacteria through direct interactions with LPS. The GBP1 microcapsule recruits caspase-4 to bacteria, a process deemed essential for caspase-4 activation. In contrast to GBP1, closely related paralog GBP2 is unable to bind bacteria on its own but requires GBP1 for direct bacterial binding. Unexpectedly, we find that GBP2 overexpression can restore gram-negative-induced pyroptosis in GBP1KO cells, without GBP2 binding to the bacterial surface. A mutant of GBP1 that lacks the triple arginine motif required for microcapsule formation also rescues pyroptosis in GBP1KO cells, showing that binding to bacteria is dispensable for GBPs to promote pyroptosis. Instead, we find that GBP2, like GBP1, directly binds and aggregates "free" LPS through protein polymerization. We demonstrate that supplementation of either recombinant polymerized GBP1 or GBP2 to an in vitro reaction is sufficient to enhance LPS-induced caspase-4 activation. This provides a revised mechanistic framework for noncanonical inflammasome activation where GBP1 or GBP2 assembles cytosol-contaminating LPS into a protein-LPS interface for caspase-4 activation as part of a coordinated host response to gram-negative bacterial infections.
Collapse
Affiliation(s)
- Mary S. Dickinson
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Miriam Kutsch
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Linda Sistemich
- Department of Physical Chemistry I, Ruhr-University Bochum, 44801Bochum, Germany
| | - Dulcemaria Hernandez
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Anthony S. Piro
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - David Needham
- Department of Mechanical Engineering and Material Science, Duke University, Durham, NC27708
| | - Cammie F. Lesser
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA02139
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Christian Herrmann
- Department of Physical Chemistry I, Ruhr-University Bochum, 44801Bochum, Germany
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
- Department of Immunology, Duke University Medical Center, Durham, NC27710
| |
Collapse
|
57
|
Lobato-Márquez D, Conesa JJ, López-Jiménez AT, Divine ME, Pruneda JN, Mostowy S. Septins and K63 ubiquitin chains are present in separate bacterial microdomains during autophagy of entrapped Shigella. J Cell Sci 2023; 136:jcs261139. [PMID: 36939083 PMCID: PMC10264824 DOI: 10.1242/jcs.261139] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/21/2023] Open
Abstract
During host cell invasion, Shigella escapes to the cytosol and polymerizes actin for cell-to-cell spread. To restrict cell-to-cell spread, host cells employ cell-autonomous immune responses including antibacterial autophagy and septin cage entrapment. How septins interact with the autophagy process to target Shigella for destruction is poorly understood. Here, we employed a correlative light and cryo-soft X-ray tomography (cryo-SXT) pipeline to study Shigella septin cage entrapment in its near-native state. Quantitative cryo-SXT showed that Shigella fragments mitochondria and enabled visualization of X-ray-dense structures (∼30 nm resolution) surrounding Shigella entrapped in septin cages. Using Airyscan confocal microscopy, we observed lysine 63 (K63)-linked ubiquitin chains decorating septin-cage-entrapped Shigella. Remarkably, septins and K63 chains are present in separate bacterial microdomains, indicating they are recruited separately during antibacterial autophagy. Cryo-SXT and live-cell imaging revealed an interaction between septins and LC3B-positive membranes during autophagy of Shigella. Together, these findings demonstrate how septin-caged Shigella are targeted for autophagy and provide fundamental insights into autophagy-cytoskeleton interactions.
Collapse
Affiliation(s)
- Damián Lobato-Márquez
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - José Javier Conesa
- MISTRAL beamline, ALBA Synchrotron Light Source, Cerdanyola del Vallès, 08290 Barcelona, Spain
| | - Ana Teresa López-Jiménez
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Michael E. Divine
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jonathan N. Pruneda
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| |
Collapse
|
58
|
Singh G, Kumar A. Japanese Encephalitis Virus Infection Causes an Imbalance in the Activation of Mitochondrial Fusion/Fission Genes and Triggers the Activation of NOX2-mediated Oxidative Stress and Neuronal Cell Death. Neurochem Res 2023; 48:2196-2205. [PMID: 36856962 DOI: 10.1007/s11064-023-03898-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/05/2023] [Accepted: 02/19/2023] [Indexed: 03/02/2023]
Abstract
Mitochondria dysfunction may be an important contributor to Japanese encephalitis (JE) viral infection disease pathogenesis. In the current study, we define whether changes in mitochondrial DNA copy number (which is the biomarker for mitochondrial function) and alteration in mitochondria dynamics (fusion and fission) contribute to the pathology of the JE virus in vivo mice model. We found decreased mitochondria copy number, reduced activation of mitochondrial fission (FIS1/DRP1), and increased activation of mitochondrial fusion (MFN1/MFN2/OPA1) genes that are associated with increased NOX2-mediated ROS generation and neuronal cell death following JE virus infection. Furthermore, we found that antioxidant glutathione level decreases. In summary, the following study demonstrates that JE viral infection causes an imbalance in mitochondrial fission/fusion gene activation and promotes NOX2-mediated oxidative stress and cell death, suggesting that intervention in mitochondrial dynamics might be a potential therapeutic strategy for combating oxidative stress and inflammatory process in JE viral infection.
Collapse
Affiliation(s)
- Gajendra Singh
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, 226014, India
| | - Alok Kumar
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, 226014, India.
| |
Collapse
|
59
|
Olive AJ, Smith CM, Baer CE, Coers J, Sassetti CM. Mycobacterium tuberculosis Evasion of Guanylate Binding Protein-Mediated Host Defense in Mice Requires the ESX1 Secretion System. Int J Mol Sci 2023; 24:2861. [PMID: 36769182 PMCID: PMC9917499 DOI: 10.3390/ijms24032861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Cell-intrinsic immune mechanisms control intracellular pathogens that infect eukaryotes. The intracellular pathogen Mycobacterium tuberculosis (Mtb) evolved to withstand cell-autonomous immunity to cause persistent infections and disease. A potent inducer of cell-autonomous immunity is the lymphocyte-derived cytokine IFNγ. While the production of IFNγ by T cells is essential to protect against Mtb, it is not capable of fully eradicating Mtb infection. This suggests that Mtb evades a subset of IFNγ-mediated antimicrobial responses, yet what mechanisms Mtb resists remains unclear. The IFNγ-inducible Guanylate binding proteins (GBPs) are key host defense proteins able to control infections with intracellular pathogens. GBPs were previously shown to directly restrict Mycobacterium bovis BCG yet their role during Mtb infection has remained unknown. Here, we examine the importance of a cluster of five GBPs on mouse chromosome 3 in controlling Mycobacterial infection. While M. bovis BCG is directly restricted by GBPs, we find that the GBPs on chromosome 3 do not contribute to the control of Mtb replication or the associated host response to infection. The differential effects of GBPs during Mtb versus M. bovis BCG infection is at least partially explained by the absence of the ESX1 secretion system from M. bovis BCG, since Mtb mutants lacking the ESX1 secretion system become similarly susceptible to GBP-mediated immune defense. Therefore, this specific genetic interaction between the murine host and Mycobacteria reveals a novel function for the ESX1 virulence system in the evasion of GBP-mediated immunity.
Collapse
Affiliation(s)
- Andrew J. Olive
- Department of Microbiology & Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Clare M. Smith
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 22710, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Christina E. Baer
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01650, USA
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 22710, USA
- Department of Immunology, Duke University Medical Center, Durham, NC 22710, USA
| | - Christopher M. Sassetti
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01650, USA
| |
Collapse
|
60
|
Panigaj M, Skelly E, Beasock D, Marriott I, Johnson MB, Salotti J, Afonin KA. Therapeutic immunomodulation by rationally designed nucleic acids and nucleic acid nanoparticles. Front Immunol 2023; 14:1053550. [PMID: 36798121 PMCID: PMC9927404 DOI: 10.3389/fimmu.2023.1053550] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 01/18/2023] [Indexed: 02/01/2023] Open
Abstract
The immune system has evolved to defend organisms against exogenous threats such as viruses, bacteria, fungi, and parasites by distinguishing between "self" and "non-self". In addition, it guards us against other diseases, such as cancer, by detecting and responding to transformed and senescent cells. However, for survival and propagation, the altered cells and invading pathogens often employ a wide range of mechanisms to avoid, inhibit, or manipulate the immunorecognition. As such, the development of new modes of therapeutic intervention to augment protective and prevent harmful immune responses is desirable. Nucleic acids are biopolymers essential for all forms of life and, therefore, delineating the complex defensive mechanisms developed against non-self nucleic acids can offer an exciting avenue for future biomedicine. Nucleic acid technologies have already established numerous approaches in therapy and biotechnology; recently, rationally designed nucleic acids nanoparticles (NANPs) with regulated physiochemical properties and biological activities has expanded our repertoire of therapeutic options. When compared to conventional therapeutic nucleic acids (TNAs), NANP technologies can be rendered more beneficial for synchronized delivery of multiple TNAs with defined stabilities, immunological profiles, and therapeutic functions. This review highlights several recent advances and possible future directions of TNA and NANP technologies that are under development for controlled immunomodulation.
Collapse
Affiliation(s)
- Martin Panigaj
- Nanoscale Science Program, Department of Chemistry, The University of North Carolina at Charlotte, Charlotte, NC, United States
- Institute of Biology & Ecology, Faculty of Science, Pavol Jozef Safarik University in Kosice, Kosice, Slovakia
| | - Elizabeth Skelly
- Nanoscale Science Program, Department of Chemistry, The University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Damian Beasock
- Nanoscale Science Program, Department of Chemistry, The University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Ian Marriott
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - M. Brittany Johnson
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Jacqueline Salotti
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Kirill A. Afonin
- Nanoscale Science Program, Department of Chemistry, The University of North Carolina at Charlotte, Charlotte, NC, United States
| |
Collapse
|
61
|
Abstract
Cellular homeostasis requires the swift and specific removal of damaged material. Selective autophagy represents a major pathway for the degradation of such cargo material. This is achieved by the sequestration of the cargo within double-membrane vesicles termed autophagosomes, which form de novo around the cargo and subsequently deliver their content to lysosomes for degradation. The importance of selective autophagy is exemplified by the various neurodegenerative diseases associated with defects in this pathway, including Parkinson's disease, amyotrophic lateral sclerosis, and frontotemporal dementia. It has become evident that cargo receptors are acting as Swiss army knives in selective autophagy by recognizing the cargo, orchestrating the recruitment of the machinery for autophagosome biogenesis, and closely aligning the membrane with the cargo. Furthermore, cargo receptors sequester ubiquitinated proteins into larger condensates upstream of autophagy induction. Here, we review recent insights into the mechanisms of action of cargo receptors in selective autophagy by focusing on the roles of sequestosome-like cargo receptors in the degradation of misfolded, ubiquitinated proteins and damaged mitochondria. We also highlight at which steps defects in their function result in the accumulation of harmful material and how this knowledge may guide the design of future therapies.
Collapse
Affiliation(s)
- Elias Adriaenssens
- Max Perutz Labs, Vienna BioCenter, University of Vienna, Dr. Bohr-Gasse 9, 1030 Vienna, Austria; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| | - Luca Ferrari
- Max Perutz Labs, Vienna BioCenter, University of Vienna, Dr. Bohr-Gasse 9, 1030 Vienna, Austria.
| | - Sascha Martens
- Max Perutz Labs, Vienna BioCenter, University of Vienna, Dr. Bohr-Gasse 9, 1030 Vienna, Austria; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| |
Collapse
|
62
|
Macrophage NFATC2 mediates angiogenic signaling during mycobacterial infection. Cell Rep 2022; 41:111817. [PMID: 36516756 PMCID: PMC9880963 DOI: 10.1016/j.celrep.2022.111817] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/05/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
During mycobacterial infections, pathogenic mycobacteria manipulate both host immune and stromal cells to establish and maintain a productive infection. In humans, non-human primates, and zebrafish models of infection, pathogenic mycobacteria produce and modify the specialized lipid trehalose 6,6'-dimycolate (TDM) in the bacterial cell envelope to drive host angiogenesis toward the site of forming granulomas, leading to enhanced bacterial growth. Here, we use the zebrafish-Mycobacterium marinum infection model to define the signaling basis of the host angiogenic response. Through intravital imaging and cell-restricted peptide-mediated inhibition, we identify macrophage-specific activation of NFAT signaling as essential to TDM-mediated angiogenesis in vivo. Exposure of cultured human cells to Mycobacterium tuberculosis results in robust induction of VEGFA, which is dependent on a signaling pathway downstream of host TDM detection and culminates in NFATC2 activation. As granuloma-associated angiogenesis is known to serve bacterial-beneficial roles, these findings identify potential host targets to improve tuberculosis disease outcomes.
Collapse
|
63
|
Farto-Vaamonde X, Diaz-Gomez L, Parga A, Otero A, Concheiro A, Alvarez-Lorenzo C. Perimeter and carvacrol-loading regulate angiogenesis and biofilm growth in 3D printed PLA scaffolds. J Control Release 2022; 352:776-792. [PMID: 36336096 DOI: 10.1016/j.jconrel.2022.10.060] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/07/2022] [Accepted: 10/30/2022] [Indexed: 11/13/2022]
Abstract
Carvacrol is a natural low-cost compound derived from oregano which presents anti-bacterial properties against both Gram-positive and Gram-negative bacteria. In this work, carvacrol-loaded PLA scaffolds were fabricated by 3D printing as platforms to support bone tissue regeneration while preventing biofilm development. Scaffolds were printed with or without a perimeter (lateral wall) mimicking the cortical structure of bone tissue to further evaluate if the lateral interconnectivity could affect the biological or antimicrobial properties of the scaffolds. Carvacrol incorporation was performed by loading either the PLA filament prior to 3D printing or the already printed PLA scaffold. The loading method determined carvacrol localization in the scaffolds and its release profile. Biphasic profiles were recorded in all cases, but scaffolds loaded post-printed released carvacrol much faster, with 50-80% released in the first day, compared to those containing carvacrol in PLA filament before printing which sustained the release for several weeks. The presence or absence of the perimeter did not affect the release rate, but total amount released. Tissue integration and vascularization of carvacrol-loaded scaffolds were evaluated in a chorioallantoic membrane model (CAM) using a novel quantitative micro-computed tomography (micro-CT) analysis approach. The obtained results confirmed the CAM tissue ingrowth and new vessel formation within the porous structure of the scaffolds after 7 days of incubation, without leading to hemorrhagic or cytotoxic effects. The absence of lateral wall facilitated lateral integration of the scaffolds in the host tissue, although increased the anisotropy of the mechanical properties. Scaffolds loaded with carvacrol post-printing showed antibiofilm activity against Staphylococcus aureus and Pseudomonas aeruginosa as observed in a decrease in CFU counting after biofilm detachment, changes in metabolic heat measured by calorimetry, and increased contact killing efficiency. In summary, this work demonstrated the feasibility of tuning carvacrol release rate and the amount released from PLA scaffolds to achieve antibiofilm protection without altering angiogenesis, which was mostly dependent on the perimeter density of the scaffolds.
Collapse
Affiliation(s)
- Xián Farto-Vaamonde
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma Group (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS), and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Luis Diaz-Gomez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma Group (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS), and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ana Parga
- Departamento de Microbiología y Parasitología, Facultad de Biología, Edificio CiBUS, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ana Otero
- Departamento de Microbiología y Parasitología, Facultad de Biología, Edificio CiBUS, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma Group (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS), and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma Group (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS), and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
64
|
YAP promotes cell-autonomous immune responses to tackle intracellular Staphylococcus aureus in vitro. Nat Commun 2022; 13:6995. [PMID: 36384856 PMCID: PMC9669043 DOI: 10.1038/s41467-022-34432-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
Transcriptional cofactors YAP/TAZ have recently been found to support autophagy and inflammation, which are part of cell-autonomous immunity and are critical in antibacterial defense. Here, we studied the role of YAP against Staphylococcus aureus using CRISPR/Cas9-mutated HEK293 cells and a primary cell-based organoid model. We found that S. aureus infection increases YAP transcriptional activity, which is required to reduce intracellular S. aureus replication. A 770-gene targeted transcriptomic analysis revealed that YAP upregulates genes involved in autophagy/lysosome and inflammation pathways in both infected and uninfected conditions. The YAP-TEAD transcriptional activity promotes autophagic flux and lysosomal acidification, which are then important for defense against intracellular S. aureus. Furthermore, the staphylococcal toxin C3 exoenzyme EDIN-B was found effective in preventing YAP-mediated cell-autonomous immune response. This study provides key insights on the anti-S. aureus activity of YAP, which could be conserved for defense against other intracellular bacteria.
Collapse
|
65
|
Wein T, Sorek R. Bacterial origins of human cell-autonomous innate immune mechanisms. Nat Rev Immunol 2022; 22:629-638. [PMID: 35396464 DOI: 10.1038/s41577-022-00705-4] [Citation(s) in RCA: 140] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 12/11/2022]
Abstract
The cell-autonomous innate immune system enables animal cells to resist viral infection. This system comprises an array of sensors that, after detecting viral molecules, activate the expression of antiviral proteins and the interferon response. The repertoire of immune sensors and antiviral proteins has long been considered to be derived from extensive evolutionary innovation in vertebrates, but new data challenge this dogma. Recent studies show that central components of the cell-autonomous innate immune system have ancient evolutionary roots in prokaryotic genes that protect bacteria from phages. These include the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, Toll/IL-1 receptor (TIR) domain-containing pathogen receptors, the viperin family of antiviral proteins, SAMHD1-like nucleotide-depletion enzymes, gasdermin proteins and key components of the RNA interference pathway. This Perspective details current knowledge of the elements of antiviral immunity that are conserved from bacteria to humans, and presents possible evolutionary scenarios to explain the observed conservation.
Collapse
Affiliation(s)
- Tanita Wein
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Rotem Sorek
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
66
|
Pattern Recognition Receptors of Nucleic Acids Can Cause Sublethal Activation of the Mitochondrial Apoptosis Pathway during Viral Infection. J Virol 2022; 96:e0121222. [PMID: 36069553 PMCID: PMC9517702 DOI: 10.1128/jvi.01212-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The mitochondrial apoptosis pathway has the function to kill the cell, but recent work shows that this pathway can also be activated to a sublethal level, where signal transduction can be observed but the cell survives. Intriguingly, this signaling has been shown to contribute to inflammatory activity of epithelial cells upon infection with numerous agents. This suggests that microbial recognition can generate sublethal activity in the mitochondrial apoptosis pathway. Because this recognition is achieved by pattern recognition receptors (PRRs), it also implies that PRR signals are linked to the mitochondrial apoptosis apparatus. We here test this hypothesis during infection of epithelial cells with modified vaccinia virus Ankara (MVA). MVA recognition is achieved through receptors specific for nucleic acids, and we present evidence that the three receptors, Toll-like receptor 3 (TLR3), RIG-I/MDA5, and cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING), are involved in this signaling. When stimulated directly by specific ligands, all three receptors could trigger sublethal apoptosis signals. During infection with MVA, sublethal apoptosis signals were unmasked in X-linked IAP (XIAP)-deficient cells, where apoptosis induction was observed. Deletion of any of the three signaling adapters, TRIF, MAVS, and STING, reduced the DNA damage response, a sensitive measure of sublethal apoptosis signals. Our results suggest that PRRs signal via mitochondria, where they generate sublethal signals through the BCL-2-family, which may contribute to the response to infectious agents. IMPORTANCE A contribution of the mitochondrial apoptosis apparatus, in the absence of cell death, to the reaction of nonprofessional immune cells to viruses is suggested to play a role as a broad alert system of an infected cell: the apoptosis system can be activated by many upstream signals and could therefore act as a central coordinator of viral recognition. The proapoptotic activity of PRRs has been documented in multiple situations, but this activity seems too low to be meaningful, and a physiological significance of such activity is not immediately obvious. This work suggests the alternative interpretation that PRRs do not have the primary function to induce apoptosis but to trigger sublethal signals in the apoptosis system. A number of lines of recent research suggest that mitochondria contribute to cellular reactions, and this pathway may be a way of triggering an early host response.
Collapse
|
67
|
Acharya D, Reis R, Volcic M, Liu G, Wang MK, Chia BS, Nchioua R, Groß R, Münch J, Kirchhoff F, Sparrer KMJ, Gack MU. Actin cytoskeleton remodeling primes RIG-I-like receptor activation. Cell 2022; 185:3588-3602.e21. [PMID: 36113429 PMCID: PMC9680832 DOI: 10.1016/j.cell.2022.08.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 06/17/2022] [Accepted: 08/10/2022] [Indexed: 01/26/2023]
Abstract
The current dogma of RNA-mediated innate immunity is that sensing of immunostimulatory RNA ligands is sufficient for the activation of intracellular sensors and induction of interferon (IFN) responses. Here, we report that actin cytoskeleton disturbance primes RIG-I-like receptor (RLR) activation. Actin cytoskeleton rearrangement induced by virus infection or commonly used reagents to intracellularly deliver RNA triggers the relocalization of PPP1R12C, a regulatory subunit of the protein phosphatase-1 (PP1), from filamentous actin to cytoplasmic RLRs. This allows dephosphorylation-mediated RLR priming and, together with the RNA agonist, induces effective RLR downstream signaling. Genetic ablation of PPP1R12C impairs antiviral responses and enhances susceptibility to infection with several RNA viruses including SARS-CoV-2, influenza virus, picornavirus, and vesicular stomatitis virus. Our work identifies actin cytoskeleton disturbance as a priming signal for RLR-mediated innate immunity, which may open avenues for antiviral or adjuvant design.
Collapse
Affiliation(s)
- Dhiraj Acharya
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA; Department of Microbiology, The University of Chicago, Chicago, IL 60637, USA
| | - Rebecca Reis
- Department of Microbiology, The University of Chicago, Chicago, IL 60637, USA
| | - Meta Volcic
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - GuanQun Liu
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA; Department of Microbiology, The University of Chicago, Chicago, IL 60637, USA
| | - May K Wang
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Bing Shao Chia
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Rayhane Nchioua
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Michaela U Gack
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA; Department of Microbiology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
68
|
Anes E, Pires D, Mandal M, Azevedo-Pereira JM. Spatial localization of cathepsins: Implications in immune activation and resolution during infections. Front Immunol 2022; 13:955407. [PMID: 35990632 PMCID: PMC9382241 DOI: 10.3389/fimmu.2022.955407] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/14/2022] [Indexed: 11/17/2022] Open
Abstract
Cathepsins were first described, as endolysosomal proteolytic enzymes in reference to the organelles where they degrade the bulk of endogenous and exogenous substrates in a slightly acidic environment. These substrates include pathogens internalized via endocytosis and/or marked for destruction by autophagy. However, the role of cathepsins during infection far exceeds that of direct digestion of the pathogen. Cathepsins have been extensively investigated in the context of tumour associated immune cells and chronic inflammation. Several cathepsin-dependent immune responses develop in the endocytic pathway while others take place in the cytosol, the nucleus, or in the extracellular space. In this review we highlight the spatial localization of cathepsins and their implications in immune activation and resolution pathways during infection.
Collapse
|
69
|
Chen W, Ouyang X, Chen L, Li L. Multiple functions of CALCOCO family proteins in selective autophagy. J Cell Physiol 2022; 237:3505-3516. [DOI: 10.1002/jcp.30836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/21/2022] [Accepted: 07/07/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Wei Chen
- Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology University of South China Hengyang Hunan China
| | - Xueqian Ouyang
- Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology University of South China Hengyang Hunan China
| | - Linxi Chen
- Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology University of South China Hengyang Hunan China
| | - Lanfang Li
- Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology University of South China Hengyang Hunan China
| |
Collapse
|
70
|
Spaan AN, Neehus AL, Laplantine E, Staels F, Ogishi M, Seeleuthner Y, Rapaport F, Lacey KA, Van Nieuwenhove E, Chrabieh M, Hum D, Migaud M, Izmiryan A, Lorenzo L, Kochetkov T, Heesterbeek DAC, Bardoel BW, DuMont AL, Dobbs K, Chardonnet S, Heissel S, Baslan T, Zhang P, Yang R, Bogunovic D, Wunderink HF, Haas PJA, Molina H, Van Buggenhout G, Lyonnet S, Notarangelo LD, Seppänen MRJ, Weil R, Seminario G, Gomez-Tello H, Wouters C, Mesdaghi M, Shahrooei M, Bossuyt X, Sag E, Topaloglu R, Ozen S, Leavis HL, van Eijk MMJ, Bezrodnik L, Blancas Galicia L, Hovnanian A, Nassif A, Bader-Meunier B, Neven B, Meyts I, Schrijvers R, Puel A, Bustamante J, Aksentijevich I, Kastner DL, Torres VJ, Humblet-Baron S, Liston A, Abel L, Boisson B, Casanova JL. Human OTULIN haploinsufficiency impairs cell-intrinsic immunity to staphylococcal α-toxin. Science 2022; 376:eabm6380. [PMID: 35587511 PMCID: PMC9233084 DOI: 10.1126/science.abm6380] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The molecular basis of interindividual clinical variability upon infection with Staphylococcus aureus is unclear. We describe patients with haploinsufficiency for the linear deubiquitinase OTULIN, encoded by a gene on chromosome 5p. Patients suffer from episodes of life-threatening necrosis, typically triggered by S. aureus infection. The disorder is phenocopied in patients with the 5p- (Cri-du-Chat) chromosomal deletion syndrome. OTULIN haploinsufficiency causes an accumulation of linear ubiquitin in dermal fibroblasts, but tumor necrosis factor receptor-mediated nuclear factor κB signaling remains intact. Blood leukocyte subsets are unaffected. The OTULIN-dependent accumulation of caveolin-1 in dermal fibroblasts, but not leukocytes, facilitates the cytotoxic damage inflicted by the staphylococcal virulence factor α-toxin. Naturally elicited antibodies against α-toxin contribute to incomplete clinical penetrance. Human OTULIN haploinsufficiency underlies life-threatening staphylococcal disease by disrupting cell-intrinsic immunity to α-toxin in nonleukocytic cells.
Collapse
Affiliation(s)
- András N Spaan
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Anna-Lena Neehus
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Institute of Experimental Hematology, REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Emmanuel Laplantine
- Centre d'Immunologie et des Maladies Infectieuses, INSERM U1135, CNRS ERL8255, Sorbonne University, 75724 Paris, France
- Institut de Recherche St. Louis, Hôpital St. Louis, INSERM U944, CNRS U7212, Paris Cité University, 75010 Paris, France
| | - Frederik Staels
- Laboratory for Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
| | - Franck Rapaport
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Keenan A Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Erika Van Nieuwenhove
- Laboratory for Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Department of Pediatric Rheumatology and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Maya Chrabieh
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
| | - David Hum
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Mélanie Migaud
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
| | - Araksya Izmiryan
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Laboratory of Genetic Skin Diseases, INSERM U1163, 75015 Paris, France
| | - Lazaro Lorenzo
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
| | - Tatiana Kochetkov
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Dani A C Heesterbeek
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Bart W Bardoel
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Ashley L DuMont
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kerry Dobbs
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, NIAID, NIH, Bethesda, MD 20852, USA
| | - Solenne Chardonnet
- Plateforme Post-génomique de la Pitié-Salpêtrière, P3S, UMS Production et Analyse de données en Sciences de la vie et en Santé, PASS, INSERM, Sorbonne University, 75013 Paris, France
| | - Søren Heissel
- Proteomics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Timour Baslan
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Peng Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Rui Yang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Dusan Bogunovic
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Herman F Wunderink
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Pieter-Jan A Haas
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Henrik Molina
- Proteomics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Griet Van Buggenhout
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- Center for Human Genetics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Stanislas Lyonnet
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Laboratory Embryology and Genetics of Malformations, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, NIAID, NIH, Bethesda, MD 20852, USA
| | - Mikko R J Seppänen
- Rare Disease and Pediatric Research Centers, Children and Adolescents, University of Helsinki and HUS Helsinki University Hospital, 00260 Helsinki, Finland
| | - Robert Weil
- Centre d'Immunologie et des Maladies Infectieuses, INSERM U1135, CNRS ERL8255, Sorbonne University, 75724 Paris, France
| | - Gisela Seminario
- Center for Clinical Immunology, Immunology Group Children's Hospital Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Héctor Gomez-Tello
- Immunology Department, Poblano Children's Hospital, 72190 Puebla, Mexico
| | - Carine Wouters
- Laboratory for Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Department of Pediatrics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Mehrnaz Mesdaghi
- Department of Allergy and Clinical Immunology, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, 15468-155514 Tehran, Iran
| | - Mohammad Shahrooei
- Clinical and Diagnostic Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Specialized Immunology Laboratory of Dr. Shahrooei, Sina Medical Complex, 15468-155514 Ahvaz, Iran
| | - Xavier Bossuyt
- Clinical and Diagnostic Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Erdal Sag
- Department of Pediatric Rheumatology, Hacettepe University, 06230 Ankara, Turkey
| | - Rezan Topaloglu
- Department of Pediatric Nephrology, Hacettepe University School of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Seza Ozen
- Department of Pediatric Rheumatology, Hacettepe University, 06230 Ankara, Turkey
| | - Helen L Leavis
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Maarten M J van Eijk
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Liliana Bezrodnik
- Center for Clinical Immunology, Immunology Group Children's Hospital Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | | | - Alain Hovnanian
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Laboratory of Genetic Skin Diseases, INSERM U1163, 75015 Paris, France
- Department of Genetics, Necker Hospital for Sick Children, AP-HP, 75015 Paris, France
| | - Aude Nassif
- Centre Médical, Institut Pasteur, 75724 Paris, France
| | - Brigitte Bader-Meunier
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Pediatric Immunology, Hematology and Rheumatology Unit, Necker Hospital for Sick Children, AP-HP, 75015 Paris, France
- Laboratory of Immunogenetics of Pediatric Autoimmunity, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
| | - Bénédicte Neven
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Pediatric Immunology, Hematology and Rheumatology Unit, Necker Hospital for Sick Children, AP-HP, 75015 Paris, France
- Laboratory of Immunogenetics of Pediatric Autoimmunity, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
| | - Isabelle Meyts
- Laboratory of Inborn Errors of Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Department of Pediatrics, Jeffrey Modell Diagnostic and Research Network Center, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Rik Schrijvers
- Allergy and Clinical Immunology Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Anne Puel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
| | - Jacinta Bustamante
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, AP-HP, 75015 Paris, France
| | - Ivona Aksentijevich
- Inflammatory Disease Section, National Human Genome Research Institute, Bethesda, MD 20892, USA
| | - Daniel L Kastner
- Inflammatory Disease Section, National Human Genome Research Institute, Bethesda, MD 20892, USA
| | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Stéphanie Humblet-Baron
- Laboratory for Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Adrian Liston
- Laboratory for Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Brain and Disease Research, Leuven 3000, Belgium
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, 75015 Paris, France
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
71
|
Artificially sporulated Escherichia coli cells as a robust cell factory for interfacial biocatalysis. Nat Commun 2022; 13:3142. [PMID: 35668090 PMCID: PMC9170730 DOI: 10.1038/s41467-022-30915-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 05/06/2022] [Indexed: 12/13/2022] Open
Abstract
The natural bacterial spores have inspired the development of artificial spores, through coating cells with protective materials, for durable whole-cell catalysis. Despite attractiveness, artificial spores developed to date are generally limited to a few microorganisms with their natural endogenous enzymes, and they have never been explored as a generic platform for widespread synthesis. Here, we report a general approach to designing artificial spores based on Escherichia coli cells with recombinant enzymes. The artificial spores are simply prepared by coating cells with polydopamine, which can withstand UV radiation, heating and organic solvents. Additionally, the protective coating enables living cells to stabilize aqueous-organic emulsions for efficient interfacial biocatalysis ranging from single reactions to multienzyme cascades. Furthermore, the interfacial system can be easily expanded to chemoenzymatic synthesis by combining artificial spores with metal catalysts. Therefore, this artificial-spore-based platform technology is envisioned to lay the foundation for next-generation cell factory engineering.
Collapse
|
72
|
Li M, Huang J, Zhu Y, Huang Z, Zhang G, Huang J. Anti-L1 antibody-bound HPV16 pseudovirus is degraded intracellularly via TRIM21/proteasomal pathway. Virol J 2022; 19:90. [PMID: 35619167 PMCID: PMC9137102 DOI: 10.1186/s12985-022-01826-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022] Open
Abstract
Background Persistent HPV16 infection is the leading risk factor for developing cervical cancer. Anti-L1 antibodies against HPV16 produced in HPV16 infections play diverse roles in the clearance of virus infection and prevention of persistence. It has been implicated that the cervicovaginal squamous epithelial cells actually express TRIM21 and that some HPV16 particles could escape leaky endosomal compartment into the cytosol and that Fc receptor TRIM21 directly neutralize infection by targeting antibody-opsonized viruses for proteasomal degradation. We explored whether anti-L1 antibody opsonized HPV16 pseudovirus (PsV) entered into the cytosol could be neutralized by TRIM21-mediated activation of a proteasomal pathway to reduce the chance of persistent HPV16 infection. Methods HPV16 PsV were generated and extracted in HEK 293FT cells co-transfected with pcDNA3.1-eGFP and p16sheLL plasmids according to the standard protocol. The HPV16 PsV with capsid protein L1 was characterized by fluorescence microscopy and western blot, and the HPV16 PsV titer and anti-L1-bound PsV entry efficiency were detected by flow cytometry. The expressions of transcription factors (TF) and cytokines elicited by the TRIM21-activated proteasomal pathway were confirmed by dual-luciferase reporter assay and RT-qPCR. The changes in HPV16 PsV load with or without inhibitors in the infected HEK 293FT cells were determinated by qPCR. Results Simultaneous transfection with pcDNA3.1-eGFP and p16sheLL plasmids into the HEK 293FT cells resulted in the self-assembly of HPV16 PsV with capsid protein L1. Both HPV16 PsV and anti-L1-bound HPV16 PsV could infect HEK 293FT cells. Anti-L1-bound PsV up-regulated TRIM21 mediated-activation of proteasome and increased expressions of TF and cytokines in the infected cells where HPV16 PsV load reduced by ~ 1000-fold in the presence of anti-L1 antibody, but inhibition of proteasomal activity increased HPV16 PsV load. Conclusion Our preliminary results indicate that anti-L1 antibody entered with HPV16 PsV into the cells could mediate degradation of HPV16 PsV by TRIM21-activated proteasomal pathway intracellularly, giving anti-capsid protein L1 antibody a role in host defense of persistent HPV16 infection.
Collapse
Affiliation(s)
- Meiying Li
- Department of Biochemistry and Molecular Biology, Sichuan Cancer Institute, Chengdu, 610041, People's Republic of China
| | - Jianmei Huang
- Department of Gynecologic Oncology, Sichuan Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, People's Republic of China
| | - Yi Zhu
- Department of Gynecologic Oncology, Sichuan Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, People's Republic of China.,Department of Ultrasound, Sichuan Cancer Hospital, Chengdu, 610041, People's Republic of China
| | - Ziyi Huang
- Department of Bioinformatics, Basic Medical College of Chongqing Medical University, Chongqing, People's Republic of China
| | - Guonan Zhang
- Department of Gynecologic Oncology, Sichuan Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, People's Republic of China
| | - Jianming Huang
- Department of Biochemistry and Molecular Biology, Sichuan Cancer Institute, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
73
|
Basu S, Huynh L, Zhang S, Rabara R, Nguyen H, Velásquez Guzmán J, Hao G, Miles G, Shi Q, Stover E, Gupta G. Two Liberibacter Proteins Combine to Suppress Critical Innate Immune Defenses in Citrus. FRONTIERS IN PLANT SCIENCE 2022; 13:869178. [PMID: 35586217 PMCID: PMC9108871 DOI: 10.3389/fpls.2022.869178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/04/2022] [Indexed: 06/15/2023]
Abstract
We adopted a systems-based approach to determine the role of two Candidatus Liberibacter asiaticus (CLas) proteins, LasP 235 and Effector 3, in Huanglongbing (HLB) pathogenesis. While a published work suggests the involvement of these CLas proteins HLB pathogenesis, the exact structure-based mechanism of their action has not been elucidated. We conducted the following experiments to determine the structure-based mechanisms of action. First, we immunoprecipitated the interacting citrus protein partners of LasP 235 and Effector 3 from the healthy and CLas-infected Hamlin extracts and identified them by Liquid Chromatography with tandem mass spectrometry (LC-MS/MS). Second, we performed a split green fluorescent protein (GFP) assay in tobacco to validate that the interactions observed in vitro are also retained in planta. The notable in planta citrus targets of LasP 235 and Effector 3 include citrus innate immune proteins. Third, in vitro and in planta studies were performed to show that LasP 235 and Effector 3 interact with and inhibit the functions of multiple citrus proteins belonging to the innate immune pathways. These inhibitory interactions led to a high level of reactive oxygen species, blocking of bactericidal lipid transfer protein (LTP), and induction of premature programed cell death (PCD), all of which are beneficial to CLas lifecycle and HLB pathogenesis. Finally, we performed molecular dynamics simulations to visualize the interactions of LasP 235 and Effector 3, respectively, with LTP and Kunitz protease inhibitor. This led to the design of an LTP mimic, which sequestered and blocked LasP 235 and rescued the bactericidal activity of LTP thereby proving that LasP 235 , indeed, participates in HLB pathogenesis.
Collapse
Affiliation(s)
- Supratim Basu
- Biolab, New Mexico Consortium, Los Alamos, NM, United States
| | - Loan Huynh
- Biolab, New Mexico Consortium, Los Alamos, NM, United States
| | - Shujian Zhang
- Biolab, New Mexico Consortium, Los Alamos, NM, United States
| | - Roel Rabara
- Biolab, New Mexico Consortium, Los Alamos, NM, United States
| | - Hau Nguyen
- Biolab, New Mexico Consortium, Los Alamos, NM, United States
| | | | - Guixia Hao
- Horticulture and Breeding, U. S. Horticultural Research Laboratory, Fort Pierce, FL, United States
| | - Godfrey Miles
- Horticulture and Breeding, U. S. Horticultural Research Laboratory, Fort Pierce, FL, United States
| | - Qingchun Shi
- Horticulture and Breeding, U. S. Horticultural Research Laboratory, Fort Pierce, FL, United States
| | - Ed Stover
- Horticulture and Breeding, U. S. Horticultural Research Laboratory, Fort Pierce, FL, United States
| | - Goutam Gupta
- Biolab, New Mexico Consortium, Los Alamos, NM, United States
| |
Collapse
|
74
|
Skariah S, Sultan AA, Mordue DG. IFN-induced cell-autonomous immune mechanisms in the control of intracellular protozoa. Parasitol Res 2022; 121:1559-1571. [DOI: 10.1007/s00436-022-07514-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 04/04/2022] [Indexed: 10/18/2022]
|
75
|
Torp MK, Ranheim T, Schjalm C, Hjorth M, Heiestad C, Dalen KT, Nilsson PH, Mollnes TE, Pischke SE, Lien E, Vaage J, Yndestad A, Stensløkken KO. Intracellular Complement Component 3 Attenuated Ischemia-Reperfusion Injury in the Isolated Buffer-Perfused Mouse Heart and Is Associated With Improved Metabolic Homeostasis. Front Immunol 2022; 13:870811. [PMID: 35432387 PMCID: PMC9011808 DOI: 10.3389/fimmu.2022.870811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/08/2022] [Indexed: 12/25/2022] Open
Abstract
The innate immune system is rapidly activated during myocardial infarction and blockade of extracellular complement system reduces infarct size. Intracellular complement, however, appears to be closely linked to metabolic pathways and its role in ischemia-reperfusion injury is unknown and may be different from complement activation in the circulation. The purpose of the present study was to investigate the role of intracellular complement in isolated, retrogradely buffer-perfused hearts and cardiac cells from adult male wild type mice (WT) and from adult male mice with knockout of complement component 3 (C3KO). Main findings: (i) Intracellular C3 protein was expressed in isolated cardiomyocytes and in whole hearts, (ii) after ischemia-reperfusion injury, C3KO hearts had larger infarct size (32 ± 9% in C3KO vs. 22 ± 7% in WT; p=0.008) and impaired post-ischemic relaxation compared to WT hearts, (iii) C3KO cardiomyocytes had lower basal oxidative respiration compared to WT cardiomyocytes, (iv) blocking mTOR decreased Akt phosphorylation in WT, but not in C3KO cardiomyocytes, (v) after ischemia, WT hearts had higher levels of ATP, but lower levels of both reduced and oxidized nicotinamide adenine dinucleotide (NADH and NAD+, respectively) compared to C3KO hearts. Conclusion: intracellular C3 protected the heart against ischemia-reperfusion injury, possibly due to its role in metabolic pathways important for energy production and cell survival.
Collapse
Affiliation(s)
- M-K. Torp
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- *Correspondence: M-K. Torp,
| | - T. Ranheim
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Division of Surgery, Inflammatory Diseases and Transplantation, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - C. Schjalm
- Department of Immunology, Institute of Clinical Medicine University of Oslo, Oslo, Norway
| | - M. Hjorth
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - C.M. Heiestad
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - K. T. Dalen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - P. H. Nilsson
- Department of Immunology, Institute of Clinical Medicine University of Oslo, Oslo, Norway
- Linnaeus Centre for Biomaterials Chemistry, and the Department of Chemistry and Biomedicine, Linnaeus University, Kalmar, Sweden
| | - T. E. Mollnes
- Department of Immunology, Institute of Clinical Medicine University of Oslo, Oslo, Norway
- Stiftelsen Kristian Gerhard Jebsen (K.G. Jebsen) Inflammation Research Center (IRC), University of Oslo, Oslo, Norway
- Research Laboratory, Nordland Hospital, Bodø, and Faculty of Health Sciences, Stiftelsen Kristian Gerhard Jebsen (K.G. Jebsen) Thrombosis Research and Expertise Center (TREC), University of Tromsø, Tromsø, Norway
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - S. E. Pischke
- Department of Immunology, Institute of Clinical Medicine University of Oslo, Oslo, Norway
- Department of Research & Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
| | - E. Lien
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Department of Medicine, UMass Medical School, Worchester, MA, United States
| | - J. Vaage
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Research & Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - A. Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - K-O. Stensløkken
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
76
|
|
77
|
Guey B, Ablasser A. Emerging dimensions of cellular cGAS-STING signaling. Curr Opin Immunol 2022; 74:164-171. [DOI: 10.1016/j.coi.2022.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 01/11/2022] [Accepted: 01/15/2022] [Indexed: 12/14/2022]
|
78
|
Petit TJ, Lebreton A. Adaptations of intracellular bacteria to vacuolar or cytosolic niches. Trends Microbiol 2022; 30:736-748. [DOI: 10.1016/j.tim.2022.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 12/28/2022]
|
79
|
Zhang J, Zhang L, Shi H, Feng S, Feng T, Chen J, Zhang X, Han Y, Liu J, Wang Y, Ji Z, Jing Z, Liu D, Shi D, Feng L. Swine acute diarrhea syndrome coronavirus replication is reduced by inhibition of the extracellular signal-regulated kinase (ERK) signaling pathway. Virology 2022; 565:96-105. [PMID: 34768113 PMCID: PMC8556614 DOI: 10.1016/j.virol.2021.10.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 01/04/2023]
Abstract
Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a newly discovered enteric coronavirus. We have previously shown that the caspase-dependent FASL-mediated and mitochondrion-mediated apoptotic pathways play a central role in SADS-CoV-induced apoptosis, which facilitates viral replication. However, the roles of intracellular signaling pathways in SADS-CoV-mediated cell apoptosis and the relative advantages that such pathways confer on the host or virus remain largely unknown. In this study, we show that SADS-CoV induces the activation of ERK during infection, irrespective of viral biosynthesis. The knockdown or chemical inhibition of ERK1/2 significantly suppressed viral protein expression and viral progeny production. The inhibition of ERK activation also circumvented SADS-CoV-induced apoptosis. Taken together, these data suggest that ERK activation is important for SADS-CoV replication, and contributes to the virus-mediated changes in host cells. Our findings demonstrate the takeover of a particular host signaling mechanism by SADS-CoV and identify a potential approach to inhibiting viral spread.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Da Shi
- Corresponding author. Harbin Veterinary Research Institute, CAAS, 678 Haping Road Xiangfang District, Harbin, 150069, China
| | - Li Feng
- Corresponding author. Harbin Veterinary Research Institute, CAAS, 678 Haping Road Xiangfang District, Harbin, 150069, China
| |
Collapse
|
80
|
Abstract
Virus entry, consisting of attachment to and penetration into the host target cell, is the first step of the virus life cycle and is a critical 'do or die' event that governs virus emergence in host populations. Most antiviral vaccines induce neutralizing antibodies that prevent virus entry into cells. However, while the prevention of virus invasion by humoral immunity is well appreciated, considerably less is known about the immune defences present within cells (known as intrinsic immunity) that interfere with virus entry. The interferon-induced transmembrane (IFITM) proteins, known for inhibiting fusion between viral and cellular membranes, were once the only factors known to restrict virus entry. However, the progressive development of genetic and pharmacological screening platforms and the onset of the COVID-19 pandemic have galvanized interest in how viruses infiltrate cells and how cells defend against it. Several host factors with antiviral potential are now implicated in the regulation of virus entry, including cholesterol 25-hydroxylase (CH25H), lymphocyte antigen 6E (LY6E), nuclear receptor co-activator protein 7 (NCOA7), interferon-γ-inducible lysosomal thiol reductase (GILT), CD74 and ARFGAP with dual pleckstrin homology domain-containing protein 2 (ADAP2). This Review summarizes what is known and what remains to be understood about the intrinsic factors that form the first line of defence against virus infection.
Collapse
|
81
|
Abstract
Toxoplasma gondii is a parasitic protist infecting a wide group of warm-blooded animals, ranging from birds to humans. While this infection is usually asymptomatic in healthy individuals, it can also lead to severe ocular or neurological outcomes in immunocompromised individuals or in developing fetuses. This obligate intracellular parasite has the ability to infect a considerable range of nucleated cells and can propagate in the intermediate host. Yet, under the pressure of the immune system it transforms into an encysted persistent form residing primarily in the brain and muscle tissues. Encysted parasites, which are resistant to current medication, may reactivate and give rise to an acute infection. The clinical outcome of toxoplasmosis depends on a complex balance between the host immune response and parasite virulence factors. Susceptibility to the disease is thus determined by both parasite strains and host species. Recent advances on our understanding of host cell-parasite interactions and parasite virulence have brought new insights into the pathophysiology of T. gondii infection and are summarized here.
Collapse
|
82
|
Wang S, Hao M, Pan Z, Lei J, Zou X. Data-driven multi-scale mathematical modeling of SARS-CoV-2 infection reveals heterogeneity among COVID-19 patients. PLoS Comput Biol 2021; 17:e1009587. [PMID: 34818337 PMCID: PMC8654229 DOI: 10.1371/journal.pcbi.1009587] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 12/08/2021] [Accepted: 10/25/2021] [Indexed: 12/17/2022] Open
Abstract
Patients with coronavirus disease 2019 (COVID-19) often exhibit diverse disease progressions associated with various infectious ability, symptoms, and clinical treatments. To systematically and thoroughly understand the heterogeneous progression of COVID-19, we developed a multi-scale computational model to quantitatively understand the heterogeneous progression of COVID-19 patients infected with severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2). The model consists of intracellular viral dynamics, multicellular infection process, and immune responses, and was formulated using a combination of differential equations and stochastic modeling. By integrating multi-source clinical data with model analysis, we quantified individual heterogeneity using two indexes, i.e., the ratio of infected cells and incubation period. Specifically, our simulations revealed that increasing the host antiviral state or virus induced type I interferon (IFN) production rate can prolong the incubation period and postpone the transition from asymptomatic to symptomatic outcomes. We further identified the threshold dynamics of T cell exhaustion in the transition between mild-moderate and severe symptoms, and that patients with severe symptoms exhibited a lack of naïve T cells at a late stage. In addition, we quantified the efficacy of treating COVID-19 patients and investigated the effects of various therapeutic strategies. Simulations results suggested that single antiviral therapy is sufficient for moderate patients, while combination therapies and prevention of T cell exhaustion are needed for severe patients. These results highlight the critical roles of IFN and T cell responses in regulating the stage transition during COVID-19 progression. Our study reveals a quantitative relationship underpinning the heterogeneity of transition stage during COVID-19 progression and can provide a potential guidance for personalized therapy in COVID-19 patients. Coronavirus disease 2019 (COVID-19) is currently destroying both lives and economies. However, patients infected with severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2) usually present heterogeneous and complicated progressions, such as different incubation periods (short and long), symptoms (asymptomatic and symptomatic) and severity (mild-moderate and severe). Currently, various clinical data and experimental data are available from different countries, which has great significance for integrating different types of data to comprehensively understand the diverse disease progression in COVID-19 patients and guide individual treatment strategies. Here, we developed a multi-scale computational model to describe the dynamical process of patients infected with SARS-CoV-2, including intracellular viral dynamics, multicellular infection process, and immune responses. By combining data integration, stochastic simulation and quantitative analysis based on the multi-scale mathematical model, we addressed an important question regarding how IFN response and T cell exhaustion quantitatively affect heterogeneous progression in patients with respect to incubation periods, symptoms and severity. Furthermore, the efficacy of various therapeutic strategies for treating COVID-19 patients with different severity degrees was evaluated and validated. The computational framework in this study can also be extended to explore the dynamical process of other coronavirus infections.
Collapse
Affiliation(s)
- Shun Wang
- School of Mathematics and Statistics, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, China
| | - Mengqian Hao
- School of Mathematics and Statistics, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, China
| | - Zishu Pan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Jinzhi Lei
- School of Mathematical Sciences, Center for Applied Mathematics, Tiangong University, Tianjin, China
- * E-mail: (JL); (XZ)
| | - Xiufen Zou
- School of Mathematics and Statistics, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, China
- * E-mail: (JL); (XZ)
| |
Collapse
|
83
|
Wang M, Fan Z, Han H. Autophagy in Staphylococcus aureus Infection. Front Cell Infect Microbiol 2021; 11:750222. [PMID: 34692566 PMCID: PMC8529010 DOI: 10.3389/fcimb.2021.750222] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/23/2021] [Indexed: 11/23/2022] Open
Abstract
Staphylococcus aureus is an invasive, facultative intracellular pathogen that can colonize niches in various host organisms, making it difficult for the host immune system to completely eliminate. Host autophagy is an intracellular clearance pathway involved in degrading S. aureus. Whereas the accessory gene regulatory system of S. aureus that controls virulence factors could resist the host immune defenses by evading and even utilizing autophagy. This article reviews the interaction between autophagy and S. aureus, providing insights on how to use these mechanisms to improve S. aureus infection control.
Collapse
Affiliation(s)
- Mengyao Wang
- Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China.,National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ziyao Fan
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Hongbing Han
- Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China.,National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
84
|
Burbidge K, Rademacher DJ, Mattick J, Zack S, Grillini A, Bousset L, Kwon O, Kubicki K, Simon A, Melki R, Campbell EM. LGALS3 (galectin 3) mediates an unconventional secretion of SNCA/α-synuclein in response to lysosomal membrane damage by the autophagic-lysosomal pathway in human midbrain dopamine neurons. Autophagy 2021; 18:1020-1048. [PMID: 34612142 PMCID: PMC9196737 DOI: 10.1080/15548627.2021.1967615] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Numerous lines of evidence support the premise that the misfolding and subsequent accumulation of SNCA/α-synuclein (synuclein alpha) is responsible for the underlying neuronal pathology observed in Parkinson disease (PD) and other synucleinopathies. Moreover, the cell-to-cell transfer of these misfolded SNCA species is thought to be responsible for disease progression and the spread of cellular pathology throughout the brain. Previous work has shown that when exogenous, misfolded SNCA fibrils enter cells through endocytosis, they can damage and rupture the membranes of their endocytotic vesicles in which they are trafficked. Rupture of these vesicular membranes exposes intralumenal glycans leading to galectin protein binding, subsequent autophagic protein recruitment, and, ultimately, their introduction into the autophagic-lysosomal pathway. Increasing evidence indicates that both pathological and non-pathological SNCA species undergo autophagy-dependent unconventional secretion. While other proteins have also been shown to be secreted from cells by autophagy, what triggers this release process and how these specific proteins are recruited to a secretory autophagic pathway is largely unknown. Here, we use a human midbrain dopamine (mDA) neuronal culture model to provide evidence in support of a cellular mechanism that explains the cell-to-cell transfer of pathological forms of SNCA that are observed in PD. We demonstrate that LGALS3 (galectin 3) mediates the release of SNCA following vesicular damage. SNCA release is also dependent on TRIM16 (tripartite motif containing 16) and ATG16L1 (autophagy related 16 like 1), providing evidence that secretion of SNCA is mediated by an autophagic secretory pathway.
Collapse
Affiliation(s)
- Kevin Burbidge
- Graduate Program in Neuroscience, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA
| | - David J Rademacher
- Core Imaging Facility and Department of Microbiology and Immunology, Loyola University of Chicago, Maywood, Illinois, USA
| | - Jessica Mattick
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University, Chicago, Maywood, Illinois, USA
| | - Stephanie Zack
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University, Chicago, Maywood, Illinois, USA
| | - Andrea Grillini
- Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA
| | - Luc Bousset
- Institut Francois Jacob (Mircen), Cea and Laboratory of Neurodegenerative Diseases, Cnrs, Fontenay-Aux-Roses Cedex, France
| | - Ochan Kwon
- Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA
| | - Konrad Kubicki
- Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA
| | - Alexander Simon
- Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA
| | - Ronald Melki
- Institut Francois Jacob (Mircen), Cea and Laboratory of Neurodegenerative Diseases, Cnrs, Fontenay-Aux-Roses Cedex, France
| | - Edward M Campbell
- Graduate Program in Neuroscience, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA.,Core Imaging Facility and Department of Microbiology and Immunology, Loyola University of Chicago, Maywood, Illinois, USA
| |
Collapse
|
85
|
Hassan Z, Kumar ND, Reggiori F, Khan G. How Viruses Hijack and Modify the Secretory Transport Pathway. Cells 2021; 10:2535. [PMID: 34685515 PMCID: PMC8534161 DOI: 10.3390/cells10102535] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/28/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022] Open
Abstract
Eukaryotic cells contain dynamic membrane-bound organelles that are constantly remodeled in response to physiological and environmental cues. Key organelles are the endoplasmic reticulum, the Golgi apparatus and the plasma membrane, which are interconnected by vesicular traffic through the secretory transport route. Numerous viruses, especially enveloped viruses, use and modify compartments of the secretory pathway to promote their replication, assembly and cell egression by hijacking the host cell machinery. In some cases, the subversion mechanism has been uncovered. In this review, we summarize our current understanding of how the secretory pathway is subverted and exploited by viruses belonging to Picornaviridae, Coronaviridae, Flaviviridae,Poxviridae, Parvoviridae and Herpesviridae families.
Collapse
Affiliation(s)
- Zubaida Hassan
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates;
- Department of Microbiology, School of Life Sciences, Modibbo Adama University, Yola PMB 2076, Nigeria
| | - Nilima Dinesh Kumar
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands; (N.D.K.); (F.R.)
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands; (N.D.K.); (F.R.)
| | - Gulfaraz Khan
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates;
| |
Collapse
|
86
|
Turco E, Savova A, Gere F, Ferrari L, Romanov J, Schuschnig M, Martens S. Reconstitution defines the roles of p62, NBR1 and TAX1BP1 in ubiquitin condensate formation and autophagy initiation. Nat Commun 2021; 12:5212. [PMID: 34471133 PMCID: PMC8410870 DOI: 10.1038/s41467-021-25572-w] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 08/13/2021] [Indexed: 01/02/2023] Open
Abstract
The autophagic degradation of misfolded and ubiquitinated proteins is important for cellular homeostasis. In this process, which is governed by cargo receptors, ubiquitinated proteins are condensed into larger structures and subsequently become targets for the autophagy machinery. Here we employ in vitro reconstitution and cell biology to define the roles of the human cargo receptors p62/SQSTM1, NBR1 and TAX1BP1 in the selective autophagy of ubiquitinated substrates. We show that p62 is the major driver of ubiquitin condensate formation. NBR1 promotes condensate formation by equipping the p62-NBR1 heterooligomeric complex with a high-affinity UBA domain. Additionally, NBR1 recruits TAX1BP1 to the ubiquitin condensates formed by p62. While all three receptors interact with FIP200, TAX1BP1 is the main driver of FIP200 recruitment and thus the autophagic degradation of p62-ubiquitin condensates. In summary, our study defines the roles of all three receptors in the selective autophagy of ubiquitin condensates.
Collapse
Affiliation(s)
- Eleonora Turco
- Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC), Vienna, Austria.
| | - Adriana Savova
- Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC), Vienna, Austria
| | - Flora Gere
- Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC), Vienna, Austria
| | - Luca Ferrari
- Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC), Vienna, Austria
| | - Julia Romanov
- Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC), Vienna, Austria
| | - Martina Schuschnig
- Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC), Vienna, Austria
| | - Sascha Martens
- Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC), Vienna, Austria.
| |
Collapse
|
87
|
Oh C, Verma A, Hafeez M, Hogland B, Aachoui Y. Shigella OspC3 suppresses murine cytosolic LPS sensing. iScience 2021; 24:102910. [PMID: 34409271 PMCID: PMC8361271 DOI: 10.1016/j.isci.2021.102910] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/09/2021] [Accepted: 07/23/2021] [Indexed: 12/18/2022] Open
Abstract
Shigella flexneri, a cytosol-invasive gram-negative pathogen, deploys an array of type III-secreted effector proteins to evade host cell defenses. Caspase-11 and its human ortholog caspase-4 detect cytosolic lipopolysaccharide (LPS) and trigger gasdermin D-mediated pyroptosis to eliminate intra-cytoplasmic bacterial threats. However, the role of caspase-11 in combating S. flexneri is unclear. The Shigella T3SS effector OspC3 reportedly suppresses cytosolic LPS sensing by inhibiting caspase-4 but not caspase-11 activity. Surprisingly, we found that S. flexneri also uses OspC3 to inhibit murine caspase-11 activity. Mechanistically, we found that OspC3 binds only to primed caspase-11. Importantly, we demonstrate that S. flexneri employs OspC3 to prevent caspase-11-mediated pyroptosis in neutrophils, enabling bacteria to disseminate and evade clearance following intraperitoneal challenge. In contrast, S. flexneri lacking OspC3 is attenuated in a caspase-11- and gasdermin D-dependent fashion. Overall, our study reveals that OspC3 suppresses cytosolic LPS detection in a broad array of mammals. S. flexneri T3SS-secreted OspC3 suppresses cytosolic LPS sensing by caspase-11 OspC3 binds to caspase-11 in a priming-dependent manner S. flexneri employs OspC3 to prevent caspase-11-mediated pyroptosis in neutrophils Neutrophil caspase-11 is essential in defense against S. flexneri ΔOspC3 in vivo
Collapse
Affiliation(s)
- Changhoon Oh
- Department of Microbiology and Immunology, Center for Microbial Pathogenesis and Host Responses, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ambika Verma
- Department of Microbiology and Immunology, Center for Microbial Pathogenesis and Host Responses, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Mohib Hafeez
- Department of Microbiology and Immunology, Center for Microbial Pathogenesis and Host Responses, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,Department of Biology, University of Arkansas at Little Rock, Little Rock, AR 72204, USA
| | - Brandon Hogland
- Department of Microbiology and Immunology, Center for Microbial Pathogenesis and Host Responses, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Youssef Aachoui
- Department of Microbiology and Immunology, Center for Microbial Pathogenesis and Host Responses, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
88
|
Sixt BS. Host cell death during infection with Chlamydia: a double-edged sword. FEMS Microbiol Rev 2021; 45:5902849. [PMID: 32897321 PMCID: PMC7794043 DOI: 10.1093/femsre/fuaa043] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
The phylum Chlamydiae constitutes a group of obligate intracellular bacteria that infect a remarkably diverse range of host species. Some representatives are significant pathogens of clinical or veterinary importance. For instance, Chlamydia trachomatis is the leading infectious cause of blindness and the most common bacterial agent of sexually transmitted diseases. Chlamydiae are exceptionally dependent on their eukaryotic host cells as a consequence of their developmental biology. At the same time, host cell death is an integral part of the chlamydial infection cycle. It is therefore not surprising that the bacteria have evolved exquisite and versatile strategies to modulate host cell survival and death programs to their advantage. The recent introduction of tools for genetic modification of Chlamydia spp., in combination with our increasing awareness of the complexity of regulated cell death in eukaryotic cells, and in particular of its connections to cell-intrinsic immunity, has revived the interest in this virulence trait. However, recent advances also challenged long-standing assumptions and highlighted major knowledge gaps. This review summarizes current knowledge in the field and discusses possible directions for future research, which could lead us to a deeper understanding of Chlamydia's virulence strategies and may even inspire novel therapeutic approaches.
Collapse
Affiliation(s)
- Barbara S Sixt
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Department of Molecular Biology, Umeå University, SE-901 87 Umeå, Sweden
| |
Collapse
|
89
|
Gaudet RG, Zhu S, Halder A, Kim BH, Bradfield CJ, Huang S, Xu D, Mamiñska A, Nguyen TN, Lazarou M, Karatekin E, Gupta K, MacMicking JD. A human apolipoprotein L with detergent-like activity kills intracellular pathogens. Science 2021; 373:eabf8113. [PMID: 34437126 PMCID: PMC8422858 DOI: 10.1126/science.abf8113] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/29/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022]
Abstract
Activation of cell-autonomous defense by the immune cytokine interferon-γ (IFN-γ) is critical to the control of life-threatening infections in humans. IFN-γ induces the expression of hundreds of host proteins in all nucleated cells and tissues, yet many of these proteins remain uncharacterized. We screened 19,050 human genes by CRISPR-Cas9 mutagenesis and identified IFN-γ-induced apolipoprotein L3 (APOL3) as a potent bactericidal agent protecting multiple non-immune barrier cell types against infection. Canonical apolipoproteins typically solubilize mammalian lipids for extracellular transport; APOL3 instead targeted cytosol-invasive bacteria to dissolve their anionic membranes into human-bacterial lipoprotein nanodiscs detected by native mass spectrometry and visualized by single-particle cryo-electron microscopy. Thus, humans have harnessed the detergent-like properties of extracellular apolipoproteins to fashion an intracellular lysin, thereby endowing resident nonimmune cells with a mechanism to achieve sterilizing immunity.
Collapse
Affiliation(s)
- Ryan G Gaudet
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shiwei Zhu
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Anushka Halder
- Yale Nanobiology Institute, West Haven, CT 06477, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Bae-Hoon Kim
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Clinton J Bradfield
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shuai Huang
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Dijin Xu
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Agnieszka Mamiñska
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Thanh Ngoc Nguyen
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Michael Lazarou
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Erdem Karatekin
- Yale Nanobiology Institute, West Haven, CT 06477, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
- Saints-Pères Paris Institute for the Neurosciences, Centre National de la Recherche Scientifique (CNRS), Université de Paris, F-75006 Paris, France
| | - Kallol Gupta
- Yale Nanobiology Institute, West Haven, CT 06477, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - John D MacMicking
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA.
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
90
|
McCutcheon JP. The Genomics and Cell Biology of Host-Beneficial Intracellular Infections. Annu Rev Cell Dev Biol 2021; 37:115-142. [PMID: 34242059 DOI: 10.1146/annurev-cellbio-120219-024122] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Microbes gain access to eukaryotic cells as food for bacteria-grazing protists, for host protection by microbe-killing immune cells, or for microbial benefit when pathogens enter host cells to replicate. But microbes can also gain access to a host cell and become an important-often required-beneficial partner. The oldest beneficial microbial infections are the ancient eukaryotic organelles now called the mitochondrion and plastid. But numerous other host-beneficial intracellular infections occur throughout eukaryotes. Here I review the genomics and cell biology of these interactions with a focus on intracellular bacteria. The genomes of host-beneficial intracellular bacteria have features that span a previously unfilled gap between pathogens and organelles. Host cell adaptations to allow the intracellular persistence of beneficial bacteria are found along with evidence for the microbial manipulation of host cells, but the cellular mechanisms of beneficial bacterial infections are not well understood. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- John P McCutcheon
- Biodesign Center for Mechanisms of Evolution, School of Life Sciences, Arizona State University, Tempe, Arizona 85287, USA;
| |
Collapse
|
91
|
Nodari A, Scambi I, Peroni D, Calabria E, Benati D, Mannucci S, Manfredi M, Frontini A, Visonà S, Bozzato A, Sbarbati A, Schena F, Marengo E, Krampera M, Galiè M. Interferon regulatory factor 7 impairs cellular metabolism in aging adipose-derived stromal cells. J Cell Sci 2021; 134:jcs256230. [PMID: 34096605 DOI: 10.1242/jcs.256230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 04/26/2021] [Indexed: 11/20/2022] Open
Abstract
Dysregulated immunity and widespread metabolic dysfunctions are the most relevant hallmarks of the passing of time over the course of adult life, and their combination at midlife is strongly related to increased vulnerability to diseases; however, the causal connection between them remains largely unclear. By combining multi-omics and functional analyses of adipose-derived stromal cells established from young (1 month) and midlife (12 months) mice, we show that an increase in expression of interferon regulatory factor 7 (IRF7) during adult life drives major metabolic changes, which include impaired mitochondrial function, altered amino acid biogenesis and reduced expression of genes involved in branched-chain amino acid (BCAA) degradation. Our results draw a new paradigm of aging as the 'sterile' activation of a cell-autonomous pathway of self-defense and identify a crucial mediator of this pathway, IRF7, as driver of metabolic dysfunction with age.
Collapse
Affiliation(s)
- Alice Nodari
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| | - Ilaria Scambi
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| | - Daniele Peroni
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Elisa Calabria
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| | - Donatella Benati
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| | - Silvia Mannucci
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| | - Marcello Manfredi
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 28100 Alessandria, Italy
- Center for Translational Research on Autoimmune and Allergic Disease - CAAD, University of Piemonte Orientale, 28100 Novara, Italy
| | - Andrea Frontini
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 20121 Ancona, Italy
| | - Silvia Visonà
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy
| | - Andrea Bozzato
- Department of Biomedical Sciences and Biotechnology, University of Brescia, 25123 Brescia, Italy
| | - Andrea Sbarbati
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| | - Federico Schena
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| | - Emilio Marengo
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 28100 Alessandria, Italy
- Center for Translational Research on Autoimmune and Allergic Disease - CAAD, University of Piemonte Orientale, 28100 Novara, Italy
| | - Mauro Krampera
- Department of Medicine, Section of Hematology, Stem Cell Research Laboratory, University of Verona, 37134 Verona, Italy
| | - Mirco Galiè
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| |
Collapse
|
92
|
Huang S, Zhu S, Kumar P, MacMicking JD. A phase-separated nuclear GBPL circuit controls immunity in plants. Nature 2021; 594:424-429. [PMID: 34040255 PMCID: PMC8478157 DOI: 10.1038/s41586-021-03572-6] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 04/21/2021] [Indexed: 02/04/2023]
Abstract
Liquid-liquid phase separation (LLPS) has emerged as a central paradigm for understanding how membraneless organelles compartmentalize diverse cellular activities in eukaryotes1-3. Here we identify a superfamily of plant guanylate-binding protein (GBP)-like GTPases (GBPLs) that assemble LLPS-driven condensates within the nucleus to protect against infection and autoimmunity. In Arabidopsis thaliana, two members of this family-GBPL1 and GBPL3-undergo phase-transition behaviour to control transcriptional responses as part of an allosteric switch that is triggered by exposure to biotic stress. GBPL1, a pseudo-GTPase, sequesters catalytically active GBPL3 under basal conditions but is displaced by GBPL3 LLPS when it enters the nucleus following immune cues to drive the formation of unique membraneless organelles termed GBPL defence-activated condensates (GDACs) that we visualized by in situ cryo-electron tomography. Within these mesoscale GDAC structures, native GBPL3 directly bound defence-gene promoters and recruited specific transcriptional coactivators of the Mediator complex and RNA polymerase II machinery to massively reprogram host gene expression for disease resistance. Together, our study identifies a GBPL circuit that reinforces the biological importance of phase-separated condensates, in this case, as indispensable players in plant defence.
Collapse
Affiliation(s)
- Shuai Huang
- Howard Hughes Medical Institute, New Haven, CT, USA.,Yale Systems Biology Institute, West Haven, CT, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.,Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Shiwei Zhu
- Howard Hughes Medical Institute, New Haven, CT, USA.,Yale Systems Biology Institute, West Haven, CT, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.,Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Pradeep Kumar
- Howard Hughes Medical Institute, New Haven, CT, USA.,Yale Systems Biology Institute, West Haven, CT, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.,Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - John D. MacMicking
- Howard Hughes Medical Institute, New Haven, CT, USA.,Yale Systems Biology Institute, West Haven, CT, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.,Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA.,Correspondence and requests for materials should be addressed to J.D.M.,
| |
Collapse
|
93
|
Havrdová M, Urbančič I, Bartoň Tománková K, Malina L, Štrancar J, Bourlinos AB. Self-Targeting of Carbon Dots into the Cell Nucleus: Diverse Mechanisms of Toxicity in NIH/3T3 and L929 Cells. Int J Mol Sci 2021; 22:ijms22115608. [PMID: 34070594 PMCID: PMC8198156 DOI: 10.3390/ijms22115608] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/16/2022] Open
Abstract
It is important to understand the nanomaterials intracellular trafficking and distribution and investigate their targeting into the nuclear area in the living cells. In our previous study, we firstly observed penetration of nonmodified positively charged carbon dots decorated with quaternary ammonium groups (QCDs) into the nucleus of mouse NIH/3T3 fibroblasts. Thus, in this work, we focused on deeper study of QCDs distribution inside two healthy mouse NIH/3T3 and L929 cell lines by fluorescence microspectroscopy and performed a comprehensive cytotoxic and DNA damage measurements. Real-time penetration of QCDs across the plasma cell membrane was recorded, concentration dependent uptake was determined and endocytic pathways were characterized. We found out that the QCDs concentration of 200 µg/mL is close to saturation and subsequently, NIH/3T3 had a different cell cycle profile, however, no significant changes in viability (not even in the case with QCDs in the nuclei) and DNA damage. In the case of L929, the presence of QCDs in the nucleus evoked a cellular death. Intranuclear environment of NIH/3T3 cells affected fluorescent properties of QCDs and evoked fluorescence blue shifts. Studying the intracellular interactions with CDs is essential for development of future applications such as DNA sensing, because CDs as DNA probes have not yet been developed.
Collapse
Affiliation(s)
- Markéta Havrdová
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University, Křížkovského 511/8, 779 00 Olomouc, Czech Republic
- Correspondence: ; Tel.: +420-58-563-4384
| | - Iztok Urbančič
- Laboratory of Biophysics, Condensed Matter Physics Department, “Jozef Stefan” Institute, Jamova 39, 1000 Ljubljana, Slovenia; (I.U.); (J.Š.)
| | - Kateřina Bartoň Tománková
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Institute of Translation Medicine, Palacký University in Olomouc, Hněvotínská 3, 775 15 Olomouc, Czech Republic; (K.B.T.); (L.M.)
| | - Lukáš Malina
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Institute of Translation Medicine, Palacký University in Olomouc, Hněvotínská 3, 775 15 Olomouc, Czech Republic; (K.B.T.); (L.M.)
| | - Janez Štrancar
- Laboratory of Biophysics, Condensed Matter Physics Department, “Jozef Stefan” Institute, Jamova 39, 1000 Ljubljana, Slovenia; (I.U.); (J.Š.)
| | | |
Collapse
|
94
|
Abstract
Salmonella is an intracellular pathogen of a substantial global health concern. In order to identify key players involved in Salmonella infection, we performed a global host phosphoproteome analysis subsequent to bacterial infection. Thereby, we identified the kinase SIK2 as a central component of the host defense machinery upon Salmonella infection. SIK2 depletion favors the escape of bacteria from the Salmonella-containing vacuole (SCV) and impairs Xenophagy, resulting in a hyperproliferative phenotype. Mechanistically, SIK2 associates with actin filaments under basal conditions; however, during bacterial infection, SIK2 is recruited to the SCV together with the elements of the actin polymerization machinery (Arp2/3 complex and Formins). Notably, SIK2 depletion results in a severe pathological cellular actin nucleation and polymerization defect upon Salmonella infection. We propose that SIK2 controls the formation of a protective SCV actin shield shortly after invasion and orchestrates the actin cytoskeleton architecture in its entirety to control an acute Salmonella infection after bacterial invasion.
Collapse
|
95
|
Zhu GD, Cao XJ, Li YP, Li JX, Leng ZJ, Xie LM, Guo XG. Identification of differentially expressed genes and signaling pathways in human conjunctiva and reproductive tract infected with Chlamydia trachomatis. Hum Genomics 2021; 15:22. [PMID: 33875006 PMCID: PMC8056519 DOI: 10.1186/s40246-021-00313-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Currently, Chlamydia trachomatis-specific host defense mechanisms in humans remain poorly defined. To study the characteristics of host cells infected early with Chlamydia trachomatis, we used bioinformatics methods to analyze the RNA transcription profiles of the conjunctiva, fallopian tubes, and endometrium in humans infected with Chlamydia trachomatis. METHOD The gene expression profiles of GSE20430, GSE20436, GSE26692, and GSE41075 were downloaded from the Gene Expression Synthesis (GEO) database. Then, we obtained the differentially expressed genes (DEGs) through the R 4.0.1 software. STRING was used to construct protein-protein interaction (PPI) networks; then, the Cytoscape 3.7.2 software was used to visualize the PPI and screen hub genes. GraphPad Prism 8.0 software was used to verify the expression of the hub gene. In addition, the gene-miRNA interaction was constructed on the NetworkAnalyst 3.0 platform using the miRTarBase v8.0 database. RESULTS A total of 600 and 135 DEGs were screened out in the conjunctival infection group and the reproductive tract infection group, respectively. After constructing a PPI network and verifying the hub genes, CSF2, CD40, and CSF3 in the reproductive tract infection group proved to have considerable statistical significance. CONCLUSION In our research, the key genes in the biological process of reproductive tract infection with Chlamydia trachomatis were clarified through bioinformatics analysis. These hub genes may be further used in clinical treatment and clinical diagnosis.
Collapse
Affiliation(s)
- Guo-Dong Zhu
- Departments of Geriatrics and Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Xun-Jie Cao
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Ya-Ping Li
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
- Department of Clinical Medicine, The Second Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Jia-Xin Li
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Zi-Jian Leng
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Li-Min Xie
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Xu-Guang Guo
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China.
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, ,510150, China.
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
96
|
Soe YM, Bedoui S, Stinear TP, Hachani A. Intracellular Staphylococcus aureus and host cell death pathways. Cell Microbiol 2021; 23:e13317. [PMID: 33550697 DOI: 10.1111/cmi.13317] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 12/13/2022]
Abstract
Staphylococcus aureus is a major opportunistic human pathogen that is globally prevalent. Although S. aureus and humans may have co-evolved to the point of commensalism, the bacterium is equipped with virulence factors causing devastating infections. The adoption of an intracellular lifestyle by S. aureus is an important facet of its pathogenesis. Occupying a privileged intracellular compartment permits evasion from the bactericidal actions of host immunity and antibiotics. However, this localization exposes S. aureus to cell-intrinsic processes comprising autophagy, metabolic challenges and clearance mechanisms orchestrated by host programmed cell death pathways (PCDs), including apoptosis, pyroptosis and necroptosis. Mounting evidence suggests that S. aureus deploys pathoadaptive mechanisms that modulate the expression of its virulence factors to prevent elimination through PCD pathways. In this review, we critically analyse the current literature on the interplay between S. aureus virulence factors with the key, intertwined nodes of PCD. We discuss how S. aureus adaptation to the human host plays an essential role in the evasion of PCD, and we consider future directions to study S. aureus-PCD interactions.
Collapse
Affiliation(s)
- Ye Mon Soe
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Abderrahman Hachani
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
97
|
Xu L, Ma Z, Li Y, Pang Z, Xiao S. Antibody dependent enhancement: Unavoidable problems in vaccine development. Adv Immunol 2021; 151:99-133. [PMID: 34656289 PMCID: PMC8438590 DOI: 10.1016/bs.ai.2021.08.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In some cases, antibodies can enhance virus entry and replication in cells. This phenomenon is called antibody-dependent infection enhancement (ADE). ADE not only promotes the virus to be recognized by the target cell and enters the target cell, but also affects the signal transmission in the target cell. Early formalin-inactivated virus vaccines such as aluminum adjuvants (RSV and measles) have been shown to induce ADE. Although there is no direct evidence that there is ADE in COVID-19, this potential risk is a huge challenge for prevention and vaccine development. This article focuses on the virus-induced ADE phenomenon and its molecular mechanism. It also summarizes various attempts in vaccine research and development to eliminate the ADE phenomenon, and proposes to avoid ADE in vaccine development from the perspective of antigens and adjuvants.
Collapse
|
98
|
Dellac S, Ben-Dov H, Raanan A, Saleem H, Zamostiano R, Semyatich R, Lavi S, Witz IP, Bacharach E, Ehrlich M. Constitutive low expression of antiviral effectors sensitizes melanoma cells to a novel oncolytic virus. Int J Cancer 2020; 148:2321-2334. [PMID: 33197301 DOI: 10.1002/ijc.33401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 12/15/2022]
Abstract
STAT1 is a critical effector and a target gene of interferon (IFN) signaling, and thus a central mediator of antiviral responses. As both a mediator and a target of IFN signals, STAT1 expression reports on, and determines IFN activity. Gene expression analyses of melanoma patient samples revealed varied levels of STAT1 expression, which highly correlated with expression of >700 genes. The ability of oncolytic viruses to exploit tumor-induced defects to antiviral responses suggests that oncolytic viruses may efficiently target a subset of melanomas, yet these should be defined. We modeled this scenario with murine B16F10 melanomas, immortalized skin fibroblasts as controls and a novel oncolytic virus, EHDV-TAU. In B16F10 cells, constitutive low expression of STAT1 and its target genes, which included intracellular pattern recognition receptors (PRRs), correlated with their inability to mount IFN-based antiviral responses upon EHDV-TAU challenge, and with potency of EHDV-TAU-induced oncolysis. This underexpression of interferon stimulated genes (ISGs) and PRRs, and the inability of EHDV-TAU to induce their expression, were reversed by epigenetic modifiers, suggesting epigenetic silencing as a basis for their underexpression. Despite their inability to mount IFN/STAT-based responses upon viral infection, EHDV-TAU infected B16F10 cells secreted immune-stimulatory chemokines. Accordingly, in vivo, EHDV-TAU enhanced intratumoral infiltration of cytotoxic T-cells and reduced growth of local and distant tumors. We propose that "STAT1 signatures" should guide melanoma virotherapy treatments, and that oncolytic viruses such as EHDV-TAU have the potential to exploit the cellular context of low-STAT1 tumors.
Collapse
Affiliation(s)
- Sarah Dellac
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Hamutal Ben-Dov
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ayala Raanan
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Hanna Saleem
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Rachel Zamostiano
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Rinat Semyatich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Sara Lavi
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Isaac P Witz
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Eran Bacharach
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
99
|
Fischer TD, Wang C, Padman BS, Lazarou M, Youle RJ. STING induces LC3B lipidation onto single-membrane vesicles via the V-ATPase and ATG16L1-WD40 domain. J Cell Biol 2020; 219:211541. [PMID: 33201170 PMCID: PMC7716379 DOI: 10.1083/jcb.202009128] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 02/08/2023] Open
Abstract
Following the detection of cytosolic double-stranded DNA from viral or bacterial infection in mammalian cells, cyclic dinucleotide activation of STING induces interferon β expression to initiate innate immune defenses. STING activation also induces LC3B lipidation, a classical but equivocal marker of autophagy, that promotes a cell-autonomous antiviral response that arose before evolution of the interferon pathway. We report that STING activation induces LC3B lipidation onto single-membrane perinuclear vesicles mediated by ATG16L1 via its WD40 domain, bypassing the requirement of canonical upstream autophagy machinery. This process is blocked by bafilomycin A1 that binds and inhibits the vacuolar ATPase (V-ATPase) and by SopF, a bacterial effector that catalytically modifies the V-ATPase to inhibit LC3B lipidation via ATG16L1. These results indicate that activation of the cGAS-STING pathway induces V-ATPase-dependent LC3B lipidation that may mediate cell-autonomous host defense, an unanticipated mechanism that is distinct from LC3B lipidation onto double-membrane autophagosomes.
Collapse
Affiliation(s)
- Tara D. Fischer
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Chunxin Wang
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Benjamin S. Padman
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Michael Lazarou
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Richard J. Youle
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD,Correspondence to Richard J. Youle:
| |
Collapse
|
100
|
Grogan LF, Humphries JE, Robert J, Lanctôt CM, Nock CJ, Newell DA, McCallum HI. Immunological Aspects of Chytridiomycosis. J Fungi (Basel) 2020; 6:jof6040234. [PMID: 33086692 PMCID: PMC7712659 DOI: 10.3390/jof6040234] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 12/27/2022] Open
Abstract
Amphibians are currently the most threatened vertebrate class, with the disease chytridiomycosis being a major contributor to their global declines. Chytridiomycosis is a frequently fatal skin disease caused by the fungal pathogens Batrachochytrium dendrobatidis (Bd) and Batrachochytrium salamandrivorans (Bsal). The severity and extent of the impact of the infection caused by these pathogens across modern Amphibia are unprecedented in the history of vertebrate infectious diseases. The immune system of amphibians is thought to be largely similar to that of other jawed vertebrates, such as mammals. However, amphibian hosts are both ectothermic and water-dependent, which are characteristics favouring fungal proliferation. Although amphibians possess robust constitutive host defences, Bd/Bsal replicate within host cells once these defences have been breached. Intracellular fungal localisation may contribute to evasion of the induced innate immune response. Increasing evidence suggests that once the innate defences are surpassed, fungal virulence factors suppress the targeted adaptive immune responses whilst promoting an ineffectual inflammatory cascade, resulting in immunopathology and systemic metabolic disruption. Thus, although infections are contained within the integument, crucial homeostatic processes become compromised, leading to mortality. In this paper, we present an integrated synthesis of amphibian post-metamorphic immunological responses and the corresponding outcomes of infection with Bd, focusing on recent developments within the field and highlighting future directions.
Collapse
Affiliation(s)
- Laura F. Grogan
- Environmental Futures Research Institute and School of Environment and Science, Griffith University, Southport, QLD 4222, Australia;
- Forest Research Centre, School of Environment, Science and Engineering, Southern Cross University, Lismore, NSW 2480, Australia; (J.E.H.); (D.A.N.)
- Correspondence:
| | - Josephine E. Humphries
- Forest Research Centre, School of Environment, Science and Engineering, Southern Cross University, Lismore, NSW 2480, Australia; (J.E.H.); (D.A.N.)
| | - Jacques Robert
- University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Chantal M. Lanctôt
- Australian Rivers Institute, Griffith University, Southport, QLD 4222, Australia;
| | - Catherine J. Nock
- Southern Cross Plant Science, Southern Cross University, Lismore, NSW 2480, Australia;
| | - David A. Newell
- Forest Research Centre, School of Environment, Science and Engineering, Southern Cross University, Lismore, NSW 2480, Australia; (J.E.H.); (D.A.N.)
| | - Hamish I. McCallum
- Environmental Futures Research Institute and School of Environment and Science, Griffith University, Southport, QLD 4222, Australia;
| |
Collapse
|