1
|
Mahmoud ME, Tingley D, Faizal A, Ghaffar A, Azhar M, Salman D, Isham IM, Abdul-Careem MF. Cyclooxygenase-2/Prostaglandin E2 Pathway Facilitates Infectious Bronchitis Virus-Induced Necroptosis in Chicken Macrophages, a Caspase-Independent Cell Death. Viruses 2025; 17:503. [PMID: 40284946 PMCID: PMC12030959 DOI: 10.3390/v17040503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/15/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Infectious bronchitis virus (IBV) poses a major challenge to poultry health and productivity. This study examined how inflammatory cell death pathways influence the replication and pathogenesis of two IBV strains-respiratory Connecticut (Conn) A5968 and nephropathogenic Delmarva (DMV)/1639-in chicken macrophages. Low serum conditions enhanced viral replication, reduced cell viability, and promoted apoptosis and necroptosis, with DMV/1639 showing more pronounced effects. Modulation of the cyclooxygenase-2/prostaglandin E2 (COX-2/PGE2) pathway displayed strain-specific effects, mitigating necroptosis in DMV/1639-infected cells but exacerbating apoptosis and necroptosis in Conn A5968-infected cells. Broad caspase inhibition (z-VAD-FMK) reduced necroptosis, while selective caspase-1/4 inhibition heightened apoptotic responses. Caspase-8 inhibition selectively reduced necroptosis in DMV/1639 infections but increased apoptosis and necroptosis in Conn A5968 infections. NLRP3 inflammasome and RIPK1 inhibition decreased cell viability and increased apoptosis in both strains but had distinct effects on necroptosis. These findings reveal the strain-specific regulation of viral replication, apoptosis, and necroptosis, underscoring the intricate interplay between IBV and host inflammatory pathways. Understanding these mechanisms provides novel insights into IBV pathogenesis and highlights potential therapeutic strategies to mitigate its impact on poultry health.
Collapse
Affiliation(s)
- Motamed Elsayed Mahmoud
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
- Department of Animal Husbandry, Faculty of Veterinary Medicine, Sohag University, Sohag 84524, Egypt
| | - Dylan Tingley
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
| | - Akeel Faizal
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
| | - Awais Ghaffar
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
| | - Muhammed Azhar
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
| | - Doaa Salman
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
| | - Ishara M. Isham
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
| | - Mohamed Faizal Abdul-Careem
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
| |
Collapse
|
2
|
St. Louis BM, Quagliato SM, Lee PC. Bacterial effector kinases and strategies to identify their target host substrates. Front Microbiol 2023; 14:1113021. [PMID: 36846793 PMCID: PMC9950578 DOI: 10.3389/fmicb.2023.1113021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/25/2023] [Indexed: 02/12/2023] Open
Abstract
Post-translational modifications (PTMs) are critical in regulating protein function by altering chemical characteristics of proteins. Phosphorylation is an integral PTM, catalyzed by kinases and reversibly removed by phosphatases, that modulates many cellular processes in response to stimuli in all living organisms. Consequently, bacterial pathogens have evolved to secrete effectors capable of manipulating host phosphorylation pathways as a common infection strategy. Given the importance of protein phosphorylation in infection, recent advances in sequence and structural homology search have significantly expanded the discovery of a multitude of bacterial effectors with kinase activity in pathogenic bacteria. Although challenges exist due to complexity of phosphorylation networks in host cells and transient interactions between kinases and substrates, approaches are continuously being developed and applied to identify bacterial effector kinases and their host substrates. In this review, we illustrate the importance of exploiting phosphorylation in host cells by bacterial pathogens via the action of effector kinases and how these effector kinases contribute to virulence through the manipulation of diverse host signaling pathways. We also highlight recent developments in the identification of bacterial effector kinases and a variety of techniques to characterize kinase-substrate interactions in host cells. Identification of host substrates provides new insights for regulation of host signaling during microbial infection and may serve as foundation for developing interventions to treat infection by blocking the activity of secreted effector kinases.
Collapse
Affiliation(s)
- Brendyn M. St. Louis
- Department of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, MI, United States
| | - Sydney M. Quagliato
- Department of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, MI, United States
| | | |
Collapse
|
3
|
Witkop EM, Wikfors GH, Proestou DA, Lundgren KM, Sullivan M, Gomez-Chiarri M. Perkinsus marinus suppresses in vitro eastern oyster apoptosis via IAP-dependent and caspase-independent pathways involving TNFR, NF-kB, and oxidative pathway crosstalk. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 129:104339. [PMID: 34998862 DOI: 10.1016/j.dci.2022.104339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/29/2021] [Accepted: 01/01/2022] [Indexed: 06/14/2023]
Abstract
The protozoan parasite Perkinsus marinus causes Dermo disease in eastern oysters, Crassostrea virginica, and can suppress apoptosis of infected hemocytes using incompletely understood mechanisms. This study challenged hemocytes in vitro with P. marinus for 1 h in the presence or absence of caspase inhibitor Z-VAD-FMK or Inhibitor of Apoptosis protein (IAP) inhibitor GDC-0152. Hemocytes exposure to P. marinus significantly reduced granulocyte apoptosis, and pre-incubation with Z-VAD-FMK did not affect P. marinus-induced apoptosis suppression. Hemocyte pre-incubation with GDC-0152 prior to P. marinus challenge further reduced apoptosis of granulocytes with engulfed parasite, but not mitochondrial permeabilization. This suggests P. marinus-induced apoptosis suppression may be caspase-independent, affect an IAP-involved pathway, and occur downstream of mitochondrial permeabilization. P. marinus challenge stimulated hemocyte differential expression of oxidation-reduction, TNFR, and NF-kB pathways. WGCNA analysis of P. marinus expression in response to hemocyte exposure revealed correlated protease, kinase, and hydrolase expression that could contribute to P. marinus-induced apoptosis suppression.
Collapse
Affiliation(s)
- Erin M Witkop
- University of Rhode Island, Department of Fisheries, Animal and Veterinary Science, 120 Flagg Rd, Kingston, RI, USA
| | - Gary H Wikfors
- NOAA Northeast Fisheries Science Center Milford Laboratory, 212 Rogers Ave, Milford, CT, USA
| | - Dina A Proestou
- USDA ARS NEA NCWMAC Shellfish Genetics Program, 120 Flagg Rd, Kingston, RI, USA
| | | | - Mary Sullivan
- USDA ARS NEA NCWMAC Shellfish Genetics Program, 120 Flagg Rd, Kingston, RI, USA
| | - Marta Gomez-Chiarri
- University of Rhode Island, Department of Fisheries, Animal and Veterinary Science, 120 Flagg Rd, Kingston, RI, USA.
| |
Collapse
|
4
|
Freitas IL, Teixeira A, Loureiro I, Lisboa J, Saraiva A, dos Santos NMS, do Vale A. Susceptibility of Sea Bream (Sparus aurata) to AIP56, an AB-Type Toxin Secreted by Photobacterium damselae subsp. piscicida. Toxins (Basel) 2022; 14:toxins14020119. [PMID: 35202146 PMCID: PMC8875918 DOI: 10.3390/toxins14020119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/24/2022] [Accepted: 02/02/2022] [Indexed: 11/16/2022] Open
Abstract
Photobacterium damselae subsp. piscicida (Phdp) is a Gram-negative bacterium that infects a large number of marine fish species in Europe, Asia, and America, both in aquacultures and in the natural environment. Among the affected hosts are economically important cultured fish, such as sea bream (Sparus aurata), sea bass (Dicentrarchus labrax), yellowtail (Seriola quinqueradiata), and cobia (Rachycentron canadum). The best characterized virulence factor of Phdp is the Apoptosis-Inducing Protein of 56 kDa (AIP56), a secreted AB-type toxin that has been shown to induce apoptosis of sea bass phagocytes during infection. AIP56 has an A subunit that displays metalloprotease activity against NF-kB p65 and a B subunit that mediates binding and internalization of the A subunit in susceptible cells. Despite the fact that the aip56 gene is highly prevalent in Phdp isolates from different fish species, the toxicity of AIP56 has only been studied in sea bass. In the present study, the toxicity of AIP56 for sea bream was evaluated. Ex vivo assays showed that sea bream phagocytes are resistant to AIP56 cytotoxicity and that resistance was associated with an inefficient internalization of the toxin by those cells. Accordingly, in vivo intoxication assays revealed that sea bream is much more resistant to AIP56-induced lethality than sea bass. These findings, showing that the effect of AIP56 is different in sea bass and sea bream, set the basis for future studies to characterize the effects of AIP56 and to fully elucidate its virulence role in different Phdp susceptible hosts.
Collapse
Affiliation(s)
- Inês Lua Freitas
- Fish Immunology and Vaccinology Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal; (I.L.F.); (A.T.); (I.L.); (J.L.)
- Fish Immunology and Vaccinology Group, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Alexandra Teixeira
- Fish Immunology and Vaccinology Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal; (I.L.F.); (A.T.); (I.L.); (J.L.)
- Fish Immunology and Vaccinology Group, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Inês Loureiro
- Fish Immunology and Vaccinology Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal; (I.L.F.); (A.T.); (I.L.); (J.L.)
- Fish Immunology and Vaccinology Group, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Johnny Lisboa
- Fish Immunology and Vaccinology Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal; (I.L.F.); (A.T.); (I.L.); (J.L.)
- Fish Immunology and Vaccinology Group, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Aurélia Saraiva
- Biology Department, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal;
- CIIMAR—Interdisciplinary Center of Marine and Environmental Research of the University of Porto, 4450-208 Matosinhos, Portugal
| | - Nuno Miguel Simões dos Santos
- Fish Immunology and Vaccinology Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal; (I.L.F.); (A.T.); (I.L.); (J.L.)
- Fish Immunology and Vaccinology Group, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Correspondence: (N.M.S.d.S.); (A.d.V.); Tel.: +351-226-074-941 (N.M.S.d.S.); +351-220-408-800 (A.d.V.)
| | - Ana do Vale
- Fish Immunology and Vaccinology Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal; (I.L.F.); (A.T.); (I.L.); (J.L.)
- Fish Immunology and Vaccinology Group, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Correspondence: (N.M.S.d.S.); (A.d.V.); Tel.: +351-226-074-941 (N.M.S.d.S.); +351-220-408-800 (A.d.V.)
| |
Collapse
|
5
|
McDaniel MM, Meibers HE, Pasare C. Innate control of adaptive immunity and adaptive instruction of innate immunity: bi-directional flow of information. Curr Opin Immunol 2021; 73:25-33. [PMID: 34425435 PMCID: PMC8648974 DOI: 10.1016/j.coi.2021.07.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 07/14/2021] [Accepted: 07/28/2021] [Indexed: 12/21/2022]
Abstract
The ability of the innate and adaptive immune systems to communicate with each other is central to protective immune responses and maintenance of host health. Myeloid cells of the innate immune system are able to sense microbial ligands, perturbations in cellular homeostasis, and virulence factors, thereby allowing them to relay distinct pathogen-specific information to naïve T cells in the form of pathogen-derived peptides and a unique cytokine milieu. Once primed, effector T helper cells produce lineage-defining cytokines to help combat the original pathogen, and a subset of these cells persist as memory or effector-memory populations. These memory T cells then play a dual role in host protection by not only responding rapidly to reinfection, but by also directly instructing myeloid cells to express licensing cytokines. This means there is a bi-directional flow of information first from the innate to the adaptive immune system, and then from the adaptive back to innate immune system. Here, we focus on how signals, first from pathogens and then from primed effector and memory T cells, are integrated by myeloid cells and its consequences for protective immunity or systemic inflammation.
Collapse
Affiliation(s)
- Margaret M McDaniel
- Immunology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Hannah E Meibers
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45220, United States
| | - Chandrashekhar Pasare
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45220, United States.
| |
Collapse
|
6
|
Cellular and molecular level host-pathogen interactions in Francisella tularensis: A microbial gene network study. Comput Biol Chem 2021; 96:107601. [PMID: 34801846 DOI: 10.1016/j.compbiolchem.2021.107601] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/12/2021] [Accepted: 11/09/2021] [Indexed: 01/17/2023]
Abstract
Due to the high infectivity and fatal effect on human population, Francisella tularensis (F. tularensis) is classified as a potential biological warfare agent. The interaction between host and pathogen behind the successful establishment of F. tularensis infection within the human host is largely unknown. In our present work, we have studied the molecular level interactions between the host cellular components and F. tularensis genes to understand the interplay between the host and pathogen. Interestingly, we have identified the pathways associated with the pathogen offensive strategies that help in invasion of host defensive systems. The F. tularensis genes purL, katG, proS, rpoB and fusA have displayed high number of interactions with the host genes and thus play a crucial role in vital pathogen pathways. The pathways identified were involved in adaptation to different stress conditions within the host and might be crucial for designing new therapeutic interventions against tularemia.
Collapse
|
7
|
Liu Y, Fu K, Wier EM, Lei Y, Hodgson A, Xu D, Xia X, Zheng D, Ding H, Sears CL, Yang J, Wan F. Bacterial genotoxin accelerates transient infection-driven murine colon tumorigenesis. Cancer Discov 2021; 12:236-249. [PMID: 34479870 DOI: 10.1158/2159-8290.cd-21-0912] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/19/2021] [Accepted: 08/24/2021] [Indexed: 12/24/2022]
Abstract
Chronic and low-grade inflammation associated with persistent bacterial infections has been linked to colon tumor development; however, the impact of transient and self-limited infections in bacterially-driven colon tumorigenesis has remained enigmatic. Here we report that UshA is a novel genotoxin in attaching/effacing (A/E) pathogens, which includes the human pathogens enteropathogenic Escherichia coli (EPEC), enterohemorrhagic E. coli (EHEC), and their murine equivalent Citrobacter rodentium (CR). UshA harbors direct DNA digestion activity with a catalytic histidine-aspartic acid dyad. Injected via the Type III Secretion System (T3SS) into host cells, UshA triggers DNA damage and initiates tumorigenic transformation during infections in vitro and in vivo. Moreover, UshA plays an indispensable role in CR infection-accelerated colon tumorigenesis in genetically susceptible ApcMinΔ716/+ mice. Collectively, our results reveal that UshA, functioning as a bacterial T3SS-dependant genotoxin, plays a critical role in prompting transient and noninvasive bacterial infection-accelerated colon tumorigenesis in mice.
Collapse
Affiliation(s)
- Yue Liu
- Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health
| | - Kai Fu
- Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health
| | - Eric M Wier
- Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health
| | - Yifan Lei
- Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health
| | - Andrea Hodgson
- Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health
| | - Dongqing Xu
- Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health
| | - Xue Xia
- Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health
| | - Dandan Zheng
- Chinese Academy of Medical Sciences and Peking Union Medical College
| | - Hua Ding
- Johns Hopkins Bloomberg School of Public Health
| | | | - Jian Yang
- Chinese Academy of Medical Sciences and Peking Union Medical College
| | - Fengyi Wan
- Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health
| |
Collapse
|
8
|
Trxlp, a thioredoxin-like effector from Edwardsiella piscicida inhibits cellular redox signaling and nuclear translocation of NF-κB. Int J Biol Macromol 2020; 148:89-101. [PMID: 31945434 DOI: 10.1016/j.ijbiomac.2020.01.114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/06/2020] [Accepted: 01/11/2020] [Indexed: 11/23/2022]
Abstract
Redox signaling and homeostasis are essential for cell survival and the immune response. Peroxiredoxin (Prx) modulates the level of H2O2 as a redox signal through H2O2 decomposition. The redox activity of thioredoxin (Trx) is required as a reducing equivalent to regenerate Prx. Edwardsiella piscicida is an opportunistic Gram-negative enteric pathogen that secretes a novel Trx-like effector protein, ETAE_2186 (Trxlp). Trxlp has unique structural properties compared with other Trx proteins. In enzymatic and binding assays, we confirmed Trxlp to be redox-inactive due to the low reactivity and flexibility of the resolving cysteine residue, C35, at the active site motif "31WCXXC35". We identified key residues near the active site that are critical for reactivity and flexibility of C35 by site-directed mutagenesis analysis. NMR titration experiment demonstrated prolong inhibitory interaction of Trxlp with Prx1 resulting in the repression of Prx1-mediated H2O2 decomposition leading to increased ROS accumulation in infected host cells. Increased ROS in turn prevented nuclear translocation of NF-κB and inhibition of NF-κB target genes, leading to bacterial survival and enhanced replication inside host cells. Targeting Trxlp-mediated virulence promises to attenuate E. piscicida infection.
Collapse
|
9
|
Chlamydia psittaci PmpD-N Exacerbated Chicken Macrophage Function by Triggering Th2 Polarization and the TLR2/MyD88/NF-κB Signaling Pathway. Int J Mol Sci 2020; 21:ijms21062003. [PMID: 32183481 PMCID: PMC7139469 DOI: 10.3390/ijms21062003] [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: 01/30/2020] [Revised: 02/29/2020] [Accepted: 03/12/2020] [Indexed: 12/25/2022] Open
Abstract
The polymorphic membrane protein D (PmpD) is a highly conserved outer membrane protein which plays an important role in pathogenesis during Chlamydia psittaci infection. In this study, we evaluated the ability of the N-terminus of PmpD (PmpD-N) to modulate the functions of chicken macrophages and the signaling pathway(s) involved in PmpD-N-induced Toll-like receptors (TLRs), as well as interleukin (IL)-6 and IL-10 cytokine secretions. Thus, HD11 macrophages were treated with exogenous and intracellular PmpD-N of C. psittaci. The chlamydial growth was evaluated by enumeration of chlamydial loads in the infected macrophages. The phagocytic function of macrophages following PmpD-N treatment was detected by fluorescein-labeled Escherichia coli (E. coli). The concentration of nitric oxide (NO) secreted by HD11 macrophages was measured by the amount of NO2- in the culture supernatant using the Griess method. The cytokine secretions were assessed using multiplex cytokine ELISA kits. Expression levels of TLRs, myeloid differentiation factor 88 (MyD88), and nuclear factor kappa B (NF-κB) were analyzed by a Western blotting assay, as well as a luciferase assay, while NF-κB p65 nuclear translocation was assessed by confocal microscopy. The nuclear translocation of the transcription factor NF-κB was confirmed by evaluating its ability to combine with the corresponding promoter using the electrophoretic mobility shift assay (EMSA). After treatment with exogenous or endogenous PmpD-N, chlamydial loads and phagocytic functions were reduced significantly compared with those of the plasmid vector group, while NO secretions were reduced significantly compared with those of the lipopolysaccharide (LPS) treatment. Stimulation of HD11 cells with PmpD-N provoked the secretion of the Th2 cytokines, IL-6, and IL-10 and upregulated the expression of TLR2, TLR4, MyD88, and NF-κB. Furthermore, inhibition of TLR2, MyD88, and NF-κB in HD11 cells significantly decreased IL-6 and IL-10 cytokine levels, while NO production and phagocytosis increased significantly, strongly suggesting their involvement in PmpD-N-induced Th2 cytokine secretion and macrophage dysfunction. Our data indicate that C. psittaci PmpD-N inhibited macrophage functions by activating the Th2 immune response and the TLR2/MyD88/NF-κB signaling pathway.
Collapse
|
10
|
Xia X, Liu Y, Hodgson A, Xu D, Guo W, Yu H, She W, Zhou C, Lan L, Fu K, Vallance BA, Wan F. EspF is crucial for Citrobacter rodentium-induced tight junction disruption and lethality in immunocompromised animals. PLoS Pathog 2019; 15:e1007898. [PMID: 31251784 PMCID: PMC6623547 DOI: 10.1371/journal.ppat.1007898] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 07/11/2019] [Accepted: 06/05/2019] [Indexed: 12/17/2022] Open
Abstract
Attaching/Effacing (A/E) bacteria include human pathogens enteropathogenic Escherichia coli (EPEC), enterohemorrhagic E. coli (EHEC), and their murine equivalent Citrobacter rodentium (CR), of which EPEC and EHEC are important causative agents of foodborne diseases worldwide. While A/E pathogen infections cause mild symptoms in the immunocompetent hosts, an increasing number of studies show that they produce more severe morbidity and mortality in immunocompromised and/or immunodeficient hosts. However, the pathogenic mechanisms and crucial host-pathogen interactions during A/E pathogen infections under immunocompromised conditions remain elusive. We performed a functional screening by infecting interleukin-22 (IL-22) knockout (Il22-/-) mice with a library of randomly mutated CR strains. Our screen reveals that interruption of the espF gene, which encodes the Type III Secretion System effector EspF (E. coli secreted protein F) conserved among A/E pathogens, completely abolishes the high mortality rates in CR-infected Il22-/- mice. Chromosomal deletion of espF in CR recapitulates the avirulent phenotype without impacting colonization and proliferation of CR, and EspF complement in ΔespF strain fully restores the virulence in mice. Moreover, the expression levels of the espF gene are elevated during CR infection and CR induces disruption of the tight junction (TJ) strands in colonic epithelium in an EspF-dependent manner. Distinct from EspF, chromosomal deletion of other known TJ-damaging effector genes espG and map failed to impede CR virulence in Il22-/- mice. Hence our findings unveil a critical pathophysiological function for EspF during CR infection in the immunocompromised host and provide new insights into the complex pathogenic mechanisms of A/E pathogens.
Collapse
Affiliation(s)
- Xue Xia
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Yue Liu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Andrea Hodgson
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Dongqing Xu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Wenxuan Guo
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Hongbing Yu
- Division of Gastroenterology, Department of Pediatrics, BC's Children's Hospital and Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Weifeng She
- Eudowood Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Chenxing Zhou
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu, PR China
| | - Lei Lan
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu, PR China
| | - Kai Fu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
- Institute of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Bruce A. Vallance
- Division of Gastroenterology, Department of Pediatrics, BC's Children's Hospital and Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Fengyi Wan
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States of America
| |
Collapse
|
11
|
Dorrington MG, Fraser IDC. NF-κB Signaling in Macrophages: Dynamics, Crosstalk, and Signal Integration. Front Immunol 2019; 10:705. [PMID: 31024544 PMCID: PMC6465568 DOI: 10.3389/fimmu.2019.00705] [Citation(s) in RCA: 480] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/14/2019] [Indexed: 12/12/2022] Open
Abstract
The nuclear factor-κB (NF-κB) signaling pathway is one of the best understood immune-related pathways thanks to almost four decades of intense research. NF-κB signaling is activated by numerous discrete stimuli and is a master regulator of the inflammatory response to pathogens and cancerous cells, as well as a key regulator of autoimmune diseases. In this regard, the role of NF-κB signaling in immunity is not unlike that of the macrophage. The dynamics by which NF-κB proteins shuttle between the cytoplasm and the nucleus to initiate transcription have been studied rigorously in fibroblasts and other non-hematopoietic cells, but many questions remain as to how current models of NF-κB signaling and dynamics can be translated to innate immune cells such as macrophages. In this review, we will present recent research on the dynamics of NF-κB signaling and focus especially on how these dynamics vary in different cell types, while discussing why these characteristics may be important. We will end by looking ahead to how new techniques and technologies should allow us to analyze these signaling processes with greater clarity, bringing us closer to a more complete understanding of inflammatory transcription factor dynamics and how different cellular contexts might allow for appropriate control of innate immune responses.
Collapse
Affiliation(s)
- Michael G Dorrington
- Signaling Systems Section, Laboratory of Immune System Biology, NIAID, DIR, NIH, Bethesda, MD, United States
| | - Iain D C Fraser
- Signaling Systems Section, Laboratory of Immune System Biology, NIAID, DIR, NIH, Bethesda, MD, United States
| |
Collapse
|
12
|
Sun Y, Detchemendy TW, Pajerowska-Mukhtar KM, Mukhtar MS. NPR1 in JazzSet with Pathogen Effectors. TRENDS IN PLANT SCIENCE 2018; 23:469-472. [PMID: 29753632 DOI: 10.1016/j.tplants.2018.04.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/20/2018] [Accepted: 04/24/2018] [Indexed: 05/26/2023]
Abstract
NON-EXPRESSOR OF PATHOGENESIS-RELATED GENES 1 (NPR1) is a master regulator of salicylic acid (SA)-mediated systemic acquired resistance (SAR), a broad-spectrum disease resistance mechanism in plants. NPR1 controls approximately 90% of SA-dependent transcriptome in Arabidopsis. Here, we discuss how pathogen effectors manipulate NPR1 functions in different cellular compartments to establish disease.
Collapse
Affiliation(s)
- Yali Sun
- Department of Biology, University of Alabama at Birmingham, AL 35294, USA
| | | | | | - M Shahid Mukhtar
- Department of Biology, University of Alabama at Birmingham, AL 35294, USA; Nutrition Obesity Research Center, University of Alabama at Birmingham, AL 35294, USA.
| |
Collapse
|
13
|
Pan Q, Zhang Q, Chu J, Pais R, Liu S, He C, Eko FO. Chlamydia abortus Pmp18.1 Induces IL-1β Secretion by TLR4 Activation through the MyD88, NF-κB, and Caspase-1 Signaling Pathways. Front Cell Infect Microbiol 2017; 7:514. [PMID: 29326885 PMCID: PMC5741698 DOI: 10.3389/fcimb.2017.00514] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/29/2017] [Indexed: 12/16/2022] Open
Abstract
The polymorphic membrane protein D (Pmp18D) is a 160-kDa outer membrane protein that is conserved and plays an important role in Chlamydia abortus pathogenesis. We have identified an N-terminal fragment of Pmp18D (designated Pmp18.1) as a possible subunit vaccine antigen. In this study, we evaluated the vaccine potential of Pmp18.1 by investigating its ability to induce innate immune responses in dendritic cells and the signaling pathway(s) involved in rPmp18.1-induced IL-1β secretion. We next investigated the immunomodulatory impact of VCG, in comparison with the more established Th1-promoting adjuvants, CpG and FL, on rPmp18.1-mediated innate immune activation. Finally, the effect of siRNA targeting TLR4, MyD88, NF-κB p50, and Caspase-1 mRNA in DCs on IL-1β cytokine secretion was also investigated. Bone marrow-derived dendritic cells (BMDCs) were stimulated with rPmp18.1 in the presence or absence of VCG or CpG or FL and the magnitude of cytokines produced was assessed using a multiplex cytokine ELISA assay. Expression of costimulatory molecules and Toll-like receptors (TLRs) was analyzed by flow cytometry. Quantitation of intracellular levels of myeloid differentiation factor 88 (MyD88), nuclear factor kappa beta (NF-κB p50/p65), and Caspase-1 was evaluated by Western immunoblotting analysis while NF-κB p65 nuclear translocation was assessed by confocal microscopy. The results showed DC stimulation with rPmp18.1 provoked the secretion of proinflammatory cytokines and upregulated expression of TLRs and co-stimulatory molecules associated with DC maturation. These responses were significantly (p ≤ 0.001) enhanced by VCG but not CpG or FL. In addition, rPmp18.1 activated the expression of MyD88, NF-κB p50, and Caspase-1 as well as the nuclear expression of NF-κB p65 in treated DCs. Furthermore, targeting TLR4, MyD88, NF-κB p50, and Caspase-1 mRNA in BMDCs with siRNA significantly reduced their expression levels, resulting in decreased IL-1β cytokine secretion, strongly suggesting their involvement in the rPmp18.1-induced IL-1β cytokine secretion. Taken together, these results indicate that C. abortus Pmp18.1 induces IL-1β secretion by TLR4 activation through the MyD88, NF-κB as well as the Caspase-1 signaling pathways and may be a potential C. abortus vaccine candidate. The vaccine potential of Pmp18.1 will subsequently be evaluated in an appropriate animal model, using VCG as an immunomodulator, following immunization and challenge.
Collapse
Affiliation(s)
- Qing Pan
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States.,Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Qiang Zhang
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jun Chu
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Roshan Pais
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Shanshan Liu
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Cheng He
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Francis O Eko
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
14
|
Zolotarenko A, Chekalin E, Mehta R, Baranova A, Tatarinova TV, Bruskin S. Identification of Transcriptional Regulators of Psoriasis from RNA-Seq Experiments. Methods Mol Biol 2017; 1613:355-370. [PMID: 28849568 DOI: 10.1007/978-1-4939-7027-8_14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Psoriasis is a common inflammatory skin disease with complex etiology and chronic progression. To provide novel insights into the molecular mechanisms of regulation of the disease we performed RNA sequencing (RNA-Seq) analysis of 14 pairs of skin samples collected from psoriatic patients. Subsequent pathway analysis and an extraction of transcriptional regulators governing psoriasis-associated pathways was executed using a combination of MetaCore Interactome enrichment tool and cisExpress algorithm, and followed by comparison to a set of previously described psoriasis response elements. A comparative approach has allowed us to identify 42 core transcriptional regulators of the disease associated with inflammation (NFkB, IRF9, JUN, FOS, SRF), activity of T-cells in the psoriatic lesions (STAT6, FOXP3, NFATC2, GATA3, TCF7, RUNX1, etc.), hyperproliferation and migration of keratinocytes (JUN, FOS, NFIB, TFAP2A, TFAP2C), and lipid metabolism (TFAP2, RARA, VDR). After merging the ChIP-seq and RNA-seq data, we conclude that the atypical expression of FOXA1 transcriptional factor is an important player in psoriasis, as it inhibits maturation of naive T cells into this Treg subpopulation (CD4+FOXA1+CD47+CD69+PD-L1(hi)FOXP3-), therefore contributing to the development of psoriatic skin lesions.
Collapse
Affiliation(s)
- Alena Zolotarenko
- Laboratory of Functional Genomics, Vavilov Institute of General Genetics RAS, Gubkina Street, 3119991, Moscow, Russia
| | - Evgeny Chekalin
- Laboratory of Functional Genomics, Vavilov Institute of General Genetics RAS, Gubkina Street, 3119991, Moscow, Russia
| | - Rohini Mehta
- The Center of the Study of Chronic Metabolic and Rare Diseases, School of Systems Biology, George Mason University, Fairfax, VA, USA
| | - Ancha Baranova
- The Center of the Study of Chronic Metabolic and Rare Diseases, School of Systems Biology, George Mason University, Fairfax, VA, USA
- Research Centre for Medical Genetics RAMS, Moscow, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow, Russia
- Atlas Biomed Group, Moscow, Russia
| | - Tatiana V Tatarinova
- Atlas Biomed Group, Moscow, Russia
- Center for Personalized Medicine, Children's Hospital Los Angeles and Spatial Sciences Institute, University of Southern California, Los Angeles, CA, USA
- A.A. Kharkevich Institute for Information Transmission Problems RAS, Moscow, Russia
| | - Sergey Bruskin
- Laboratory of Functional Genomics, Vavilov Institute of General Genetics RAS, Gubkina Street, 3119991, Moscow, Russia.
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow, Russia.
| |
Collapse
|
15
|
Integrated computational approach to the analysis of RNA-seq data reveals new transcriptional regulators of psoriasis. Exp Mol Med 2016; 48:e268. [PMID: 27811935 PMCID: PMC5133374 DOI: 10.1038/emm.2016.97] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 05/06/2016] [Accepted: 05/24/2016] [Indexed: 02/07/2023] Open
Abstract
Psoriasis is a common inflammatory skin disease with complex etiology and chronic progression. To provide novel insights into the regulatory molecular mechanisms of the disease, we performed RNA sequencing analysis of 14 pairs of skin samples collected from patients with psoriasis. Subsequent pathway analysis and extraction of the transcriptional regulators governing psoriasis-associated pathways was executed using a combination of the MetaCore Interactome enrichment tool and the cisExpress algorithm, followed by comparison to a set of previously described psoriasis response elements. A comparative approach allowed us to identify 42 core transcriptional regulators of the disease associated with inflammation (NFκB, IRF9, JUN, FOS, SRF), the activity of T cells in psoriatic lesions (STAT6, FOXP3, NFATC2, GATA3, TCF7, RUNX1), the hyperproliferation and migration of keratinocytes (JUN, FOS, NFIB, TFAP2A, TFAP2C) and lipid metabolism (TFAP2, RARA, VDR). In addition to the core regulators, we identified 38 transcription factors previously not associated with the disease that can clarify the pathogenesis of psoriasis. To illustrate these findings, we analyzed the regulatory role of one of the identified transcription factors (TFs), FOXA1. Using ChIP-seq and RNA-seq data, we concluded that the atypical expression of the FOXA1 TF is an important player in the disease as it inhibits the maturation of naive T cells into the (CD4+FOXA1+CD47+CD69+PD-L1(hi)FOXP3-) regulatory T cell subpopulation, therefore contributing to the development of psoriatic skin lesions.
Collapse
|
16
|
Kjærner-Semb E, Ayllon F, Furmanek T, Wennevik V, Dahle G, Niemelä E, Ozerov M, Vähä JP, Glover KA, Rubin CJ, Wargelius A, Edvardsen RB. Atlantic salmon populations reveal adaptive divergence of immune related genes - a duplicated genome under selection. BMC Genomics 2016; 17:610. [PMID: 27515098 PMCID: PMC4982270 DOI: 10.1186/s12864-016-2867-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 06/30/2016] [Indexed: 12/31/2022] Open
Abstract
Background Populations of Atlantic salmon display highly significant genetic differences with unresolved molecular basis. These differences may result from separate postglacial colonization patterns, diversifying natural selection and adaptation, or a combination. Adaptation could be influenced or even facilitated by the recent whole genome duplication in the salmonid lineage which resulted in a partly tetraploid species with duplicated genes and regions. Results In order to elucidate the genes and genomic regions underlying the genetic differences, we conducted a genome wide association study using whole genome resequencing data from eight populations from Northern and Southern Norway. From a total of ~4.5 million sequencing-derived SNPs, more than 10 % showed significant differentiation between populations from these two regions and ten selective sweeps on chromosomes 5, 10, 11, 13–15, 21, 24 and 25 were identified. These comprised 59 genes, of which 15 had one or more differentiated missense mutation. Our analysis showed that most sweeps have paralogous regions in the partially tetraploid genome, each lacking the high number of significant SNPs found in the sweeps. The most significant sweep was found on Chr 25 and carried several missense mutations in the antiviral mx genes, suggesting that these populations have experienced differing viral pressures. Interestingly the second most significant sweep, found on Chr 5, contains two genes involved in the NF-KB pathway (nkap and nkrf), which is also a known pathogen target that controls a large number of processes in animals. Conclusion Our results show that natural selection acting on immune related genes has contributed to genetic divergence between salmon populations in Norway. The differences between populations may have been facilitated by the plasticity of the salmon genome. The observed signatures of selection in duplicated genomic regions suggest that the recently duplicated genome has provided raw material for evolutionary adaptation. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2867-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Erik Kjærner-Semb
- Institute of Marine Research, Bergen, Norway. .,Department of Biology, University of Bergen, Bergen, Norway.
| | | | | | | | - Geir Dahle
- Institute of Marine Research, Bergen, Norway
| | - Eero Niemelä
- Natural Resources Institute Finland, Helsinki, Finland
| | - Mikhail Ozerov
- Kevo Subarctic Research Institute, University of Turku, Turku, Finland
| | - Juha-Pekka Vähä
- Kevo Subarctic Research Institute, University of Turku, Turku, Finland.,Association for Water and Environment of Western Uusimaa, Uusimaa, Finland
| | - Kevin A Glover
- Institute of Marine Research, Bergen, Norway.,Department of Biology, University of Bergen, Bergen, Norway
| | - Carl J Rubin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | | | | |
Collapse
|