1
|
Zhang T, Chien RC, Budachetri K, Lin M, Boyaka P, Huang W, Rikihisa Y. Ehrlichia effector TRP120 manipulates bacteremia to facilitate tick acquisition. mBio 2024; 15:e0047624. [PMID: 38501870 PMCID: PMC11005420 DOI: 10.1128/mbio.00476-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 03/03/2024] [Indexed: 03/20/2024] Open
Abstract
Ehrlichia species are obligatory intracellular bacteria that cause a potentially fatal disease, human ehrlichiosis. The biomolecular mechanisms of tick acquisition of Ehrlichia and transmission between ticks and mammals are poorly understood. Ehrlichia japonica infection of mice recapitulates the full spectrum of human ehrlichiosis. We compared the pathogenicity and host acquisition of wild-type E. japonica with an isogenic transposon mutant of E. japonica that lacks tandem repeat protein 120 (TRP120) (ΔTRP120). Both wild-type and ΔTRP120 E. japonica proliferated similarly in cultures of mammalian and tick cells. Upon inoculation into mice, both wild-type and ΔTRP120 E. japonica multiplied to high levels in various tissues, with similar clinical chemistry and hematologic changes, proinflammatory cytokine induction, and fatal disease. However, the blood levels of ΔTRP120 E. japonica were almost undetectable within 24 h, whereas the levels of the wild type increased exponentially. Greater than 90% of TRP120 was released from infected cells into the culture medium. Mouse blood monocytes exposed to native TRP120 from culture supernatants showed significantly reduced cell surface expression of the transmigration-related markers Ly6C and CD11b. Larval ticks attached to mice infected with either wild-type or ΔTRP120 E. japonica imbibed similar amounts of blood and subsequently molted to nymphs at similar rates. However, unlike wild-type E. japonica, the ΔTRP120 mutant was minimally acquired by larval ticks and subsequent molted nymphs and, thus, failed to transmit to naïve mice. Thus, TRP120 is required for bacteremia but not disease. These findings suggest a novel mechanism whereby an obligatory intracellular bacterium manipulates infected blood monocytes to sustain the tick-mammal transmission cycle. IMPORTANCE Effective prevention of tick-borne diseases such as human ehrlichiosis requires an understanding of how disease-causing organisms are acquired. Ehrlichia species are intracellular bacteria that require infection of both mammals and ticks, involving cycles of transmission between them. Mouse models of ehrlichiosis and tick-mouse transmission can advance our fundamental understanding of the pathogenesis and prevention of ehrlichiosis. Herein, a mutant of Ehrlichia japonica was used to investigate the role of a single Ehrlichia factor, named tandem repeat protein 120 (TRP120), in infection of mammalian and tick cells in culture, infection and disease progression in mice, and tick acquisition of E. japonica from infected mice. Our results suggest that TRP120 is necessary only for Ehrlichia proliferation in circulating mouse blood and ongoing bacteremia to permit Ehrlichia acquisition by ticks. This study provides new insights into the importance of bacterial factors in regulating bacteremia, which may facilitate tick acquisition of pathogens.
Collapse
Affiliation(s)
- Tsian Zhang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Rory C. Chien
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Khemraj Budachetri
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Mingqun Lin
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Prosper Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Weiyan Huang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Yasuko Rikihisa
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
2
|
Sharma AK, Ismail N. Non-Canonical Inflammasome Pathway: The Role of Cell Death and Inflammation in Ehrlichiosis. Cells 2023; 12:2597. [PMID: 37998332 PMCID: PMC10670716 DOI: 10.3390/cells12222597] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Activating inflammatory caspases and releasing pro-inflammatory mediators are two essential functions of inflammasomes which are triggered in response to pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs). The canonical inflammasome pathway involves the activation of inflammasome and its downstream pathway via the adaptor ASC protein, which causes caspase 1 activation and, eventually, the cleavage of pro-IL-1b and pro-IL-18. The non-canonical inflammasome pathway is induced upon detecting cytosolic lipopolysaccharide (LPS) by NLRP3 inflammasome in Gram-negative bacteria. The activation of NLRP3 triggers the cleavage of murine caspase 11 (human caspase 4 or caspase 5), which results in the formation of pores (via gasdermin) to cause pyroptosis. Ehrlichia is an obligately intracellular bacterium which is responsible for causing human monocytic ehrlichiosis (HME), a potentially lethal disease similar to toxic shock syndrome and septic shock syndrome. Several studies have indicated that canonical and non-canonical inflammasome activation is a crucial pathogenic mechanism that induces dysregulated inflammation and host cellular death in the pathophysiology of HME. Mechanistically, the activation of canonical and non-canonical inflammasome pathways affected by virulent Ehrlichia infection is due to a block in autophagy. This review aims to explore the significance of non-canonical inflammasomes in ehrlichiosis, and how the pathways involving caspases (with the exception of caspase 1) contribute to the pathophysiology of severe and fatal ehrlichiosis. Improving our understanding of the non-canonical inflammatory pathway that cause cell death and inflammation in ehrlichiosis will help the advancement of innovative therapeutic, preventative, and diagnostic approaches to the treatment of ehrlichiosis.
Collapse
Affiliation(s)
| | - Nahed Ismail
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA;
| |
Collapse
|
3
|
Teymournejad O, Sharma AK, Abdelwahed M, Kader M, Ahmed I, Elkafas H, Ismail N. Hepatocyte-specific regulation of autophagy and inflammasome activation via MyD88 during lethal Ehrlichia infection. Front Immunol 2023; 14:1212167. [PMID: 38022511 PMCID: PMC10662044 DOI: 10.3389/fimmu.2023.1212167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Hepatocytes play a crucial role in host response to infection. Ehrlichia is an obligate intracellular bacterium that causes potentially life-threatening human monocytic ehrlichiosis (HME) characterized by an initial liver injury followed by sepsis and multi-organ failure. We previously showed that infection with highly virulent Ehrlichia japonica (E. japonica) induces liver damage and fatal ehrlichiosis in mice via deleterious MyD88-dependent activation of CASP11 and inhibition of autophagy in macrophage. While macrophages are major target cells for Ehrlichia, the role of hepatocytes (HCs) in ehrlichiosis remains unclear. We investigated here the role of MyD88 signaling in HCs during infection with E. japonica using primary cells from wild-type (WT) and MyD88-/- mice, along with pharmacologic inhibitors of MyD88 in a murine HC cell line. Similar to macrophages, MyD88 signaling in infected HCs led to deleterious CASP11 activation, cleavage of Gasdermin D, secretion of high mobility group box 1, IL-6 production, and inflammatory cell death, while controlling bacterial replication. Unlike macrophages, MyD88 signaling in Ehrlichia-infected HCs attenuated CASP1 activation but activated CASP3. Mechanistically, active CASP1/canonical inflammasome pathway negatively regulated the activation of CASP3 in infected MyD88-/- HCs. Further, MyD88 promoted autophagy induction in HCs, which was surprisingly associated with the activation of the mammalian target of rapamycin complex 1 (mTORC1), a known negative regulator of autophagy. Pharmacologic blocking mTORC1 activation in E. japonica-infected WT, but not infected MyD88-/- HCs, resulted in significant induction of autophagy, suggesting that MyD88 promotes autophagy during Ehrlichia infection not only in an mTORC1-indpenedent manner, but also abrogates mTORC1-mediated inhibition of autophagy in HCs. In conclusion, this study demonstrates that hepatocyte-specific regulation of autophagy and inflammasome pathway via MyD88 is distinct than MyD88 signaling in macrophages during fatal ehrlichiosis. Understanding hepatocyte-specific signaling is critical for the development of new therapeutics against liver-targeting pathogens such as Ehrlichia.
Collapse
Affiliation(s)
- Omid Teymournejad
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Aditya Kumar Sharma
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Mohammed Abdelwahed
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Hofstra School of Medicine, North Well Health, New York, NY, United States
| | - Muhamuda Kader
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ibrahim Ahmed
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Hoda Elkafas
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Nahed Ismail
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
4
|
Ismail N, Sharma A, Soong L, Walker DH. Review: Protective Immunity and Immunopathology of Ehrlichiosis. ZOONOSES (BURLINGTON, MASS.) 2022; 2:10.15212/zoonoses-2022-0009. [PMID: 35876763 PMCID: PMC9300479 DOI: 10.15212/zoonoses-2022-0009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Human monocytic ehrlichiosis, a tick transmitted infection, ranges in severity from apparently subclinical to a fatal toxic shock-like fatal disease. Models in immunocompetent mice range from an abortive infection to uniformly lethal depending on the infecting Ehrlichia species, dose of inoculum, and route of inoculation. Effective immunity is mediated by CD4+ T lymphocytes and gamma interferon. Lethal infection occurs with early overproduction of proinflammatory cytokines and overproduction of TNF alpha and IL-10 by CD8+ T lymphocytes. Furthermore, fatal ehrlichiosis is associated with signaling via TLR 9/MyD88 with upregulation of several inflammasome complexes and secretion of IL-1 beta, IL-1 alpha, and IL-18 by hepatic mononuclear cells, suggesting activation of canonical and noncanonical inflammasome pathways, a deleterious role for IL-18, and the protective role for caspase 1. Autophagy promotes ehrlichial infection, and MyD88 signaling hinders ehrlichial infection by inhibiting autophagy induction and flux. Activation of caspase 11 during infection of hepatocytes by the lethal ehrlichial species after interferon alpha receptor signaling results in the production of inflammasome-dependent IL-1 beta, extracellular secretion of HMGB1, and pyroptosis. The high level of HMGB1 in lethal ehrlichiosis suggests a role in toxic shock. Studies of primary bone marrow-derived macrophages infected by highly avirulent or mildly avirulent ehrlichiae reveal divergent M1 and M2 macrophage polarization that links with generation of pathogenic CD8 T cells, neutrophils, and excessive inflammation or with strong expansion of protective Th1 and NKT cells, resolution of inflammation and clearance of infection, respectively.
Collapse
Affiliation(s)
- Nahed Ismail
- Clinical Microbiology, Laboratory Medicine, University of Illinois at Chicago-College of Medicine, University of Illinois Hospitals & Health Science System, Chicago, IL
| | - Aditya Sharma
- Clinical Microbiology, Laboratory Medicine, University of Illinois at Chicago-College of Medicine, University of Illinois Hospitals & Health Science System, Chicago, IL
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
- Department of Pathology, Center for Biodefense & Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX
| | - David H. Walker
- Department of Pathology, Center for Biodefense & Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
5
|
Böning MAL, Parzmair GP, Jeron A, Düsedau HP, Kershaw O, Xu B, Relja B, Schlüter D, Dunay IR, Reinhold A, Schraven B, Bruder D. Enhanced Susceptibility of ADAP-Deficient Mice to Listeria monocytogenes Infection Is Associated With an Altered Phagocyte Phenotype and Function. Front Immunol 2021; 12:724855. [PMID: 34659211 PMCID: PMC8515145 DOI: 10.3389/fimmu.2021.724855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/03/2021] [Indexed: 12/04/2022] Open
Abstract
The adhesion and degranulation-promoting adaptor protein (ADAP) serves as a multifunctional scaffold and is involved in the formation of immune signaling complexes. To date, only limited data exist regarding the role of ADAP in pathogen-specific immunity during in vivo infection, and its contribution in phagocyte-mediated antibacterial immunity remains elusive. Here, we show that mice lacking ADAP (ADAPko) are highly susceptible to the infection with the intracellular pathogen Listeria monocytogenes (Lm) by showing enhanced immunopathology in infected tissues together with increased morbidity, mortality, and excessive infiltration of neutrophils and monocytes. Despite high phagocyte numbers in the spleen and liver, ADAPko mice only inefficiently controlled pathogen growth, hinting at a functional impairment of infection-primed phagocytes in the ADAP-deficient host. Flow cytometric analysis of hallmark pro-inflammatory mediators and unbiased whole genome transcriptional profiling of neutrophils and inflammatory monocytes uncovered broad molecular alterations in the inflammatory program in both phagocyte subsets following their activation in the ADAP-deficient host. Strikingly, ex vivo phagocytosis assay revealed impaired phagocytic capacity of neutrophils derived from Lm-infected ADAPko mice. Together, our data suggest that an alternative priming of phagocytes in ADAP-deficient mice during Lm infection induces marked alterations in the inflammatory profile of neutrophils and inflammatory monocytes that contribute to enhanced immunopathology while limiting their capacity to eliminate the pathogen and to prevent the fatal outcome of the infection.
Collapse
Affiliation(s)
- Martha A L Böning
- Infection Immunology, Institute of Medical Microbiology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany.,Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Gerald P Parzmair
- Infection Immunology, Institute of Medical Microbiology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany.,Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Andreas Jeron
- Infection Immunology, Institute of Medical Microbiology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany.,Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Henning P Düsedau
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Olivia Kershaw
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität, Berlin, Germany
| | - Baolin Xu
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Dirk Schlüter
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany.,Institute of Medical Microbiology and Hospital Hygiene, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Otto-von-Guericke University, Magdeburg, Germany
| | - Annegret Reinhold
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Dunja Bruder
- Infection Immunology, Institute of Medical Microbiology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany.,Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
6
|
Lin M, Xiong Q, Chung M, Daugherty SC, Nagaraj S, Sengamalay N, Ott S, Godinez A, Tallon LJ, Sadzewicz L, Fraser C, Dunning Hotopp JC, Rikihisa Y. Comparative Analysis of Genome of Ehrlichia sp. HF, a Model Bacterium to Study Fatal Human Ehrlichiosis. BMC Genomics 2021; 22:11. [PMID: 33407096 PMCID: PMC7789307 DOI: 10.1186/s12864-020-07309-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The genus Ehrlichia consists of tick-borne obligatory intracellular bacteria that can cause deadly diseases of medical and agricultural importance. Ehrlichia sp. HF, isolated from Ixodes ovatus ticks in Japan [also referred to as I. ovatus Ehrlichia (IOE) agent], causes acute fatal infection in laboratory mice that resembles acute fatal human monocytic ehrlichiosis caused by Ehrlichia chaffeensis. As there is no small laboratory animal model to study fatal human ehrlichiosis, Ehrlichia sp. HF provides a needed disease model. However, the inability to culture Ehrlichia sp. HF and the lack of genomic information have been a barrier to advance this animal model. In addition, Ehrlichia sp. HF has several designations in the literature as it lacks a taxonomically recognized name. RESULTS We stably cultured Ehrlichia sp. HF in canine histiocytic leukemia DH82 cells from the HF strain-infected mice, and determined its complete genome sequence. Ehrlichia sp. HF has a single double-stranded circular chromosome of 1,148,904 bp, which encodes 866 proteins with a similar metabolic potential as E. chaffeensis. Ehrlichia sp. HF encodes homologs of all virulence factors identified in E. chaffeensis, including 23 paralogs of P28/OMP-1 family outer membrane proteins, type IV secretion system apparatus and effector proteins, two-component systems, ankyrin-repeat proteins, and tandem repeat proteins. Ehrlichia sp. HF is a novel species in the genus Ehrlichia, as demonstrated through whole genome comparisons with six representative Ehrlichia species, subspecies, and strains, using average nucleotide identity, digital DNA-DNA hybridization, and core genome alignment sequence identity. CONCLUSIONS The genome of Ehrlichia sp. HF encodes all known virulence factors found in E. chaffeensis, substantiating it as a model Ehrlichia species to study fatal human ehrlichiosis. Comparisons between Ehrlichia sp. HF and E. chaffeensis will enable identification of in vivo virulence factors that are related to host specificity, disease severity, and host inflammatory responses. We propose to name Ehrlichia sp. HF as Ehrlichia japonica sp. nov. (type strain HF), to denote the geographic region where this bacterium was initially isolated.
Collapse
Affiliation(s)
- Mingqun Lin
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA.
| | - Qingming Xiong
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
| | - Matthew Chung
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Sean C Daugherty
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Sushma Nagaraj
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Naomi Sengamalay
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Sandra Ott
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Al Godinez
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Luke J Tallon
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Lisa Sadzewicz
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Claire Fraser
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
- Department of Medicine, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Julie C Dunning Hotopp
- Institute for Genome Sciences, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
- Greenebaum Cancer Center, University of Maryland School of Medicine, 801 W. Baltimore St, Baltimore, MD, 21201, USA
| | - Yasuko Rikihisa
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA.
| |
Collapse
|
7
|
Kader M, El Andaloussi A, Vorhaour J, Tamama K, Nieto N, Scott MJ, Ismail N. Interferon Type I Regulates Inflammasome Activation and High Mobility Group Box 1 Translocation in Hepatocytes During Ehrlichia-Induced Acute Liver Injury. Hepatol Commun 2021; 5:33-51. [PMID: 33437899 PMCID: PMC7789844 DOI: 10.1002/hep4.1608] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 08/13/2020] [Accepted: 08/27/2020] [Indexed: 12/12/2022] Open
Abstract
Inflammasomes are an important innate immune host defense against intracellular microbial infection. Activation of inflammasomes by microbial or host ligands results in cleavage of caspase-1 (canonical pathway) or caspase-11 (noncanonical pathway), release of interleukin (IL)-1β, IL-18, high mobility group box 1 (HMGB1), and inflammatory cell death known as pyroptosis. Ehrlichia are obligate, intracellular, gram-negative bacteria that lack lipopolysaccharide but cause potentially life-threatening monocytic ehrlichiosis in humans and mice that is characterized by liver injury followed by sepsis and multiorgan failure. Employing murine models of mild and fatal ehrlichiosis caused by infection with mildly and highly virulent Ehrlichia muris (EM) and Ixodes ovatus Ehrlichia (IOE), respectively, we have previously shown that IOE infection triggers type I interferon (IFN-I) response and deleterious caspase-11 activation in liver tissues, which promotes liver injury and sepsis. In this study, we examined the contribution of IFN-I signaling in hepatocytes (HCs) to Ehrlichia-induced liver injury. Compared to EM infection, we found that IOE enter and replicate in vitro cultured primary murine HCs and induce secretion of IFNβ and several chemokines, including regulated upon activation, normal T-cell expressed, and secreted (RANTES), monocyte chemoattractant protein 1 (MCP1), monokine induced by gamma (MIG)/chemokine (C-X-C motif) ligand 9 (CXCL9), macrophage inflammatory protein 1 alpha (MIP1α), keratinocyte-derived chemokine (KC), and granulocyte-macrophage colony-stimulating factor (GM-CSF). Notably, in vitro stimulation of uninfected and Ehrlichia-infected HCs with recombinant IFNβ triggered activation of caspase-1/11, cytosolic translocation of HMGB1, and enhanced autophagy and intracellular bacterial replication. Secretion of HMGB1 by IOE-infected HCs was dependent on caspase-11. Primary HCs from IOE- but not EM-infected mice also expressed active caspase-1/11. Conclusion: HC-specific IFN-I signaling may exacerbate liver pathology during infection with obligate intracellular Ehrlichia by promoting bacterial replication and detrimental caspase-11-mediated inflammasome activation.
Collapse
Affiliation(s)
- Muhamuda Kader
- Department of PathologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
| | | | - Jennie Vorhaour
- Department of PathologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
| | - Kenichi Tamama
- Department of PathologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
| | - Natalia Nieto
- Department of PathologyCollege of MedicineUniversity of Illinois at ChicagoChicagoILUSA
| | - Melanie J Scott
- Department of SurgerySchool of MedicineUniversity of PittsburghPittsburghPAUSA
| | - Nahed Ismail
- Department of PathologyCollege of MedicineUniversity of Illinois at ChicagoChicagoILUSA
| |
Collapse
|
8
|
Shi C, Han W, Zhang M, Zang R, Du K, Li L, Xu X, Li C, Wang S, Qiu P, Guan H, Yang J, Xiao S, Wang X. Sulfated polymannuroguluronate TGC161 ameliorates leukopenia by inhibiting CD4 + T cell apoptosis. Carbohydr Polym 2020; 247:116728. [PMID: 32829850 PMCID: PMC7336955 DOI: 10.1016/j.carbpol.2020.116728] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/23/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023]
Abstract
Polysaccharides have aroused considerable interest due to their diverse biological activities and low toxicity. In this study, we evaluated the effect of marine polysaccharide sulfated polymannuroguluronate (TGC161) on the leukopenia induced by chemotherapy. It is found that TGC161 ameliorates the leukopenia. Besides, TGC161 would promote CD4+ T cell differentiation and maturation in the thymus, but does not have a significant effect on precursor cells in bone marrow. Furthermore, TGC161 inhibits CD4+ T cell apoptosis in vitro. Collectively, our findings offer a natural and harmless polysaccharide to ameliorate leukopenia.
Collapse
Affiliation(s)
- Chuanqin Shi
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China
| | - Wenwei Han
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China
| | - Meifang Zhang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China
| | - Ruochen Zang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China
| | - Kaixin Du
- Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Li Li
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Ximing Xu
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Chunxia Li
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Shixin Wang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Peiju Qiu
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Huashi Guan
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Jinbo Yang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Shuai Xiao
- Department of Gastrointestinal Surgery and Institute of Clinical Medicine, the First Affiliated Hospital, University of South China, Hengyang, 421001, China.
| | - Xin Wang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China.
| |
Collapse
|
9
|
Bekebrede H, Lin M, Teymournejad O, Rikihisa Y. Discovery of in vivo Virulence Genes of Obligatory Intracellular Bacteria by Random Mutagenesis. Front Cell Infect Microbiol 2020; 10:2. [PMID: 32117791 PMCID: PMC7010607 DOI: 10.3389/fcimb.2020.00002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/06/2020] [Indexed: 11/13/2022] Open
Abstract
Ehrlichia spp. are emerging tick-borne obligatory intracellular bacteria that cause febrile and sometimes fatal diseases with abnormal blood cell counts and signs of hepatitis. Ehrlichia HF strain provides an excellent mouse disease model of fatal human ehrlichiosis. We recently obtained and established stable culture of Ehrlichia HF strain in DH82 canine macrophage cell line, and obtained its whole genome sequence and annotation. To identify genes required for in vivo virulence of Ehrlichia, we constructed random insertional HF strain mutants by using Himar1 transposon-based mutagenesis procedure. Of total 158 insertional mutants isolated via antibiotic selection in DH82 cells, 74 insertions were in the coding regions of 55 distinct protein-coding genes, including TRP120 and multi-copy genes, such as p28/omp-1, virB2, and virB6. Among 84 insertions mapped within the non-coding regions, seven are located in the putative promoter region since they were within 50 bp upstream of the seven distinct genes. Using limited dilution methods, nine stable clonal mutants that had no apparent defect for multiplication in DH82 cells, were obtained. Mouse virulence of seven mutant clones was similar to that of wild-type HF strain, whereas two mutant clones showed significantly retarded growth in blood, livers, and spleens, and the mice inoculated with them lived longer than mice inoculated with wild-type. The two clones contained mutations in genes encoding a conserved hypothetical protein and a staphylococcal superantigen-like domain protein, respectively, and both genes are conserved among Ehrlichia spp., but lack homology to other bacterial genes. Inflammatory cytokine mRNA levels in the liver of mice infected with the two mutants were significantly diminished than those infected with HF strain wild-type, except IL-1β and IL-12 p40 in one clone. Thus, we identified two Ehrlichia virulence genes responsible for in vivo infection, but not for infection and growth in macrophages.
Collapse
Affiliation(s)
| | | | | | - Yasuko Rikihisa
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
10
|
Böning MAL, Trittel S, Riese P, van Ham M, Heyner M, Voss M, Parzmair GP, Klawonn F, Jeron A, Guzman CA, Jänsch L, Schraven B, Reinhold A, Bruder D. ADAP Promotes Degranulation and Migration of NK Cells Primed During in vivo Listeria monocytogenes Infection in Mice. Front Immunol 2020; 10:3144. [PMID: 32038647 PMCID: PMC6987423 DOI: 10.3389/fimmu.2019.03144] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 12/27/2019] [Indexed: 12/18/2022] Open
Abstract
The adhesion and degranulation-promoting adaptor protein (ADAP) serves as a multifunctional scaffold and is involved in the formation of immune signaling complexes. To date only limited and moreover conflicting data exist regarding the role of ADAP in NK cells. To extend existing knowledge we investigated ADAP-dependency of NK cells in the context of in vivo infection with the intracellular pathogen Listeria monocytogenes (Lm). Ex vivo analysis of infection-primed NK cells revealed impaired cytotoxic capacity in NK cells lacking ADAP as indicated by reduced CD107a surface expression and inefficient perforin production. However, ADAP-deficiency had no global effect on NK cell morphology or intracellular distribution of CD107a-containing vesicles. Proteomic definition of ADAPko and wild type NK cells did not uncover obvious differences in protein composition during the steady state and moreover, similar early response patterns were induced in NK cells upon infection independent of the genotype. In line with protein network analyses that suggested an altered migration phenotype in naïve ADAPko NK cells, in vitro migration assays uncovered significantly reduced migration of both naïve as well as infection-primed ADAPko NK cells compared to wild type NK cells. Notably, this migration defect was associated with a significantly reduced expression of the integrin CD11a on the surface of splenic ADAP-deficient NK cells 1 day post-Lm infection. We propose that ADAP-dependent alterations in integrin expression might account at least in part for the fact that during in vivo infection significantly lower numbers of ADAPko NK cells accumulate in the spleen i.e., the site of infection. In conclusion, we show here that during systemic Lm infection in mice ADAP is essential for efficient cytotoxic capacity and migration of NK cells.
Collapse
Affiliation(s)
- Martha A L Böning
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stephanie Trittel
- Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Peggy Riese
- Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Marco van Ham
- Cellular Proteome Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Maxi Heyner
- Cellular Proteome Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Martin Voss
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Gerald P Parzmair
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Frank Klawonn
- Cellular Proteome Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Andreas Jeron
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Carlos A Guzman
- Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lothar Jänsch
- Cellular Proteome Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Annegret Reinhold
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
11
|
l-Serine Lowers the Inflammatory Responses during Pasteurella multocida Infection. Infect Immun 2019; 87:IAI.00677-19. [PMID: 31570555 PMCID: PMC6867830 DOI: 10.1128/iai.00677-19] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 09/23/2019] [Indexed: 12/15/2022] Open
Abstract
Pasteurella multocida causes a variety of infectious diseases in various species of mammals and birds, resulting in enormous economic loss to the modern livestock and poultry industry. However, the mechanism of host-pathogen interaction is unclear. Here, we found that l-serine levels were significantly decreased in murine lungs infected with P. multocida. Pasteurella multocida causes a variety of infectious diseases in various species of mammals and birds, resulting in enormous economic loss to the modern livestock and poultry industry. However, the mechanism of host-pathogen interaction is unclear. Here, we found that l-serine levels were significantly decreased in murine lungs infected with P. multocida. Exogenous l-serine supplementation significantly increased the survival rate of mice and decreased the colonization of P. multocida in the lungs of mice. Notably, l-serine decreased the macrophage- and neutrophil-mediated inflammatory responses in mice during P. multocida infection.
Collapse
|
12
|
Haloul M, Oliveira ERA, Kader M, Wells JZ, Tominello TR, El Andaloussi A, Yates CC, Ismail N. mTORC1-mediated polarization of M1 macrophages and their accumulation in the liver correlate with immunopathology in fatal ehrlichiosis. Sci Rep 2019; 9:14050. [PMID: 31575880 PMCID: PMC6773708 DOI: 10.1038/s41598-019-50320-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/02/2019] [Indexed: 12/13/2022] Open
Abstract
A polarized macrophage response into inflammatory (M1) or regenerative/anti-inflammatory (M2) phenotypes is critical in host response to multiple intracellular bacterial infections. Ehrlichia is an obligate Gram-negative intracellular bacterium that causes human monocytic ehrlichiosis (HME): a febrile illness that may progress to fatal sepsis with multi-organ failure. We have shown that liver injury and Ehrlichia-induced sepsis occur due to dysregulated inflammation. Here, we investigated the contribution of macrophages to Ehrlichia-induced sepsis using murine models of mild and fatal ehrlichiosis. Lethally-infected mice showed accumulation of M1 macrophages (iNOS-positive) in the liver. In contrast, non-lethally infected mice showed polarization of M2 macrophages and their accumulation in peritoneum, but not in the liver. Predominance of M1 macrophages in lethally-infected mice was associated with expansion of IL-17-producing T, NK, and NKT cells. Consistent with the in vivo data, infection of bone marrow-derived macrophages (BMM) with lethal Ehrlichia polarized M0 macrophages into M1 phenotype under an mTORC1-dependent manner, while infection with non-lethal Ehrlichia polarized these cells into M2 types. This work highlights that mTORC1-mediated polarization of macrophages towards M1 phenotype may contribute to induction of pathogenic immune responses during fatal ehrlichiosis. Targeting mTORC1 pathway may provide a novel aproach for treatment of HME.
Collapse
Affiliation(s)
- Mohamed Haloul
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Children's Cancer Hospital Egypt, 57357, Cairo, Egypt
| | - Edson R A Oliveira
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Muhamuda Kader
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jakob Z Wells
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tyler R Tominello
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Abdeljabar El Andaloussi
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Cecelia C Yates
- Nursing School, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nahed Ismail
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
13
|
Tominello TR, Oliveira ERA, Hussain SS, Elfert A, Wells J, Golden B, Ismail N. Emerging Roles of Autophagy and Inflammasome in Ehrlichiosis. Front Immunol 2019; 10:1011. [PMID: 31134081 PMCID: PMC6517498 DOI: 10.3389/fimmu.2019.01011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/23/2019] [Indexed: 12/16/2022] Open
Abstract
Human monocytic ehrlichiosis (HME) is a potentially life-threatening tick-borne rickettsial disease (TBRD) caused by the obligate intracellular Gram-negative bacteria, Ehrlichia. Fatal HME presents with acute ailments of sepsis and toxic shock-like symptoms that can evolve to multi-organ failure and death. Early clinical and laboratory diagnosis of HME are problematic due to non-specific flu-like symptoms and limitations in the current diagnostic testing. Several studies in murine models showed that cell-mediated immunity acts as a “double-edged sword” in fatal ehrlichiosis. Protective components are mainly formed by CD4 Th1 and NKT cells, in contrast to deleterious effects originated from neutrophils and TNF-α-producing CD8 T cells. Recent research has highlighted the central role of the inflammasome and autophagy as part of innate immune responses also leading to protective or pathogenic scenarios. Recognition of pathogen-associated molecular patterns (PAMPS) or damage-associated molecular patterns (DAMPS) triggers the assembly of the inflammasome complex that leads to multiple outcomes. Recognition of PAMPs or DAMPs by such complexes can result in activation of caspase-1 and -11, secretion of the pro-inflammatory cytokines IL-1β and IL-18 culminating into dysregulated inflammation, and inflammatory cell death known as pyroptosis. The precise functions of inflammasomes and autophagy remain unexplored in infections with obligate intracellular rickettsial pathogens, such as Ehrlichia. In this review, we discuss the intracellular innate immune surveillance in ehrlichiosis involving the regulation of inflammasome and autophagy, and how this response influences the innate and adaptive immune responses against Ehrlichia. Understanding such mechanisms would pave the way in research for novel diagnostic, preventative and therapeutic approaches against Ehrlichia and other rickettsial diseases.
Collapse
Affiliation(s)
- Tyler R Tominello
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Edson R A Oliveira
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Shah S Hussain
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Amr Elfert
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Jakob Wells
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brandon Golden
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Nahed Ismail
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
14
|
Uyangaa E, Choi JY, Patil AM, Hossain FMA, Park SO, Kim B, Kim K, Eo SK. Dual TLR2/9 Recognition of Herpes Simplex Virus Infection Is Required for Recruitment and Activation of Monocytes and NK Cells and Restriction of Viral Dissemination to the Central Nervous System. Front Immunol 2018; 9:905. [PMID: 29760708 PMCID: PMC5936768 DOI: 10.3389/fimmu.2018.00905] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 04/11/2018] [Indexed: 12/24/2022] Open
Abstract
The importance of TLR2 and TLR9 in the recognition of infection with herpes simplex virus (HSV) and HSV-caused diseases has been described, but some discrepancies remain concerning the benefits of these responses. Moreover, the impact of TLR2/9 on innate and adaptive immune responses within relevant mucosal tissues has not been elucidated using natural mucosal infection model of HSV. Here, we demonstrate that dual TLR2/9 recognition is essential to provide resistance against mucosal infection with HSV via an intravaginal route. Dual TLR2/9 ablation resulted in the highly enhanced mortality with exacerbated symptoms of encephalitis compared with TLR2 or TLR9 deficiency alone, coinciding with highly increased viral load in central nervous system tissues. TLR2 appeared to play a minor role in providing resistance against mucosal infection with HSV, since TLR2-ablated mice showed higher survival rate compared with TLR9-ablated mice. Also, the high mortality in dual TLR2/9-ablated mice was closely associated with the reduction in early monocyte and NK cell infiltration in the vaginal tract (VT), which was likely to correlate with low expression of cytokines and CCR2 ligands (CCL2 and CCL7). More interestingly, our data revealed that dual TLR2/9 recognition of HSV infection plays an important role in the functional maturation of TNF-α and iNOS-producing dendritic cells (Tip-DCs) from monocytes as well as NK cell activation in VT. TLR2/9-dependent maturation of Tip-DCs from monocytes appeared to specifically present cognate Ag, which effectively provided functional effector CD4+ and CD8+ T cells specific for HSV Ag in VT and its draining lymph nodes. TLR2/9 expressed in monocytes was likely to directly facilitate Tip-DC-like features after HSV infection. Also, dual TLR2/9 recognition of HSV infection directly activated NK cells without the aid of dendritic cells through activation of p38 MAPK pathway. Taken together, these results indicate that dual TLR2/9 recognition plays a critical role in providing resistance against mucosal infection with HSV, which may involve a direct regulation of Tip-DCs and NK cells in VT. Therefore, our data provide a more detailed understanding of TLR2/9 role in conferring antiviral immunity within relevant mucosal tissues after mucosal infection with HSV.
Collapse
Affiliation(s)
- Erdenebileg Uyangaa
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, South Korea
| | - Jin Young Choi
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, South Korea
| | - Ajit Mahadev Patil
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, South Korea
| | - Ferdaus Mohd Altaf Hossain
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, South Korea.,Faculty of Veterinary, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Sung Ok Park
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, South Korea
| | - Bumseok Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, South Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, South Korea
| | - Seong Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, South Korea
| |
Collapse
|
15
|
Cangussu ASR, Mariúba LAM, Lalwani P, Pereira KDES, Astolphi-Filho S, Orlandi PP, Epiphanio S, Viana KF, Ribeiro MFB, Silva HM, Marinho CRF, Nogueira PA. A hybrid protein containing MSP1a repeats and Omp7, Omp8 and Omp9 epitopes protect immunized BALB/c mice against anaplasmosis. Vet Res 2018; 49:6. [PMID: 29351812 PMCID: PMC5775529 DOI: 10.1186/s13567-018-0503-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 12/20/2017] [Indexed: 12/23/2022] Open
Abstract
Anaplasma marginale (A. marginale) has a remarkable impact on livestock production, and an effective vaccine is not currently available due to the inexistence of a small animal model. Recently, BALB/c mice were successfully infected with A. marginale, resulting in an acute and persistent anaplasmosis infection. Here, we designed a hybrid protein containing repeats of polypeptide 1a from major surface protein-1 complex (MSP1a) repeats and common epitopes of outer membrane proteins (OMPs) OMP7, OMP8 and OMP9 expressed in Escherichia coli. Our proof-of-concept assessed vaccinal effectiveness against a challenge with live bacteria. The MSP1a/OMP7/8/9 immunized BALB/C mice exhibited a strong reduction in rickettsemia and had no signs of anaplasmosis or hepatic lesions. In contrast, the non-immunized mice exhibited signs of anaplasmosis and a body weight loss associated with increases in monocyte and neutrophil counts. Furthermore, the non-immunized mice displayed atrophies with chronic inflammatory infiltrates in the spleen and increased binucleation and hydropic degeneration in the hepatocytes. Our findings demonstrated that immunization with our hybrid protein induced a strong reduction in rickettsemia and conferred protection against anaplasmosis. Therefore, given the strong evidence of the protective effect against anaplasmosis, hybrid protein designs are potential candidates for the rational design of vaccinal subunits.
Collapse
Affiliation(s)
- Alex Sander R Cangussu
- Universidade Federal do Amazonas-Programa de pós-graduação em Rede de Biodiversidade e Biotecnologia da Amazônia Legal, Manaus, AM/Brasil, Brazil. .,Universidade Federal do Tocantins-Engenharia de Bioprocessos e Biotecnologia, Gurupi, TO/Brazil, Brazil.
| | - Luis André M Mariúba
- Instituto Leônidas e Maria Deane-Fiocruz Amazônia, Manaus, Amazonas/Brazil, Brazil
| | - Pritesh Lalwani
- Instituto Leônidas e Maria Deane-Fiocruz Amazônia, Manaus, Amazonas/Brazil, Brazil
| | | | - Spartaco Astolphi-Filho
- Universidade Federal do Amazonas-Programa de pós-graduação em Rede de Biodiversidade e Biotecnologia da Amazônia Legal, Manaus, AM/Brasil, Brazil
| | - Patricia P Orlandi
- Instituto Leônidas e Maria Deane-Fiocruz Amazônia, Manaus, Amazonas/Brazil, Brazil
| | - Sabrina Epiphanio
- Departamento de Análises Clínicas e Toxicológicas da Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, São Paulo/Brazil, Brazil
| | - Kelvison F Viana
- Universidade Federal do Tocantins-Engenharia de Bioprocessos e Biotecnologia, Gurupi, TO/Brazil, Brazil
| | - Mucio Flavio B Ribeiro
- Departamento de Parasitologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais/Brazil, Brazil
| | - Hidelberto M Silva
- Faculdade de Medicina, Centro Universitário Unirg, Gurupi, Tocantins/Brazil, Brazil
| | - Claudio R F Marinho
- Departamento de Parasitologia do Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Paulo A Nogueira
- Instituto Leônidas e Maria Deane-Fiocruz Amazônia, Manaus, Amazonas/Brazil, Brazil
| |
Collapse
|
16
|
Nefefe T, Liebenberg J, van Kleef M, Steyn HC, Pretorius A. Innate immune transcriptomic evaluation of PBMC isolated from sheep after infection with E. ruminantium Welgevonden strain. Mol Immunol 2017; 91:238-248. [PMID: 28988038 DOI: 10.1016/j.molimm.2017.09.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 08/18/2017] [Accepted: 09/27/2017] [Indexed: 12/30/2022]
Abstract
Heartwater is a tick-borne non-infectious fatal disease of wild and domestic ruminants caused by the bacterium Ehrlichia ruminantium, transmitted by Amblyomma ticks. Although there is evidence that interferon-gamma (IFN-γ) controls E. ruminantium growth and that cellular immune responses could be protective, an effective recombinant vaccine for this disease is lacking. An overall analysis of which immune pathways are up- or down-regulated in sheep peripheral blood mononuclear cells is expected to lead to a better understanding of the global immune response of sheep to E. ruminantium infection. Therefore, a systems biology oriented approach following the infection with E. ruminantium was investigated from peripheral blood mononuclear cells to aid recombinant vaccine development. In this study, heartwater naïve sheep were infected and challenged by allowing E. ruminantium infected ticks to feed on them. After primary infection, all the animals were treated with antibiotic during the resulting febrile response. Blood was collected daily for E. ruminantium detection by qPCR (pCS20 assay). The pCS20 assay only detected the pathogen in the blood one day prior to and during the febrile stage of infection confirming infection of the sheep. IFN-γ real-time PCR indicated that this cytokine was expressed at specific time points: post infection, during the febrile stage of the disease and after challenge. These were used as a guide to select samples for transcriptome sequencing. This paper focuses on transcripts that are associated with innate activating pathways that were identified to be up- and down-regulated after primary infection and the subsequent challenge. These included the CD14 monocyte marker, toll-like receptor (TLR), nod-like receptor, chemokine, cytosolic and cytokine-cytokine interaction receptor pathways. In particular, TLR4, TLR9 and CD14 were activated together with DNA detection pathways, suggesting that vaccine formulations may be improved if CpG motifs and lipopolysaccharides are included. This data indicates that innate immune activation, perhaps by using adjuvants, should be an important component for consideration during future heartwater recombinant vaccine development.
Collapse
Affiliation(s)
- T Nefefe
- Agricultural Research Council - Onderstepoort Veterinary Research, Private Bag X5, Onderstepoort, 0110, South Africa; Department of Veterinary Tropical Diseases, University of Pretoria, Private Bag X04, Onderstepoort, 0110, South Africa.
| | - J Liebenberg
- Agricultural Research Council - Onderstepoort Veterinary Research, Private Bag X5, Onderstepoort, 0110, South Africa
| | - M van Kleef
- Agricultural Research Council - Onderstepoort Veterinary Research, Private Bag X5, Onderstepoort, 0110, South Africa; Department of Veterinary Tropical Diseases, University of Pretoria, Private Bag X04, Onderstepoort, 0110, South Africa
| | - H C Steyn
- Agricultural Research Council - Onderstepoort Veterinary Research, Private Bag X5, Onderstepoort, 0110, South Africa
| | - A Pretorius
- Agricultural Research Council - Onderstepoort Veterinary Research, Private Bag X5, Onderstepoort, 0110, South Africa; Department of Veterinary Tropical Diseases, University of Pretoria, Private Bag X04, Onderstepoort, 0110, South Africa
| |
Collapse
|
17
|
Kader M, Alaoui-EL-Azher M, Vorhauer J, Kode BB, Wells JZ, Stolz D, Michalopoulos G, Wells A, Scott M, Ismail N. MyD88-dependent inflammasome activation and autophagy inhibition contributes to Ehrlichia-induced liver injury and toxic shock. PLoS Pathog 2017; 13:e1006644. [PMID: 29049365 PMCID: PMC5663626 DOI: 10.1371/journal.ppat.1006644] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 10/31/2017] [Accepted: 09/11/2017] [Indexed: 01/19/2023] Open
Abstract
Severe hepatic inflammation is a common cause of acute liver injury following systemic infection with Ehrlichia, obligate Gram-negative intracellular bacteria that lack lipopolysaccharide (LPS). We have previously shown that type I IFN (IFN-I) and inflammasome activation are key host-pathogenic mediators that promote excessive inflammation and liver damage following fatal Ehrlichia infection. However, the underlying signals and mechanisms that regulate protective immunity and immunopathology during Ehrlichia infection are not well understood. To address this issue, we compared susceptibility to lethal Ixodes ovatus Ehrlichia (IOE) infection between wild type (WT) and MyD88-deficient (MyD88-/-) mice. We show here that MyD88-/- mice exhibited decreased inflammasome activation, attenuated liver injury, and were more resistant to lethal infection than WT mice, despite suppressed protective immunity and increased bacterial burden in the liver. MyD88-dependent inflammasome activation was also dependent on activation of the metabolic checkpoint kinase mammalian target of rapamycin complex 1 (mTORC1), inhibition of autophagic flux, and defective mitophagy in macrophages. Blocking mTORC1 signaling in infected WT mice and primary macrophages enhanced bacterial replication and attenuated inflammasome activation, suggesting autophagy promotes bacterial replication while inhibiting inflammasome activation. Finally, our data suggest TLR9 and IFN-I are upstream signaling mechanisms triggering MyD88-mediated mTORC1 and inflammasome activation in macrophages following Ehrlichia infection. This study reveals that Ehrlichia-induced liver injury and toxic shock are mediated by MyD88-dependent inflammasome activation and autophagy inhibition.
Collapse
MESH Headings
- Animals
- Autophagy/immunology
- Blotting, Western
- Disease Models, Animal
- Ehrlichia/immunology
- Ehrlichiosis/immunology
- Ehrlichiosis/metabolism
- Enzyme-Linked Immunosorbent Assay
- Female
- Flow Cytometry
- Fluorescent Antibody Technique
- Image Processing, Computer-Assisted
- In Situ Nick-End Labeling
- Inflammasomes/immunology
- Inflammasomes/metabolism
- Liver Failure, Acute/immunology
- Liver Failure, Acute/metabolism
- Liver Failure, Acute/microbiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Confocal
- Microscopy, Electron, Transmission
- Myeloid Differentiation Factor 88/immunology
- Myeloid Differentiation Factor 88/metabolism
- Real-Time Polymerase Chain Reaction
- Shock, Septic/immunology
- Shock, Septic/metabolism
Collapse
Affiliation(s)
- Muhamuda Kader
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mounia Alaoui-EL-Azher
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jennie Vorhauer
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Bhushan B Kode
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jakob Z. Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Donna Stolz
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, United States of America
| | - George Michalopoulos
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Melanie Scott
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Nahed Ismail
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
18
|
Weis S, Rubio I, Ludwig K, Weigel C, Jentho E. Hormesis and Defense of Infectious Disease. Int J Mol Sci 2017; 18:E1273. [PMID: 28617331 PMCID: PMC5486095 DOI: 10.3390/ijms18061273] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/16/2017] [Accepted: 05/20/2017] [Indexed: 12/22/2022] Open
Abstract
Infectious diseases are a global health burden and remain associated with high social and economic impact. Treatment of affected patients largely relies on antimicrobial agents that act by directly targeting microbial replication. Despite the utility of host specific therapies having been assessed in previous clinical trials, such as targeting the immune response via modulating the cytokine release in sepsis, results have largely been frustrating and did not lead to the introduction of new therapeutic tools. In this article, we will discuss current evidence arguing that, by applying the concept of hormesis, already approved pharmacological agents could be used therapeutically to increase survival of patients with infectious disease via improving disease tolerance, a defense mechanism that decreases the extent of infection-associated tissue damage without directly targeting pathogenic microorganisms.
Collapse
Affiliation(s)
- Sebastian Weis
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Jena, Jena 07747, Germany.
- Center for Infectious Diseases and Infection Control, University Hospital Jena, Jena 07747, Germany.
- Center for Sepsis Control and Care, University Hospital Jena, Jena 07747, Germany.
| | - Ignacio Rubio
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, Jena 07745, Germany.
| | - Kristin Ludwig
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, Jena 07745, Germany.
| | - Cynthia Weigel
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Jena, Jena 07747, Germany.
- Fritz Lipmann Institute, Leibniz Institute on Aging, Jena 07745, Germany.
| | - Elisa Jentho
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Jena, Jena 07747, Germany.
| |
Collapse
|
19
|
Puerta-Arias JD, Pino-Tamayo PA, Arango JC, González Á. Depletion of Neutrophils Promotes the Resolution of Pulmonary Inflammation and Fibrosis in Mice Infected with Paracoccidioides brasiliensis. PLoS One 2016; 11:e0163985. [PMID: 27690127 PMCID: PMC5045199 DOI: 10.1371/journal.pone.0163985] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 09/16/2016] [Indexed: 12/11/2022] Open
Abstract
Chronic stages of paracoccidioidomycosis (PCM) are characterized by granulomatous lesions which promote the development of pulmonary fibrosis leading to the loss of respiratory function in 50% of patients; in addition, it has been observed that neutrophils predominate during these chronic stages of P. brasiliensis infection. The goal of this study was to evaluate the role of the neutrophil during the chronic stages of experimental pulmonary PCM and during the fibrosis development and tissue repair using a monoclonal specific to this phagocytic cell. Male BALB/c mice were inoculated intranasally with 1.5x106 P. brasiliensis yeast cells. A monoclonal antibody specific to neutrophils was administered at 4 weeks post-inoculation followed by doses every 48h during two weeks. Mice were sacrificed at 8 and 12 weeks post-inoculation to assess cellularity, fungal load, cytokine/chemokine levels, histopathological analysis, collagen and expression of genes related to fibrosis development. Depletion of neutrophils was associated with a significant decrease in the number of eosinophils, dendritic cells, B cells, CD4-T cells, MDSCs and Treg cells, fungal load and levels of most of the pro-inflammatory cytokines/chemokines evaluated, including IL-17, TNF-α and TGF-β1. Recovery of lung architecture was also associated with reduced levels of collagen, high expression of TGF-β3, matrix metalloproteinase (MMP)-12 and -14, and decreased expression of tissue inhibitor metalloproteinase (TIMP)-2, and MMP-8. Depletion of neutrophils might attenuate lung fibrosis and inflammation through down-regulating TGF-β1, TNF-α, IL-17, MMP-8 and TIMP-2. These results suggest that neutrophil could be considered as a therapeutic target in pulmonary fibrosis induced by P. brasiliensis.
Collapse
Affiliation(s)
- Juan David Puerta-Arias
- Medical and Experimental Mycology Unit, Corporación para Investigaciones Biológicas (CIB), Medellín, Colombia
| | - Paula Andrea Pino-Tamayo
- Medical and Experimental Mycology Unit, Corporación para Investigaciones Biológicas (CIB), Medellín, Colombia
| | - Julián Camilo Arango
- Medical and Experimental Mycology Unit, Corporación para Investigaciones Biológicas (CIB), Medellín, Colombia
- School of Microbiology, Universidad de Antioquia, Medellín, Colombia
| | - Ángel González
- School of Microbiology, Universidad de Antioquia, Medellín, Colombia
- Basic and Applied Microbiology Research Group (MICROBA), Universidad de Antioquia, Medellín, Colombia
- * E-mail:
| |
Collapse
|
20
|
Müller S, Wolf AJ, Iliev ID, Berg BL, Underhill DM, Liu GY. Poorly Cross-Linked Peptidoglycan in MRSA Due to mecA Induction Activates the Inflammasome and Exacerbates Immunopathology. Cell Host Microbe 2016; 18:604-12. [PMID: 26567511 DOI: 10.1016/j.chom.2015.10.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/23/2015] [Accepted: 10/20/2015] [Indexed: 10/22/2022]
Abstract
Methicillin-resistant S. aureus (MRSA) is a leading health problem. Compared to methicillin-sensitive S. aureus, MRSA infections are associated with greater morbidity and mortality, but the mechanisms underlying MRSA pathogenicity are unclear. Here we show that the protein conferring β-lactam antibiotic resistance, penicillin-binding protein 2A (encoded by the mecA gene), directly contributes to pathogenicity during MRSA infection. MecA induction leads to a reduction in peptidoglycan cross-linking that allows for enhanced degradation and detection by phagocytes, resulting in robust IL-1β production. Peptidoglycan isolated from β-lactam-challenged MRSA strongly induces the NLRP3 inflammasome in macrophages, but these effects are lost upon peptidoglycan solubilization. Mutant MRSA bacteria with naturally occurring reduced peptidoglycan cross-links induce high IL-1β levels in vitro and cause increased pathology in vivo. β-lactam treatment of MRSA skin infection exacerbates immunopathology, which is IL-1 dependent. Thus, antibiotic-induced expression of mecA during MRSA skin infection contributes to immunopathology by altering peptidoglycan structure.
Collapse
Affiliation(s)
- Sabrina Müller
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Andrea J Wolf
- Division of Biomedical Sciences and the F. Widjaja Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Iliyan D Iliev
- Division of Biomedical Sciences and the F. Widjaja Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Bethany L Berg
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - David M Underhill
- Division of Biomedical Sciences and the F. Widjaja Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | - George Y Liu
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
21
|
Popova TG, Teunis A, Espina V, Liotta LA, Popov SG. Chemokine-Releasing Microparticles Improve Bacterial Clearance and Survival of Anthrax Spore-Challenged Mice. PLoS One 2016; 11:e0163163. [PMID: 27632537 PMCID: PMC5025034 DOI: 10.1371/journal.pone.0163163] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 09/02/2016] [Indexed: 11/18/2022] Open
Abstract
In this study the hydrogel microparticles (MPs) were used to enhance migration of neutrophils in order to improve outcome of anthrax infection in a mouse model. Two MP formulations were tested. In the first one the polyacrylamide gel MPs were chemically coupled with Cibacron Blue (CB) affinity bait. In the second one the bait molecules within the MPs were additionally loaded with neutrophil-attracting chemokines (CKs), human CXCL8 and mouse CCL3. A non-covalent interaction of the bait with the CKs provided their gradual release after administration of the MPs to the host. Mice were challenged into footpads with Bacillus anthracis Sterne spores and given a dose of MPs a few hours before and/or after the spores. Pre-treatment with a single dose of CK-releasing MPs without any additional intervention was able to induce influx of neutrophils to the site of spore inoculation and regional lymph nodes correlating with reduced bacterial burden and decreased inflammatory response in footpads. On average, in two independent experiments, up to 53% of mice survived over 13 days. All control spore-challenged but MP-untreated mice died. The CB-coupled particles were also found to improve survival likely due to the capacity to stimulate release of endogenous CKs, but were less potent at decreasing the inflammatory host response than the CK-releasing MPs. The CK post-treatment did not improve survival compared to the untreated mice which died within 4 to 6 days with a strong inflammation of footpads, indicating quick dissemination of spores though the lymphatics after challenge. This is the first report on the enhanced innate host resistance to anthrax in response to CKs delivered and/or endogenously induced by the MPs.
Collapse
Affiliation(s)
- Taissia G. Popova
- Center for Applied Proteomics and Molecular Medicine, Department of Molecular Microbiology, School of Systems Biology, George Mason University, Manassas, VA, 20110, United States of America
| | - Allison Teunis
- National Center for Biodefense and Infectious Diseases, Department of Molecular Microbiology, School of Systems Biology, George Mason University, Manassas, VA, 20110, United States of America
| | - Virginia Espina
- Center for Applied Proteomics and Molecular Medicine, Department of Molecular Microbiology, School of Systems Biology, George Mason University, Manassas, VA, 20110, United States of America
| | - Lance A. Liotta
- Center for Applied Proteomics and Molecular Medicine, Department of Molecular Microbiology, School of Systems Biology, George Mason University, Manassas, VA, 20110, United States of America
| | - Serguei G. Popov
- National Center for Biodefense and Infectious Diseases, Department of Molecular Microbiology, School of Systems Biology, George Mason University, Manassas, VA, 20110, United States of America
- * E-mail:
| |
Collapse
|
22
|
Ligocki AJ, Niederkorn JY. Natural Killer T Cells Contribute to Neutrophil Recruitment and Ocular Tissue Damage in a Model of Intraocular Tumor Rejection. Invest Ophthalmol Vis Sci 2016; 57:813-23. [PMID: 26934137 PMCID: PMC4777277 DOI: 10.1167/iovs.15-18786] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE Immune privilege of the eye protects the nonregenerative ocular tissues from innate and adaptive immune-mediated inflammation. In the case of intraocular tumors, immune privilege can be arrested to allow for immune-mediated rejection. Activation of innate immune cells can contribute to necrosis of the intraocular tumor and bystander ocular tissue. Identifying the cellular components of the innate immune system that contribute to ocular destruction, but are not needed for tumor rejection, provides insights into the immunopathological sequelae in intraocular tumor rejection. METHODS Wild-type (WT), Jα18 knockout (KO) mice lacking type I natural killer T (NKT) cells, and CD1d KO mice lacking all NKT cells, were used to identify the role of type II NKT cells in intraocular tumor rejection immunopathology. RESULTS CD1d KO mice had significantly lowered rates of necrotic eye destruction during tumor rejection compared to WT or Jα18 KO mice. Transcriptome and protein analyses revealed that CD1d KO mice had significantly lower expression of CXCL3 compared to WT or Jα18 KO mice, and this was associated with decreased neutrophil recruitment. The presence of type II NKT cells in WT or Jα18 KO mice led to increased CXCL3, which attracted neutrophils to the intraocular tumor and culminated in destruction of the eye. CONCLUSIONS We found that type II NKT cells are critical in initiating a damaging inflammatory antitumor response involving the recruitment of neutrophils that compromises the integrity of the eye. Loss of type II NKT cells or depleting neutrophils allows for a productive intraocular tumor response that converts the rejection phenotype to preserve the eye.
Collapse
|
23
|
Attenuated viral hepatitis in Trem1-/- mice is associated with reduced inflammatory activity of neutrophils. Sci Rep 2016; 6:28556. [PMID: 27328755 PMCID: PMC4916511 DOI: 10.1038/srep28556] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/20/2016] [Indexed: 12/22/2022] Open
Abstract
TREM1 (Triggering Receptor Expressed on Myeloid Cells 1) is a pro-inflammatory receptor expressed by phagocytes, which can also be released as a soluble molecule (sTREM1). The roles of TREM1 and sTREM1 in liver infection and inflammation are not clear. Here we show that patients with hepatitis B virus (HBV) or hepatitis C virus (HCV) infection manifest elevated serum levels of sTREM1. In mice, experimental viral hepatitis induced by infection with Lymphocytic Choriomeningitis Virus (LCMV)-WE was likewise associated with increased sTREM1 in serum and urine, and with increased TREM1 and its associated adapter molecule DAP12 in the liver. Trem1−/− mice showed accelerated clearance of LCMV-WE and manifested attenuated liver inflammation and injury. TREM1 expression in the liver of wild-type mice was mostly confined to infiltrating neutrophils, which responded to LCMV by secretion of CCL2 and TNF-α, and release of sTREM1. Accordingly, the production of CCL2 and TNF-α was decreased in the livers of LCMV-infected Trem1−/− mice, as compared to LCMV-infected wildtype mice. These findings indicate that TREM1 plays a role in viral hepatitis, in which it seems to aggravate the immunopathology associated with viral clearance, mainly by increasing the inflammatory activity of neutrophils.
Collapse
|
24
|
Endoplasmic reticulum stress in bone marrow-derived cells prevents acute cardiac inflammation and injury in response to angiotensin II. Cell Death Dis 2016; 7:e2258. [PMID: 27277680 PMCID: PMC5143392 DOI: 10.1038/cddis.2016.164] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/05/2016] [Accepted: 05/06/2016] [Indexed: 12/16/2022]
Abstract
Inflammation plays an important role in hypertensive cardiac injury. The endoplasmic reticulum (ER) stress pathway is involved in the inflammatory response. However, the role of ER stress in elevated angiotensin II (Ang II)-induced cardiac injury remains unclear. In this study, we investigated the role of ER stress in Ang II-induced hypertensive cardiac injury. Transcriptome analysis and quantitative real-time PCR showed that Ang II infusion in mice increased ER stress-related genes expression in the heart. C/EBP homologous protein (CHOP) deficiency, a key mediator of ER stress, increased infiltration of inflammatory cells, especially neutrophils, the production of inflammatory cytokines, chemokines in Ang II-infused mouse hearts. CHOP deficiency increased Ang II-induced cardiac fibrotic injury: (1) Masson trichrome staining showed increased fibrotic areas, (2) immunohistochemistry staining showed increased expression of α-smooth muscle actin, transforming growth factor β1 and (3) quantitative real-time PCR showed increased expression of collagen in CHOP-deficient mouse heart. Bone marrow transplantation experiments indicated that CHOP deficiency in bone marrow cells was responsible for Ang II-induced cardiac fibrotic injury. Moreover, TUNEL staining and flow cytometry revealed that CHOP deficiency decreased neutrophil apoptosis in response to Ang II. Taken together, our study demonstrated that hypertension induced ER stress after Ang II infusion. ER stress in bone marrow-derived cells protected acute cardiac inflammation and injury in response to Ang II.
Collapse
|
25
|
Habib S, El Andaloussi A, Hisham A, Ismail N. NK Cell-Mediated Regulation of Protective Memory Responses against Intracellular Ehrlichial Pathogens. PLoS One 2016; 11:e0153223. [PMID: 27092553 PMCID: PMC4836677 DOI: 10.1371/journal.pone.0153223] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/25/2016] [Indexed: 11/18/2022] Open
Abstract
Ehrlichiae are gram-negative obligate intracellular bacteria that cause potentially fatal human monocytic ehrlichiosis. We previously showed that natural killer (NK) cells play a critical role in host defense against Ehrlichia during primary infection. However, the contribution of NK cells to the memory response against Ehrlichia remains elusive. Primary infection of C57BL/6 mice with Ehrlichia muris provides long-term protection against a second challenge with the highly virulent Ixodes ovatus Ehrlichia (IOE), which ordinarily causes fatal disease in naïve mice. Here, we show that the depletion of NK cells in E. muris-primed mice abrogates the protective memory response against IOE. Approximately, 80% of NK cell-depleted E. muris-primed mice succumbed to lethal IOE infection on days 8-10 after IOE infection, similar to naïve mice infected with the same dose of IOE. The lack of a recall response in NK cell-depleted mice correlated with an increased bacterial burden, extensive liver injury, decreased frequency of Ehrlichia-specific IFN-γ-producing memory CD4+ and CD8+ T-cells, and a low titer of Ehrlichia-specific antibodies. Intraperitoneal infection of mice with E. muris resulted in the production of IL-15, IL-12, and IFN-γ as well as an expansion of activated NKG2D+ NK cells. The adoptive transfer of purified E. muris-primed hepatic and splenic NK cells into Rag2-/-Il2rg-/- recipient mice provided protective immunity against challenge with E. muris. Together, these data suggest that E. muris-induced memory-like NK cells, which contribute to the protective, recall response against Ehrlichia.
Collapse
Affiliation(s)
- Samar Habib
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Abdeljabar El Andaloussi
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Ahmed Hisham
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Nahed Ismail
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
26
|
Wu B, Capilato J, Pham MP, Walker J, Spur B, Rodriguez A, Perez LJ, Yin K. Lipoxin A4 augments host defense in sepsis and reduces Pseudomonas aeruginosa virulence through quorum sensing inhibition. FASEB J 2016; 30:2400-10. [PMID: 26965685 DOI: 10.1096/fj.201500029r] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 02/29/2016] [Indexed: 12/13/2022]
Abstract
Bacterial infections can quickly turn into sepsis, with its attendant clinical sequelae of inflammation, tissue injury, and organ failure. Paradoxically, sustained inflammation in sepsis may lead to immune suppression, because of which the host is unable to clear the existing infection. Use of agents that suppress the inflammatory response may accelerate host immune suppression, whereas use of traditional antibiotics does not significantly affect inflammation. In this study, we investigated whether lipoxin A4 (LXA4), a specialized, proresolution lipid mediator, could increase neutrophil phagocytic activity as well as reduce bacterial virulence. Using the mouse cecal ligation and puncture (CLP) model of sepsis, the administration of LXA4 (7 μg/kg i.v.) 1 h after surgery increased neutrophil phagocytic ability and Fcγ receptor I (CD64) expression. Ex vivo studies have confirmed that the direct addition of LXA4 to CLP neutrophils increased phagocytic ability but not CD64 expression. LXA4 did not affect neutrophils taken from control mice in which CD64 expression was minimal. Taken together with in vivo data, these results suggest that LXA4 directly augments CD64-mediated neutrophil phagocytic ability but does not directly increase neutrophil CD64 expression. Bacterial communication and virulence is regulated by quorum sensing inducers. In Pseudomonas aeruginosa, virulence is induced with release of various virulence factors, by N-3-oxododecanolyl homoserine lactone binding to the quorum sensing receptor, LasR. We show that LXA4 is an inhibitor of LasR in P. aeruginosa and that it decreases the release of pyocyanin exotoxin. These results suggest that LXA4 has the novel dual properties of increasing host defense and decreasing pathogen virulence by inhibiting quorum sensing.-Wu, B., Capilato, J., Pham, M. P., Walker, J., Spur, B., Rodriguez, A., Perez, L. J., Yin, K. Lipoxin A4 augments host defense in sepsis and reduces Pseudomonas aeruginosa virulence through quorum sensing inhibition.
Collapse
Affiliation(s)
- Benedict Wu
- Department of Cell Biology, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA; and
| | - Joseph Capilato
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey, USA
| | - Michelle P Pham
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey, USA
| | - Jean Walker
- Department of Cell Biology, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA; and
| | - Bernd Spur
- Department of Cell Biology, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA; and
| | - Ana Rodriguez
- Department of Cell Biology, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA; and
| | - Lark J Perez
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey, USA
| | - Kingsley Yin
- Department of Cell Biology, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA; and
| |
Collapse
|
27
|
Uyangaa E, Kim JH, Patil AM, Choi JY, Kim SB, Eo SK. Distinct Upstream Role of Type I IFN Signaling in Hematopoietic Stem Cell-Derived and Epithelial Resident Cells for Concerted Recruitment of Ly-6Chi Monocytes and NK Cells via CCL2-CCL3 Cascade. PLoS Pathog 2015; 11:e1005256. [PMID: 26618488 PMCID: PMC4664252 DOI: 10.1371/journal.ppat.1005256] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 10/09/2015] [Indexed: 12/24/2022] Open
Abstract
Type I interferon (IFN-I)-dependent orchestrated mobilization of innate cells in inflamed tissues is believed to play a critical role in controlling replication and CNS-invasion of herpes simplex virus (HSV). However, the crucial regulators and cell populations that are affected by IFN-I to establish the early environment of innate cells in HSV-infected mucosal tissues are largely unknown. Here, we found that IFN-I signaling promoted the differentiation of CCL2-producing Ly-6Chi monocytes and IFN-γ/granzyme B-producing NK cells, whereas deficiency of IFN-I signaling induced Ly-6Clo monocytes producing CXCL1 and CXCL2. More interestingly, recruitment of Ly-6Chi monocytes preceded that of NK cells with the levels peaked at 24 h post-infection in IFN-I–dependent manner, which was kinetically associated with the CCL2-CCL3 cascade response. Early Ly-6Chi monocyte recruitment was governed by CCL2 produced from hematopoietic stem cell (HSC)-derived leukocytes, whereas NK cell recruitment predominantly depended on CC chemokines produced by resident epithelial cells. Also, IFN-I signaling in HSC-derived leukocytes appeared to suppress Ly-6Ghi neutrophil recruitment to ameliorate immunopathology. Finally, tissue resident CD11bhiF4/80hi macrophages and CD11chiEpCAM+ dendritic cells appeared to produce initial CCL2 for migration-based self-amplification of early infiltrated Ly-6Chi monocytes upon stimulation by IFN-I produced from infected epithelial cells. Ultimately, these results decipher a detailed IFN-I–dependent pathway that establishes orchestrated mobilization of Ly-6Chi monocytes and NK cells through CCL2-CCL3 cascade response of HSC-derived leukocytes and epithelium-resident cells. Therefore, this cascade response of resident–to-hematopoietic–to-resident cells that drives cytokine–to-chemokine–to-cytokine production to recruit orchestrated innate cells is critical for attenuation of HSV replication in inflamed tissues. Herpes simplex virus type 1 and 2 (HSV-1 and HSV-2) are the most common cause of genital ulceration in humans worldwide with lifelong latent infection after peripheral replication in mucosal tissues. Furthermore, acquisition of human immunodeficiency virus (HIV) is increased in HSV-infected individuals, underscoring the contribution of this virus in facilitating increased susceptibility to other microbial pathogens. Therefore, it is imperative to characterize the host defense to HSV infection and identify key components that regulate virus resistance, in order to devise therapeutic strategy. Although type I interferon (IFN-I)-dependent orchestrated mobilization of innate cells in inflamed tissues is considered a key player to control replication and CNS-invasion of HSV, the regulators and cell population that are affected by IFN-I to establish the orchestrated environment of innate cells in HSV-infected tissues are largely unknown. In the present study, we demonstrate that IFN-I signal governs the sequential recruitment of Ly-6Chi monocytes and then NK cells into mucosal tissues, depending on CCL2-CCL3 cascade mediated by HSC-derived leukocytes and epithelial resident cells, respectively. Also, tissue resident CD11bhiF4/80hi macrophages and CD11chiEpCAM+ dendritic cells were involved in producing the initial CCL2 for migration-based self-amplification of rapidly infiltrated Ly-6Chi monocytes through stimulation by IFN-I produced from infected epithelial cells. This study deciphers detailed IFN-I-dependent pathway that establishes orchestrated mobilization of Ly-6Chi monocytes and NK cells through CCL2-CCL3 cascade.
Collapse
Affiliation(s)
- Erdenebileg Uyangaa
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Republic of Korea
| | - Jin Hyoung Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Republic of Korea
| | - Ajit Mahadev Patil
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Republic of Korea
| | - Jin Young Choi
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Republic of Korea
| | - Seong Bum Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Republic of Korea
| | - Seong Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Republic of Korea
- Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, Republic of Korea
- * E-mail:
| |
Collapse
|
28
|
Lin BY, Zhou L, Geng L, Zheng ZY, Jia JJ, Zhang J, Yao J, Zheng SS. High neutrophil-lymphocyte ratio indicates poor prognosis for acute-on-chronic liver failure after liver transplantation. World J Gastroenterol 2015; 21:3317-3324. [PMID: 25805939 PMCID: PMC4363762 DOI: 10.3748/wjg.v21.i11.3317] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 10/14/2014] [Accepted: 12/22/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the significance of pre-transplant neutrophil-lymphocyte ratio (NLR) in determining the prognosis of liver transplant (LT) recipients with acute-on-chronic liver failure (ACLF).
METHODS: Data were collected from the liver transplantation data bank. The NLR values and other conventional inflammatory markers were evaluated for their ability to predict the prognosis of 153 patients with ACLF after LT. The NLR cut-off value was based on a receiver operating characteristic curve analysis. A Kaplan-Meier curve analysis and univariate and multivariate Cox regression models were used to define the independent risk factors for poor outcomes.
RESULTS: The optimal NLR cut-off value was 4.6. Out of 153 patients, 83 (54.2%) had an NLR ≥ 4.6. The 1-, 3-, and 5-year overall survival rates were 94.3%, 92.5% and 92.5%, respectively, in the normal NLR group and 74.7%, 71.8% and 69.8%, respectively, in patients with high NLRs (P < 0.001). Furthermore, there was a significant difference in infectious complications after LT between the high and normal NLR groups. There were no significant differences for other complications. In the multivariate Cox regression model, a high NLR was defined as a significant predictor of poor outcomes for LT.
CONCLUSION: A high NLR is a convenient and available predictor for prognosis of LT patients and can potentially optimize the current criteria for LT in ACLF.
Collapse
|
29
|
Wu B, Walker J, Spur B, Rodriguez A, Yin K. Effects of Lipoxin A4 on antimicrobial actions of neutrophils in sepsis. Prostaglandins Leukot Essent Fatty Acids 2015; 94:55-64. [PMID: 25476955 DOI: 10.1016/j.plefa.2014.11.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 11/13/2014] [Accepted: 11/15/2014] [Indexed: 01/19/2023]
Abstract
In sepsis, hyperactivation of neutrophils can lead to tissue injury. Later, neutrophil dysregulation with reduced levels of migration, decreased apoptosis and inadequate phagocytosis may impair the host׳s ability to clear infection. Lipoxin A4 (LXA4) is a pro-resolution lipid mediator which reduces neutrophil migration and inflammatory mediator expression. As neutrophil migration and activation are important in bacterial clearance, the role of LXA4 in regulating neutrophil function for bacterial clearance is unclear. Using the cecal ligation and puncture (CLP) rat model of sepsis, LXA4 given after 1h reduced blood bacterial load at 24h. LXA4 treatment decreased neutrophil migration to the peritoneum but augmented blood neutrophil phagocytic ability and promoted apoptosis without affecting free radical production. In contrast, LXA4 increased peritoneal neutrophil phagocytic ability without affecting apoptosis or free radical production suggesting that in vivo effects of LXA4 were compartment specific. To investigate if LXA4 acted directly on neutrophils, blood and peritoneal leukocytes were taken from CLP rats 1h after surgery and incubated ex vivo with and without LXA4. LXA4 (1nM) increased phagocytosis in blood neutrophils without affecting apoptosis or free radical production. Ex vivo LXA4 had no effect on peritoneal neutrophils which suggests that LXA4 enhanced peritoneal neutrophil phagocytic ability in vivo by an indirect mechanism. The results suggest that LXA4 reduced neutrophil migration, but increased neutrophil bacteria clearing function without excessive free radical production. This phenotype was associated with reduced blood bacteria load.
Collapse
Affiliation(s)
- B Wu
- Department of Cell Biology, Rowan University - SOM, Stratford, NJ, USA
| | - J Walker
- Department of Cell Biology, Rowan University - SOM, Stratford, NJ, USA
| | - B Spur
- Department of Cell Biology, Rowan University - SOM, Stratford, NJ, USA
| | - A Rodriguez
- Department of Cell Biology, Rowan University - SOM, Stratford, NJ, USA
| | - K Yin
- Department of Cell Biology, Rowan University - SOM, Stratford, NJ, USA.
| |
Collapse
|
30
|
Yang Q, Stevenson HL, Scott MJ, Ismail N. Type I interferon contributes to noncanonical inflammasome activation, mediates immunopathology, and impairs protective immunity during fatal infection with lipopolysaccharide-negative ehrlichiae. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:446-61. [PMID: 25481711 DOI: 10.1016/j.ajpath.2014.10.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 09/18/2014] [Accepted: 10/09/2014] [Indexed: 12/29/2022]
Abstract
Ehrlichia species are intracellular bacteria that cause fatal ehrlichiosis, mimicking toxic shock syndrome in humans and mice. Virulent ehrlichiae induce inflammasome activation leading to caspase-1 cleavage and IL-18 secretion, which contribute to development of fatal ehrlichiosis. We show that fatal infection triggers expression of inflammasome components, activates caspase-1 and caspase-11, and induces host-cell death and secretion of IL-1β, IL-1α, and type I interferon (IFN-I). Wild-type and Casp1(-/-) mice were highly susceptible to fatal ehrlichiosis, had overwhelming infection, and developed extensive tissue injury. Nlrp3(-/-) mice effectively cleared ehrlichiae, but displayed acute mortality and developed liver injury similar to wild-type mice. By contrast, Ifnar1(-/-) mice were highly resistant to fatal disease and had lower bacterial burden, attenuated pathology, and prolonged survival. Ifnar1(-/-) mice also had improved protective immune responses mediated by IFN-γ and CD4(+) Th1 and natural killer T cells, with lower IL-10 secretion by T cells. Importantly, heightened resistance of Ifnar1(-/-) mice correlated with improved autophagosome processing, and attenuated noncanonical inflammasome activation indicated by decreased activation of caspase-11 and decreased IL-1β, compared with other groups. Our findings demonstrate that IFN-I signaling promotes host susceptibility to fatal ehrlichiosis, because it mediates ehrlichia-induced immunopathology and supports bacterial replication, perhaps via activation of noncanonical inflammasomes, reduced autophagy, and suppression of protective CD4(+) T cells and natural killer T-cell responses against ehrlichiae.
Collapse
Affiliation(s)
- Qin Yang
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Heather L Stevenson
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Melanie J Scott
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nahed Ismail
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
31
|
TRPV1 antagonism by capsazepine modulates innate immune response in mice infected with Plasmodium berghei ANKA. Mediators Inflamm 2014; 2014:506450. [PMID: 25242870 PMCID: PMC4158567 DOI: 10.1155/2014/506450] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 07/08/2014] [Indexed: 12/17/2022] Open
Abstract
Thousands of people suffer from severe malaria every year. The innate immune response plays a determinant role in host's defence to malaria. Transient receptor potential vanilloid 1 (TRPV1) modulates macrophage-mediated responses in sepsis, but its role in other pathogenic diseases has never been addressed. We investigated the effects of capsazepine, a TRPV1 antagonist, in malaria. C57BL/6 mice received 10(5) red blood cells infected with Plasmodium berghei ANKA intraperitoneally. Noninfected mice were used as controls. Capsazepine or vehicle was given intraperitoneally for 6 days. Mice were culled on day 7 after infection and blood and spleen cell phenotype and activation were evaluated. Capsazepine decreased circulating but not spleen F4/80(+)Ly6G(+) cell numbers as well as activation of both F4/80(+)and F4/80(+)Ly6G(+) cells in infected animals. In addition, capsazepine increased circulating but not spleen GR1(+) and natural killer (NK) population, without interfering with natural killer T (NKT) cell numbers and blood NK and NKT activation. However, capsazepine diminished CD69 expression in spleen NKT but not NK cells. Infection increased lipid peroxidation and the release of TNFα and IFNγ, although capsazepine-treated group exhibited lower levels of lipid peroxidation and TNFα. Capsazepine treatment did not affect parasitaemia. Overall, TRPV1 antagonism modulates the innate immune response to malaria.
Collapse
|
32
|
Type I interferons promote severe disease in a mouse model of lethal ehrlichiosis. Infect Immun 2014; 82:1698-709. [PMID: 24491580 DOI: 10.1128/iai.01564-13] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human monocytic ehrlichiosis (HME) is caused by a tick-borne obligate intracellular pathogen of the order Rickettsiales. HME disease can range from mild to a fatal, toxic shock-like syndrome, yet the mechanisms regulating pathogenesis are not well understood. We define a central role for type I interferons (alpha interferon [IFN-α] and IFN-β) in severe disease in a mouse model of fatal ehrlichiosis caused by Ixodes ovatus Ehrlichia (IOE). IFN-α and IFN-β were induced by IOE infection but not in response to a less virulent strain, Ehrlichia muris. The major sources of type I IFNs during IOE infection were plasmacytoid dendritic cells and monocytes. Mice lacking the receptor for type I IFNs (Ifnar deficient) or neutralization of IFN-α and IFN-β resulted in a reduced bacterial burden. Ifnar-deficient mice exhibited significantly increased survival after IOE infection, relative to that of wild-type (WT) mice, that correlated with increased type II IFN (IFN-γ) production. Pathogen-specific antibody responses were also elevated in Ifnar-deficient mice, and this required IFN-γ. Remarkably, increased IFN-γ and IgM were not essential for protection in the absence of type I IFN signaling. The direct effect of type I IFNs on hematopoietic and nonhematopoietic cells was evaluated in bone marrow chimeric mice. We observed that chimeric mice containing Ifnar-deficient hematopoietic cells succumbed to infection early, whereas Ifnar-deficient mice containing WT hematopoietic cells exhibited increased survival, despite having a higher bacterial burden. These data demonstrate that IFN-α receptor signaling in nonhematopoietic cells is important for pathogenesis. Thus, type I IFNs are induced during a rickettsial infection in vivo and promote severe disease.
Collapse
|