1
|
Ciepłucha HD, Bożejko M, Piesiak P, Serafińska S, Szetela B. Bacterial Pneumonia and Cryptogenic Pleuritis after Probable Monkeypox Virus Infection: A Case Report. Infect Dis Rep 2023; 15:795-805. [PMID: 38131884 PMCID: PMC10742663 DOI: 10.3390/idr15060071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
A large number of monkeypox (MPOX) cases have been reported in Europe and North America in 2022, and a new outbreak of this disease was declared. We describe a case of a patient with probable monkeypox during the height of this epidemic in Poland. The patient's symptoms resolved within two weeks, but over the next two months, he developed community-acquired pneumonia requiring hospitalization and, subsequently, non-specific pleuritis. The simultaneous occurrence of such severe infections in a previously healthy young man is not typical and suggests a potential underlying cause. We believe the potential association of these diseases with probable monkeypox virus infection is very likely. Cases of monkeypox pneumonia, both viral and secondary bacterial, have already been reported in the literature. Cases of viral pleuritis in the course of MPOX in animals have also been described; however, to our knowledge, no similar cases have been described in humans to date. Our case indicates that it is important to monitor patients after MPOX in order to respond promptly to potentially life-threatening but, as of yet, not fully understood complications.
Collapse
Affiliation(s)
- Hubert Dawid Ciepłucha
- Department of Infectious Diseases, Liver Diseases and Acquired Immune Deficiencies, Wroclaw Medical University, 51-149 Wroclaw, Poland
| | - Mateusz Bożejko
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Paweł Piesiak
- Lower Silesian Oncology, Pulmonology and Hematology Center, 53-439 Wroclaw, Poland
| | - Sylwia Serafińska
- Department of Infectious Diseases and Hepatology, Wroclaw Medical University, 51-149 Wroclaw, Poland
| | - Bartosz Szetela
- Department of Infectious Diseases, Liver Diseases and Acquired Immune Deficiencies, Wroclaw Medical University, 51-149 Wroclaw, Poland
| |
Collapse
|
2
|
Szulc-Dąbrowska L, Wojtyniak P, Struzik J, Toka FN, Winnicka A, Gieryńska M. ECTV Abolishes the Ability of GM-BM Cells to Stimulate Allogeneic CD4 T Cells in a Mouse Strain-Independent Manner. Immunol Invest 2019; 48:392-409. [PMID: 30884992 DOI: 10.1080/08820139.2019.1569676] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Ectromelia virus (ECTV) is the etiological agent of mousepox, an acute and systemic disease with high mortality rates in susceptible strains of mice. Resistance and susceptibility to mousepox are triggered by the dichotomous T-helper (Th) immune response generated in infected animals, with strong protective Th1 or nonprotective Th2 profile, respectively. Th1/Th2 balance is influenced by dendritic cells (DCs), which were shown to differ in their ability to polarize naïve CD4+ T cells in different mouse strains. Therefore, we have studied the inner-strain differences in the ability of conventional DCs (cDCs), generated from resistant (C57BL/6) and susceptible (BALB/c) mice, to stimulate proliferation and activation of Th cells upon ECTV infection. We found that ECTV infection of GM-CSF-derived bone marrow (GM-BM) cells, composed of cDCs and macrophages, affected initiation of allogeneic CD4+ T cells proliferation in a mouse strain-independent manner. Moreover, infected GM-BM cells from both mouse strains failed to induce and even inhibited the production of Th1 (IFN-γ and IL-2), Th2 (IL-4 and IL-10) and Th17 (IL-17A) cytokines by allogeneic CD4+ T cells. These results indicate that in in vitro conditions ECTV compromises the ability of cDCs to initiate/polarize adaptive antiviral immune response independently of the host strain resistance/susceptibility to lethal infection.
Collapse
Affiliation(s)
- Lidia Szulc-Dąbrowska
- a Department of Preclinical Sciences, Faculty of Veterinary Medicine , Warsaw University of Life Sciences , Warsaw , Poland
| | - Piotr Wojtyniak
- a Department of Preclinical Sciences, Faculty of Veterinary Medicine , Warsaw University of Life Sciences , Warsaw , Poland
| | - Justyna Struzik
- a Department of Preclinical Sciences, Faculty of Veterinary Medicine , Warsaw University of Life Sciences , Warsaw , Poland
| | - Felix N Toka
- a Department of Preclinical Sciences, Faculty of Veterinary Medicine , Warsaw University of Life Sciences , Warsaw , Poland.,b Center for Integrative Mammalian Research , Ross University School of Veterinary Medicine , Basseterre, St. Kitts , West Indies
| | - Anna Winnicka
- c Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine , Warsaw University of Life Sciences , Warsaw , Poland
| | - Małgorzata Gieryńska
- a Department of Preclinical Sciences, Faculty of Veterinary Medicine , Warsaw University of Life Sciences , Warsaw , Poland
| |
Collapse
|
3
|
Szulc-Dąbrowska L, Palusiński M, Struzik J, Gregorczyk-Zboroch KP, Toka FN, Schollenberger A, Gieryńska M. Ectromelia virus induces tubulin cytoskeletal rearrangement in immune cells accompanied by a loss of the microtubule organizing center and increased α-tubulin acetylation. Arch Virol 2018; 164:559-565. [PMID: 30374707 PMCID: PMC6373239 DOI: 10.1007/s00705-018-4030-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 09/09/2018] [Indexed: 01/06/2023]
Abstract
Ectromelia virus (ECTV) is an orthopoxvirus that productively replicates in dendritic cells (DCs), but its influence on the microtubule (MT) cytoskeleton in DCs is not known. Here, we show that ECTV infection of primary murine
granulocyte-macrophage colony stimulating factor-derived bone marrow cells (GM-BM) downregulates numerous genes engaged in MT cytoskeleton organization and dynamics. In infected cells, the MT cytoskeleton undergoes dramatic rearrangement and relaxation, accompanied by disappearance of the microtubule organizing centre (MTOC) and increased acetylation and stabilization of MTs, which are exploited by progeny virions for intracellular transport. This indicates a strong ability of ECTV to subvert the MT cytoskeleton of highly specialized immune cells.
Collapse
Affiliation(s)
- Lidia Szulc-Dąbrowska
- Division of Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Ciszewskiego 8, 02-786, Warsaw, Poland.
| | - Mateusz Palusiński
- Division of Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Ciszewskiego 8, 02-786, Warsaw, Poland
| | - Justyna Struzik
- Division of Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Ciszewskiego 8, 02-786, Warsaw, Poland
| | - Karolina P Gregorczyk-Zboroch
- Division of Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Ciszewskiego 8, 02-786, Warsaw, Poland
| | - Felix N Toka
- Division of Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Ciszewskiego 8, 02-786, Warsaw, Poland.,Center for Integrative Mammalian Research, Ross University School of Veterinary Medicine, PO Box 334, Basseterre, St. Kitts and Nevis
| | - Ada Schollenberger
- Division of Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Ciszewskiego 8, 02-786, Warsaw, Poland
| | - Małgorzata Gieryńska
- Division of Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Ciszewskiego 8, 02-786, Warsaw, Poland
| |
Collapse
|
4
|
Lauron EJ, Yang L, Elliott JI, Gainey MD, Fremont DH, Yokoyama WM. Cross-priming induces immunodomination in the presence of viral MHC class I inhibition. PLoS Pathog 2018; 14:e1006883. [PMID: 29444189 PMCID: PMC5812664 DOI: 10.1371/journal.ppat.1006883] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/17/2018] [Indexed: 01/07/2023] Open
Abstract
Viruses have evolved mechanisms of MHCI inhibition in order to evade recognition by cytotoxic CD8+ T cells (CTLs), which is well-illustrated by our prior studies on cowpox virus (CPXV) that encodes potent MHCI inhibitors. Deletion of CPXV viral MHCI inhibitors markedly attenuated in vivo infection due to effects on CTL effector function, not priming. However, the CTL response to CPXV in C57BL/6 mice is dominated by a single peptide antigen presented by H-2Kb. Here we evaluated the effect of viral MHCI inhibition on immunodominant (IDE) and subdominant epitopes (SDE) as this has not been thoroughly examined. We found that cross-priming, but not cross-dressing, is the main mechanism driving IDE and SDE CTL responses following CPXV infection. Secretion of the immunodominant antigen was not required for immunodominance. Instead, immunodominance was caused by CTL interference, known as immunodomination. Both immunodomination and cross-priming of SDEs were not affected by MHCI inhibition. SDE-specific CTLs were also capable of exerting immunodomination during primary and secondary responses, which was in part dependent on antigen abundance. Furthermore, CTL responses directed solely against SDEs protected against lethal CPXV infection, but only in the absence of the CPXV MHCI inhibitors. Thus, both SDE and IDE responses can contribute to protective immunity against poxviruses, implying that these principles apply to poxvirus-based vaccines. The use of vaccinia virus (VACV) to eradicate smallpox is the arguably the most successful demonstration of vaccination. The VACV vaccine also provides cross-protection against related zoonotic orthopoxviruses, including monkey poxvirus (MXPV) and CPXV, which circulate between various animal hosts and humans. Interestingly, Edward Jenner first demonstrated the concept of vaccination against smallpox in the late 1700s using CPXV. He also made the curious observation that CPXV vaccination did not always protect against recurrent exposure to CPXV. Jenner’s observations may be explained by the ability for CPXV to evade antiviral CD8+ T cell immune responses. To evade CD8+ T cells, CPXV inhibits MHCI antigen presentation, which is required to prime CD8+ T cells. Importantly, CPXV is the only orthopoxvirus that inhibits MHCI and thus provides a unique opportunity to investigate the effects of viral MHCI inhibition on CD8+ T cell priming. Here, we examine the factors that contribute to priming of CPXV-specific CD8+ T cells and show that viral MHCI inhibition does not affect CD8+ T cell priming, but prior CPXV immunization does inhibit priming during subsequent exposure to CPXV. The effects of pre-existing poxvirus immunity are therefore important to consider if poxvirus-based vaccines against various diseases are to be widely used.
Collapse
Affiliation(s)
- Elvin J. Lauron
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Liping Yang
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jabari I. Elliott
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Maria D. Gainey
- Department of Biology, Western Carolina University, Cullowhee, North Carolina, United States of America
| | - Daved H. Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Wayne M. Yokoyama
- Division of Rheumatology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
5
|
Szulc-Dąbrowska L, Struzik J, Cymerys J, Winnicka A, Nowak Z, Toka FN, Gieryńska M. The in Vitro Inhibitory Effect of Ectromelia Virus Infection on Innate and Adaptive Immune Properties of GM-CSF-Derived Bone Marrow Cells Is Mouse Strain-Independent. Front Microbiol 2017; 8:2539. [PMID: 29312229 PMCID: PMC5742134 DOI: 10.3389/fmicb.2017.02539] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/06/2017] [Indexed: 11/29/2022] Open
Abstract
Ectromelia virus (ECTV) belongs to the Orthopoxvirus genus of the Poxviridae family and is a natural pathogen of mice. Certain strains of mice are highly susceptible to ECTV infection and develop mousepox, a lethal disease similar to smallpox of humans caused by variola virus. Currently, the mousepox model is one of the available small animal models for investigating pathogenesis of generalized viral infections. Resistance and susceptibility to ECTV infection in mice are controlled by many genetic factors and are associated with multiple mechanisms of immune response, including preferential polarization of T helper (Th) immune response toward Th1 (protective) or Th2 (non-protective) profile. We hypothesized that viral-induced inhibitory effects on immune properties of conventional dendritic cells (cDCs) are more pronounced in ECTV-susceptible than in resistant mouse strains. To this extent, we confronted the cDCs from resistant (C57BL/6) and susceptible (BALB/c) mice with ECTV, regarding their reactivity and potential to drive T cell responses following infection. Our results showed that in vitro infection of granulocyte-macrophage colony-stimulating factor-derived bone marrow cells (GM-BM—comprised of cDCs and macrophages) from C57BL/6 and BALB/c mice similarly down-regulated multiple genes engaged in DC innate and adaptive immune functions, including antigen uptake, processing and presentation, chemokines and cytokines synthesis, and signal transduction. On the contrary, ECTV infection up-regulated Il10 in GM-BM derived from both strains of mice. Moreover, ECTV similarly inhibited surface expression of major histocompatibility complex and costimulatory molecules on GM-BM, explaining the inability of the cells to attain full maturation after Toll-like receptor (TLR)4 agonist treatment. Additionally, cells from both strains of mice failed to produce cytokines and chemokines engaged in T cell priming and Th1/Th2 polarization after TLR4 stimulation. These data strongly suggest that in vitro modulation of GM-BM innate and adaptive immune functions by ECTV occurs irrespective of whether the mouse strain is susceptible or resistant to infection. Moreover, ECTV limits the GM-BM (including cDCs) capacity to stimulate protective Th1 immune response. We cannot exclude that this may be an important factor in the generation of non-protective Th2 immune response in susceptible BALB/c mice in vivo.
Collapse
Affiliation(s)
- Lidia Szulc-Dąbrowska
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Justyna Struzik
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Joanna Cymerys
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Anna Winnicka
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Zuzanna Nowak
- Department of Genetics and Animal Breeding, Faculty of Animal Sciences, Warsaw University of Life Sciences, Warsaw, Poland
| | - Felix N Toka
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
| | - Małgorzata Gieryńska
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| |
Collapse
|
6
|
Dynamics of Pathological and Virological Findings During Experimental Calpox Virus Infection of Common Marmosets (Callithrix jacchus). Viruses 2017; 9:v9120363. [PMID: 29182537 PMCID: PMC5744138 DOI: 10.3390/v9120363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 12/26/2022] Open
Abstract
Experimental intranasal infection of marmosets (Callithrix jacchus) with calpox virus results in fatal disease. Route and dose used for viral inoculation of the test animals mimics the natural transmission of smallpox, thus representing a suitable model to study pathogenesis and to evaluate new vaccines against orthopoxvirus infection. However, the pathogenic mechanisms leading to death are still unclear. Therefore, our study aimed at investigating the kinetics of pathological alterations to clarify the pathogenesis in calpox virus infection. Following intranasal inoculation with two different viral doses, common marmosets were sacrificed on days 3, 5, 7, 10 and 12 post inoculation. Collected tissue was screened using histopathology, immunohistochemistry, transmission electron microscopy, and virological assays. Our data suggest that primary replication took place in nasal and bronchial epithelia followed by secondary replication in submandibular lymph nodes and spleen. Parallel to viremia at day 7, virus was detectable in many organs, mainly located in epithelial cells and macrophages, as well as in endothelial cells. Based on the onset of clinical signs, the histological and ultrastructural lesions and the immunohistochemical distribution pattern of the virus, the incubation period was defined to last 11 days, which resembles human smallpox. In conclusion, the data indicate that the calpox model is highly suitable for studying orthopoxvirus-induced disease.
Collapse
|
7
|
Szulc-Dąbrowska L, Struzik J, Ostrowska A, Guzera M, Toka FN, Bossowska-Nowicka M, Gieryńska MM, Winnicka A, Nowak Z, Niemiałtowski MG. Functional paralysis of GM-CSF-derived bone marrow cells productively infected with ectromelia virus. PLoS One 2017; 12:e0179166. [PMID: 28604814 PMCID: PMC5467855 DOI: 10.1371/journal.pone.0179166] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/24/2017] [Indexed: 12/31/2022] Open
Abstract
Ectromelia virus (ECTV) is an orthopoxvirus responsible for mousepox, a lethal disease of certain strains of mice that is similar to smallpox in humans, caused by variola virus (VARV). ECTV, similar to VARV, exhibits a narrow host range and has co-evolved with its natural host. Consequently, ECTV employs sophisticated and host-specific strategies to control the immune cells that are important for induction of antiviral immune response. In the present study we investigated the influence of ECTV infection on immune functions of murine GM-CSF-derived bone marrow cells (GM-BM), comprised of conventional dendritic cells (cDCs) and macrophages. Our results showed for the first time that ECTV is able to replicate productively in GM-BM and severely impaired their innate and adaptive immune functions. Infected GM-BM exhibited dramatic changes in morphology and increased apoptosis during the late stages of infection. Moreover, GM-BM cells were unable to uptake and process antigen, reach full maturity and mount a proinflammatory response. Inhibition of cytokine/chemokine response may result from the alteration of nuclear translocation of NF-κB, IRF3 and IRF7 transcription factors and down-regulation of many genes involved in TLR, RLR, NLR and type I IFN signaling pathways. Consequently, GM-BM show inability to stimulate proliferation of purified allogeneic CD4+ T cells in a primary mixed leukocyte reaction (MLR). Taken together, our data clearly indicate that ECTV induces immunosuppressive mechanisms in GM-BM leading to their functional paralysis, thus compromising their ability to initiate downstream T-cell activation events.
Collapse
Affiliation(s)
- Lidia Szulc-Dąbrowska
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Warsaw, Poland
- * E-mail:
| | - Justyna Struzik
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Warsaw, Poland
| | | | - Maciej Guzera
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Felix N. Toka
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Warsaw, Poland
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre, St. Kitts, West Indies
| | - Magdalena Bossowska-Nowicka
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Warsaw, Poland
| | - Małgorzata M. Gieryńska
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Warsaw, Poland
| | - Anna Winnicka
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Warsaw, Poland
| | - Zuzanna Nowak
- Department of Genetics and Animal Breeding, Faculty of Animal Sciences, Warsaw University of Life Sciences-SGGW, Warsaw, Poland
| | - Marek G. Niemiałtowski
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Warsaw, Poland
| |
Collapse
|
8
|
Nagata N, Saijo M, Kataoka M, Ami Y, Suzaki Y, Sato Y, Iwata-Yoshikawa N, Ogata M, Kurane I, Morikawa S, Sata T, Hasegawa H. Pathogenesis of fulminant monkeypox with bacterial sepsis after experimental infection with West African monkeypox virus in a cynomolgus monkey. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:4359-70. [PMID: 25120821 PMCID: PMC4129056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 07/01/2014] [Indexed: 06/03/2023]
Abstract
The pathogenesis of severe human monkeypox, which causes systemic and fulminant infections, is not clear. This study presents a case repot of fulminant monkeypox with bacterial sepsis after experimental infection with monkeypox virus in a cynomolgus monkey (Macaca fascicularis). In our previous study (Saijo et al., 2009, J Gen Virol), two cynomolgus monkeys became moribund after experimental infection with monkeypox virus Liberia strain, West African strain. One exhibited typical monkeypox-related papulovesicular lesions. The other monkey presented fulminant clinical symptoms with a characteristic flat red rash similar to that found in smallpox, which is associated with extremely high fatality rates. In this study, we found that the monkey with flat red rash had high levels of viremia and neutropenia, as well as high plasma levels of pro-inflammatory cytokines and chemokines compared with the other monkey. Monkeypox virus replicates in epithelial cells and macrophages in various organs. Sepsis due to Gram-positive cocci was confirmed histopathologically in the monkey with flat red rash. The lack of inflammatory response in the lesion suggested that the monkey with sepsis experienced strong immune suppression during the viral infection. The neutropenia and excessive inflammatory cytokine responses indicate that neutrophils play key roles in the pathogenesis of systemic and fulminant human monkeypox virus infections with sepsis.
Collapse
Affiliation(s)
- Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Masayuki Saijo
- Department of Virology 1, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Michiyo Kataoka
- Department of Pathology, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Yasushi Ami
- Department of Division of Experimental Animals Research, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Yuriko Suzaki
- Department of Division of Experimental Animals Research, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Yuko Sato
- Department of Pathology, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Naoko Iwata-Yoshikawa
- Department of Pathology, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Momoko Ogata
- Department of Virology 1, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Ichiro Kurane
- Department of Virology 1, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Shigeru Morikawa
- Department of Veterinary Science, National Institute of Infectious Diseases1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Tetsutaro Sata
- Department of Pathology, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| |
Collapse
|
9
|
Szulc-Dąbrowska L, Gieryńska M, Boratyńska-Jasińska A, Martyniszyn L, Winnicka A, Niemiałtowski MG. Quantitative immunophenotypic analysis of antigen-presenting cells involved in ectromelia virus antigen presentation in BALB/c and C57BL/6 mice. Pathog Dis 2013; 68:105-15. [PMID: 23776161 DOI: 10.1111/2049-632x.12054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 06/04/2013] [Accepted: 06/09/2013] [Indexed: 11/27/2022] Open
Abstract
During mousepox in resistant (C57BL/6) or susceptible (BALB/c) strains of mice, stimulation of Th1 or Th2 cytokine immune response, respectively, is observed. Because mechanisms of different polarization of T cells remain elusive, in this study, we quantitatively assessed the phenotype of antigen-presenting cells (APCs) involved in ectromelia virus (ECTV) antigen presentation and cluster formation with effector cells in secondary lymphoid organs of BALB/c and C57BL/6 mice. We showed that both strains of mice display similar dynamics and kinetics of viral antigen presentation by CD11c(+) , CD11b(+) , and CD19(+) cells. CD11c(+) and CD11b(+) cells highly participated in viral antigen presentation during all stages of mousepox, whereas CD19(+) cells presented viral peptides later in infection. The main population of dendritic cells (DCs) engaged in ECTV antigen presentation and cell junction formation with effector cells was a population of myeloid CD11b(+) DCs (mDCs). We suggest that, on the one hand, ECTV may differentially affect the functions of APCs depending on the strain of mice. On the other hand, we suggest that some types of APCs, such as mDCs or other DCs subsets, have different abilities to direct the shape of immune response depending on the host resistance to mousepox.
Collapse
Affiliation(s)
- Lidia Szulc-Dąbrowska
- Division of Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Warsaw, Poland.
| | | | | | | | | | | |
Collapse
|
10
|
Walline CC, Sehra S, Fisher AJ, Guindon LM, Kratzke IM, Montgomery JB, Lipking KP, Glosson NL, Benson HL, Sandusky GE, Wilkes DS, Brutkiewicz RR, Kaplan MH, Blum JS. Allergic airway disease in mice alters T and B cell responses during an acute respiratory poxvirus infection. PLoS One 2013; 8:e62222. [PMID: 23620814 PMCID: PMC3631162 DOI: 10.1371/journal.pone.0062222] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 03/19/2013] [Indexed: 11/18/2022] Open
Abstract
Pulmonary viral infections can exacerbate or trigger the development of allergic airway diseases via multiple mechanisms depending upon the infectious agent. Respiratory vaccinia virus transmission is well established, yet the effects of allergic airway disease on the host response to intra-pulmonary vaccinia virus infection remain poorly defined. As shown here BALB/c mice with preexisting airway disease infected with vaccinia virus developed more severe pulmonary inflammation, higher lung virus titers and greater weight loss compared with mice inoculated with virus alone. This enhanced viremia was observed despite increased pulmonary recruitment of CD8+ T effectors, greater IFNγ production in the lung, and high serum levels of anti-viral antibodies. Notably, flow cytometric analyses of lung CD8+ T cells revealed a shift in the hierarchy of immunodominant viral epitopes in virus inoculated mice with allergic airway disease compared to mice treated with virus only. Pulmonary IL-10 production by T cells and antigen presenting cells was detected following virus inoculation of animals and increased dramatically in allergic mice exposed to virus. IL-10 modulation of host responses to this respiratory virus infection was greatly influenced by the localized pulmonary microenvironment. Thus, blocking IL-10 signaling in virus-infected mice with allergic airway disease enhanced pulmonary CD4+ T cell production of IFNγ and increased serum anti-viral IgG1 levels. In contrast, pulmonary IFNγ and virus-specific IgG1 levels were reduced in vaccinia virus-treated mice with IL-10 receptor blockade. These observations demonstrate that pre-existing allergic lung disease alters the quality and magnitude of immune responses to respiratory poxviruses through an IL-10-dependent mechanism.
Collapse
Affiliation(s)
- Crystal C. Walline
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Sarita Sehra
- Department of Pediatrics, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Amanda J. Fisher
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Lynette M. Guindon
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Ian M. Kratzke
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Jessica B. Montgomery
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Kelsey P. Lipking
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Nicole L. Glosson
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Pediatrics, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Heather L. Benson
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - George E. Sandusky
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - David S. Wilkes
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Randy R. Brutkiewicz
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Mark H. Kaplan
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Pediatrics, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Janice S. Blum
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
11
|
Immune regulation and evasion of Mammalian host cell immunity during viral infection. INDIAN JOURNAL OF VIROLOGY : AN OFFICIAL ORGAN OF INDIAN VIROLOGICAL SOCIETY 2013; 24:1-15. [PMID: 24426252 DOI: 10.1007/s13337-013-0130-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 02/15/2013] [Indexed: 12/18/2022]
Abstract
The mammalian host immune system has wide array of defence mechanisms against viral infections. Depending on host immunity and the extent of viral persistence, either the host immune cells might clear/restrict the viral load and disease progression or the virus might evade host immunity by down regulating host immune effector response(s). Viral antigen processing and presentation in the host cells through major histocompatibility complex (MHC) elicit subsequent anti-viral effector T cell response(s). However, modulation of such response(s) might generate one of the important viral immune evasion strategies. Viral peptides are mostly generated by proteolytic cleavage in the cytosol of the infected host cells. CD8(+) T lymphocytes play critical role in the detection of viral infection by recognizing these peptides displayed at the plasma membrane by MHC-I molecules. The present review summarises the current knowledge on the regulation of mammalian host innate and adaptive immune components, which are operative in defence mechanisms against viral infections and the variety of strategies that viruses have evolved to escape host cell immunity. The understanding of viral immune evasion strategies is important for designing anti-viral immunotherapies.
Collapse
|
12
|
Kuri T, Sörensen AS, Thomas S, Karlsson Hedestam GB, Normark S, Henriques-Normark B, McInerney GM, Plant L. Influenza A virus-mediated priming enhances cytokine secretion by human dendritic cells infected with Streptococcus pneumoniae. Cell Microbiol 2013; 15:1385-400. [PMID: 23421931 PMCID: PMC3798092 DOI: 10.1111/cmi.12122] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 01/23/2013] [Accepted: 02/04/2013] [Indexed: 12/22/2022]
Abstract
Secondary infections with Streptococcus pneumoniae (SP) are frequently observed following influenza A virus (IAV) infection and have a substantial impact on global health. Despite this, the basis for the disease progression is incompletely understood. To investigate the effect of co-infection on human monocyte-derived dendritic cells (MDDCs) we analysed the expression of clinically important pro-inflammatory and immune-modulatory cytokines. IAV infection or treatment with supernatants from IAV-infected cell cultures resulted in priming of the DCs which subsequently influenced the production of IL-12p70, as well as IL-6, following SP infection. Co-infection of the same cell was not required but this effect was dependent on the time, dose and duration of the infections, as well as pathogen viability, bacterial uptake and endosome acidification. Bacterially infected cells were characterized as the main producers of IL-12p70. Finally, we showed that type I interferons were primarily responsible for the priming of IL-12p70 that was observed by infection with IAV. These results provide a probable mechanism for the elevated levels of particular cytokines observed in IAV and SP co-infected cell cultures with implications for the pathogenic outcome observed during in vivo infection.
Collapse
Affiliation(s)
- Thomas Kuri
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|