51
|
TAK1 inhibition elicits mitochondrial ROS to block intracellular bacterial colonization. Proc Natl Acad Sci U S A 2021; 118:2023647118. [PMID: 34161265 DOI: 10.1073/pnas.2023647118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Mitogen-activated protein kinase kinase kinase 7 (MAP3K7), known as TAK1, is an intracellular signaling intermediate of inflammatory responses. However, a series of mouse Tak1 gene deletion analyses have revealed that ablation of TAK1 does not prevent but rather elicits inflammation, which is accompanied by elevation of reactive oxygen species (ROS). This has been considered a consequence of impaired TAK1-dependent maintenance of tissue integrity. Contrary to this view, here we propose that TAK1 inhibition-induced ROS are an active cellular process that targets intracellular bacteria. Intracellular bacterial effector proteins such as Yersinia's outer membrane protein YopJ are known to inhibit TAK1 to circumvent the inflammatory host responses. We found that such TAK1 inhibition induces mitochondrial-derived ROS, which effectively destroys intracellular bacteria. Two cell death-signaling molecules, caspase 8 and RIPK3, cooperatively participate in TAK1 inhibition-induced ROS and blockade of intracellular bacterial growth. Our results reveal a previously unrecognized host defense mechanism, which is initiated by host recognition of pathogen-induced impairment in a host protein, TAK1, but not directly of pathogens.
Collapse
|
52
|
Yang J, Zhou M, Zhong Y, Xu L, Zeng C, Zhao X, Zhang M. Gene duplication and adaptive evolution of Toll-like receptor genes in birds. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 119:103990. [PMID: 33422554 DOI: 10.1016/j.dci.2020.103990] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/27/2020] [Accepted: 12/27/2020] [Indexed: 06/12/2023]
Abstract
Toll-like receptors (TLRs) play an important role in innate immune through recognizes pathogens. In order to reveal the evolutionary patterns and adaptive evolution of avian TLRs, we examined 66 representative bird species in 26 orders. Phylogenetic results indicated that TLR1A and TLR1B may have differentiated functionally. Evolutionary analysis showed that the TLR genes in birds under strong Purification selection (0.165-0.4265). A total of 126 common positively selected codons were identified in 10 TLR genes of avian, and most sites were located in the extracellular leucine-rich repeat (LRR) functional domains, and both environment and feeding habits were external factors driving the evolution of avian TLR genes. Environmental pressures had a greater effect on TLR1B, TLR2B, TLR3 and TLR4, while feeding habits were active in affecting TLR2A, TLR2B, TLR15 and TLR21. Our data suggested that TLR genes have been subjected to different selective pressures in the diversification of birds and that these changes enabled them to respond differently to pathogens from diverse sources.
Collapse
Affiliation(s)
- Jiandong Yang
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Southwest China Wildlife Rsources Conservation (Ministry of Education), China West Normal University, Nanchong, 637009, PR China.
| | - Ming Zhou
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, 611130, China; Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Yun Zhong
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Liqun Xu
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Changjun Zeng
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiaoling Zhao
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Ming Zhang
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| |
Collapse
|
53
|
Zhang X, Harding BW, Aggad D, Courtine D, Chen JX, Pujol N, Ewbank JJ. Antagonistic fungal enterotoxins intersect at multiple levels with host innate immune defences. PLoS Genet 2021; 17:e1009600. [PMID: 34166401 PMCID: PMC8263066 DOI: 10.1371/journal.pgen.1009600] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 07/07/2021] [Accepted: 05/12/2021] [Indexed: 12/25/2022] Open
Abstract
Animals and plants need to defend themselves from pathogen attack. Their defences drive innovation in virulence mechanisms, leading to never-ending cycles of co-evolution in both hosts and pathogens. A full understanding of host immunity therefore requires examination of pathogen virulence strategies. Here, we take advantage of the well-studied innate immune system of Caenorhabditis elegans to dissect the action of two virulence factors from its natural fungal pathogen Drechmeria coniospora. We show that these two enterotoxins have strikingly different effects when expressed individually in the nematode epidermis. One is able to interfere with diverse aspects of host cell biology, altering vesicle trafficking and preventing the key STAT-like transcription factor STA-2 from activating defensive antimicrobial peptide gene expression. The second increases STA-2 levels in the nucleus, modifies the nucleolus, and, potentially as a consequence of a host surveillance mechanism, causes increased defence gene expression. Our results highlight the remarkably complex and potentially antagonistic mechanisms that come into play in the interaction between co-evolved hosts and pathogens.
Collapse
Affiliation(s)
- Xing Zhang
- Aix Marseille Univ, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
| | - Benjamin W. Harding
- Aix Marseille Univ, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
| | - Dina Aggad
- Aix Marseille Univ, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
| | - Damien Courtine
- Aix Marseille Univ, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
| | | | - Nathalie Pujol
- Aix Marseille Univ, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
| | - Jonathan J. Ewbank
- Aix Marseille Univ, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
| |
Collapse
|
54
|
Alhazmi HA, Najmi A, Javed SA, Sultana S, Al Bratty M, Makeen HA, Meraya AM, Ahsan W, Mohan S, Taha MME, Khalid A. Medicinal Plants and Isolated Molecules Demonstrating Immunomodulation Activity as Potential Alternative Therapies for Viral Diseases Including COVID-19. Front Immunol 2021; 12:637553. [PMID: 34054806 PMCID: PMC8155592 DOI: 10.3389/fimmu.2021.637553] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/26/2021] [Indexed: 12/19/2022] Open
Abstract
Plants have been extensively studied since ancient times and numerous important chemical constituents with tremendous therapeutic potential are identified. Attacks of microorganisms including viruses and bacteria can be counteracted with an efficient immune system and therefore, stimulation of body's defense mechanism against infections has been proven to be an effective approach. Polysaccharides, terpenoids, flavonoids, alkaloids, glycosides, and lactones are the important phytochemicals, reported to be primarily responsible for immunomodulation activity of the plants. These phytochemicals may act as lead molecules for the development of safe and effective immunomodulators as potential remedies for the prevention and cure of viral diseases. Natural products are known to primarily modulate the immune system in nonspecific ways. A number of plant-based principles have been identified and isolated with potential immunomodulation activity which justify their use in traditional folklore medicine and can form the basis of further specified research. The aim of the current review is to describe and highlight the immunomodulation potential of certain plants along with their bioactive chemical constituents. Relevant literatures of recent years were searched from commonly employed scientific databases on the basis of their ethnopharmacological use. Most of the plants displaying considerable immunomodulation activity are summarized along with their possible mechanisms. These discussions shall hopefully elicit the attention of researchers and encourage further studies on these plant-based immunomodulation products as potential therapy for the management of infectious diseases, including viral ones such as COVID-19.
Collapse
Affiliation(s)
- Hassan A. Alhazmi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Asim Najmi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Sadique A. Javed
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Shahnaz Sultana
- Department of Pharmacognosy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Mohammed Al Bratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Hafiz A. Makeen
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Abdulkarim M. Meraya
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Waquar Ahsan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Manal M. E. Taha
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
55
|
Abstract
Commensal microbial communities have immense effects on their vertebrate hosts, contributing to a number of physiological functions, as well as host fitness. In particular, host immunity is strongly linked to microbiota composition through poorly understood bi-directional links. Gene expression may be a potential mediator of these links between microbial communities and host function. However, few studies have investigated connections between microbiota composition and expression of host immune genes in complex systems. Here, we leverage a large study of laboratory-raised fish from the species Gasterosteus aculeatus (three-spined stickleback) to document correlations between gene expression and microbiome composition. First, we examined correlations between microbiome alpha diversity and gene expression. Our results demonstrate robust positive associations between microbial alpha diversity and expression of host immune genes. Next, we examined correlations between host gene expression and abundance of microbial taxa. We identified 15 microbial families that were highly correlated with host gene expression. These families were all tightly correlated with host expression of immune genes and processes, falling into one of three categories—those positively correlated, negatively correlated, and neutrally related to immune processes. Furthermore, we highlight several important immune processes that are commonly associated with the abundance of these taxa, including both macrophage and B cell functions. Further functional characterization of microbial taxa will help disentangle the mechanisms of the correlations described here. In sum, our study supports prevailing hypotheses of intimate links between host immunity and gut microbiome composition.
Collapse
|
56
|
Abstract
Some bacterial pathogens utilize cell-cell communication systems, such as quorum sensing (QS), to coordinate genetic programs during host colonization and infection. The human-restricted pathosymbiont Streptococcus pyogenes (group A streptococcus [GAS]) uses the Rgg2/Rgg3 QS system to modify the bacterial surface, enabling biofilm formation and lysozyme resistance. Here, we demonstrate that innate immune cell responses to GAS are substantially altered by the QS status of the bacteria. We found that macrophage activation, stimulated by multiple agonists and assessed by cytokine production and NF-κB activity, was substantially suppressed upon interaction with QS-active GAS but not QS-inactive bacteria. Neither macrophage viability nor bacterial adherence, internalization, or survival were altered by the QS activation status, yet tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), and interferon beta (IFN-β) levels and NF-κB reporter activity were drastically lower following infection with QS-active GAS. Suppression required contact between viable bacteria and macrophages. A QS-regulated biosynthetic gene cluster (BGC) in the GAS genome, encoding several putative enzymes, was also required for macrophage modulation. Our findings suggest a model wherein upon contact with macrophages, QS-active GAS produce a BGC-derived factor capable of suppressing inflammatory responses. The suppressive capability of QS-active GAS is abolished after treatment with a specific QS inhibitor. These observations suggest that interfering with the ability of bacteria to collaborate via QS can serve as a strategy to counteract microbial efforts to manipulate host defenses.
Collapse
|
57
|
Prediction and Activity of a Cationic α-Helix Antimicrobial Peptide ZM-804 from Maize. Int J Mol Sci 2021; 22:ijms22052643. [PMID: 33807972 PMCID: PMC7961353 DOI: 10.3390/ijms22052643] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
Antimicrobial peptides (AMPs) are small molecules consisting of less than fifty residues of amino acids. Plant AMPs establish the first barrier of defense in the innate immune system in response to invading pathogens. The purpose of this study was to isolate new AMPs from the Zea mays L. inbred line B73 and investigate their antimicrobial activities and mechanisms against certain essential plant pathogenic bacteria. In silico, the Collection of Anti-Microbial Peptides (CAMPR3), a computational AMP prediction server, was used to screen a cDNA library for AMPs. A ZM-804 peptide, isolated from the Z. mays L. inbred line B73 cDNA library, was predicted as a new cationic AMP with high prediction values. ZM-804 was tested against eleven pathogens of Gram-negative and Gram-positive bacteria and exhibited high antimicrobial activities as determined by the minimal inhibitory concentrations (MICs) and the minimum bactericidal concentrations (MBCs). A confocal laser scanning microscope observation showed that the ZM-804 AMP targets bacterial cell membranes. SEM and TEM images revealed the disruption and damage of the cell membrane morphology of Clavibacter michiganensis subsp. michiganensis and Pseudomonas syringae pv. tomato (Pst) DC3000 caused by ZM-804. In planta, ZM-804 demonstrated antimicrobial activity and prevented the infection of tomato plants by Pst DC3000. Moreover, four virulent phytopathogenic bacteria were prevented from inducing hypersensitive response (HR) in tobacco leaves in response to low ZM-804 concentrations. ZM-804 exhibits low hemolytic activity against mouse red blood cells (RBCs) and is relatively safe for mammalian cells. In conclusion, the ZM-804 peptide has a strong antibacterial activity and provides an alternative tool for plant disease control. Additionally, the ZM-804 peptide is considered a promising candidate for human and animal drug development.
Collapse
|
58
|
Merselis LC, Rivas ZP, Munson GP. Breaching the Bacterial Envelope: The Pivotal Role of Perforin-2 (MPEG1) Within Phagocytes. Front Immunol 2021; 12:597951. [PMID: 33692780 PMCID: PMC7937864 DOI: 10.3389/fimmu.2021.597951] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 01/04/2021] [Indexed: 11/13/2022] Open
Abstract
The membrane attack complex (MAC) of the complement system and Perforin-1 are well characterized innate immune effectors. MAC is composed of C9 and other complement proteins that target the envelope of gram-negative bacteria. Perforin-1 is deployed when killer lymphocytes degranulate to destroy virally infected or cancerous cells. These molecules polymerize with MAC-perforin/cholesterol-dependent cytolysin (MACPF/CDC) domains of each monomer deploying amphipathic β-strands to form pores through target lipid bilayers. In this review we discuss one of the most recently discovered members of this family; Perforin-2, the product of the Mpeg1 gene. Since their initial description more than 100 years ago, innumerable studies have made macrophages and other phagocytes some of the best understood cells of the immune system. Yet remarkably it was only recently revealed that Perforin-2 underpins a pivotal function of phagocytes; the destruction of phagocytosed microbes. Several studies have established that phagocytosed bacteria persist and in some cases flourish within phagocytes that lack Perforin-2. When challenged with either gram-negative or gram-positive pathogens Mpeg1 knockout mice succumb to infectious doses that the majority of wild-type mice survive. As expected by their immunocompromised phenotype, bacterial pathogens replicate and disseminate to deeper tissues of Mpeg1 knockout mice. Thus, this evolutionarily ancient gene endows phagocytes with potent bactericidal capability across taxa spanning sponges to humans. The recently elucidated structures of mammalian Perforin-2 reveal it to be a homopolymer that depends upon low pH, such as within phagosomes, to transition to its membrane-spanning pore conformation. Clinical manifestations of Mpeg1 missense mutations further highlight the pivotal role of Perforin-2 within phagocytes. Controversies and gaps within the field of Perforin-2 research are also discussed as well as animal models that may be used to resolve the outstanding issues. Our review concludes with a discussion of bacterial counter measures against Perforin-2.
Collapse
Affiliation(s)
- Leidy C Merselis
- Department of Microbiology and Immunology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Zachary P Rivas
- Department of Microbiology and Immunology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - George P Munson
- Department of Microbiology and Immunology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
59
|
Bastounis EE, Serrano-Alcalde F, Radhakrishnan P, Engström P, Gómez-Benito MJ, Oswald MS, Yeh YT, Smith JG, Welch MD, García-Aznar JM, Theriot JA. Mechanical competition triggered by innate immune signaling drives the collective extrusion of bacterially infected epithelial cells. Dev Cell 2021; 56:443-460.e11. [PMID: 33621492 PMCID: PMC7982222 DOI: 10.1016/j.devcel.2021.01.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/02/2020] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Abstract
Intracellular pathogens alter their host cells' mechanics to promote dissemination through tissues. Conversely, host cells may respond to the presence of pathogens by altering their mechanics to limit infection. Here, we monitored epithelial cell monolayers infected with intracellular bacterial pathogens, Listeria monocytogenes or Rickettsia parkeri, over days. Under conditions in which these pathogens trigger innate immune signaling through NF-κB and use actin-based motility to spread non-lytically intercellularly, we found that infected cell domains formed three-dimensional mounds. These mounds resulted from uninfected cells moving toward the infection site, collectively squeezing the softer and less contractile infected cells upward and ejecting them from the monolayer. Bacteria in mounds were less able to spread laterally in the monolayer, limiting the growth of the infection focus, while extruded infected cells underwent cell death. Thus, the coordinated forceful action of uninfected cells actively eliminates large domains of infected cells, consistent with this collective cell response representing an innate immunity-driven process.
Collapse
Affiliation(s)
- Effie E Bastounis
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | | | - Prathima Radhakrishnan
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Patrik Engström
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - María J Gómez-Benito
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza 50009, Spain
| | - Mackenzi S Oswald
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Yi-Ting Yeh
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jason G Smith
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Matthew D Welch
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - José M García-Aznar
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza 50009, Spain
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
60
|
Mishra R, Krishnamoorthy P, Kumar H. MicroRNA-30e-5p Regulates SOCS1 and SOCS3 During Bacterial Infection. Front Cell Infect Microbiol 2021; 10:604016. [PMID: 33585275 PMCID: PMC7873556 DOI: 10.3389/fcimb.2020.604016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Host innate immunity is the major player against continuous microbial infection. Various pathogenic bacteria adopt the strategies to evade the immunity and show resistance toward the various established therapies. Despite the advent of many antibiotics for bacterial infections, there is a substantial need for the host-directed therapies (HDTs) to combat the infection. HDTs are recently being adopted to be useful in eradicating intracellular bacterial infection. Changing the innate immune responses of the host cells alters pathogen's ability to reside inside the cell. MicroRNAs are the small non-coding endogenous molecules and post-transcriptional regulators to target the 3'UTR of the messenger RNA. They are reported to modulate the host's immune responses during bacterial infections. Exploiting microRNAs as a therapeutic candidate in HDTs upon bacterial infection is still in its infancy. Here, initially, we re-analyzed the publicly available transcriptomic dataset of macrophages, infected with different pathogenic bacteria and identified significant genes and microRNAs common to the differential infections. We thus identified and miR-30e-5p, to be upregulated in different bacterial infections which enhances innate immunity to combat bacterial replication by targeting key negative regulators such as SOCS1 and SOCS3 of innate immune signaling pathways. Therefore, we propose miR-30e-5p as one of the potential candidates to be considered for additional clinical validation toward HDTs.
Collapse
Affiliation(s)
- Richa Mishra
- Laboratory of Immunology and Infectious Disease Biology, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, India
| | - Pandikannan Krishnamoorthy
- Laboratory of Immunology and Infectious Disease Biology, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, India
| | - Himanshu Kumar
- Laboratory of Immunology and Infectious Disease Biology, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, India.,WPI Immunology, Frontier Research Centre, Osaka University, Osaka, Japan
| |
Collapse
|
61
|
Zhang Z, Wang M, Zhang Y, Zhang Y, Bartkuhn M, Markmann M, Hossain H, Chakraborty T, Hake SB, Jia Z, Meinhardt A, Bhushan S. Uropathogenic Escherichia coli Virulence Factor α-Hemolysin Reduces Histone Acetylation to Inhibit Expression of Proinflammatory Cytokine Genes. J Infect Dis 2021; 223:1040-1051. [PMID: 33453118 DOI: 10.1093/infdis/jiab018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022] Open
Abstract
Urinary tract infections are common and costly diseases affecting millions of people. Uropathogenic Escherichia coli (UPEC) is a primary cause of these infections and has developed multiple strategies to avoid the host immune response. Here, we dissected the molecular mechanisms underpinning UPEC inhibition of inflammatory cytokine in vitro and in vivo. We found that UPEC infection simulates nuclear factor-κB activation but does not result in transcription of cytokine genes. Instead, UPEC-mediated suppression of the metabolic enzyme ATP citrate lyase results in decreased acetyl-CoA levels, leading to reduced H3K9 histone acetylation in the promotor region of CXCL8. These effects were dependent on the UPEC virulence factor α-hemolysin and were reversed by exogenous acetate. In a murine cystitis model, prior acetate supplementation rapidly resolved UPEC-elicited immune responses and improved tissue recovery. Thus, upon infection, UPEC rearranges host cell metabolism to induce chromatin remodeling processes that subvert expression of host innate immune response genes.
Collapse
Affiliation(s)
- Zhengguo Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Ming Wang
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University of Giessen, Giessen, Germany.,Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiming Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Marek Bartkuhn
- Institute for Genetics, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Melanie Markmann
- Institute of Medical Microbiology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Hamid Hossain
- Klinikum St Marien Amberg, Amberg and Kliniken Nordoberpfalz AG, Weiden, Germany
| | - Trinad Chakraborty
- Institute of Medical Microbiology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Sandra B Hake
- Institute for Genetics, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Zhankui Jia
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Andreas Meinhardt
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Sudhanshu Bhushan
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University of Giessen, Giessen, Germany
| |
Collapse
|
62
|
C7ORF41 Regulates Inflammation by Inhibiting NF- κB Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2021; 2021:7413605. [PMID: 33506033 PMCID: PMC7806384 DOI: 10.1155/2021/7413605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 12/02/2020] [Accepted: 12/18/2020] [Indexed: 11/18/2022]
Abstract
Inflammation is an important biological process for eliciting immune responses against physiological and pathological stimuli. Inflammation must be efficiently regulated to ensure homeostasis in the body. Nuclear factor-kappa B (NF-κB) signaling is crucial for inflammatory and immune responses. Aberrant activation of NF-κB signaling leads to development of numerous human diseases. In this study, we investigated the function of chromosome 7 open reading frame 41 (C7ORF41) in NF-κB signaling during inflammation. C7ORF41 was upregulated in cells stimulated with tumor necrosis factor-alpha or lipopolysaccharide. Moreover, overexpression of C7ORF41 inhibited the activation of NF-κB and decreased the expression of its downstream target genes. Notably, small hairpin RNA-mediated depletion of C7ORF41 increased the levels of downstream genes and enabled the activation of NF-κB. In conclusion, C7ORF41 negatively regulated inflammation via NF-κB signaling and p65 phosphorylation in vitro. These findings may help to diagnose and prognosticate inflammatory conditions and may help develop new strategies for the management of inflammation-related diseases.
Collapse
|
63
|
Kloc M, Uosef A, Kubiak JZ, Ghobrial RM. Macrophage Proinflammatory Responses to Microorganisms and Transplanted Organs. Int J Mol Sci 2020; 21:ijms21249669. [PMID: 33352942 PMCID: PMC7766629 DOI: 10.3390/ijms21249669] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/09/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023] Open
Abstract
Tissue-resident macrophages and those conscripted from the blood/bone marrow are professional phagocytes. They play a role in tissue homeostasis, replacement, and healing, and are the first-line responders to microbial (viral, bacterial, and fungi) infections. Intrinsic ameboid-type motility allows non-resident macrophages to move to the site of inflammation or injury, where, in response to the inflammatory milieu they perform the anti-microbial and/or tissue repair functions. Depending on the need and the signaling from the surrounding tissue and other immune cells, macrophages acquire morphologically and functionally different phenotypes, which allow them to play either pro-inflammatory or anti-inflammatory functions. As such, the macrophages are also the major players in the rejection of the transplanted organs making an excellent target for the novel anti-rejection therapies in clinical transplantation. In this review, we describe some of the less covered aspects of macrophage response to microbial infection and organ transplantation.
Collapse
Affiliation(s)
- Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX 77030, USA; (A.U.); (R.M.G.)
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
- MD Anderson Cancer Center, Department of Genetics Houston, The University of Texas, Austin, TX 77030, USA
- Correspondence:
| | - Ahmed Uosef
- The Houston Methodist Research Institute, Houston, TX 77030, USA; (A.U.); (R.M.G.)
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
| | - Jacek Z. Kubiak
- Laboratory of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology (WIHE), 01-163 Warsaw, Poland;
- Cell Cycle Group, Faculty of Medicine, Institute of Genetics and Development of Rennes (IGDR), University Rennes, UMR 6290, CNRS, 35043 Rennes, France
| | - Rafik M. Ghobrial
- The Houston Methodist Research Institute, Houston, TX 77030, USA; (A.U.); (R.M.G.)
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
64
|
de Groot NS, Torrent Burgas M. Bacteria use structural imperfect mimicry to hijack the host interactome. PLoS Comput Biol 2020; 16:e1008395. [PMID: 33275611 PMCID: PMC7744059 DOI: 10.1371/journal.pcbi.1008395] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/16/2020] [Accepted: 09/23/2020] [Indexed: 12/25/2022] Open
Abstract
Bacteria use protein-protein interactions to infect their hosts and hijack fundamental pathways, which ensures their survival and proliferation. Hence, the infectious capacity of the pathogen is closely related to its ability to interact with host proteins. Here, we show that hubs in the host-pathogen interactome are isolated in the pathogen network by adapting the geometry of the interacting interfaces. An imperfect mimicry of the eukaryotic interfaces allows pathogen proteins to actively bind to the host's target while preventing deleterious effects on the pathogen interactome. Understanding how bacteria recognize eukaryotic proteins may pave the way for the rational design of new antibiotic molecules.
Collapse
Affiliation(s)
- Natalia Sanchez de Groot
- Gene Function and Evolution Lab, Centre for Genomic Regulation (CRG), Dr. Aiguader 88, Barcelona, Spain
- * E-mail: (NSdG); (MTB)
| | - Marc Torrent Burgas
- Systems Biology of Infection Lab, Department of Biochemistry and Molecular Biology, Biosciences Faculty, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- * E-mail: (NSdG); (MTB)
| |
Collapse
|
65
|
Toshchakov VY, Javmen A. Targeting the TLR signalosome with TIR domain-derived cell-permeable decoy peptides: the current state and perspectives. Innate Immun 2020; 26:35-47. [PMID: 31955621 PMCID: PMC6974878 DOI: 10.1177/1753425919844310] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The ability to engineer pharmaceuticals that target the signal-dependent
interactions of signaling proteins should revolutionize drug development. One
approach to the rational design of protein interaction inhibitors uses decoy
peptides, i.e. segments of protein primary sequence, which are derived from
interfaces that mediate functional protein interactions. Decoy peptides often
retain the ability of the full-length prototype to bind the docking site of the
folded protein and thereby block the signal transduction. This review summarizes
advances made in the last decade in the development of cell-permeable decoy
peptide (CPDP) inhibitors to target the Toll/IL-1R resistance (TIR)
domain-mediated protein interactions in TLR signaling, in connection with the
recent progress in understanding of the TLR signalosome assembly mechanisms. We
present a large collection of currently available, TIR-targeting CPDPs and
propose their classification based on the types of TIR–TIR interactions they
target. The binding behavior of different CPDP-TIR pairs, studied in cell-based
assays and in binary in vitro systems using recombinant TIR
domains, is also reviewed. The available affinity data provide benchmarks for
rapid preliminary evaluation of future inhibitors. We review literature that
evaluates the in vivo potency of select CPDPs and attempt to
outline the areas of forthcoming progress, towards the development of CPDP-based
TLR inhibitors of pharmaceutical grade.
Collapse
Affiliation(s)
- Vladimir Y Toshchakov
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Artur Javmen
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
66
|
Mei L, Qiu X, Jiang C, Yang A. Host Delipidation Mediated by Bacterial Effectors. Trends Microbiol 2020; 29:238-250. [PMID: 33092951 DOI: 10.1016/j.tim.2020.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/19/2022]
Abstract
Protein lipidation, the covalent attachment of a lipid moiety to a target protein, plays a critical role in many cellular processes in eukaryotic cells. Bacterial pathogens secrete various effectors to subvert the host signaling pathway as a mechanism of microbial pathogenesis. An increasing number of effectors from diverse bacterial pathogens function as cysteine proteases to cause irreversible delipidation of host lipidated proteins. This in turn results in disruption of crucial lipidation-mediated host signal transduction, thereby enabling pathogen survival and replication. In this review, we discuss the role of the bacterial effectors in interactions with the host and highlight our knowledge of irreversible host delipidation, with a focus on the common concerted biochemical mechanisms of the bacterial effectors.
Collapse
Affiliation(s)
- Ligang Mei
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaofeng Qiu
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Chen Jiang
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing 401331, China; Ultrafast Transient Materials Science Center, Institute of Advanced Interdisciplinary Studies, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
67
|
Khan FA, Nasim N, Wang Y, Alhazmi A, Sanam M, Ul-Haq Z, Yalamati D, Ulanova M, Jiang ZH. Amphiphilic desmuramyl peptides for the rational design of new vaccine adjuvants: Synthesis, in vitro modulation of inflammatory response and molecular docking studies. Eur J Med Chem 2020; 209:112863. [PMID: 33032082 DOI: 10.1016/j.ejmech.2020.112863] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 01/23/2023]
Abstract
Nucleotide-binding oligomerization domain 2 (NOD2) is cytosolic surveillance receptor of the innate immune system capable of recognizing the bacterial and viral infections. Muramyl dipeptide (MDP) is the minimal immunoreactive unit of murein. NOD2 perceives MDP as pathogen-associated molecular pattern, thereby triggering an immune response with undesirable side-effects. Beneficial properties of MDP, such as pro-inflammatory characteristics for the rational design of new vaccine adjuvants, can be harnessed by strategically re-designing the molecule. In this work, a new class of amphiphilic desmuramylpeptides (DMPs) were synthesized by replacing the carbohydrate moiety (muramic acid) of the parent molecule with hydrophilic arenes. A lipophilic chain was also introduced at the C-terminus of dipeptide moiety (alanine-isoglutamine), while conserving its L-D configuration. These novel DMPs were found to set off the release of higher levels of tumour necrosis factor alpha (TNF-α) than Murabutide, which is a well-known NOD2 agonist. Molecular docking studies indicate that all these DMPs bind well to NOD2 receptor with similar dock scores (binding energy) through a number of hydrogen bonding and hydrophobic/π interactions with several crucial residues of the receptor. More studies are needed to further assess their immunomodulatory therapeutic potential, as well as the possible involvement of NOD2 activation.
Collapse
Affiliation(s)
- Farooq-Ahmad Khan
- H.E.J. Research Institute of Chemistry, International Center for Chemical & Biological Sciences, University of Karachi, 75270, Pakistan.
| | - Nourina Nasim
- H.E.J. Research Institute of Chemistry, International Center for Chemical & Biological Sciences, University of Karachi, 75270, Pakistan
| | - Yan Wang
- H.E.J. Research Institute of Chemistry, International Center for Chemical & Biological Sciences, University of Karachi, 75270, Pakistan
| | - Alaa Alhazmi
- Medical Sciences Division, Northern Ontario School of Medicine, Lakehead University, 955 Oliver Road, Thunder Bay, Ontario, P7B 5E1, Canada; Medical Laboratory Technology Department, Jazan University, Jazan, Saudi Arabia
| | - Mehar Sanam
- H.E.J. Research Institute of Chemistry, International Center for Chemical & Biological Sciences, University of Karachi, 75270, Pakistan
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Damayanthi Yalamati
- Alberta Research Chemicals Inc., 11421 Saskatchewan Drive, Edmonton, Alberta, T6G 2M9, Canada
| | - Marina Ulanova
- Medical Sciences Division, Northern Ontario School of Medicine, Lakehead University, 955 Oliver Road, Thunder Bay, Ontario, P7B 5E1, Canada
| | - Zi-Hua Jiang
- Department of Chemistry, Lakehead University, 955 Oliver Road, Thunder Bay, Ontario, P7B 5E1, Canada.
| |
Collapse
|
68
|
Chen M, Sun H, Boot M, Shao L, Chang SJ, Wang W, Lam TT, Lara-Tejero M, Rego EH, Galán JE. Itaconate is an effector of a Rab GTPase cell-autonomous host defense pathway against Salmonella. Science 2020; 369:450-455. [PMID: 32703879 DOI: 10.1126/science.aaz1333] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 01/26/2020] [Accepted: 05/15/2020] [Indexed: 11/02/2022]
Abstract
The guanosine triphosphatase (GTPase) Rab32 coordinates a cell-intrinsic host defense mechanism that restricts the replication of intravacuolar pathogens such as Salmonella Here, we show that this mechanism requires aconitate decarboxylase 1 (IRG1), which synthesizes itaconate, a metabolite with antimicrobial activity. We find that Rab32 interacts with IRG1 on Salmonella infection and facilitates the delivery of itaconate to the Salmonella-containing vacuole. Mice defective in IRG1 rescued the virulence defect of a S. enterica serovar Typhimurium mutant specifically defective in its ability to counter the Rab32 defense mechanism. These studies provide a link between a metabolite produced in the mitochondria after stimulation of innate immune receptors and a cell-autonomous defense mechanism that restricts the replication of an intracellular bacterial pathogen.
Collapse
Affiliation(s)
- Meixin Chen
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Hui Sun
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Maikel Boot
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Lin Shao
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Shu-Jung Chang
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Weiwei Wang
- WM Keck Foundation Biotechnology Resource Laboratory, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Tukiet T Lam
- WM Keck Foundation Biotechnology Resource Laboratory, Yale University School of Medicine, New Haven, CT 06536, USA.,Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Maria Lara-Tejero
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06536, USA
| | - E Hesper Rego
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Jorge E Galán
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06536, USA.
| |
Collapse
|
69
|
Destoumieux-Garzón D, Canesi L, Oyanedel D, Travers MA, Charrière GM, Pruzzo C, Vezzulli L. Vibrio-bivalve interactions in health and disease. Environ Microbiol 2020; 22:4323-4341. [PMID: 32363732 DOI: 10.1111/1462-2920.15055] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/12/2022]
Abstract
In the marine environment, bivalve mollusks constitute habitats for bacteria of the Vibrionaceae family. Vibrios belong to the microbiota of healthy oysters and mussels, which have the ability to concentrate bacteria in their tissues and body fluids, including the hemolymph. Remarkably, these important aquaculture species respond differently to infectious diseases. While oysters are the subject of recurrent mass mortalities at different life stages, mussels appear rather resistant to infections. Thus, Vibrio species are associated with the main diseases affecting the worldwide oyster production. Here, we review the current knowledge on Vibrio-bivalve interaction in oysters (Crassostrea sp.) and mussels (Mytilus sp.). We discuss the transient versus stable associations of vibrios with their bivalve hosts as well as technical issues limiting the monitoring of these bacteria in bivalve health and disease. Based on the current knowledge of oyster/mussel immunity and their interactions with Vibrio species pathogenic for oyster, we discuss how differences in immune effectors could contribute to the higher resistance of mussels to infections. Finally, we review the multiple strategies evolved by pathogenic vibrios to circumvent the potent immune defences of bivalves and how key virulence mechanisms could have been positively or negatively selected in the marine environment through interactions with predators.
Collapse
Affiliation(s)
| | - Laura Canesi
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genoa, Genoa, Italy
| | - Daniel Oyanedel
- IHPE, Université de Montpellier, CNRS, Ifremer, Université de Perpignan Via Domitia, Montpellier, France
| | - Marie-Agnès Travers
- IHPE, Université de Montpellier, CNRS, Ifremer, Université de Perpignan Via Domitia, Montpellier, France
| | - Guillaume M Charrière
- IHPE, Université de Montpellier, CNRS, Ifremer, Université de Perpignan Via Domitia, Montpellier, France
| | - Carla Pruzzo
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genoa, Genoa, Italy
| | - Luigi Vezzulli
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genoa, Genoa, Italy
| |
Collapse
|
70
|
Gur-Arie L, Eitan-Wexler M, Weinberger N, Rosenshine I, Livnah O. The bacterial metalloprotease NleD selectively cleaves mitogen-activated protein kinases that have high flexibility in their activation loop. J Biol Chem 2020; 295:9409-9420. [PMID: 32404367 DOI: 10.1074/jbc.ra120.013590] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/07/2020] [Indexed: 01/07/2023] Open
Abstract
Microbial pathogens often target the host mitogen-activated protein kinase (MAPK) network to suppress host immune responses. We previously identified a bacterial type III secretion system effector, termed NleD, a metalloprotease that inactivates MAPKs by specifically cleaving their activation loop. Here, we show that NleDs form a growing family of virulence factors harbored by human and plant pathogens as well as insect symbionts. These NleDs disable specifically Jun N-terminal kinases (JNKs) and p38s that are required for host immune response, whereas extracellular signal-regulated kinase (ERK), which is essential for host cell viability, remains intact. We investigated the mechanism that makes ERK resistant to NleD cleavage. Biochemical and structural analyses revealed that NleD exclusively targets activation loops with high conformational flexibility. Accordingly, NleD cleaved the flexible loops of JNK and p38 but not the rigid loop of ERK. Our findings elucidate a compelling mechanism of native substrate proteolysis that is promoted by entropy-driven specificity. We propose that such entropy-based selectivity is a general attribute of proteolytic enzymes.
Collapse
Affiliation(s)
- Lihi Gur-Arie
- Department Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maayan Eitan-Wexler
- Department of Biological Chemistry, Alexander Silverman Institute of Life Sciences, The Wolfson Centre for Applied Structural Biology, The Edmond J. Safra Campus, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nina Weinberger
- Department of Biological Chemistry, Alexander Silverman Institute of Life Sciences, The Wolfson Centre for Applied Structural Biology, The Edmond J. Safra Campus, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ilan Rosenshine
- Department Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Oded Livnah
- Department of Biological Chemistry, Alexander Silverman Institute of Life Sciences, The Wolfson Centre for Applied Structural Biology, The Edmond J. Safra Campus, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
71
|
Liu Y, Jia Y, Yang K, Wang Z. Heterogeneous Strategies to Eliminate Intracellular Bacterial Pathogens. Front Microbiol 2020; 11:563. [PMID: 32390959 PMCID: PMC7192003 DOI: 10.3389/fmicb.2020.00563] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 03/16/2020] [Indexed: 12/21/2022] Open
Abstract
Antibiotic tolerance in bacterial pathogens that are genetically susceptible, but phenotypically tolerant to treatment, represents a growing crisis for public health. In particular, the intracellular bacteria-mediated antibiotic tolerance by acting as “Trojan horses” play a critical and underappreciated role in the disease burden of bacterial infections. Thus, more intense efforts are required to tackle this problem. In this review, we firstly provide a brief overview of modes of action of bacteria invasion and survival in macrophage or non-professional phagocytic cells. Furthermore, we summarize our current knowledge about promising strategies to eliminate these intracellular bacterial pathogens, including direct bactericidal agents, antibiotic delivery to infection sites by various carriers, and activation of host immune functions. Finally, we succinctly discuss the challenges faced by bringing them into clinical trials and our constructive perspectives.
Collapse
Affiliation(s)
- Yuan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Yuqian Jia
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Kangni Yang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhiqiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
72
|
Blötz C, Singh N, Dumke R, Stülke J. Characterization of an Immunoglobulin Binding Protein (IbpM) From Mycoplasma pneumoniae. Front Microbiol 2020; 11:685. [PMID: 32373096 PMCID: PMC7176901 DOI: 10.3389/fmicb.2020.00685] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/24/2020] [Indexed: 01/30/2023] Open
Abstract
Bacteria evolved many ways to invade, colonize and survive in the host tissue. Such complex infection strategies of other bacteria are not present in the cell-wall less Mycoplasmas. Due to their strongly reduced genomes, these bacteria have only a minimal metabolism. Mycoplasma pneumoniae is a pathogenic bacterium using its virulence repertoire very efficiently, infecting the human lung. M. pneumoniae can cause a variety of conditions including fever, inflammation, atypical pneumoniae, and even death. Due to its strongly reduced metabolism, M. pneumoniae is dependent on nutrients from the host and aims to persist as long as possible, resulting in chronic diseases. Mycoplasmas evolved strategies to subvert the host immune system which involve proteins fending off immunoglobulins (Igs). In this study, we investigated the role of MPN400 as the putative factor responsible for Ig-binding and host immune evasion. MPN400 is a cell-surface localized protein which binds strongly to human IgG, IgA, and IgM. We therefore named the protein MPN400 immunoglobulin binding protein of Mycoplasma (IbpM). A strain devoid of IbpM is slightly compromised in cytotoxicity. Taken together, our study indicates that M. pneumoniae uses a refined mechanism for immune evasion.
Collapse
Affiliation(s)
- Cedric Blötz
- Department of General Microbiology, Göttingen Center for Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Neil Singh
- Department of General Microbiology, Göttingen Center for Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Roger Dumke
- Medical Faculty Carl Gustav Carus, Institute of Medical Microbiology and Hygiene, Technical University Dresden, Dresden, Germany
| | - Jörg Stülke
- Department of General Microbiology, Göttingen Center for Molecular Biosciences, University of Göttingen, Göttingen, Germany
| |
Collapse
|
73
|
Ismail S, Ahmad S, Azam SS. Vaccinomics to design a novel single chimeric subunit vaccine for broad-spectrum immunological applications targeting nosocomial Enterobacteriaceae pathogens. Eur J Pharm Sci 2020; 146:105258. [PMID: 32035109 DOI: 10.1016/j.ejps.2020.105258] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/09/2020] [Accepted: 02/04/2020] [Indexed: 12/21/2022]
Abstract
Healthcare associated infections (HAIs) are major cause of elevated mortality, morbidity, and high healthcare costs. Development of a vaccine targeting these pathogens could benefit in reducing HAIs count and excessive use of antibiotics. This work aimed to design a multi-epitope based prophylactic/ therapeutic vaccine directing against carbapenem resistant Enterobacter cloacae and other leading nosocomial members of Enterobacteriaceae group. Based on subtractive proteomics and immunoinformatics in-depth investigation of E. cloacae reference proteome, we prioritize four targets: outer membrane usher protein-lpfC, putative outer membrane protein A-OmpA, putative outer membrane protein-FimD, and arginine transporter fulfilling criteria of vaccine candidacy. A multi-epitope peptide vaccine construct is then formulated comprising predicted epitopes with potential to evoke both innate and adaptive immunity and B-subunit of cholera toxin as an adjuvant. The construct is modelled, loop refined, improved for stability via disulfide engineering and optimized for codon usage as per Escherichia coli expression system to ensure its maximum expression. Cross-conservation analysis carried out to evaluate broad-spectrum applicability by providing cross protection against nosocomial pathogens. A blind docking method is applied further to predict predominant binding mode of the construct with TLR4 innate immune receptor, followed by molecular dynamics simulation protocol to probe complex dynamics and exposed topology of the construct epitopes for recognition and immune processing by the host. Towards the end, binding free energies of the vaccine construct-TLR4 receptor were estimated to test docking predictions and affirm complex stability. We believe these findings to be highly useful for vaccinologists in making a highly effective vaccine for E. cloacae specifically, and other notorious Enterobacteriaceae nosocomial pathogens in general.
Collapse
Affiliation(s)
- Saba Ismail
- Computational Biology Lab, National Center for Bioinformatics (NCB), Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Sajjad Ahmad
- Computational Biology Lab, National Center for Bioinformatics (NCB), Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Syed Sikander Azam
- Computational Biology Lab, National Center for Bioinformatics (NCB), Quaid-i-Azam University, Islamabad, 45320, Pakistan.
| |
Collapse
|
74
|
Deramaudt TB, Ali M, Vinit S, Bonay M. Sulforaphane reduces intracellular survival of Staphylococcus aureus in macrophages through inhibition of JNK and p38 MAPK‑induced inflammation. Int J Mol Med 2020; 45:1927-1941. [PMID: 32323751 PMCID: PMC7169961 DOI: 10.3892/ijmm.2020.4563] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 02/11/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophages are active contributors to the innate immune defense system. As macrophage activation is clearly affected by the surrounding microenvironment, the present study investigated the effect of sulforaphane (SFN) on the bactericidal activity of macrophages and the underlying molecular mechanisms involved in this process. Human THP-1-derived macrophages, primary human peripheral blood mononuclear cell-derived macrophages, and primary mouse bone marrow derived-macrophages (BMDMs) pretreated with SFN or DMSO were utilized in a model of Staphylococcus aureus infection. The results suggested that SFN pretreatment of macrophages effectively repressed the intracellular survival of S. aureus through modulation of p38/JNK signaling and decreased S. aureus-induced caspases-3/7-dependent cell apoptosis, potentially through downregulation of microRNA (miR)-142-5p and miR-146a-5p. As SFN is a well-known activator of nuclear factor erythroid 2-related factor 2 (Nrf2), Nrf2−/− BMDMs were used to demonstrate that the SFN-mediated inhibitory effect was independent of Nrf2. Nevertheless, an increase in intracellular bacterial survival in Nrf2-deficient macrophages was observed. In addition, SFN pretreatment suppressed S. aureus-induced transcriptional expression of genes coding for the proinflammatory cytokines interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α), as well as for the M1 markers C-C motif chemokine receptor 7, IL-23 and inducible nitric oxide synthase (iNOS). Western blot analysis indicated that S. aureus challenge activated p38 mitogen-activated protein kinase (MAPK) (p38) and c-Jun N-terminal kinase (JNK) MAPK signaling pathways, while SFN pretreatment prevented p38 and JNK phosphorylation. Pretreatment with 2 specific inhibitors of p38 and JNK, SB203580 and SP600125, respectively, resulted in a decrease in S. aureus-induced proinflammatory gene expression levels compared with those observed in the SFN-pretreated macrophages. Furthermore, THP-1-derived macrophages pretreated with SB203580 or SP600125 prior to bacterial infection exhibited a significant inhibition in intracellular S. aureus survival. In conclusion, we hypothesize that concomitant targeting of the p38/JNK-inflammatory response and the S. aureus-induced apoptosis with SFN may be a promising therapeutic approach in S. aureus infection.
Collapse
Affiliation(s)
- Therese B Deramaudt
- Department of Neuromuscular Handicap: Biotherapies and Therapeutic Innovations, National Institute of Health and Medical Research, University of Versailles‑Saint‑Quentin‑en‑Yvelines, 78180 Montigny‑le‑Bretonneux, France
| | - Malika Ali
- Department of Neuromuscular Handicap: Biotherapies and Therapeutic Innovations, National Institute of Health and Medical Research, University of Versailles‑Saint‑Quentin‑en‑Yvelines, 78180 Montigny‑le‑Bretonneux, France
| | - Stephane Vinit
- Department of Neuromuscular Handicap: Biotherapies and Therapeutic Innovations, National Institute of Health and Medical Research, University of Versailles‑Saint‑Quentin‑en‑Yvelines, 78180 Montigny‑le‑Bretonneux, France
| | - Marcel Bonay
- Department of Neuromuscular Handicap: Biotherapies and Therapeutic Innovations, National Institute of Health and Medical Research, University of Versailles‑Saint‑Quentin‑en‑Yvelines, 78180 Montigny‑le‑Bretonneux, France
| |
Collapse
|
75
|
Krishnakumari V, Binny TM, Adicherla H, Nagaraj R. Escherichia coli Lipopolysaccharide Modulates Biological Activities of Human-β-Defensin Analogues but Not Non-Ribosomally Synthesized Peptides. ACS OMEGA 2020; 5:6366-6375. [PMID: 32258871 PMCID: PMC7114172 DOI: 10.1021/acsomega.9b03770] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 03/04/2020] [Indexed: 06/11/2023]
Abstract
Human-β-defensins (HBD1-3) are antibacterial peptides containing three disulphide bonds. In the present study, the effect of Escherichia coli lipopolysaccharide (LPS) on the antibacterial activities of HBD2-3, C-terminal analogues having a single disulphide bond, Phd1-3, and their corresponding myristoylated analogues MPhd1-3 were investigated. The effect of LPS on the activities of linear amphipathic peptides melittin, LL37 and non-ribosomally synthesized peptides, polymyxin B, alamethicin, gramicidin A, and gramicidin S was also examined. The antibacterial activity of HBD 2-3, Phd1-3, and MPhd1-3 in the presence of LPS against E. coli and Staphylococcus aureus was inhibited. While LPS inhibited the antibacterial activity of LL37, the inhibition of melittin activity was partial. The hemolytic activity exhibited by MPhd1, MPhd3, melittin, and LL37 was inhibited in the presence of LPS. HBD2-3, Phd1-3, and MPhd1-3 also showed endotoxin neutralizing activity. The antibacterial and hemolytic activities of polymyxin B, alamethicin, gramicidin A, and gramicidin S were not inhibited in the presence of LPS. Fluorescence assays employing dansyl cadaverine showed that HBD2-3 and defensin analogues bind to LPS more strongly as compared to alamethicin, gramicidin A, and gramicidin S. Electron microscopy images indicated that peptides disintegrate the structure of LPS. The inhibition of the antibacterial activity of native defensins and analogues in the presence of LPS indicates that the initial interaction with the bacterial surface is similar. The native defensin sequence or structure is also not essential, although cationic charges are necessary for binding to LPS. Hydrophobic interaction is the main driving force for association of non-ribosomally synthesized polymyxin B, alamethicin, gramicidin A, and gramicidin S with LPS. It is likely that these peptides rapidly insert into membranes and do not interact with the bacterial cell surface, whereas cationic peptides such as β-defensin and their analogues, melittin and LL37, first interact with the bacterial cell surface and then the membrane. Our results suggest that evaluating interaction of antibacterial and hemolytic peptides with LPS is a compelling way of elucidating the mechanism of bacterial killing or hemolysis.
Collapse
|
76
|
Muendlein HI, Jetton D, Connolly WM, Eidell KP, Magri Z, Smirnova I, Poltorak A. cFLIP L protects macrophages from LPS-induced pyroptosis via inhibition of complex II formation. Science 2020; 367:1379-1384. [PMID: 32193329 DOI: 10.1126/science.aay3878] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/16/2019] [Accepted: 02/20/2020] [Indexed: 12/16/2022]
Abstract
Cell death and inflammation are interdependent host responses to infection. During pyroptotic cell death, interleukin-1β (IL-1β) release occurs through caspase-1 and caspase-11-mediated gasdermin D pore formation. In vivo, responses to lipopolysaccharide (LPS) result in IL-1β secretion. In vitro, however, murine macrophages require a second "danger signal" for the inflammasome-driven maturation of IL-1β. Recent reports have shown caspase-8-mediated pyroptosis in LPS-activated macrophages but have provided conflicting evidence regarding the release of IL-1β under these conditions. Here, to further characterize the mechanism of LPS-induced secretion in vitro, we reveal an important role for cellular FLICE-like inhibitory protein (cFLIP) in the regulation of the inflammatory response. Specifically, we show that deficiency of the long isoform cFLIPL promotes complex II formation, driving pyroptosis, and the secretion of IL-1β in response to LPS alone.
Collapse
Affiliation(s)
- Hayley I Muendlein
- Graduate Program in Genetics, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - David Jetton
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Wilson M Connolly
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Keith P Eidell
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Zoie Magri
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Irina Smirnova
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Alexander Poltorak
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA. .,Laboratory of Genetics of Innate Immunity, Petrozavodsk State University, Petrozavodsk, Republic of Karelia 185910, Russia
| |
Collapse
|
77
|
Jun JI, Lau LF. CCN1 is an opsonin for bacterial clearance and a direct activator of Toll-like receptor signaling. Nat Commun 2020; 11:1242. [PMID: 32144270 PMCID: PMC7060279 DOI: 10.1038/s41467-020-15075-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 02/17/2020] [Indexed: 12/30/2022] Open
Abstract
Expression of the matricellular protein CCN1 (CYR61) is associated with inflammation and is required for successful wound repair. Here, we show that CCN1 binds bacterial pathogen-associated molecular patterns including peptidoglycans of Gram-positive bacteria and lipopolysaccharides of Gram-negative bacteria. CCN1 opsonizes methicillin-resistant Staphylococcus aureus (MRSA) and Pseudomonas aeruginosa and accelerates their removal by phagocytosis and increased production of bactericidal reactive oxygen species in macrophages through the engagement of integrin αvβ3. Mice with myeloid-specific Ccn1 deletion and knock-in mice expressing CCN1 unable to bind αvβ3 are more susceptible to infection by S. aureus or P. aeruginosa, resulting in increased mortality and organ colonization. Furthermore, CCN1 binds directly to TLR2 and TLR4 to activate MyD88-dependent signaling, cytokine expression and neutrophil mobilization. CCN1 is therefore a pattern recognition receptor that opsonizes bacteria for clearance and functions as a damage-associated molecular pattern to activate inflammatory responses, activities that contribute to wound healing and tissue repair. CCN1 is a matricellular protein with a variety of functions, including an effect on wound healing and an association with inflammation. Here, the authors identify a possible mechanism by showing that CCN1 mediates the clearance of bacterial infections in mice and activates TLR signalling.
Collapse
Affiliation(s)
- Joon-Il Jun
- Department of Biochemistry and Molecular Genetics, College of Medicine, The University of Illinois at Chicago, 900 South Ashland Avenue, Chicago, IL, 60607, USA
| | - Lester F Lau
- Department of Biochemistry and Molecular Genetics, College of Medicine, The University of Illinois at Chicago, 900 South Ashland Avenue, Chicago, IL, 60607, USA.
| |
Collapse
|
78
|
Mursalin MH, Coburn PS, Livingston E, Miller FC, Astley R, Flores-Mireles AL, Callegan MC. Bacillus S-Layer-Mediated Innate Interactions During Endophthalmitis. Front Immunol 2020; 11:215. [PMID: 32117322 PMCID: PMC7028758 DOI: 10.3389/fimmu.2020.00215] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/27/2020] [Indexed: 12/14/2022] Open
Abstract
Bacillus endophthalmitis is a severe intraocular infection. Hallmarks of Bacillus endophthalmitis include robust inflammation and rapid loss of vision. We reported that the absence of Bacillus surface layer protein (SLP) significantly blunted endophthalmitis severity. Here, we further investigated SLP in the context of Bacillus-retinal cell interactions and innate immune pathways to explore the mechanisms by which SLP contributes to intraocular inflammation. We compared phenotypes of Wild-type (WT) and SLP deficient (ΔslpA) Bacillus thuringiensis by analyzing bacterial adherence to and phagocytosis by human retinal Muller cells and phagocytosis by mouse neutrophils. Innate immune receptor activation by the Bacillus envelope and purified SLP was analyzed using TLR2/4 reporter cell lines. A synthetic TLR2/4 inhibitor was used as a control for this receptor activation. To induce endophthalmitis, mouse eyes were injected intravitreally with 100 CFU WT or ΔslpA B. thuringiensis. A group of WT infected mice was treated intravitreally with a TLR2/4 inhibitor at 4 h postinfection. At 10 h postinfection, infected eyes were analyzed for viable bacteria, inflammation, and retinal function. We observed that B. thuringiensis SLPs contributed to retinal Muller cell adherence, and protected this pathogen from Muller cell- and neutrophil-mediated phagocytosis. We found that B. thuringiensis envelope activated TLR2 and, surprisingly, TLR4, suggesting the presence of a surface-associated TLR4 agonist in Bacillus. Further investigation showed that purified SLP from B. thuringiensis activated TLR4, as well as TLR2 in vitro. Growth of WT B. thuringiensis was significantly higher and caused greater inflammation in untreated eyes than in eyes treated with the TLR2/4 inhibitor. Retinal function analysis also showed greater retention of A-wave and B-wave function in infected eyes treated with the TLR2/4 inhibitor. The TLR2/4 inhibitor was not antibacterial in vitro, and did not cause inflammation when injected into uninfected eyes. Taken together, these results suggest a potential role for Bacillus SLP in host-bacterial interactions, as well as in endophthalmitis pathogenesis via TLR2- and TLR4-mediated pathways.
Collapse
Affiliation(s)
- Md Huzzatul Mursalin
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Phillip S. Coburn
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Dean McGee Eye Institute, Oklahoma City, OK, United States
| | - Erin Livingston
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Frederick C. Miller
- Department of Cell Biology and Department of Family and Preventive Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Roger Astley
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Dean McGee Eye Institute, Oklahoma City, OK, United States
| | - Ana L. Flores-Mireles
- Department of Biological Sciences, University of Notre Dame, South Bend, IN, United States
| | - Michelle C. Callegan
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Dean McGee Eye Institute, Oklahoma City, OK, United States
| |
Collapse
|
79
|
Di Pietro M, Filardo S, Frasca F, Scagnolari C, Manera M, Sessa V, Antonelli G, Sessa R. Interferon-γ Possesses Anti-Microbial and Immunomodulatory Activity on a Chlamydia Trachomatis Infection Model of Primary Human Synovial Fibroblasts. Microorganisms 2020; 8:E235. [PMID: 32050567 PMCID: PMC7074713 DOI: 10.3390/microorganisms8020235] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 11/16/2022] Open
Abstract
Chlamydia trachomatis, an obligate intracellular pathogen, is the most common cause of bacterial sexually transmitted diseases, and it is potentially responsible for severe chronic sequelae, such as reactive arthritis. To date, details of the mechanisms by which Chlamydiae induce innate antimicrobial pathways in synovial fibroblasts, are not well characterized; therefore, herein, we investigated the effects of interferon (IFN)α, IFNβ, and IFNγ on the infection, and replication phases of the C. trachomatis developmental cycle, as well as on the induction of pattern recognition receptors (PRRs) and IFN-related pathways. To do so, we set up an in vitro chlamydial-infection model of primary human synovial cells treated with IFNs before or after the infection. We then determined the number of chlamydial inclusion forming units and inclusion size, as well as the expression of toll like receptor (TLR)2, TLR3, TLR4, cyclic GMP-AMP synthase (cGAS), stimulator of IFN gene (STING), IRF9, ISG56, and GBP1. The main result of our study is the significant inhibition of C. trachomatis infection and replication in human synovial cells following the treatment with IFNγ, whereas IFN-I proved to be ineffective. Furthermore, IFNγ greatly upregulated all the PRRs and ISGs examined. In conclusion, IFNγ exhibited a potent anti-Chlamydia activity in human synovial cells as well as the ability to induce a strong increase of innate immune pathways.
Collapse
Affiliation(s)
- Marisa Di Pietro
- Section of Microbiology, Department of Public Health and Infectious Diseases, Sapienza University, 00185 Rome, Italy; (M.D.P.); (M.M.); (R.S.)
| | - Simone Filardo
- Section of Microbiology, Department of Public Health and Infectious Diseases, Sapienza University, 00185 Rome, Italy; (M.D.P.); (M.M.); (R.S.)
| | - Federica Frasca
- Laboratory of Virology, Department of Molecular Medicine, affiliated to Istituto Pasteur Italia–Cenci Bolognetti Foundation, Sapienza University, 00185 Rome, Italy; (F.F.); (C.S.); (G.A.)
| | - Carolina Scagnolari
- Laboratory of Virology, Department of Molecular Medicine, affiliated to Istituto Pasteur Italia–Cenci Bolognetti Foundation, Sapienza University, 00185 Rome, Italy; (F.F.); (C.S.); (G.A.)
| | - Martina Manera
- Section of Microbiology, Department of Public Health and Infectious Diseases, Sapienza University, 00185 Rome, Italy; (M.D.P.); (M.M.); (R.S.)
| | - Vincenzo Sessa
- Department of Orthopedics, San Giovanni Calibita-Fatebenefratelli Hospital, 00186 Rome, Italy;
| | - Guido Antonelli
- Laboratory of Virology, Department of Molecular Medicine, affiliated to Istituto Pasteur Italia–Cenci Bolognetti Foundation, Sapienza University, 00185 Rome, Italy; (F.F.); (C.S.); (G.A.)
- Microbiology and Virology Unit, Hospital “Policlinico Umberto I”, Sapienza University, 00185 Rome, Italy
| | - Rosa Sessa
- Section of Microbiology, Department of Public Health and Infectious Diseases, Sapienza University, 00185 Rome, Italy; (M.D.P.); (M.M.); (R.S.)
| |
Collapse
|
80
|
Zheng F, Xu N, Zhang Y. TRIM27 Promotes Hepatitis C Virus Replication by Suppressing Type I Interferon Response. Inflammation 2020; 42:1317-1325. [PMID: 30847745 DOI: 10.1007/s10753-019-00992-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Type I interferon (IFN) response is central for host defense against viral infection. Tripartite motif 27 (TRIM27) is implicated in antiviral innate immune response; however, whether it affects the replication of hepatitis C virus (HCV) and the underlying mechanisms remain uncharacterized. Here, we show that TRIM27 expression is induced in Huh7.5 human hepatoma cells infected with HCV or stimulated with type I IFNs in vitro. In addition, TRIM27 overexpression increases and its knockdown decreases viral RNA and protein levels, suggesting that TRIM27 positively regulates HCV replication. Mechanistically, TRIM27 inhibits type I IFN response against HCV infection through inhibiting IRF3 and NF-κB pathways, since TRIM27 mutant unable to inhibit these two inflammatory pathways fails to promote HCV replication. Taken together, this study identifies TRIM27 as a novel positive regulator of HCV replication, and also implicates that targeting TRIM27 may serve as a therapeutic strategy for controlling HCV replication.
Collapse
Affiliation(s)
- Feng Zheng
- Department of Infectious Disease, Qilu Hospital of Shandong University, 107# West Wenhua Road, Jinan, 250012, Shandong province, People's Republic of China.
| | - Nannan Xu
- Department of Infectious Disease, Qilu Hospital of Shandong University, 107# West Wenhua Road, Jinan, 250012, Shandong province, People's Republic of China
| | - Yajun Zhang
- Department of Infectious Disease, Qilu Hospital of Shandong University, 107# West Wenhua Road, Jinan, 250012, Shandong province, People's Republic of China
| |
Collapse
|
81
|
Jalili M, Amraei M, Sadeghifard N, Ghafourian S. Evaluation of Biofilm Formation and Anti-biofilm Properties of Peganum Harmala and Crocus Sativus in Shigella Flexneri Clinical Isolates. Open Microbiol J 2019. [DOI: 10.2174/1874285801913010297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background:
Biofilm formation causes many serious problems in the treatment of bacterial infections. In addition, chronic infections due to biofilm formation can pose a huge burden to the health care systems. Also, many bacteria are biofilm producers as an important strategy for pathogenicity. Furthermore, the traditional use of herbal medicines such as Peganum harmala and Crocus sativus in Iran is interesting.
Objective:
The purpose of the current study was to investigate the biofilm formation in Shigella flexneri clinical isolates and to evaluate the anti-biofilm properties of P. harmala and C. sativus on Shigella flexneri clinical isolates.
Methods:
For the study purpose, Thirty S.flexneri clinical isolates were collected from Ahvaz, Iran. Then, the collected bacteria were subjected to biofilm formation assay. Afterward, P. harmala and C. sativus were applied as an anti-biofilm formation in S. flexneri.
Results & Conclusion:
Our results demonstrated that a significant number of samples were identified as strong biofilm producers. Then, P. harmala and C . sativus in a concentration of 30μg/ml and 60μg/ml were able to eradicate a strong biofilm formation in S. flexneri, respectively. In addition, it seems that more extensive studies and in vivo research should be done to confirm their properties.
Collapse
|
82
|
Saha G, Khamar BM, Prerna K, Kumar M, Dubey VK. BLIMP-1 Plays Important Role in the Regulation of Macrophage Pyroptosis for the Growth and Multiplication of Leishmania donovani. ACS Infect Dis 2019; 5:2087-2095. [PMID: 31618572 DOI: 10.1021/acsinfecdis.9b00186] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Visceral leishmaniasis, one of the fatal forms of the disease, is caused by Leishmania donovani and presents morbid clinical manifestations. The parasite evades pro-inflammatory immune responses by several reported mechanisms and modulates the host immune system to cause fatal symptoms. A plethora of reports related to the role of BLIMP-1 and its involvement in suppressing the immune response in various infectious diseases have been documented. Higher parasitic burden due to increased BLIMP-1 production has been reported earlier for malaria and leishmaniasis with no detailed information. We report for the first time the role of BLIMP-1 in suppressing macrophage pyroptosis during L. donovani infection and thereby tweaking the tight regulation of the NFκβ-NLRP3 signaling pathway. Expression analyses of BLIMP-1 and NFκβ have been measured using real-time PCR and Western blotting. The importance of BLIMP-1 has been validated using a siRNA-mediated experiment along with caspase 1 activity, LDH release assay, and infectivity index analyses. An inverse relationship between BLIMP-1 and NFκβ expression has been highlighted during L. donovani infection, which is reversed in blimp-1 deficient cells infected with promastigotes. The above fact has been further validated with caspase 1 activity assay, and LDH release along with IFNγ and TNF-α release assay. Finally, resumption of pyroptosis has been concluded in infected blimp-1 deficient cells in contrast to wild type infected cells. We conjecture that parasites modulate the NFκβ-NLRP3 signaling pathway by taking advantage of BLIMP-1 dependent IL-10 production and finally disrupting an inflammation-mediated pyroptosis cell death pathway in infected cells.
Collapse
Affiliation(s)
- Gundappa Saha
- Department of Biosciences & Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | | | - Kumari Prerna
- School of Biochemical Engineering, Indian Institute of Technology BHU, Varanasi, Uttar Pradesh 221005, India
| | - Manish Kumar
- Department of Biosciences & Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Vikash Kumar Dubey
- Department of Biosciences & Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
- School of Biochemical Engineering, Indian Institute of Technology BHU, Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
83
|
Bravo-Santano N, Capilla-Lasheras P, Mateos LM, Calle Y, Behrends V, Letek M. Identification of novel targets for host-directed therapeutics against intracellular Staphylococcus aureus. Sci Rep 2019; 9:15435. [PMID: 31659191 PMCID: PMC6817851 DOI: 10.1038/s41598-019-51894-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/09/2019] [Indexed: 01/26/2023] Open
Abstract
During patient colonization, Staphylococcus aureus is able to invade and proliferate within human cells to evade the immune system and last resort drugs such as vancomycin. Hijacking specific host molecular factors and/or pathways is necessary for pathogens to successfully establish an intracellular infection. In this study, we employed an unbiased shRNA screening coupled with ultra-fast sequencing to screen 16,000 human genes during S. aureus infection and we identified several host genes important for this intracellular pathogen. In addition, we interrogated our screening results to find novel host-targeted therapeutics against intracellular S. aureus. We found that silencing the human gene TRAM2 resulted in a significant reduction of intracellular bacterial load while host cell viability was restored, showing its importance during intracellular infection. Furthermore, TRAM2 is an interactive partner of the endoplasmic reticulum SERCA pumps and treatment with the SERCA-inhibitor Thapsigargin halted intracellular MRSA survival. Our results suggest that Thapsigargin could be repurposed to tackle S. aureus host cell infection in combination with conventional antibiotics.
Collapse
Affiliation(s)
| | | | - Luis M Mateos
- Department of Molecular Biology, Area of Microbiology, University of León, León, Spain
| | - Yolanda Calle
- Health Sciences Research Centre, University of Roehampton, London, UK
| | - Volker Behrends
- Health Sciences Research Centre, University of Roehampton, London, UK.
| | - Michal Letek
- Health Sciences Research Centre, University of Roehampton, London, UK.
| |
Collapse
|
84
|
Clark GC, Essex-Lopresti A, Moore KA, Williamson ED, Lukaszewski R, Paszkiewicz K, David J. Common Host Responses in Murine Aerosol Models of Infection Caused by Highly Virulent Gram-Negative Bacteria from the Genera Burkholderia, Francisella and Yersinia. Pathogens 2019; 8:pathogens8040159. [PMID: 31546628 PMCID: PMC6963870 DOI: 10.3390/pathogens8040159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/06/2019] [Accepted: 09/18/2019] [Indexed: 11/16/2022] Open
Abstract
Highly virulent bacterial pathogens cause acute infections which are exceptionally difficult to treat with conventional antibiotic therapies alone. Understanding the chain of events that are triggered during an infection of a host has the potential to lead to new therapeutic strategies. For the first time, the transcriptomic responses within the lungs of Balb/C mice have been compared during an acute infection with the intracellular pathogens Burkholderia pseudomallei, Francisella tularensis and Yersinia pestis. Temporal changes were determined using RNAseq and a bioinformatics pipeline; expression of protein was also studied from the same sample. Collectively it was found that early transcriptomic responses within the infected host were associated with the (a) slowing down of critical cellular functions, (b) production of circulatory system components, (c) lung tissue integrity, and (d) intracellular regulatory processes. One common molecule was identified, Errfi1 (ErbB receptor feedback inhibitor 1); upregulated in response to all three pathogens and a potential novel marker of acute infection. Based upon the pro-inflammatory responses observed, we sought to synchronise each infection and report that 24 h p.i. of B. pseudomallei infection closely aligned with 48 h p.i. of infection with F. tularensis and Y. pestis. Post-transcriptional modulation of RANTES expression occurred across all pathogens, suggesting that these infections directly or indirectly modulate cell trafficking through chemokine expression/detection. Collectively, this unbiased NGS approach has provided an in-depth characterisation of the host transcriptome following infection with these highly virulent pathogens ultimately aiding in the development of host-directed therapies as adjuncts or alternatives to antibiotic treatment.
Collapse
Affiliation(s)
- Graeme C Clark
- Chemical, Biological and Radiological Division, DSTL Porton Down, Salisbury SP4 0JQ, UK.
| | - Angela Essex-Lopresti
- Chemical, Biological and Radiological Division, DSTL Porton Down, Salisbury SP4 0JQ, UK.
| | - Karen A Moore
- Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK.
| | - E Diane Williamson
- Chemical, Biological and Radiological Division, DSTL Porton Down, Salisbury SP4 0JQ, UK.
| | - Roman Lukaszewski
- Chemical, Biological and Radiological Division, DSTL Porton Down, Salisbury SP4 0JQ, UK.
| | - Konrad Paszkiewicz
- Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK.
| | - Jonathan David
- Chemical, Biological and Radiological Division, DSTL Porton Down, Salisbury SP4 0JQ, UK.
| |
Collapse
|
85
|
Yang D, Liu X, Xu W, Gu Z, Yang C, Zhang L, Tan J, Zheng X, Wang Z, Quan S, Zhang Y, Liu Q. The Edwardsiella piscicida thioredoxin-like protein inhibits ASK1-MAPKs signaling cascades to promote pathogenesis during infection. PLoS Pathog 2019; 15:e1007917. [PMID: 31314784 PMCID: PMC6636751 DOI: 10.1371/journal.ppat.1007917] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/14/2019] [Indexed: 12/02/2022] Open
Abstract
It is important that bacterium can coordinately deliver several effectors into host cells to disturb the cellular progress during infection, however, the precise role of effectors in host cell cytosol remains to be resolved. In this study, we identified a new bacterial virulence effector from pathogenic Edwardsiella piscicida, which presents conserved crystal structure to thioredoxin family members and is defined as a thioredoxin-like protein (Trxlp). Unlike the classical bacterial thioredoxins, Trxlp can be translocated into host cells, mimicking endogenous thioredoxin to abrogate ASK1 homophilic interaction and phosphorylation, then suppressing the phosphorylation of downstream Erk1/2- and p38-MAPK signaling cascades. Moreover, Trxlp-mediated inhibition of ASK1-Erk/p38-MAPK axis promotes the pathogenesis of E. piscicida in zebrafish larvae infection model. Taken together, these data provide insights into the mechanism underlying the bacterial thioredoxin as a virulence effector in downmodulating the innate immune responses during E. piscicida infection. Thioredoxin (Trx) is universally conserved thiol-oxidoreductase that regulates numerous cellular pathways under thiol-based redox control in both prokaryotic and eukaryotic organisms. Despite its central importance, the mechanism of bacterial Trx recognizes its target proteins in host cellular signaling remains unknown. Here, we uncover a bacterial thioredoxin-like protein that can be translocated into host cells and mimic the endogenous TRX1 to target ASK1-MAPK signaling, finally facilitating bacterial pathogenesis. This work expands our understanding of bacterial thioredoxins in manipulating host innate immunity.
Collapse
Affiliation(s)
- Dahai Yang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, China
| | - Xiaohong Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, China
| | - Wenting Xu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Zhaoyan Gu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Cuiting Yang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Lingzhi Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Jinchao Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Xin Zheng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Zhuang Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Shu Quan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yuanxing Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, China
| | - Qin Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- * E-mail:
| |
Collapse
|
86
|
Griewahn L, Köser A, Maurer U. Keeping Cell Death in Check: Ubiquitylation-Dependent Control of TNFR1 and TLR Signaling. Front Cell Dev Biol 2019; 7:117. [PMID: 31316982 PMCID: PMC6609852 DOI: 10.3389/fcell.2019.00117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/11/2019] [Indexed: 01/05/2023] Open
Abstract
Pro-inflammatory signaling pathways, induced by pathogens, tissue damage or cytokines, depend on the ubiquitylation of various subunits of receptor signaling complexes, controlled by ubiquitin ligases and deubiquitinases. Ubiquitylation sets the stage for the activation of kinases within these receptor complexes, which ultimately regulate pro-inflammatory gene expression. The receptors, which transduce pro-inflammatory signals, can often induce cell death, which is controlled by ubiquitylation as well. In this review, we discuss the key role of ubiquitylation in pro-inflammatory signaling by TNFR1 and TLRs and its role in setting the threshold for cell death induced by these pro-inflammatory triggers.
Collapse
Affiliation(s)
- Laura Griewahn
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg im Breisgau, Germany.,Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg im Breisgau, Germany.,Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Aaron Köser
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg im Breisgau, Germany
| | - Ulrich Maurer
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg im Breisgau, Germany.,Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg im Breisgau, Germany.,BIOSS Centre for Biological Signalling Studies, Freiburg im Breisgau, Germany
| |
Collapse
|
87
|
Species-specific mechanisms of cytotoxicity toward immune cells determine the successful outcome of Vibrio infections. Proc Natl Acad Sci U S A 2019; 116:14238-14247. [PMID: 31221761 DOI: 10.1073/pnas.1905747116] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Vibrio species cause infectious diseases in humans and animals, but they can also live as commensals within their host tissues. How Vibrio subverts the host defenses to mount a successful infection remains poorly understood, and this knowledge is critical for predicting and managing disease. Here, we have investigated the cellular and molecular mechanisms underpinning infection and colonization of 2 virulent Vibrio species in an ecologically relevant host model, oyster, to study interactions with marine Vibrio species. All Vibrio strains were recognized by the immune system, but only nonvirulent strains were controlled. We showed that virulent strains were cytotoxic to hemocytes, oyster immune cells. By analyzing host and bacterial transcriptional responses to infection, together with Vibrio gene knock-outs, we discovered that Vibrio crassostreae and Vibrio tasmaniensis use distinct mechanisms to cause hemocyte lysis. Whereas V. crassostreae cytotoxicity is dependent on a direct contact with hemocytes and requires an ancestral gene encoding a protein of unknown function, r5.7, V. tasmaniensis cytotoxicity is dependent on phagocytosis and requires intracellular secretion of T6SS effectors. We conclude that proliferation of commensal vibrios is controlled by the host immune system, preventing systemic infections in oysters, whereas the successful infection of virulent strains relies on Vibrio species-specific molecular determinants that converge to compromise host immune cell function, allowing evasion of the host immune system.
Collapse
|
88
|
Zhou R, Chen Z, Hao D, Wang Y, Zhang Y, Yi X, Lyu LD, Liu H, Zou Q, Chu Y, Ge B, Yan D. Enterohemorrhagic Escherichia coli Tir inhibits TAK1 activation and mediates immune evasion. Emerg Microbes Infect 2019; 8:734-748. [PMID: 31130074 PMCID: PMC6542180 DOI: 10.1080/22221751.2019.1620589] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Many pathogens infect hosts through various immune evasion strategies. However, the molecular mechanisms by which pathogen proteins modulate and evade the host immune response remain unclear. Enterohemorrhagic Escherichia coli (EHEC) is a pathological strain that can induce mitogen-activated protein (MAP) kinase (Erk, Jnk and p38 MAPK) and NF-κB pathway activation and proinflammatory cytokine production, which then causes diarrheal diseases such as hemorrhagic colitis and hemolytic uremic syndrome. Transforming growth factor β-activated kinase-1 (TAK1) is a key regulator involved in distinct innate immune signalling pathways. Here we report that EHEC translocated intimin receptor (Tir) protein inhibits the expression of EHEC-induced proinflammatory cytokines by interacting with the host tyrosine phosphatase SHP-1, which is dependent on the phosphorylation of immunoreceptor tyrosine-based inhibition motifs (ITIMs). Mechanistically, the association of EHEC Tir with SHP-1 facilitated the recruitment of SHP-1 to TAK1 and inhibited TAK1 phosphorylation, which then negatively regulated K63-linked polyubiquitination of TAK1 and downstream signal transduction. Taken together, these results suggest that EHEC Tir negatively regulates proinflammatory responses by inhibiting the activation of TAK1, which is essential for immune evasion and could be a potential target for the treatment of bacterial infection.
Collapse
Affiliation(s)
- Ruixue Zhou
- a Department of Immunology, School of Basic Medical Sciences & Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH , Fudan University , Shanghai , People's Republic of China
| | - Zijuan Chen
- a Department of Immunology, School of Basic Medical Sciences & Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH , Fudan University , Shanghai , People's Republic of China
| | - Doudou Hao
- a Department of Immunology, School of Basic Medical Sciences & Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH , Fudan University , Shanghai , People's Republic of China
| | - Yu Wang
- b Department of Microbiology and Biochemical Pharmacy, National Engineering Research Centre of Immunological Products, College of Pharmacy , Army Medical University , Chongqing , People's Republic of China
| | - Yihua Zhang
- a Department of Immunology, School of Basic Medical Sciences & Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH , Fudan University , Shanghai , People's Republic of China
| | - Xianfu Yi
- c School of Biomedical Engineering , Tianjin Medical University , Tianjin , People's Republic of China
| | - Liang-Dong Lyu
- a Department of Immunology, School of Basic Medical Sciences & Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH , Fudan University , Shanghai , People's Republic of China
| | - Haipeng Liu
- d Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital , Tongji University School of Medicine , Shanghai , People's Republic of China
| | - Quanming Zou
- b Department of Microbiology and Biochemical Pharmacy, National Engineering Research Centre of Immunological Products, College of Pharmacy , Army Medical University , Chongqing , People's Republic of China
| | - Yiwei Chu
- a Department of Immunology, School of Basic Medical Sciences & Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH , Fudan University , Shanghai , People's Republic of China
| | - Baoxue Ge
- d Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital , Tongji University School of Medicine , Shanghai , People's Republic of China.,e Department of Microbiology and Immunology , Tongji University School of Medicine , Shanghai , People's Republic of China
| | - Dapeng Yan
- a Department of Immunology, School of Basic Medical Sciences & Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH , Fudan University , Shanghai , People's Republic of China
| |
Collapse
|
89
|
Wiles TJ, Guillemin K. The Other Side of the Coin: What Beneficial Microbes Can Teach Us about Pathogenic Potential. J Mol Biol 2019; 431:2946-2956. [PMID: 31078557 DOI: 10.1016/j.jmb.2019.05.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/19/2019] [Accepted: 05/01/2019] [Indexed: 02/07/2023]
Abstract
Koch's postulates and molecular Koch's postulates have made an indelible mark on how we study and classify microbes, particularly pathogens. However, rigid adherence to these historic postulates constrains our view of not only microbial pathogenesis but also host-microbe relationships in general. Collectively, the postulates imply that a "microbial pathogen" is a clearly identifiable organism with the exclusive capacity to elicit disease through an arsenal of pathogen-specific "virulence factors." This narrow definition has been repeatedly contradicted. Advances in DNA sequencing technologies and new experimental systems have revealed that the outcomes of host-microbe interactions are highly contextual and dynamic, especially those involving resident microbiota and variable aspects of host biology. Clarifying what differentiates pathogenic from non-pathogenic microbes, including their paradoxical ability to masquerade as one another, is critical to developing targeted diagnostics and treatments for infectious disease. Such endeavors will also inform the design of therapeutic strategies based on microbiome engineering by providing insights into how manipulating entire host-microbe systems may directly or indirectly alter the pathogenic potential of microbial communities. With these goals in mind, we discuss the need to develop experimental models that better capture the contexts that determine the nature of host-microbe relationships. To demonstrate the potential of one such model-the zebrafish and its resident microbiota-we describe recent work that has revealed the thin line between pathogenic and mutualistic relationships, how the intestine physically shapes bacterial populations and inflammation, and the ability of microbial transmission to override the host's innate immune system.
Collapse
Affiliation(s)
- Travis J Wiles
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA.
| | - Karen Guillemin
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA; Humans and the Microbiome Program, CIFAR, Toronto, Ontario, Canada.
| |
Collapse
|
90
|
Wang W, Chen J, Liao B, Xia L, Hou S, Wang Z, Lu Y. Identification and functional characterization of Histone-like DNA-binding protein in Nocardia seriolae (NsHLP) involved in cell apoptosis. JOURNAL OF FISH DISEASES 2019; 42:657-666. [PMID: 30854666 DOI: 10.1111/jfd.12962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/29/2018] [Accepted: 12/31/2018] [Indexed: 06/09/2023]
Abstract
Nocardia seriolae, a facultative intracellular bacterium, is the main pathogen of fish nocardiosis. Bioinformatic analysis showed that the histone-like DNA-binding protein (HLP) gene of N. seriolae (nshlp) encoded a secreted protein and might target the mitochondria in the host cell. To further study the preliminary function of HLP in N. seriolae (NsHLP), the gene cloning, extracellular products identification, subcellular localization, overexpression and apoptosis detection assay were carried out in this study. Mass spectrometry analysis of the extracellular products from N. seriolae showed that NsHLP was a secreted protein. Subcellular localization of HLP-GFP fusion proteins mainly assembled in the nucleus, which indicated that the NsHLP was co-located with the nucleus rather than mitochondria in fathead minnow (FHM) cells. Notably, the expression of NsHLP had changed the distribution of mitochondria into lumps in the FHM cell. In addition, apoptotic features were found in the transfected FHM cells by overexpression of NsHLP. Quantitative assays of mitochondrial membrane potential value, caspase-3 activity and pro-apoptotic genes mRNA (Bad, Bid and Bax) expression level demonstrated that the cell apoptosis was induced in the transfected FHM cells. All the results presented in this study provided insight on the function of NsHLP, which suggested that it may participate in the cell apoptosis regulation and play an important role in the pathogenesis of N. seriolae.
Collapse
Affiliation(s)
- Wenji Wang
- Shenzhen Research Institute of Guangdong Ocean University, Shenzhen, China
- Fisheries College of Guangdong Ocean University, Zhanjiang, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China
| | - Jianlin Chen
- Shenzhen Research Institute of Guangdong Ocean University, Shenzhen, China
- Fisheries College of Guangdong Ocean University, Zhanjiang, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China
| | - Baoshan Liao
- Fisheries College of Guangdong Ocean University, Zhanjiang, China
| | - Liqun Xia
- Shenzhen Research Institute of Guangdong Ocean University, Shenzhen, China
- Fisheries College of Guangdong Ocean University, Zhanjiang, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China
| | - Suying Hou
- Shenzhen Research Institute of Guangdong Ocean University, Shenzhen, China
- Fisheries College of Guangdong Ocean University, Zhanjiang, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China
| | - Zhiwen Wang
- Shenzhen Research Institute of Guangdong Ocean University, Shenzhen, China
- Fisheries College of Guangdong Ocean University, Zhanjiang, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China
| | - Yishan Lu
- Shenzhen Research Institute of Guangdong Ocean University, Shenzhen, China
- Fisheries College of Guangdong Ocean University, Zhanjiang, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China
| |
Collapse
|
91
|
Isendahl J, Giske CG, Tegmark Wisell K, Ternhag A, Nauclér P. Risk factors for community-onset bloodstream infection with extended-spectrum β-lactamase-producing Enterobacteriaceae: national population-based case-control study. Clin Microbiol Infect 2019; 25:1408-1414. [PMID: 30986557 DOI: 10.1016/j.cmi.2019.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 02/24/2019] [Accepted: 04/03/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVES The aim was to investigate risk factors for community-onset bloodstream infections with extended-spectrum β-lactamase-producing Enterobacteriaceae (EPE BSI). METHODS It is mandatory to report EPE BSI to a national register at the Public Health Agency of Sweden. Using this register, we performed a population-based case-control study from 2007 to 2012 of 945 cases and 9390 controls. Exposure data on comorbidity, hospitalization, in- and outpatient antibiotic consumption and socio-economic status were collected from hospital and health registers. RESULTS The overall incidence of EPE BSI was 1.7 per 100 000 person-years. The 30-day mortality was 11.3%. Urological disorders inferred the highest EPE BSI risk, adjusted odds ratio (aOR) 4.32 (95% Confidence Interval (CI) 3.41-5.47), followed by immunological disorders, aOR 3.54 (CI 2.01-6.23), haematological malignancy, aOR 2.77 (CI 1.57-4.87), solid tumours, aOR 2.28 (1.76-2.94) and diabetes, aOR 2.03 (1.58-2.61). Consumption of fluoroquinolones or mostly non-EPE-active antibiotics with selective Gram-negative spectrum of activity within the previous 3 months was associated with EPE BSI, aORs 5.52 (CI 2.8-11.0) and 3.8, CI 1.9-7.7) respectively. There was a dose-response relationship in EPE BSI risk with increasing number of consecutive regimens. Antibiotic consumption >3 months before EPE BSI was not associated with increased risk. Higher age, malignancies and education ≤12 years (aORs >2) were associated with increased 30-day mortality. CONCLUSIONS Targeted interventions should be directed towards improving care for patients with immunosuppression, urological disorders and subjects with lower socio-economic status. Antibiotic stewardship should focus on reduction of fluoroquinolones.
Collapse
Affiliation(s)
- J Isendahl
- Department of Medicine Solna, Unit for Infectious Diseases, Karolinska Institutet, Stockholm, Sweden.
| | - C G Giske
- Department of Clinical Microbiology, Karolinska Institutet, Karolinska University Hospital Laboratory, Stockholm, Sweden
| | | | - A Ternhag
- Department of Medicine Solna, Unit for Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - P Nauclér
- Department of Medicine Solna, Unit for Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
92
|
Curto P, Riley SP, Simões I, Martinez JJ. Macrophages Infected by a Pathogen and a Non-pathogen Spotted Fever Group Rickettsia Reveal Differential Reprogramming Signatures Early in Infection. Front Cell Infect Microbiol 2019; 9:97. [PMID: 31024862 PMCID: PMC6467950 DOI: 10.3389/fcimb.2019.00097] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 03/22/2019] [Indexed: 12/22/2022] Open
Abstract
Despite their high degree of genomic similarity, different spotted fever group (SFG) Rickettsia are often associated with very different clinical presentations. For example, Rickettsia conorii causes Mediterranean spotted fever, a life-threatening disease for humans, whereas Rickettsia montanensis is associated with limited or no pathogenicity to humans. However, the molecular basis responsible for the different pathogenicity attributes are still not understood. Although killing microbes is a critical function of macrophages, the ability to survive and/or proliferate within phagocytic cells seems to be a phenotypic feature of several intracellular pathogens. We have previously shown that R. conorii and R. montanensis exhibit different intracellular fates within macrophage-like cells. By evaluating early macrophage responses upon insult with each of these rickettsial species, herein we demonstrate that infection with R. conorii results in a profound reprogramming of host gene expression profiles. Transcriptional programs generated upon infection with this pathogenic bacteria point toward a sophisticated ability to evade innate immune signals, by modulating the expression of several anti-inflammatory molecules. Moreover, R. conorii induce the expression of several pro-survival genes, which may result in the ability to prolong host cell survival, thus protecting its replicative niche. Remarkably, R. conorii-infection promoted a robust modulation of different transcription factors, suggesting that an early manipulation of the host gene expression machinery may be key to R. conorii proliferation in THP-1 macrophages. This work provides new insights into the early molecular processes hijacked by a pathogenic SFG Rickettsia to establish a replicative niche in macrophages, opening several avenues of research in host-rickettsiae interactions.
Collapse
Affiliation(s)
- Pedro Curto
- Ph.D. Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Sean P. Riley
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Isaura Simões
- CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Juan J. Martinez
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
93
|
Ding J, Pan X, Du L, Yao Q, Xue J, Yao H, Wang DC, Li S, Shao F. Structural and Functional Insights into Host Death Domains Inactivation by the Bacterial Arginine GlcNAcyltransferase Effector. Mol Cell 2019; 74:922-935.e6. [PMID: 30979585 DOI: 10.1016/j.molcel.2019.03.028] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 12/27/2018] [Accepted: 03/22/2019] [Indexed: 01/10/2023]
Abstract
Enteropathogenic E. coli NleB and related type III effectors catalyze arginine GlcNAcylation of death domain (DD) proteins to block host defense, but the underlying mechanism is unknown. Here we solve crystal structures of NleB alone and in complex with FADD-DD, UDP, and Mn2+ as well as NleB-GlcNAcylated DDs of TRADD and RIPK1. NleB adopts a GT-A fold with a unique helix-pair insertion to hold FADD-DD; the interface contacts explain the selectivity of NleB for certain DDs. The acceptor arginine is fixed into a cleft, in which Glu253 serves as a base to activate the guanidinium. Analyses of the enzyme-substrate complex and the product structures reveal an inverting sugar-transfer reaction and a detailed catalytic mechanism. These structural insights are validated by mutagenesis analyses of NleB-mediated GlcNAcylation in vitro and its function in mouse infection. Our study builds a structural framework for understanding of NleB-catalyzed arginine GlcNAcylation of host death domain.
Collapse
Affiliation(s)
- Jingjin Ding
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; National Institute of Biological Sciences, Beijing 102206, China.
| | - Xing Pan
- Bio-Medical Center, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Lijie Du
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Qing Yao
- National Institute of Biological Sciences, Beijing 102206, China
| | - Juan Xue
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Hongwei Yao
- College of Chemistry and Chemical Engineering, High-Field Nuclear Magnetic Resonance Center, Xiamen University, Xiamen, Fujian 361005, China
| | - Da-Cheng Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Shan Li
- Bio-Medical Center, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Feng Shao
- National Institute of Biological Sciences, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China.
| |
Collapse
|
94
|
Staphylococcus aureus Biofilm-Conditioned Medium Impairs Macrophage-Mediated Antibiofilm Immune Response by Upregulating KLF2 Expression. Infect Immun 2019; 87:IAI.00643-18. [PMID: 30692179 DOI: 10.1128/iai.00643-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 01/20/2019] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus infections associated with the formation of biofilms on medical implants or host tissue play a critical role in the persistence of chronic infections. One critical mechanism of biofilm infection that leads to persistent infection lies in the capacity of biofilms to evade the macrophage-mediated innate immune response. It is now increasingly apparent that microorganisms exploit the negative regulatory mechanisms of the pattern recognition receptor (PRR)-mediated inflammatory response to subvert host cell functions by using various virulence factors. However, the detailed molecular mechanism, along with the identity of a target molecule, underlying the evasion of the macrophage-mediated innate immune response against S. aureus infection associated with biofilm formation remains to be elucidated. Here, using an in vitro culture model of murine macrophage-like RAW 264.7 cells, we demonstrate that S. aureus biofilm-conditioned medium significantly attenuated the capacity for macrophage bactericidal and proinflammatory responses. Importantly, the responses were associated with attenuated activation of NF-κB and increased expression of Kruppel-like factor 2 (KLF2) in RAW 264.7 cells. Small interfering RNA (siRNA)-mediated silencing of KLF2 in RAW 264.7 cells could restore the activation of NF-κB toward the bactericidal activity and generation of proinflammatory cytokines in the presence of S. aureus biofilm-conditioned medium. Collectively, our results suggest that factors secreted from S. aureus biofilms might exploit the KLF2-dependent negative regulatory mechanism to subvert macrophage-mediated innate immune defense against S. aureus biofilms.
Collapse
|
95
|
Adibzadeh Sereshgi MM, Abdollahpour-Alitappeh M, Mahdavi M, Ranjbar R, Ahmadi K, Taheri RA, Fasihi-Ramandi M. Immunologic balance of regulatory T cell/T helper 17 responses in gastrointestinal infectious diseases: Role of miRNAs. Microb Pathog 2019; 131:135-143. [PMID: 30914387 DOI: 10.1016/j.micpath.2019.03.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 03/18/2019] [Accepted: 03/22/2019] [Indexed: 12/22/2022]
Abstract
Gastrointestinal Infectious diseases (GIDs) are the second cause of death worldwide. T helper17 cells (Th17) play an important role in GIDs through production of IL-17A, IL-17F, and IL-22 cytokines. Because of their increased activities in GID, Th17 and its inflammatory cytokines can inhibit the progression and eliminate the infection. Actually, although Th17 have the best performance in the acute phase, regulatory T cells (Treg cells) are enhanced in the chronic phase and infection progress through its suppressive function. In addition, Treg cells prevent undesirable inflammatory damages developed by immune system components. On the other hand, miRNAs have important roles in the regulation of immune responses to eliminate bacterial infections and protect host organisms from harmful effects. Actually, miRNAs can reinforce innate and adaptive immunity to remove infections. Of note, miRNAs can develop a regulatory network with the immune system. Additionally, miRNAs can also serve in favor of bacteria to reduce immune responses. Therefore, balance of immune responses in Treg and Th17 cells can influence outcome of many infectious diseases. In conclusion, there is an imbalance in the Treg/Th17 ratio in GIDs; importantly, sets of miRNAs, particularly miR155 and miR146, were determined to be involved clearly in GIDs.
Collapse
Affiliation(s)
| | | | - Mehdi Mahdavi
- Immunotherapy Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran; Recombinant Vaccine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Ranjbar
- Molecular Biology Research Center, System Biology and Poisoning Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Kazem Ahmadi
- Molecular Biology Research Center, System Biology and Poisoning Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ramezan Ali Taheri
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahdi Fasihi-Ramandi
- Molecular Biology Research Center, System Biology and Poisoning Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
96
|
Curto P, Santa C, Allen P, Manadas B, Simões I, Martinez JJ. A Pathogen and a Non-pathogen Spotted Fever Group Rickettsia Trigger Differential Proteome Signatures in Macrophages. Front Cell Infect Microbiol 2019; 9:43. [PMID: 30895174 PMCID: PMC6414445 DOI: 10.3389/fcimb.2019.00043] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/11/2019] [Indexed: 12/13/2022] Open
Abstract
We have previously reported that Rickettsia conorii and Rickettsia montanensis have distinct intracellular fates within THP-1 macrophages, suggesting that the ability to proliferate within macrophages may be a distinguishable factor between pathogenic and non-pathogenic Spotted fever group (SFG) members. To start unraveling the molecular mechanisms underlying the capacity (or not) of SFG Rickettsia to establish their replicative niche in macrophages, we have herein used quantitative proteomics by SWATH-MS to profile the alterations resulted by the challenge of THP-1 macrophages with R. conorii and R. montanensis. We show that the pathogenic, R. conorii, and the non-pathogenic, R. montanensis, member of SFG Rickettsia trigger differential proteomic signatures in macrophage-like cells upon infection. R. conorii specifically induced the accumulation of several enzymes of the tricarboxylic acid cycle, oxidative phosphorylation, fatty acid β-oxidation, and glutaminolysis, as well as of several inner and outer membrane mitochondrial transporters. These results suggest a profound metabolic rewriting of macrophages by R. conorii toward a metabolic signature of an M2-like, anti-inflammatory activation program. Moreover, several subunits forming the proteasome and immunoproteasome are found in lower abundance upon infection with both rickettsial species, which may help bacteria to escape immune surveillance. R. conorii-infection specifically induced the accumulation of several host proteins implicated in protein processing and quality control in ER, suggesting that this pathogenic Rickettsia may be able to increase the ER protein folding capacity. This work reveals novel aspects of macrophage-Rickettsia interactions, expanding our knowledge of how pathogenic rickettsiae explore host cells to their advantage.
Collapse
Affiliation(s)
- Pedro Curto
- PhD Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Cátia Santa
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Paige Allen
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Bruno Manadas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Isaura Simões
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Juan J. Martinez
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
97
|
Wong J, Choi SYC, Liu R, Xu E, Killam J, Gout PW, Wang Y. Potential Therapies for Infectious Diseases Based on Targeting Immune Evasion Mechanisms That Pathogens Have in Common With Cancer Cells. Front Cell Infect Microbiol 2019; 9:25. [PMID: 30809511 PMCID: PMC6379255 DOI: 10.3389/fcimb.2019.00025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/24/2019] [Indexed: 12/18/2022] Open
Abstract
Many global infectious diseases are not well-controlled, underlining a critical need for new, more effective therapies. Pathogens and pathogen-infected host cells, like cancer cells, evade immune surveillance via immune evasion mechanisms. The present study indicates that pathogenic bacteria, endoparasites, and virus-infected host cells can have immune evasion mechanisms in common with cancers. These include entry into dormancy and metabolic reprogramming to aerobic glycolysis leading to excessive secretion of lactic acid and immobilization of local host immunity. The latter evasion tactic provides a therapeutic target for cancer, as shown by our recent finding that patient-derived cancer xenografts can be growth-arrested, without major host toxicity, by inhibiting their lactic acid secretion (as mediated by the MCT4 transporter)-with evidence of host immunity restoration. Accordingly, the multiplication of bacteria, endoparasites, and viruses that primarily depend on metabolic reprogramming to aerobic glycolysis for survival may be arrested using cancer treatment strategies that inhibit their lactic acid secretion. Immune evasion mechanisms shared by pathogens and cancer cells likely represent fundamental, evolutionarily-conserved mechanisms that may be particularly critical to their welfare. As such, their targeting may lead to novel therapies for infectious diseases.
Collapse
Affiliation(s)
- Jodi Wong
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada
| | - Stephen Yiu Chuen Choi
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver, BC, Canada.,Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Rongrong Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Eddie Xu
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver, BC, Canada.,Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - James Killam
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Peter W Gout
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver, BC, Canada.,Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
98
|
Unterholzner L, Almine JF. Camouflage and interception: how pathogens evade detection by intracellular nucleic acid sensors. Immunology 2018; 156:217-227. [PMID: 30499584 PMCID: PMC6376273 DOI: 10.1111/imm.13030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/24/2018] [Accepted: 11/26/2018] [Indexed: 12/16/2022] Open
Abstract
Intracellular DNA and RNA sensors play a vital part in the innate immune response to viruses and other intracellular pathogens, causing the secretion of type I interferons, cytokines and chemokines from infected cells. Pathogen RNA can be detected by retinoic-acid inducible gene I-like receptors in the cytosol, whereas cytosolic DNA is recognized by DNA sensors such as cyclic GMP-AMP synthase (cGAS). The resulting local immune response, which is initiated within hours of infection, is able to eliminate many pathogens before they are able to establish an infection in the host. For this reason, all viruses, and some intracellular bacteria and protozoa, need to evade detection by nucleic acid sensors. Immune evasion strategies include the sequestration and modification of nucleic acids, and the inhibition or degradation of host factors involved in innate immune signalling. Large DNA viruses, such as herpesviruses, often use multiple viral proteins to inhibit signalling cascades at several different points; for instance herpes simplex virus 1 targets both DNA sensors cGAS and interferon-γ-inducible protein 16, as well as the adaptor protein STING (stimulator of interferon genes) and other signalling factors in the pathway. Viruses with a small genome encode only a few immunomodulatory proteins, but these are often multifunctional, such as the NS1 protein from influenza A virus, which inhibits RNA sensing in multiple ways. Intracellular bacteria and protozoa can also be detected by nucleic acid sensors. However, as the type I interferon response is not always beneficial for the host under these circumstances, some bacteria subvert, rather than evade, these signalling cascades for their own gain.
Collapse
Affiliation(s)
- Leonie Unterholzner
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, UK
| | - Jessica F Almine
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, UK
| |
Collapse
|
99
|
Świderská Z, Šmídová A, Buchtová L, Bryjová A, Fabiánová A, Munclinger P, Vinkler M. Avian Toll-like receptor allelic diversity far exceeds human polymorphism: an insight from domestic chicken breeds. Sci Rep 2018; 8:17878. [PMID: 30552359 PMCID: PMC6294777 DOI: 10.1038/s41598-018-36226-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 11/14/2018] [Indexed: 02/07/2023] Open
Abstract
Immune genes show remarkable levels of adaptive variation shaped by pathogen-mediated selection. Compared to humans, however, population polymorphism in animals has been understudied. To provide an insight into immunogenetic diversity in birds, we sequenced complete protein-coding regions of all Toll-like receptor (TLR) genes with direct orthology between mammals and birds (TLR3, TLR4, TLR5 and TLR7) in 110 domestic chickens from 25 breeds and compared their variability with a corresponding human dataset. Chicken TLRs (chTLRs) exhibit on average nine-times higher nucleotide diversity than human TLRs (hTLRs). Increased potentially functional non-synonymous variability is found in chTLR ligand-binding ectodomains. While we identified seven sites in chTLRs under positive selection and found evidence for convergence between alleles, no selection or convergence was detected in hTLRs. Up to six-times more alleles were identified in fowl (70 chTLR4 alleles vs. 11 hTLR4 alleles). In chTLRs, high numbers of alleles are shared between the breeds and the allelic frequencies are more equal than in hTLRs. These differences may have an important impact on infectious disease resistance and host-parasite co-evolution. Though adaptation through high genetic variation is typical for acquired immunity (e.g. MHC), our results show striking levels of intraspecific polymorphism also in poultry innate immune receptors.
Collapse
Grants
- 504214 Grantová Agentura, Univerzita Karlova (Charles University Grant Agency)
- 504214 Grantová Agentura, Univerzita Karlova (Charles University Grant Agency)
- 204069 Univerzita Karlova v Praze (Charles University)
- 204069 Univerzita Karlova v Praze (Charles University)
- PRIMUS/17/SCI/12 Univerzita Karlova v Praze (Charles University)
- SVV 260434/2018 Ministerstvo Školství, Mládeže a Tělovýchovy (Ministry of Education, Youth and Sports)
- INTER-COST LTC18060 Ministerstvo Školství, Mládeže a Tělovýchovy (Ministry of Education, Youth and Sports)
- SVV 260434/2018 Ministerstvo Školství, Mládeže a Tělovýchovy (Ministry of Education, Youth and Sports)
- P502/12/P179 Grantová Agentura České Republiky (Grant Agency of the Czech Republic)
- Grantová Agentura, Univerzita Karlova (Charles University Grant Agency)
- Ministerstvo Školství, Mládeže a Tělovýchovy (Ministry of Education, Youth and Sports)
- Grantová Agentura České Republiky (Grant Agency of the Czech Republic)
Collapse
Affiliation(s)
- Zuzana Świderská
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, Prague, 12843, Czech Republic
- Charles University, Faculty of Science, Department of Cell Biology, Viničná 7, Prague, 12843, Czech Republic
| | - Adéla Šmídová
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, Prague, 12843, Czech Republic
| | - Lucie Buchtová
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, Prague, 12843, Czech Republic
| | - Anna Bryjová
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, Prague, 12843, Czech Republic
- The Czech Academy of Sciences, Institute of Vertebrate Biology, v.v.i., Květná 8, Brno, 60365, Czech Republic
| | - Anežka Fabiánová
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, Prague, 12843, Czech Republic
| | - Pavel Munclinger
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, Prague, 12843, Czech Republic
| | - Michal Vinkler
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, Prague, 12843, Czech Republic.
| |
Collapse
|
100
|
Brock DA, Haselkorn TS, Garcia JR, Bashir U, Douglas TE, Galloway J, Brodie F, Queller DC, Strassmann JE. Diversity of Free-Living Environmental Bacteria and Their Interactions With a Bactivorous Amoeba. Front Cell Infect Microbiol 2018; 8:411. [PMID: 30533398 PMCID: PMC6266680 DOI: 10.3389/fcimb.2018.00411] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/05/2018] [Indexed: 01/06/2023] Open
Abstract
A small subset of bacteria in soil interact directly with eukaryotes. Which ones do so can reveal what is important to a eukaryote and how eukaryote defenses might be breached. Soil amoebae are simple eukaryotic organisms and as such could be particularly good for understanding how eukaryote microbiomes originate and are maintained. One such amoeba, Dictyostelium discoideum, has both permanent and temporary associations with bacteria. Here we focus on culturable bacterial associates in order to interrogate their relationship with D. discoideum. To do this, we isolated over 250 D. discoideum fruiting body samples from soil and deer feces at Mountain Lake Biological Station. In one-third of the wild D. discoideum we tested, one to six bacterial species were found per fruiting body sorus (spore mass) for a total of 174 bacterial isolates. The remaining two-thirds of D. discoideum fruiting body samples did not contain culturable bacteria, as is thought to be the norm. A majority (71.4%) of the unique bacterial haplotypes are in Proteobacteria. The rest are in either Actinobacteria, Bacteriodetes, or Firmicutes. The highest bacterial diversity was found in D. discoideum fruiting bodies originating from deer feces (27 OTUs), greater than either of those originating in shallow (11 OTUs) or in deep soil (4 OTUs). Rarefaction curves and the Chao1 estimator for species richness indicated the diversity in any substrate was not fully sampled, but for soil it came close. A majority of the D. discoideum-associated bacteria were edible by D. discoideum and supported its growth (75.2% for feces and 81.8% for soil habitats). However, we found several bacteria genera were able to evade phagocytosis and persist in D. discoideum cells through one or more social cycles. This study focuses not on the entire D. discoideum microbiome, but on the culturable subset of bacteria that have important eukaryote interactions as prey, symbionts, or pathogens. These eukaryote and bacteria interactions may provide fertile ground for investigations of bacteria using amoebas to gain an initial foothold in eukaryotes and of the origins of symbiosis and simple microbiomes.
Collapse
Affiliation(s)
- Debra A Brock
- Queller/Strassmann Laboratory, Washington University in St. Louis, Department of Biology, St. Louis, MO, United States
| | - Tamara S Haselkorn
- Queller/Strassmann Laboratory, Washington University in St. Louis, Department of Biology, St. Louis, MO, United States
| | - Justine R Garcia
- Queller/Strassmann Laboratory, Washington University in St. Louis, Department of Biology, St. Louis, MO, United States
| | - Usman Bashir
- Queller/Strassmann Laboratory, Washington University in St. Louis, Department of Biology, St. Louis, MO, United States
| | - Tracy E Douglas
- Queller/Strassmann Laboratory, Washington University in St. Louis, Department of Biology, St. Louis, MO, United States
| | - Jesse Galloway
- Mountain Lake Biological Laboratory, University of Virginia, Mountain Lake, VA, United States
| | - Fisher Brodie
- Mountain Lake Biological Laboratory, University of Virginia, Mountain Lake, VA, United States
| | - David C Queller
- Queller/Strassmann Laboratory, Washington University in St. Louis, Department of Biology, St. Louis, MO, United States
| | - Joan E Strassmann
- Queller/Strassmann Laboratory, Washington University in St. Louis, Department of Biology, St. Louis, MO, United States
| |
Collapse
|