1
|
Zhu Y, Lu Y, Yuan L, Ling W, Jiang X, Chen S, Hu B. LincRNA-Cox2 regulates IL6/JAK3/STAT3 and NF-κB P65 pathway activation in Listeria monocytogenes-infected RAW264.7 cells. Int J Med Microbiol 2021; 311:151515. [PMID: 34146956 DOI: 10.1016/j.ijmm.2021.151515] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 05/16/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
Listeria monocytogenes (Lm) can lead to high mortality rates relative to other foodborne pathogens. Lm-induced inflammation is partly characterized by macrophage activation. Long non-coding RNAs (lncRNAs) have important roles in various biological processes. However, it is unknown how lncRNAs regulate the host response to Lm infection. To identify the role of lncRNA in Lm infection, we used in vitro and in vivo models. We found that lincRNA-Cox2 was highly expressed in Lm-infected RAW264.7 cells. LincRNA-Cox2 knockdown resulted in reduced proinflammatory cytokines, apoptosis, migration ability and enhanced phagocytosis of Lm. LincRNA-Cox2 knockdown also reduced the phosphorylation of Janus kinase 3 (JAK3) and signal transducer and activator of transcription (STAT3) and the nuclear translocation of nuclear factor (NF)-κB P65, which are known to be involved in inflammatory responses. Experimentally inhibiting the protein and phosphorylation levels of STAT3 resulted in reduced proinflammatory cytokines and enhanced phagocytosis of Lm by the RAW264.7 cells. Our research suggests that lincRNA-Cox2 plays important roles in inflammation, the phagocytic function and cell migration ability of RAW264.7 cells by activating interleukin (IL)-6/JAK3/STAT3 signaling, and lincRNA-Cox2 also regulates NF-κB P65 nuclear translocation. Our research provides new insights into the regulatory role of lincRNA-Cox2 in Lm infection.
Collapse
Affiliation(s)
- Yurong Zhu
- School of medicine, Jiangsu University, Zhenjiang, 212013, China; Department of Microbiology Laboratory, Linfen Central Hospital, Linfen, 041000, China
| | - Ye Lu
- School of medicine, Jiangsu University, Zhenjiang, 212013, China; Department of Clinical Laboratory, Yixing People's Hospital, Affiliated Jiangsu University, Wuxi, 214200, China
| | - Lin Yuan
- School of medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Wei Ling
- School of medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Xugan Jiang
- School of medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Shengxia Chen
- School of medicine, Jiangsu University, Zhenjiang, 212013, China.
| | - Bing Hu
- Department of Clinical Laboratory, Northern Jiangsu People' s Hospital, Yangzhou, 225001, China.
| |
Collapse
|
2
|
Johnson RM, Olatunde AC, Woodie LN, Greene MW, Schwartz EH. The Systemic and Cellular Metabolic Phenotype of Infection and Immune Response to Listeria monocytogenes. Front Immunol 2021; 11:614697. [PMID: 33628207 PMCID: PMC7897666 DOI: 10.3389/fimmu.2020.614697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/21/2020] [Indexed: 12/24/2022] Open
Abstract
It is widely accepted that infection and immune response incur significant metabolic demands, yet the respective demands of specific immune responses to live pathogens have not been well delineated. It is also established that upon activation, metabolic pathways undergo shifts at the cellular level. However, most studies exploring these issues at the systemic or cellular level have utilized pathogen associated molecular patterns (PAMPs) that model sepsis, or model antigens at isolated time points. Thus, the dynamics of pathogenesis and immune response to a live infection remain largely undocumented. To better quantitate the metabolic demands induced by infection, we utilized a live pathogenic infection model. Mice infected with Listeria monocytogenes were monitored longitudinally over the course of infection through clearance. We measured systemic metabolic phenotype, bacterial load, innate and adaptive immune responses, and cellular metabolic pathways. To further delineate the role of adaptive immunity in the metabolic phenotype, we utilized two doses of bacteria, one that induced both sickness behavior and protective (T cell mediated) immunity, and the other protective immunity alone. We determined that the greatest impact to systemic metabolism occurred during the early immune response, which coincided with the greatest shift in innate cellular metabolism. In contrast, during the time of maximal T cell expansion, systemic metabolism returned to resting state. Taken together, our findings demonstrate that the timing of maximal metabolic demand overlaps with the innate immune response and that when the adaptive response is maximal, the host has returned to relative metabolic homeostasis.
Collapse
Affiliation(s)
- Robert M Johnson
- Department of Biological Sciences, Auburn University, Auburn, AL, United States
| | - Adesola C Olatunde
- Department of Biological Sciences, Auburn University, Auburn, AL, United States
| | - Lauren N Woodie
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL, United States
| | - Michael W Greene
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL, United States
| | | |
Collapse
|
3
|
Yu AI, Zhao L, Eaton KA, Ho S, Chen J, Poe S, Becker J, Gonzalez A, McKinstry D, Hasso M, Mendoza-Castrejon J, Whitfield J, Koumpouras C, Schloss PD, Martens EC, Chen GY. Gut Microbiota Modulate CD8 T Cell Responses to Influence Colitis-Associated Tumorigenesis. Cell Rep 2020; 31:107471. [PMID: 32268087 PMCID: PMC7934571 DOI: 10.1016/j.celrep.2020.03.035] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 02/13/2020] [Accepted: 03/12/2020] [Indexed: 02/07/2023] Open
Abstract
There is increasing evidence that gut microbiome perturbations, also known as dysbiosis, can influence colorectal cancer development. To understand the mechanisms by which the gut microbiome modulates cancer susceptibility, we examine two wild-type mouse colonies with distinct gut microbial communities that develop significantly different tumor numbers using a mouse model of inflammation-associated tumorigenesis. We demonstrate that adaptive immune cells contribute to the different tumor susceptibilities associated with the two microbial communities. Mice that develop more tumors have increased colon lamina propria CD8+ IFNγ+ T cells before tumorigenesis but reduced CD8+ IFNγ+ T cells in tumors and adjacent tissues compared with mice that develop fewer tumors. Notably, intratumoral T cells in mice that develop more tumors exhibit increased exhaustion. Thus, these studies suggest that microbial dysbiosis can contribute to colon tumor susceptibility by hyperstimulating CD8 T cells to promote chronic inflammation and early T cell exhaustion, which can reduce anti-tumor immunity.
Collapse
Affiliation(s)
- Amy I Yu
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lili Zhao
- Department of Biostatistics, University of Michigan, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kathryn A Eaton
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sharon Ho
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jiachen Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sara Poe
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - James Becker
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Allison Gonzalez
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Delaney McKinstry
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Muneer Hasso
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Joel Whitfield
- Cancer Center Immunology Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Charles Koumpouras
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Patrick D Schloss
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eric C Martens
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Grace Y Chen
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
4
|
Lakho SA, Haseeb M, Huang J, Hasan MW, Naqvi MAUH, Zhou Z, Song X, Yan R, Xu L, Li X. Recombinant ubiquitin-conjugating enzyme of Eimeria maxima induces immunogenic maturation in chicken splenic-derived dendritic cells and drives Th1 polarization in-vitro. Microb Pathog 2020; 143:104162. [PMID: 32194180 DOI: 10.1016/j.micpath.2020.104162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/21/2022]
Abstract
Dendritic cells (DCs) are key linkages between innate immunity and acquired immunity. The antigens that promote the functions of DCs might be the effective candidates of novel vaccine. In this research, the ability of ubiquitin-conjugating enzyme (UCE), a recognized common antigens among chicken Eimeria species, to stimulate DCs of chickens were evaluated. We cloned UCE gene from Eimeria maxima (EmUCE), and its protein expression was confirmed by SDS-PAGE and western-blot. Immunofluorescence assay confirmed the binding of rEmUCE on the surface of chicken splenic-derived DCs (ChSP-DCs). Flow cytometric analysis showed that rEmUCE-treated ChSP-DCs increased MHCII, CD1.1, CD11c, CD80, and CD86 phenotypes. qRT-PCR indicated that transcript levels of maturation markers CCL5, CCR7, and CD83 in ChSP-DCs were upregulated in response to rEmUCE. Following rEmUCE treatment, chSP-DCs activated TLR signaling and inhibited Wnt signaling. Moreover, rEmUCE promoted DC-mediated T-cell proliferation in DC/T-cell co-incubation. Interestingly, CD3+/CD4+ T-cells were significantly enhanced when rEmUCE-treated chSP-DCs were co-incubated with T-cells. Cytokine secretion pattern of rEmUCE-stimulated ChSP-DCs revealed that the production of IL-12 and IFN-γ was increased whereas IL-10 and TGF-β were unchanged. Likewise, the co-incubation of ChSP-DCs with T-cells indicated increased production of IFN-γ but not IL-4. Collectively, rEmUCE could polarize DCs to immunogenic phenotype and shift the immune cells towards Th1 response. Our observations provide valuable insight for future research aimed at vaccine development against avian coccidiosis.
Collapse
Affiliation(s)
- Shakeel Ahmed Lakho
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China.
| | - Muhammad Haseeb
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China.
| | - Jianmei Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China.
| | - Muhammad Waqqas Hasan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China.
| | - Muhammad Ali-Ul-Husnain Naqvi
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China.
| | - Zhouyang Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China.
| | - XiaoKai Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China.
| | - RuoFeng Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China.
| | - Lixin Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China.
| | - XiangRui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China.
| |
Collapse
|
5
|
Montakhab-Yeganeh H, Abdossamadi Z, Zahedifard F, Taslimi Y, Badirzadeh A, Saljoughian N, Taheri T, Taghikhani M, Rafati S. Leishmania tarentolaeexpressing CXCL-10 as an efficient immunotherapy approach againstLeishmania major-infected BALB/c mice. Parasite Immunol 2017; 39. [DOI: 10.1111/pim.12461] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/14/2017] [Indexed: 02/06/2023]
Affiliation(s)
- H. Montakhab-Yeganeh
- Department of Clinical Biochemistry; Faculty of Medical Sciences; Tarbiat Modares University; Tehran Iran
| | - Z. Abdossamadi
- Department of Immunotherapy and Leishmania Vaccine Research; Pasteur Institute of Iran; Tehran Iran
| | - F. Zahedifard
- Department of Immunotherapy and Leishmania Vaccine Research; Pasteur Institute of Iran; Tehran Iran
| | - Y. Taslimi
- Department of Immunotherapy and Leishmania Vaccine Research; Pasteur Institute of Iran; Tehran Iran
| | - A. Badirzadeh
- Department of Immunotherapy and Leishmania Vaccine Research; Pasteur Institute of Iran; Tehran Iran
| | - N. Saljoughian
- Department of Immunotherapy and Leishmania Vaccine Research; Pasteur Institute of Iran; Tehran Iran
| | - T. Taheri
- Department of Immunotherapy and Leishmania Vaccine Research; Pasteur Institute of Iran; Tehran Iran
| | - M. Taghikhani
- Department of Clinical Biochemistry; Faculty of Medical Sciences; Tarbiat Modares University; Tehran Iran
| | - S. Rafati
- Department of Immunotherapy and Leishmania Vaccine Research; Pasteur Institute of Iran; Tehran Iran
| |
Collapse
|
6
|
Lukic J, Jancic I, Mirkovic N, Bufan B, Djokic J, Milenkovic M, Begovic J, Strahinic I, Lozo J. Lactococcus lactis and Lactobacillus salivarius differently modulate early immunological response of Wistar rats co-administered with Listeria monocytogenes. Benef Microbes 2017; 8:809-822. [PMID: 28856909 DOI: 10.3920/bm2017.0007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In the light of the increasing resistance of bacterial pathogens to antibiotics, one of the main global strategies in applied science is development of alternative treatments, which would be safe both for the host and from the environmental perspective. Accordingly, the aim of this study was to test whether two lactic acid bacteria (LAB) strains, Lactococcus lactis BGBU1-4 and Lactobacillus salivarius BGHO1, could be applied as safe supplements for Listeria infection. Two major research objectives were set: to compare the effects of BGBU1-4 and BGHO1 on early immune response in gut tissue of Wistar rats co-administered with Listeria monocytogenes ATCC19111 and next, to test how this applies to their usage as therapeutics in acute ATCC19111 infection. Intestinal villi (IV), Peyer's patches (PP) and mesenteric lymph nodes (MLN) were used for the analysis. The results showed that BGHO1 increased the mRNA expression of innate immune markers CD14, interleukin (IL)-1β and tumour necrosis factor (TNF)-α in PP and IV, and, in parallel, caused a decrease of listeriolysin O (LLO) mRNA expression in same tissues. In MLN of BGHO1 treated rats, LLO expression was increased, along with an increase of the expression of OX-62 mRNA and CD69, pointing to the activation of adaptive immunity. On the other hand, in BGBU1-4 treated rats, there was no reduction of LLO mRNA expression and no induction of innate immunity markers in intestinal tissue. Additionally, CD14 and IL-1β, as well as LLO, but not OX-62 mRNA and CD69 expression, were elevated in MLN of BGBU1-4 treated rats. However, when applied therapeutically, both, BGBU1-4 and BGHO1, lowered Listeria count in spleens of infected rats. Our results not only reveal the potential of LAB to ameliorate Listeria infections, but suggest different immunological effects of two different LAB strains, both of which could be effective in Listeria elimination.
Collapse
Affiliation(s)
- J Lukic
- 1 Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11010 Belgrade, Serbia
| | - I Jancic
- 2 Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11010 Belgrade, Serbia
| | - N Mirkovic
- 1 Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11010 Belgrade, Serbia
| | - B Bufan
- 2 Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11010 Belgrade, Serbia
| | - J Djokic
- 1 Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11010 Belgrade, Serbia
| | - M Milenkovic
- 2 Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11010 Belgrade, Serbia
| | - J Begovic
- 1 Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11010 Belgrade, Serbia
| | - I Strahinic
- 1 Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11010 Belgrade, Serbia
| | - J Lozo
- 1 Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11010 Belgrade, Serbia.,3 Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia
| |
Collapse
|
7
|
Olex AL, Turkett WH, Brzoza-Lewis KL, Fetrow JS, Hiltbold EM. Impact of the Type I Interferon Receptor on the Global Gene Expression Program During the Course of Dendritic Cell Maturation Induced by Polyinosinic Polycytidylic Acid. J Interferon Cytokine Res 2016; 36:382-400. [PMID: 27035059 DOI: 10.1089/jir.2014.0150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Dendritic cell (DC) maturation involves widespread changes in cellular function and gene expression. The regulatory role of IFNAR in the program of DC maturation remains incompletely defined. Thus, the time evolution impact of IFNAR on this process was evaluated. Changes in DC phenotype, function, and gene expression induced by poly I:C were measured in wild-type and IFNAR(-/-) DC at 9 time points over 24 h. Temporal gene expression profiles were filtered on consistency and response magnitude across replicates. The number of genes whose expression was altered by poly I:C treatment was greatly reduced in IFNAR(-/-) DC, including the majority of the downregulated gene expression program previously observed in wild-type (WT) DC. Furthermore, the number of genes upregulated was almost equal between WT and IFNAR(-/-) DC, yet the identities of those genes were distinct. Integrating these data with protein-protein interaction data revealed several novel subnetworks active during maturation, including nucleotide synthesis, metabolism, and repair. A subnetwork associated with redox activity was uniquely identified in IFNAR(-/-) DC. Overall, temporal gene expression and network analyses identified many genes regulated by the type I interferon response and revealed previously unidentified aspects of the DC maturation process.
Collapse
Affiliation(s)
- Amy L Olex
- 1 Department of Computer Science, Wake Forest University , Winston-Salem, North Carolina
| | - William H Turkett
- 1 Department of Computer Science, Wake Forest University , Winston-Salem, North Carolina
| | - Kristina L Brzoza-Lewis
- 2 Department of Microbiology and Immunology, Wake Forest School of Medicine , Winston-Salem, North Carolina
| | - Jacquelyn S Fetrow
- 1 Department of Computer Science, Wake Forest University , Winston-Salem, North Carolina.,3 Department of Physics, Wake Forest University , Winston-Salem, North Carolina
| | | |
Collapse
|
8
|
Enhanced survival during experimental Listeria monocytogenes sepsis in neonatal mice prophylactically treated with Th1 and macrophage immunoregulatory cytokines and mediators. Pediatr Infect Dis J 2014; 33:e330-7. [PMID: 24911893 DOI: 10.1097/inf.0000000000000442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Impairments in T-cell and macrophage-mediated host defense lead to increased infection-related morbidity and mortality in neonates, partly because of immaturity in T helper (Th)1 function. Listeria monocytogenes (Lm) is an intracellular pathogen disproportionately causing severe disease among neonates and the immunocompromised. Intact macrophage and Th1-mediated immune responses are critical for Lm clearance. We and others have previously demonstrated downregulation of Th1 and macrophage immunoregulatory cytokines in cord blood versus adult peripheral blood. We sought to determine whether therapeutic or prophylactic single agent or combination recombinant murine interleukin (rmIL)-12, rmIL-18, recombinant murine macrophage-colony stimulating factor (rmM-CSF), recombinant murine granulocyte macrophage-colony stimulating factor (rmGM-CSF) and recombinant murine interferon (rmIFN)-γ would enhance survival during experimental neonatal Lm sepsis. METHODS A 90% lethal dose (LD90) of Lm was established in C57/BL/6 neonatal mice. rmIL-12, rmIL-18, rmM-CSF, rmGM-CSF and rmIFN-γ were administered singly or sequentially, before or after LD90 Lm inoculation; ampicillin was administered 24 hours after inoculation. RESULTS Therapeutic doses of rmIL-12, rmIL-18, rmM-CSF, rmGM-CSF and rmIFN-γ as single agents and sequential therapy with rmM-CSF + (rmIL-12 and/or rmIL-18) + rmIFN-γ in addition to ampicillin were not associated with increased survival. However, prophylactic single doses of rmIL-12, rmIL-18, rmM-CSF and rmIFN-γ and prophylactic sequential doses of rmM-CSF + (rmIL-12 and/or rmIL-18) + rmIFN-γ in addition to ampicillin were associated with significantly enhanced survival compared with ampicillin alone. CONCLUSIONS These data suggest prophylactic administration of macrophage and Th1 immunoregulatory cytokines can potentially overcome deficits in neonatal immunity to protect against Lm.
Collapse
|
9
|
Dupont A, Mohamed F, Salehen N, Glenn S, Francescut L, Adib R, Byrne S, Brewin H, Elliott I, Richards L, Dimitrova P, Schwaeble W, Ivanovska N, Kadioglu A, Machado LR, Andrew PW, Stover C. Septicaemia models using Streptococcus pneumoniae and Listeria monocytogenes: understanding the role of complement properdin. Med Microbiol Immunol 2014; 203:257-71. [PMID: 24728387 PMCID: PMC4118039 DOI: 10.1007/s00430-013-0324-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 12/18/2013] [Indexed: 01/24/2023]
Abstract
Streptococcus pneumoniae and Listeria monocytogenes, pathogens which can cause severe infectious disease in human, were used to infect properdin-deficient and wildtype mice. The aim was to deduce a role for properdin, positive regulator of the alternative pathway of complement activation, by comparing and contrasting the immune response of the two genotypes in vivo. We show that properdin-deficient and wildtype mice mounted antipneumococcal serotype-specific IgM antibodies, which were protective. Properdin-deficient mice, however, had increased survival in the model of streptococcal pneumonia and sepsis. Low activity of the classical pathway of complement and modulation of FcγR2b expression appear to be pathogenically involved. In listeriosis, however, properdin-deficient mice had reduced survival and a dendritic cell population that was impaired in maturation and activity. In vitro analyses of splenocytes and bone marrow-derived myeloid cells support the view that the opposing outcomes of properdin-deficient and wildtype mice in these two infection models is likely to be due to a skewing of macrophage activity to an M2 phenotype in the properdin-deficient mice. The phenotypes observed thus appear to reflect the extent to which M2- or M1-polarised macrophages are involved in the immune responses to S. pneumoniae and L. monocytogenes. We conclude that properdin controls the strength of immune responses by affecting humoral as well as cellular phenotypes during acute bacterial infection and ensuing inflammation.
Collapse
Affiliation(s)
- Aline Dupont
- Department of Infection, Immunity and Inflammation, Maurice Shock Medical Sciences Building, University of Leicester, University Road, Leicester, LE1 9HN, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Kis-Toth K, Tsokos GC. Engagement of SLAMF2/CD48 prolongs the time frame of effective T cell activation by supporting mature dendritic cell survival. THE JOURNAL OF IMMUNOLOGY 2014; 192:4436-42. [PMID: 24670806 DOI: 10.4049/jimmunol.1302909] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Signaling lymphocyte activation molecule family (SLAMF)2/CD48 is a coactivator and adhesion molecule on cells with hematopoietic origin. It ligates mainly SLAMF4 on effector/memory CD8(+) T cells and NK cells, suggesting a potential role during viral infection, with SLAMF2 acting as a ligand to activate SLAMF4-bearing cells. The ability of SLAMF2 to signal on its own after it is engaged and the functional consequences are largely unknown. We found that cytosolic DNA-activated dendritic cells (DCs) upregulate the expression of SLAMF2 molecules. Using anti-SLAMF2 Ab and SLAMF4 recombinant protein, we found that SLAMF2 engagement activates immature DCs and, more interestingly, prolongs the survival of DNA-activated DCs by inhibiting IFN-β production and IFN-β-induced apoptosis and promotes the production of the granzyme B inhibitor protease inhibitor-9. Thus, SLAMF2 can serve as a survival molecule for DNA-activated DCs during their interaction with SLAMF4-expressing cytotoxic T cells. Based on our results, we propose that SLAMF2 engagement regulates adaptive immune responses by providing longer access of putative APCs to virus-specific effector T cells by prolonging the time frame of effective stimulation.
Collapse
Affiliation(s)
- Katalin Kis-Toth
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | | |
Collapse
|
11
|
Rothman J, Paterson Y. Live-attenuatedListeria-based immunotherapy. Expert Rev Vaccines 2014; 12:493-504. [DOI: 10.1586/erv.13.34] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
12
|
Matsumura F, Yamakita Y, Starovoytov V, Yamashiro S. Fascin confers resistance to Listeria infection in dendritic cells. THE JOURNAL OF IMMUNOLOGY 2013; 191:6156-64. [PMID: 24244012 DOI: 10.4049/jimmunol.1300498] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Ag-presenting dendritic cells (DCs) must survive bacterial infection to present Ag information to naive T cells. The greater ability of DCs' host defense is evident from the report that DCs are more resistant to Listeria monocytogenes than macrophages. However, the molecular mechanism of this resistance is unclear. We found that Listeria replicate more slowly in wild-type DCs compared with fascin1 knockout DCs. This finding is significant because fascin1, an actin-bundling protein, is specifically and greatly induced upon maturation of dendritic cells, but not other blood cells, including macrophages and neutrophils. Infection by Listeria makes phagosomes more acidic in wild-type DCs than in fascin1 knockout DCs, suggesting that fascin1 facilitates phagolysosomal fusion for killing of phagocytosed Listeria. We further found that fascin1 binds to LC3, an autophagosome marker, both in vivo and in vitro. Listeria are associated with LC3 to a greater extent in wild-type DCs than in fascin1 knockout DCs, suggesting that fascin1 facilitates autophagy for eradication of cytoplasmic Listeria. Taken together, our results suggest that fascin1 plays critical roles in the survival of DCs during Listeria infection, allowing DCs to function in innate and adaptive immunity.
Collapse
Affiliation(s)
- Fumio Matsumura
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
| | | | | | | |
Collapse
|
13
|
Laird RM, Wolf BJ, Princiotta MF, Hayes SM. γδ T cells acquire effector fates in the thymus and differentiate into cytokine-producing effectors in a Listeria model of infection independently of CD28 costimulation. PLoS One 2013; 8:e63178. [PMID: 23671671 PMCID: PMC3650071 DOI: 10.1371/journal.pone.0063178] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 03/30/2013] [Indexed: 11/18/2022] Open
Abstract
Both antigen recognition and CD28 costimulation are required for the activation of naïve αβ T cells and their subsequent differentiation into cytokine-producing or cytotoxic effectors. Notably, this two-signal paradigm holds true for all αβ T cell subsets, regardless of whether they acquire their effector function in the periphery or the thymus. Because of contradictory results, however, it remains unresolved as to whether CD28 costimulation is necessary for γδ T cell activation and differentiation. Given that γδ T cells have been recently shown to acquire their effector fates in the thymus, it is conceivable that the contradictory results may be explained, in part, by a differential requirement for CD28 costimulation in the development or differentiation of each γδ T cell effector subset. To test this, we examined the role of CD28 in γδ T cell effector fate determination and function. We report that, although IFNγ-producing γδ T (γδ-IFNγ) cells express higher levels of CD28 than IL-17-producing γδ T (γδ-17) cells, CD28-deficiency had no effect on the thymic development of either subset. Also, following Listeria infection, we found that the expansion and differentiation of γδ-17 and γδ-IFNγ effectors were comparable between CD28+/+ and CD28−/− mice. To understand why CD28 costimulation is dispensable for γδ T cell activation and differentiation, we assessed glucose uptake and utilization by γδ T cells, as CD28 costimulation is known to promote glycolysis in αβ T cells. Importantly, we found that γδ T cells express higher surface levels of glucose transporters than αβ T cells and, when activated, exhibit effector functions over a broader range of glucose concentrations than activated αβ T cells. Together, these data not only demonstrate an enhanced glucose metabolism in γδ T cells but also provide an explanation for why γδ T cells are less dependent on CD28 costimulation than αβ T cells.
Collapse
MESH Headings
- Animals
- CD28 Antigens/genetics
- CD28 Antigens/immunology
- CD28 Antigens/metabolism
- Cell Differentiation/immunology
- Cell Proliferation
- Cells, Cultured
- Cytokines/immunology
- Cytokines/metabolism
- Flow Cytometry
- Glucose/immunology
- Glucose/metabolism
- Host-Pathogen Interactions/immunology
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Listeria monocytogenes/immunology
- Listeria monocytogenes/physiology
- Mice
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/microbiology
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/metabolism
Collapse
Affiliation(s)
- Renee M. Laird
- Department of Microbiology and Immunology, State University of New York, Upstate Medical University, Syracuse, New York, United States of America
| | - Benjamin J. Wolf
- Department of Microbiology and Immunology, State University of New York, Upstate Medical University, Syracuse, New York, United States of America
| | - Michael F. Princiotta
- Department of Microbiology and Immunology, State University of New York, Upstate Medical University, Syracuse, New York, United States of America
| | - Sandra M. Hayes
- Department of Microbiology and Immunology, State University of New York, Upstate Medical University, Syracuse, New York, United States of America
- * E-mail:
| |
Collapse
|
14
|
Rowe JH, Ertelt JM, Xin L, Way SS. Listeria monocytogenes cytoplasmic entry induces fetal wastage by disrupting maternal Foxp3+ regulatory T cell-sustained fetal tolerance. PLoS Pathog 2012; 8:e1002873. [PMID: 22916020 PMCID: PMC3420962 DOI: 10.1371/journal.ppat.1002873] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 07/08/2012] [Indexed: 11/26/2022] Open
Abstract
Although the intracellular bacterium Listeria monocytogenes has an established predilection for disseminated infection during pregnancy that often results in spontaneous abortion or stillbirth, the specific host-pathogen interaction that dictates these disastrous complications remain incompletely defined. Herein, we demonstrate systemic maternal Listeria infection during pregnancy fractures fetal tolerance and triggers fetal wastage in a dose-dependent fashion. Listeria was recovered from the majority of concepti after high-dose infection illustrating the potential for in utero invasion. Interestingly with reduced inocula, fetal wastage occurred without direct placental or fetal invasion, and instead paralleled reductions in maternal Foxp3(+) regulatory T cell suppressive potency with reciprocal expansion and activation of maternal fetal-specific effector T cells. Using mutants lacking virulence determinants required for in utero invasion, we establish Listeria cytoplasmic entry is essential for disrupting fetal tolerance that triggers maternal T cell-mediated fetal resorption. Thus, infection-induced reductions in maternal Foxp3(+) regulatory T cell suppression with ensuing disruptions in fetal tolerance play critical roles in pathogenesis of immune-mediated fetal wastage.
Collapse
Affiliation(s)
| | | | | | - Sing Sing Way
- Departments of Pediatrics and Microbiology, Center for Infectious Disease and Microbiology Translational Research, University of Minnesota School of Medicine, Minneapolis, Minnesota, United States of America
| |
Collapse
|
15
|
Guirnalda P, Wood L, Paterson Y. Listeria monocytogenes and its products as agents for cancer immunotherapy. Adv Immunol 2012; 113:81-118. [PMID: 22244580 DOI: 10.1016/b978-0-12-394590-7.00004-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This review covers the use of Listeria monocytogenes and its virulence factors as cancer immunotherapeutics. We describe their development as vectors to carry protein tumor antigen and eukaryotic DNA plasmids to antigen-presenting cells and efforts to harness their tumor-homing properties. We also describe their use as vectors of angiogenic molecules to induce an immune response that will destroy tumor vasculature. The background knowledge necessary to understand the biology behind the rationale to develop Listeria as a vaccine vector for tumor immunotherapy is included as well as a brief summary of the major therapies that have used this approach thus far.
Collapse
Affiliation(s)
- Patrick Guirnalda
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
16
|
Plasmacytoid dendritic cells mature independently of MyD88 and IFN-αβR in response to Listeria and stimulate CD8 T cells. Immunol Lett 2011; 138:104-12. [DOI: 10.1016/j.imlet.2011.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 03/11/2011] [Accepted: 03/22/2011] [Indexed: 12/27/2022]
|
17
|
Campisi L, Soudja SM, Cazareth J, Bassand D, Lazzari A, Brau F, Narni-Mancinelli E, Glaichenhaus N, Geissmann F, Lauvau G. Splenic CD8α⁺ dendritic cells undergo rapid programming by cytosolic bacteria and inflammation to induce protective CD8⁺ T-cell memory. Eur J Immunol 2011; 41:1594-605. [PMID: 21469106 DOI: 10.1002/eji.201041036] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 01/08/2011] [Accepted: 03/09/2011] [Indexed: 11/09/2022]
Abstract
Memory CD8(+) T lymphocytes are critical effector cells of the adaptive immune system mediating long-lived pathogen-specific protective immunity. Three signals - antigen, costimulation and inflammation - orchestrate optimal CD8(+) T-cell priming and differentiation into effector and memory cells and shape T-cell functional fate and ability to protect against challenge infections. While among the conventional spleen DCs (cDCs), the CD8α(+) but not the CD8α(-) cDCs most efficiently mediate CD8(+) T-cell priming, it is unclear which subset, irrespective of their capacity to process MHC class I-associated antigens, is most efficient at inducing naïve CD8(+) T-cell differentiation into pathogen-specific protective memory cells in vivo. Moreover, the origin of the required signals is still unclear. Using mice infected with the intracellular bacterium Listeria monocytogenes, we show that splenic CD8α(+) cDCs become endowed with all functional features to optimally prime protective memory CD8(+) T cells in vivo within only a few hours post-immunization. Such programming requires both cytosolic signals resulting from bacterial invasion of the host cells and extracellular inflammatory mediators. Thus, these data designate these cells as the best candidates to facilitate the development of cell-based vaccine therapy.
Collapse
Affiliation(s)
- Laura Campisi
- Institut National de Santé et de Recherche Médicale Unité 924, Groupe Avenir, Valbonne, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Waite JC, Leiner I, Lauer P, Rae CS, Barbet G, Zheng H, Portnoy DA, Pamer EG, Dustin ML. Dynamic imaging of the effector immune response to listeria infection in vivo. PLoS Pathog 2011; 7:e1001326. [PMID: 21455492 PMCID: PMC3063765 DOI: 10.1371/journal.ppat.1001326] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 02/25/2011] [Indexed: 11/18/2022] Open
Abstract
Host defense against the intracellular pathogen Listeria monocytogenes (Lm) requires innate and adaptive immunity. Here, we directly imaged immune cell dynamics at Lm foci established by dendritic cells in the subcapsular red pulp (scDC) using intravital microscopy. Blood borne Lm rapidly associated with scDC. Myelomonocytic cells (MMC) swarmed around non-motile scDC forming foci from which blood flow was excluded. The depletion of scDC after foci were established resulted in a 10-fold reduction in viable Lm, while graded depletion of MMC resulted in 30-1000 fold increase in viable Lm in foci with enhanced blood flow. Effector CD8+ T cells at sites of infection displayed a two-tiered reduction in motility with antigen independent and antigen dependent components, including stable interactions with infected and non-infected scDC. Thus, swarming MMC contribute to control of Lm prior to development of T cell immunity by direct killing and sequestration from blood flow, while scDC appear to promote Lm survival while preferentially interacting with CD8+ T cells in effector sites.
Collapse
Affiliation(s)
- Janelle C. Waite
- Program in Molecular Pathogenesis, Helen L. and Martin S. Kimmel Center for Biology and Medicine of the Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York City, New York, United States of America
| | - Ingrid Leiner
- Infectious Disease Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, Immunology Program, Sloan-Kettering Institute, New York City, New York, United States of America
| | - Peter Lauer
- Aduro BioTech, Berkeley, California, United States of America
| | - Chris S. Rae
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, United States of America
| | - Gaetan Barbet
- Program in Molecular Pathogenesis, Helen L. and Martin S. Kimmel Center for Biology and Medicine of the Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York City, New York, United States of America
| | - Huan Zheng
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Daniel A. Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, United States of America
- School of Public Health, University of California, Berkeley, Berkeley, California, United State of America
| | - Eric G. Pamer
- Infectious Disease Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, Immunology Program, Sloan-Kettering Institute, New York City, New York, United States of America
| | - Michael L. Dustin
- Program in Molecular Pathogenesis, Helen L. and Martin S. Kimmel Center for Biology and Medicine of the Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York City, New York, United States of America
- * E-mail:
| |
Collapse
|
19
|
Wei Y, Wang X, Xiong X, Wen B. Coxiella burnetii antigen-stimulated dendritic cells mediated protection against Coxiella burnetii in BALB/c mice. J Infect Dis 2011; 203:283-91. [PMID: 21288829 DOI: 10.1093/infdis/jiq037] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Coxiella burnetii is the etiological agent of human Q fever. In this study, adaptive transfer of mouse bone marrow-derived dendritic cells (BMDCs) stimulated with C. burnetii antigen, phase I whole-cell antigen (PIAg), lipopolysaccharide (LPS)-removed PIAg (PIIAg), protein antigen Com1, or SecB significantly reduced coxiella burden in recipient mice compared with control mice. Mice that received PIIAg-pulsed BMDCs displayed substantially lower coxiella burden than recipient mice of PIAg-pulsed BMDCs after C burnetii challenge. The protection offered by the antigen-activated BMDCs was correlated with the increased proliferation of helper T (T(H)) T(H)1 CD4(+) cells, preferential development of T(H)17 cells, and impaired expansion of regulatory T lymphocytes. Our results suggest that PIIAg is far superior to PIAg in activating BMDCs to confer protection against C. burnetii in vivo, whereas Com1 and SecB are protective antigens because Com1- or SecB-pulsed BMDCs confer partial protection.
Collapse
Affiliation(s)
- Yan Wei
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | | | | | | |
Collapse
|
20
|
Distinct responses of splenic dendritic cell subsets to infection with Listeria monocytogenes: maturation phenotype, level of infection, and T cell priming capacity ex vivo. Cell Immunol 2011; 268:79-86. [PMID: 21457950 DOI: 10.1016/j.cellimm.2011.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 02/08/2011] [Accepted: 03/04/2011] [Indexed: 12/22/2022]
Abstract
To determine the relative contributions of DC subsets in the development of protective immunity to Listeria monocytogenes we examined the relationship between maturation, bacterial burden, and T cell priming capacity of four well characterized subsets of splenic DC following infection with Lm. CD8α(+), CD4(+), and CD8α(-)CD4(-) DC and the B220(+) plasmacytoid DC (pDC) were compared for abundance and costimulatory molecule expression at 24, 48, and 72h post i.v. infection. We further determined the bacterial burden associated with each DC subset and their relative capacities to prime CD8(+) T cells at 24hpi. The CD8α(+) DC displayed the highest level of maturation, association with live bacteria, and T cell activation potential. Second, the CD4(+) DC were also mature, yet were associated with fewer bacteria, and stimulated T cell proliferation, but not IFN-γ production. The CD8α(-)CD4(-) DC showed a modest maturation response and were associated with a high number of bacteria, but failed to induce T cell proliferation ex vivo. pDC displayed a strong maturation response, but were not associated with detectable bacteria and also failed to stimulate T cell activation. Finally, we measured the cytokine responses in these subsets and determined that IL-12 was produced predominantly by the CD8(+) DC, correlating with the ability of this subset DC to induce IFN-γ production in T cells. We conclude that Listeria-specific CD8(+) T cell activation in the spleen is most effectively achieved by infection-induced maturation of the CD8α(+) DC subset.
Collapse
|
21
|
Dexamethasone inhibits immunoreactivity of dendritic cells in patients with chronic idiopathic thrombocytopenic purpura. Blood Coagul Fibrinolysis 2011; 21:564-7. [PMID: 20581660 DOI: 10.1097/mbc.0b013e32833c2b8c] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The objective of this study was to investigate the possible effects of dexamethasone treatment on the immunoreactivity of dendritic cells in patients with chronic idiopathic thrombocytopenic purpura (ITP). Thirty-six newly diagnosed patients with chronic ITP received an oral high dose of dexamethasone (HD-DXM) at single daily doses of 40 mg for 4 consecutive days. The CD14 leukocytes isolated from the 21 remission patients and 10 normal controls were stimulated by recombinant human granulocyte-macrophage colony-stimulating factor and rhIL-4. The surface antigens of the dendritic cells were analyzed by flow cytometry and the level of IL-12p70 in the supernatant was detected by enzyme-linked immunosorbent assay. In ITP patients, the expression of both CD80 and CD86 in dendritic cells were significantly increased compared with those of the normal controls (51.60 +/- 13.47 vs. 36.03 +/- 15.43%, 61.50 +/- 15.93 vs. 40.28 +/- 11.49%, respectively; P < 0.05). After HD-DXM treatment, both CD80 and CD86 were decreased to levels comparable to normal controls (P > 0.05). The level of IL-12p70 in ITP patients was significantly higher (67.52 +/- 14.43 pg/ml) than the controls (39.78 +/- 10.03 pg/ml, P < 0.05). After treatment, IL-12p70 was reduced to 43.90 +/- 8.49 pg/ml with no significant differences between ITP group and control (P > 0.05). Dendritic cells and their cytokine secretion play important roles in ITP, and DXM may achieve its therapeutic effect on ITP by inhibiting immune responses through suppressing the function of dendritic cells.
Collapse
|
22
|
Sciaranghella G, Lakhashe SK, Ayash-Rashkovsky M, Mirshahidi S, Siddappa NB, Novembre FJ, Velu V, Amara RR, Zhou C, Li S, Li Z, Frankel FR, Ruprecht RM. A live attenuated Listeria monocytogenes vaccine vector expressing SIV Gag is safe and immunogenic in macaques and can be administered repeatedly. Vaccine 2010; 29:476-86. [PMID: 21070847 DOI: 10.1016/j.vaccine.2010.10.072] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 10/23/2010] [Accepted: 10/25/2010] [Indexed: 02/05/2023]
Abstract
Listeria monocytogenes (Lm) is known to induce strong cellular immune responses. We constructed a live-attenuated Lm vector, Lmdd-BdopSIVgag, which encodes SIVmac239 gag. Intragastric (i.g.) administration of 3 × 10(12) bacteria to rhesus macaques was safe and induced anti-Gag cellular but no humoral immune responses. Boosting of Gag-specific cellular responses was observed after i.g. administration of Lmdd-BdopSIVgag to previously vaccinated RM despite preexisting anti-Lm immunity shown by lymphoproliferative responses. Surprisingly, anti-Lm cellular responses were also detected in non-vaccinated controls, which may reflect the fact that Lm is a ubiquitous bacterium. The novel, live-attenuated Lmdd-BdopSIVgag may be an attractive platform for oral vaccine delivery.
Collapse
Affiliation(s)
- Gaia Sciaranghella
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Henry CJ, Grayson JM, Brzoza-Lewis KL, Mitchell LM, Westcott MM, Cook AS, Hiltbold EM. The roles of IL-12 and IL-23 in CD8+ T cell-mediated immunity against Listeria monocytogenes: Insights from a DC vaccination model. Cell Immunol 2010; 264:23-31. [PMID: 20483409 PMCID: PMC2902594 DOI: 10.1016/j.cellimm.2010.04.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Accepted: 04/21/2010] [Indexed: 02/02/2023]
Abstract
Listeria monocytogenes infection induces a strong inflammatory response characterized by the production of IL-12 and IFN-gamma and protective immunity against this pathogen is dependent on CD8+ T cells (CTL). Recent studies have suggested that these inflammatory cytokines affect the rate of memory CD8+ T cell generation as well as the number of short-lived effector cells generated. The role of the closely related cytokine, IL-23, in this response has not been examined. We hypothesized that IL-12 and IL-23 produced by dendritic cells collectively enhance the generation and function of memory cells. To test this hypothesis, we employed a DC vaccination approach. Mice lacking IL-12 and IL-23 were vaccinated with wild-type (WT), IL-12(-/-), or IL-12/23(-/-) DC and protection to Lm was monitored. Mice vaccinated with WT and IL-12(-/-) DC were resistant to lethal challenge with Lm. Surprisingly, mice vaccinated with IL-12/23(-/-) DC exhibited significantly reduced protection when challenged. Protection correlated with the relative size of the memory pools generated. In summary, these data indicate that IL-23 can partially compensate for the lack of IL-12 in the generation protective immunity against Lm.
Collapse
Affiliation(s)
- Curtis J. Henry
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 88010
- Integrated Department of Immunology, National Jewish Health, Denver, CO 80206
| | - Jason M. Grayson
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| | - Kristina L. Brzoza-Lewis
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| | - Latoya M. Mitchell
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
- Department of Microbiology, University of Alabama-Birmingham, Birmingham, AL 35243
| | - Marlena M. Westcott
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| | - Anne S. Cook
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| | - Elizabeth M. Hiltbold
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| |
Collapse
|
24
|
Dendritic cells inhibit the progression of Listeria monocytogenes intracellular infection by retaining bacteria in major histocompatibility complex class II-rich phagosomes and by limiting cytosolic growth. Infect Immun 2010; 78:2956-65. [PMID: 20404078 DOI: 10.1128/iai.01027-09] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Dendritic cells (DC) provide a suboptimal niche for the growth of Listeria monocytogenes, a facultative intracellular bacterial pathogen of immunocompromised and pregnant hosts. This is due in part to a failure of large numbers of bacteria to escape to the cytosol, an essential step in the intracellular life cycle that is mediated by listeriolysin O (LLO). Here, we demonstrate that wild-type bacteria that failed to enter the cytosol of bone marrow-derived DC were retained in a LAMP2+ compartment. An isogenic L. monocytogenes strain that produces an LLO protein with reduced pore-forming activity had a severe escape and growth phenotype in DC. Few mutant bacteria entered the cytosol in the first 2 h and were instead found in LAMP2+, major histocompatibility complex class II+ (MHC-II+) H2-DM vesicles characteristic of MHC-II antigen loading compartments (MIIC). In contrast, the mutant had a minor phenotype in bone marrow-derived macrophages (BMM) despite the reduced LLO activity. In the first hour, DC phagosomes acidified to a pH that was, on average, half a point higher than that of BMM phagosomes. Unlike BMM, L. monocytogenes growth in DC was minimal after 5 h, and consequently, DC remained viable and matured late in infection. Taken together, the data are consistent with a model in which phagosomal maturation events associated with the acquisition of MHC-II molecules present a suboptimal environment for L. monocytogenes escape to the DC cytosol, possibly by limiting the activity of LLO. This, in combination with an undefined mechanism that controls bacterial growth late in infection, promotes DC survival during the critical maturation response.
Collapse
|
25
|
Bahjat KS, Meyer-Morse N, Lemmens EE, Shugart JA, Dubensky TW, Brockstedt DG, Portnoy DA. Suppression of cell-mediated immunity following recognition of phagosome-confined bacteria. PLoS Pathog 2009; 5:e1000568. [PMID: 19730694 PMCID: PMC2731223 DOI: 10.1371/journal.ppat.1000568] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Accepted: 08/05/2009] [Indexed: 11/30/2022] Open
Abstract
Listeria monocytogenes is a facultative intracellular pathogen capable of inducing a robust cell-mediated immune response to sub-lethal infection. The capacity of L. monocytogenes to escape from the phagosome and enter the host cell cytosol is paramount for the induction of long-lived CD8 T cell–mediated protective immunity. Here, we show that the impaired T cell response to L. monocytogenes confined within a phagosome is not merely a consequence of inefficient antigen presentation, but is the result of direct suppression of the adaptive response. This suppression limited not only the adaptive response to vacuole-confined L. monocytogenes, but negated the response to bacteria within the cytosol. Co-infection with phagosome-confined and cytosolic L. monocytogenes prevented the generation of acquired immunity and limited expansion of antigen-specific T cells relative to the cytosolic L. monocytogenes strain alone. Bacteria confined to a phagosome suppressed the production of pro-inflammatory cytokines and led to the rapid MyD88-dependent production of IL-10. Blockade of the IL-10 receptor or the absence of MyD88 during primary infection restored protective immunity. Our studies demonstrate that the presence of microbes within a phagosome can directly impact the innate and adaptive immune response by antagonizing the signaling pathways necessary for inflammation and the generation of protective CD8 T cells. Little is understood about how the immune system distinguishes between pathogenic and non-pathogenic microbes. Limiting or preventing infections by intracellular pathogens requires the activation of innate immunity and the consequent generation of effector and memory T cells, which recognize and kill infected cells. Investigators are currently testing attenuated versions of pathogenic microbes as vaccines in an attempt to generate pathogen-specific T cells without causing disease. Unfortunately, attenuated microbes often fail to elicit long-lived protective immunity. We hypothesized that attenuated bacterial vaccines do not immunize because they fail to activate a stimulatory arm of host innate immune receptors. However, we found that these attenuated bacterial vaccines are not simply prevented from activating immunity, but rather generate a negative signal that inhibits the desired immune response. These studies may explain why the addition of an adjuvant to ineffective vaccines does not necessarily improve immunogenicity. Furthermore, these studies provide a framework for the development of attenuated vaccines that do not inhibit the desired immune responses.
Collapse
Affiliation(s)
- Keith S. Bahjat
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Research Center, Providence Cancer Center, Portland, Oregon, United States of America
- * E-mail: (KSB); (DAP)
| | - Nicole Meyer-Morse
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | | | - Jessica A. Shugart
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Research Center, Providence Cancer Center, Portland, Oregon, United States of America
| | | | | | - Daniel A. Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
- School of Public Health, University of California, Berkeley, California, United States of America
- * E-mail: (KSB); (DAP)
| |
Collapse
|
26
|
Nakayama Y, Kim SI, Kim EH, Lambris JD, Sandor M, Suresh M. C3 promotes expansion of CD8+ and CD4+ T cells in a Listeria monocytogenes infection. THE JOURNAL OF IMMUNOLOGY 2009; 183:2921-31. [PMID: 19648268 DOI: 10.4049/jimmunol.0801191] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It is known that C3 is required for optimal expansion of T cells during acute viral infections. However, it is not yet determined whether T cell responses to intracellular bacterial infections require C3. Therefore, we have investigated the requirement for C3 to elicit potent T cell responses to Listeria monocytogenes (LM). We show that expansion of Ag-specific CD8 and CD4 T cells during a primary response to LM was markedly reduced in the absence of C3 activity. Further studies indicated that, unlike in an influenza virus infection, the regulation of LM-specific T cell responses by C3 might not involve the downstream effector C5a. Moreover, reduced T cell responses to LM was not linked to defective maturation of dendritic cells or developmental anomalies in the peripheral T cell compartment of C3-deficient mice. Experiments involving adoptive transfer of C3-deficient CD8 T cells into the C3-sufficient environment of wild-type mice showed that these T cells do not have intrinsic proliferative defects, and a paracrine source of C3 will suffice for clonal expansion of CD8 T cells in vivo. However, stimulation of purified C3-deficient CD8 T cells by plastic-immobilized anti-CD3 showed that C3 promotes T cell proliferation directly, independent of its effects on APC. On the basis of these findings, we propose that diminished T cell responses to LM in C3-deficient mice might be at least in part due to lack of direct effects of C3 on T cells. These studies have furthered our understanding of C3-mediated regulation of T cell immunity to intracellular pathogens.
Collapse
Affiliation(s)
- Yumi Nakayama
- Department of Pathobiological Sciences, University of Wisconsin, Madison, WI 53706, USA
| | | | | | | | | | | |
Collapse
|
27
|
Walch M, Rampini SK, Stoeckli I, Latinovic-Golic S, Dumrese C, Sundstrom H, Vogetseder A, Marino J, Glauser DL, van den Broek M, Sander P, Groscurth P, Ziegler U. Involvement of CD252 (CD134L) and IL-2 in the expression of cytotoxic proteins in bacterial- or viral-activated human T cells. THE JOURNAL OF IMMUNOLOGY 2009; 182:7569-79. [PMID: 19494280 DOI: 10.4049/jimmunol.0800296] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Regulation of cytotoxic effector molecule expression in human CTLs after viral or bacterial activation is poorly understood. By using human autologous dendritic cells (DCs) to prime T lymphocytes, we found perforin only highly up-regulated in virus- (HSV-1, vaccinia virus) but not in intracellular bacteria- (Listeria innocua, Listeria monocytogenes, Mycobacterium tuberculosis, Chlamydophila pneumoniae) activated CTLs. In contrast, larger quantities of IFN-gamma and TNF-alpha were produced in Listeria-stimulated cultures. Granzyme B and granulysin were similarly up-regulated by all tested viruses and intracellular bacteria. DCs infected with HSV-1 showed enhanced surface expression of the costimulatory molecule CD252 (CD134L) compared with Listeria-infected DC and induced enhanced secretion of IL-2. Adding blocking CD134 or neutralizing IL-2 Abs during T cell activation reduced the HSV-dependent up-regulation of perforin. These data indicate a distinct CTL effector function in response to intracellular pathogens triggered via differing endogenous IL-2 production upon costimulation through CD252.
Collapse
Affiliation(s)
- Michael Walch
- Division of Cell Biology, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Cerovic V, McDonald V, Nassar MA, Paulin SM, Macpherson GG, Milling SWF. New insights into the roles of dendritic cells in intestinal immunity and tolerance. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2009; 272:33-105. [PMID: 19121816 DOI: 10.1016/s1937-6448(08)01602-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) play a critical key role in the initiation of immune responses to pathogens. Paradoxically, they also prevent potentially damaging immune responses being directed against the multitude of harmless antigens, to which the body is exposed daily. These roles are particularly important in the intestine, where only a single layer of epithelial cells provides a barrier against billions of commensal microorganisms, pathogens, and food antigens, over a huge surface area. In the intestine, therefore, DCs are required to perform their dual roles very efficiently to protect the body from the dual threats of invading pathogens and unwanted inflammatory reactions. In this review, we first describe the biology of DCs and their interactions with other cells types, paying particular attention to intestinal DCs. We, then, examine the ways in which this biology may become misdirected, resulting in inflammatory bowel disease. Finally, we discuss how DCs potentiate immune responses against viral, bacterial, parasitic infections, and their importance in the pathogenesis of prion diseases. We, therefore, provide an overview of the complex cellular interactions that affect intestinal DCs and control the balance between immunity and tolerance.
Collapse
Affiliation(s)
- Vuk Cerovic
- Sir William Dunn School of Pathology, Oxford University, Oxford, United Kingdom
| | | | | | | | | | | |
Collapse
|
29
|
Henry CJ, Ornelles DA, Mitchell LM, Brzoza-Lewis KL, Hiltbold EM. IL-12 produced by dendritic cells augments CD8+ T cell activation through the production of the chemokines CCL1 and CCL17. THE JOURNAL OF IMMUNOLOGY 2009; 181:8576-84. [PMID: 19050277 DOI: 10.4049/jimmunol.181.12.8576] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
IL-12 family members are an important link between innate and adaptive immunity. IL-12 drives Th1 responses by augmenting IFN-gamma production, which is key for clearance of intracellular pathogens. IL-23 promotes the development of IL-17-producing CD4(+) T cells that participate in the control of extracellular pathogens and the induction of autoimmunity. However, recent studies have shown that these cytokines can modulate lymphocyte migration and cellular interactions. Therefore, we sought to determine the individual roles of IL-12 and IL-23 in naive CD8(+) T cell activation by addressing their ability to influence IFN-gamma production and cellular interaction dynamics during priming by Listeria monocytogenes-infected dendritic cells (DC). We found that IL-12 was the major cytokine influencing the level of IFN-gamma production by CD8(+) T cells while IL-23 had little effect on this response. In addition, we observed that IL-12 promoted longer duration conjugation events between CD8(+) T cells and DC. This enhanced cognate interaction time correlated with increased production of the chemokines CCL1 and CCL17 by WT but not IL-12-deficient DC. Neutralization of both chemokines resulted in reduced interaction time and IFN-gamma production, demonstrating their importance in priming naive CD8(+) T cells. Our study demonstrates a novel mechanism through which IL-12 augments naive CD8(+) T cell activation by facilitating chemokine production, thus promoting more stable cognate interactions during priming.
Collapse
Affiliation(s)
- Curtis J Henry
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | |
Collapse
|
30
|
Loeffler DIM, Smolen K, Aplin L, Cai B, Kollmann TR. Fine-tuning the safety and immunogenicity of Listeria monocytogenes-based neonatal vaccine platforms. Vaccine 2008; 27:919-27. [PMID: 19059297 DOI: 10.1016/j.vaccine.2008.11.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2008] [Revised: 11/14/2008] [Accepted: 11/14/2008] [Indexed: 10/21/2022]
Abstract
We have developed virulence-attenuated strains of Listeria monocytogenes (Lm) that can be used as safe yet effective vaccine carriers for neonatal vaccination. Here we compare the vaccine efficacy of Lm based vaccine carrier candidates after only a single immunization in murine neonates and adults: Lm Delta(trpS actA) based strains that express and secrete multiple copies of the model antigen ovalbumin (OVA) either under the control of a phagosomal (P(hly)) or cytosolic (P(actA))-driven listerial promoter. While both strains induced high levels of antigen-specific primary and secondary CD8 and CD4 T cell responses, both neonatal and adult mice immunized with the phagosomal driven strain were significantly better protected against wildtype Lm challenge as compared to the naïve control group than mice immunized with the cytosolic driven strains. Interestingly, only neonatal mice immunized with the phagosomal driven strains generated high IgG antibody responses against OVA. Our phagosomal driven Lm-based vaccine platform presents the broadest (cellular & humoral response) and most efficient (highly protective) vaccine platform for neonatal vaccination yet described.
Collapse
Affiliation(s)
- Daniela I M Loeffler
- Department of Paediatrics, Child and Family Research Institute, Vancouver, BC, Canada.
| | | | | | | | | |
Collapse
|
31
|
Skoberne M, Yewdall A, Bahjat KS, Godefroy E, Lauer P, Lemmens E, Liu W, Luckett W, Leong M, Dubensky TW, Brockstedt DG, Bhardwaj N. KBMA Listeria monocytogenes is an effective vector for DC-mediated induction of antitumor immunity. J Clin Invest 2008; 118:3990-4001. [PMID: 19033668 PMCID: PMC2579623 DOI: 10.1172/jci31350] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Accepted: 10/01/2008] [Indexed: 01/16/2023] Open
Abstract
Vaccine strategies that utilize human DCs to enhance antitumor immunity have yet to realize their full potential. Approaches that optimally target a spectrum of antigens to DCs are urgently needed. Here we report the development of a platform for loading DCs with antigen. It is based on killed but metabolically active (KBMA) recombinant Listeria monocytogenes and facilitates both antigen delivery and maturation of human DCs. Highly attenuated KBMA L. monocytogenes were engineered to express an epitope of the melanoma-associated antigen MelanA/Mart-1 that is recognized by human CD8+ T cells when presented by the MHC class I molecule HLA-A*0201. The engineered KBMA L. monocytogenes induced human DC upregulation of costimulatory molecules and secretion of pro-Th1 cytokines and type I interferons, leading to effective priming of Mart-1-specific human CD8+ T cells and lysis of patient-derived melanoma cells. KBMA L. monocytogenes expressing full-length NY-ESO-1 protein, another melanoma-associated antigen, delivered the antigen for presentation by MHC class I and class II molecules independent of the MHC haplotype of the DC donor. A mouse therapeutic tumor model was used to show that KBMA L. monocytogenes efficiently targeted APCs in vivo to induce protective antitumor responses. Together, our data demonstrate that KBMA L. monocytogenes may be a powerful platform that can both deliver recombinant antigen to DCs for presentation and provide a potent DC-maturation stimulus, making it a potential cancer vaccine candidate.
Collapse
Affiliation(s)
- Mojca Skoberne
- Cancer Institute, New York University School of Medicine, New York, New York, USA.
Anza Therapeutics, Concord, California, USA
| | - Alice Yewdall
- Cancer Institute, New York University School of Medicine, New York, New York, USA.
Anza Therapeutics, Concord, California, USA
| | - Keith S. Bahjat
- Cancer Institute, New York University School of Medicine, New York, New York, USA.
Anza Therapeutics, Concord, California, USA
| | - Emmanuelle Godefroy
- Cancer Institute, New York University School of Medicine, New York, New York, USA.
Anza Therapeutics, Concord, California, USA
| | - Peter Lauer
- Cancer Institute, New York University School of Medicine, New York, New York, USA.
Anza Therapeutics, Concord, California, USA
| | - Edward Lemmens
- Cancer Institute, New York University School of Medicine, New York, New York, USA.
Anza Therapeutics, Concord, California, USA
| | - Weiqun Liu
- Cancer Institute, New York University School of Medicine, New York, New York, USA.
Anza Therapeutics, Concord, California, USA
| | - Will Luckett
- Cancer Institute, New York University School of Medicine, New York, New York, USA.
Anza Therapeutics, Concord, California, USA
| | - Meredith Leong
- Cancer Institute, New York University School of Medicine, New York, New York, USA.
Anza Therapeutics, Concord, California, USA
| | - Thomas W. Dubensky
- Cancer Institute, New York University School of Medicine, New York, New York, USA.
Anza Therapeutics, Concord, California, USA
| | - Dirk G. Brockstedt
- Cancer Institute, New York University School of Medicine, New York, New York, USA.
Anza Therapeutics, Concord, California, USA
| | - Nina Bhardwaj
- Cancer Institute, New York University School of Medicine, New York, New York, USA.
Anza Therapeutics, Concord, California, USA
| |
Collapse
|
32
|
Williams NL, Kloeze E, Govan BL, Käorner H, Ketheesan N. Burkholderia pseudomallei enhances maturation of bone marrow-derived dendritic cells. Trans R Soc Trop Med Hyg 2008; 102 Suppl 1:S71-5. [DOI: 10.1016/s0035-9203(08)70019-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
33
|
Maciag PC, Seavey MM, Pan ZK, Ferrone S, Paterson Y. Cancer immunotherapy targeting the high molecular weight melanoma-associated antigen protein results in a broad antitumor response and reduction of pericytes in the tumor vasculature. Cancer Res 2008; 68:8066-75. [PMID: 18829565 DOI: 10.1158/0008-5472.can-08-0287] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The high molecular weight melanoma-associated antigen (HMW-MAA), also known as melanoma chondroitin sulfate proteoglycan, has been used as a target for the immunotherapy of melanoma. This antigen is expressed on the cell surface and has a restricted distribution in normal tissues. Besides its expression in a broad range of transformed cells, this antigen is also found in pericytes, which are important for tumor angiogenesis. We generated a recombinant Listeria monocytogenes (Lm-LLO-HMW-MAA-C) that expresses and secretes a fragment of HMW-MAA (residues 2,160-2,258) fused to the first 441 residues of the listeriolysin O (LLO) protein. Immunization with Lm-LLO-HMW-MAA-C was able to impede the tumor growth of early established B16F10-HMW-MAA tumors in mice and both CD4(+) and CD8(+) T cells were required for therapeutic efficacy. Immune responses to a known HLA-A2 epitope present in the HMW-MAA(2160-2258) fragment was detected in the HLA-A2/K(b) transgenic mice immunized with Lm-LLO-HMW-MAA-C. Surprisingly, this vaccine also significantly impaired the in vivo growth of other tumorigenic cell lines, such as melanoma, renal carcinoma, and breast tumors, which were not engineered to express HMW-MAA. One hypothesis is that the vaccine could be targeting pericytes, which are important for tumor angiogenesis. In a breast tumor model, immunization with Lm-LLO-HMW-MAA-C caused CD8(+) T-cell infiltration in the tumor stroma and a significant decrease in the number of pericytes in the tumor blood vessels. In conclusion, a Lm-based vaccine against HMW-MAA can trigger cell-mediated immune responses to this antigen that can target not only tumor cells but also pericytes in the tumor vasculature.
Collapse
Affiliation(s)
- Paulo Cesar Maciag
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6076, USA
| | | | | | | | | |
Collapse
|
34
|
Shen H, Kanoh M, Maruyama S, Matsumoto A, Zhang W, Asano Y. Attenuated Listeria infection activates natural killer cell cytotoxicity to regress melanoma growth in vivo. Microbiol Immunol 2008; 52:107-17. [PMID: 18380808 DOI: 10.1111/j.1348-0421.2008.00018.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Listeria monocytogenes infection induces various types of immune responses. The Lm-induced immunity not only protects the hosts against Lm infection but also has a therapeutic effect on other diseases such as tumors and infectious diseases. In the present study, we sought to identify the cells and molecules that are primarily responsible for the Lm-induced antitumor immune response. We investigated the mechanism of the antitumor immune response induced by Lm infection using melanoma cells and various types of gene-manipulated mice and B16F10 melanoma cells. Melanoma cells were implanted into mice intrasplenically or intraperitoneally. Lm infection of mice remarkably suppressed the growth of transplanted melanoma. The suppression of melanoma growth was due to the augmented NK cytotoxicity. The Lm-induced NK activation against melanoma was type I interferon- and signal transducer and activator of transcription (STAT)1-dependent but independent of IL-12 and IFN-gamma. In contrast to avirulent Listeria innocua and hly(-) Lm failed to induce NK activation, a mutant Lm strain with minimal hemolytic activity and with normal accessibility to cytoplasm-induced NK activation. We demonstrated that the attenuated Lm entrance into the cytoplasm induces the production of type I IFN followed by the activation of NK cells, which is essential for the Lm-induced antitumor response.
Collapse
Affiliation(s)
- Hua Shen
- Department of Immunology and Host Defenses, Ehime University Graduate School of Medicine, Ehime, Japan
| | | | | | | | | | | |
Collapse
|
35
|
Orgun NN, Way SS. A critical role for phospholipase C in protective immunity conferred by listeriolysin O-deficient Listeria monocytogenes. Microb Pathog 2007; 44:159-63. [PMID: 17888620 PMCID: PMC2276143 DOI: 10.1016/j.micpath.2007.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2007] [Revised: 08/03/2007] [Accepted: 08/06/2007] [Indexed: 01/28/2023]
Abstract
Attenuated recombinant Listeria monocytogenes (Lm) strains are a promising class of vaccine vectors that trigger protective antigen-specific CD8 T cells. Listeriolysin O (LLO) is an important Lm virulence determinant allowing the bacterium to escape from the endocytic vacuole into the cell cytoplasm in phagocytic cells. However in non-phagocytic cells, Lm phospholipase C can also mediate cytoplasmic entry. The ability of LLO-deficient Lm to confer long-term protection to infection is uncertain. Herein, we demonstrate that LLO-deficient Lm mutants can prime protective immunity to subsequent Lm infection and that Lm phospholipase C is required for protective immunity conferred by LLO-deficient Lm.
Collapse
Affiliation(s)
| | - Sing Sing Way
- *For correspondence: Dr. Sing Sing Way, , Phone: 612-626-2526, Fax: 612-626-9924
| |
Collapse
|
36
|
Abstract
The immunostimulatory characteristics and intracellular niche of Listeria monocytogenes make it uniquely suitable for use as a live bacterial vaccine vector. Preclinical results supporting this idea, and current strategies to induce beneficial cell-mediated immunity to both infectious diseases and cancer with this vector, are discussed in this review.
Collapse
Affiliation(s)
- Kevin W Bruhn
- Department of Medicine, Division of Dermatology, Los Angeles Biomedical Research Institute at Harbor/UCLA Medical Center, 1124 W. Carson St., Torrance, CA 90502, USA.
| | | | | |
Collapse
|
37
|
Shaughnessy LM, Lipp P, Lee KD, Swanson JA. Localization of protein kinase C epsilon to macrophage vacuoles perforated by Listeria monocytogenes cytolysin. Cell Microbiol 2007; 9:1695-704. [PMID: 17346313 PMCID: PMC1974810 DOI: 10.1111/j.1462-5822.2007.00903.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2006] [Revised: 12/30/2006] [Accepted: 01/02/2007] [Indexed: 12/04/2022]
Abstract
Three proteins secreted by Listeria monocytogenes facilitate escape from macrophage vacuoles: the cholesterol-dependent cytolysin listeriolysin O (LLO), a phosphoinositide-specific phospholipase C (PI-PLC) and a broad-range phospholipase C (PC-PLC). LLO and PI-PLC can activate several members of the protein kinase C (PKC) family during infection. PKCepsilon is a novel PKC that contributes to macrophage activation, defence against bacterial infection, and phagocytosis; however, a role for PKCepsilon in Lm infections has not been described. To study PKCepsilon dynamics, PKCepsilon-YFP chimeras were visualized in macrophages during Lm infection. PKCepsilon-YFP was recruited to forming vacuoles during macrophage phagocytosis of Lm and again later to fully formed Lm vacuoles. The PKCepsilon-YFP localization to the fully formed Lm vacuole was LLO-dependent but independent of PI-PLC or PC-PLC. PKCepsilon-YFP recruitment often followed LLO perforation of the membrane, as indicated by localization of PKCepsilon-YFP to Lm vacuoles after they released small fluorescent dyes into the cytoplasm. PKCepsilon-YFP recruitment to vesicles also followed phagocytosis of LLO-containing liposomes or osmotic lysis of endocytic vesicles, indicating that vacuole perforation by LLO was the chief cause of the PKCepsilon response. These studies implicate PKCepsilon in a cellular mechanism for recognizing damaged membranous organelles, including the disrupted vacuoles created when Lm escapes into cytoplasm.
Collapse
Affiliation(s)
- Lee M Shaughnessy
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI 48109, USA.
| | - Peter Lipp
- Institute for Molecular Cell Biology, Saarland UniversityHomburg, Germany.
| | - Kyung-Dall Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of MichiganAnn Arbor, MI 48109, USA.
| | - Joel A Swanson
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI 48109, USA.
| |
Collapse
|
38
|
Abstract
The Gram-positive facultative intracellular bacterium Listeria monocytogenes is a model pathogen for elucidating important mechanisms of the immune response. Infection of mice with a sub-lethal dose of bacteria generates highly reproducible innate and adaptive immune responses, resulting in clearance of the bacteria and resistance to subsequent L. monocytogenes infection. Both the innate and adaptive immune systems are crucial to the recognition and elimination of this pathogen from the host.
Collapse
Affiliation(s)
- Lauren A. Zenewicz
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Hao Shen
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- *To whom correspondence should be addressed: Dr. Hao Shen, Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104. Phone: 215.573.5259; FAX: 215.573.9068; Email address:
| |
Collapse
|
39
|
Fang R, Ismail N, Soong L, Popov VL, Whitworth T, Bouyer DH, Walker DH. Differential interaction of dendritic cells with Rickettsia conorii: impact on host susceptibility to murine spotted fever rickettsiosis. Infect Immun 2007; 75:3112-23. [PMID: 17403875 PMCID: PMC1932850 DOI: 10.1128/iai.00007-07] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Spotted fever group rickettsioses are emerging and reemerging infectious diseases, some of which are life-threatening. In order to understand how dendritic cells (DCs) contribute to the host resistance or susceptibility to rickettsial diseases, we first characterized the in vitro interaction of rickettsiae with bone marrow-derived DCs (BMDCs) from resistant C57BL/6 (B6) and susceptible C3H/HeN (C3H) mice. In contrast to the exclusively cytosolic localization within endothelial cells, rickettsiae efficiently entered and localized in both phagosomes and cytosol of BMDCs from both mouse strains. Rickettsia conorii-infected BMDCs from resistant mice harbored higher bacterial loads compared to C3H mice. R. conorii infection induced maturation of BMDCs from both mouse strains as judged by upregulated expression of classical major histocompatibility complex (MHC) and costimulatory molecules. Compared to C3H counterparts, B6 BMDCs exhibited higher expression levels of MHC class II and higher interleukin-12 (IL-12) p40 production upon rickettsial infection and were more potent in priming naïve CD4(+) T cells to produce gamma interferon. In vitro DC infection and T-cell priming studies suggested a delayed CD4(+) T-cell activation and suppressed Th1/Th2 cell development in C3H mice. The suppressive CD4(+) T-cell responses seen in C3H mice were associated with a high frequency of Foxp3(+) T regulatory cells promoted by syngeneic R. conorii-infected BMDCs in the presence of IL-2. These data suggest that rickettsiae can target DCs to stimulate a protective type 1 response in resistant hosts but suppressive adaptive immunity in susceptible hosts.
Collapse
Affiliation(s)
- Rong Fang
- Center for Biodefense and Emerging Infectious Diseases, 301 University Blvd., Galveston, TX 77555-0609, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Westcott MM, Henry CJ, Cook AS, Grant KW, Hiltbold EM. Differential susceptibility of bone marrow-derived dendritic cells and macrophages to productive infection with Listeria monocytogenes. Cell Microbiol 2007; 9:1397-411. [PMID: 17250592 DOI: 10.1111/j.1462-5822.2006.00880.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Dendritic cells (DC) are required for the immune response against Listeria monocytogenes and are permissive for infection in vivo and in vitro. However, it is unclear if DC provide a desirable intracellular niche for bacterial growth. To address this issue, we have compared the behaviour of L. monocytogenes in murine bone marrow-derived DC and macrophages (BMM). Similar to BMM, bacteria escaped to the cytosol in DC, replicated, and spread to adjacent cells. However, DC infection was less robust in terms of intracellular doubling time and total increase in bacterial numbers. Immunofluorescence analysis using a strain of L. monocytogenes that expresses green fluorescent protein upon bacterial entry into the cytosol suggested that a subpopulation of DC restricted bacteria to vacuoles, a finding that was confirmed by electron microscopy. In unstimulated DC cultures, L. monocytogenes replicated preferentially in phenotypically immature cells. Furthermore, DC that were induced to mature prior to infection were poor hosts for bacterial growth. We conclude that DC provide a suboptimal niche for L. monocytogenes growth, and this is at least in part a function of the DC maturation state. Therefore, the generation of an effective T cell response may be a net effect of both productive and non-productive infection of DC.
Collapse
Affiliation(s)
- Marlena M Westcott
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | | | | | | | | |
Collapse
|
41
|
Bahjat KS, Liu W, Lemmens EE, Schoenberger SP, Portnoy DA, Dubensky TW, Brockstedt DG. Cytosolic entry controls CD8+-T-cell potency during bacterial infection. Infect Immun 2006; 74:6387-97. [PMID: 16954391 PMCID: PMC1695486 DOI: 10.1128/iai.01088-06] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Interaction with host immunoreceptors during microbial infection directly impacts the magnitude of the ensuing innate immune response. How these signals affect the quality of the adaptive T-cell response remains poorly understood. Utilizing an engineered strain of the intracellular pathogen Listeria monocytogenes that infects cells but fails to escape from the phagosome, we demonstrate the induction of long-lived memory T cells that are capable of secondary expansion and effector function but are incapable of providing protective immunity. We demonstrate that microbial invasion of the cytosol is required for dendritic cell activation and integration of CD40 signaling, ultimately determining the ability of the elicited CD8+-T-cell pool to protect against lethal wild-type L. monocytogenes challenge. These results reveal a crucial role for phagosomal escape, not for delivery of antigen to the class I major histocompatibility complex pathway but for establishing the appropriate cellular context during CD8+-T-cell priming.
Collapse
Affiliation(s)
- Keith S Bahjat
- Cerus Corporation, 2411 Stanwell Drive, Concord, CA 94520, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Tam MA, Wick MJ. Differential expansion, activation and effector functions of conventional and plasmacytoid dendritic cells in mouse tissues transiently infected with Listeria monocytogenes. Cell Microbiol 2006; 8:1172-87. [PMID: 16819969 DOI: 10.1111/j.1462-5822.2006.00700.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Dendritic cells (DC) are crucial in generating immunity to infection. Here we characterize changes in DC in terms of number, activation and effector functions, focusing on conventional DC (cDC) and plasmacytoid DC (pDC), in Listeria-infected mice. Kinetic studies showed a subset- and tissue-specific expansion of cDC and upregulation of CD80 and CD86 on splenic and mesenteric lymph node (MLN) cDC after intragastric infection. Expansion of pDC was more prolonged than cDC, and pDC upregulated CD86 and MHC-II, but not CD80, in both the spleen and MLN. cDC were an important source of IL-12 but not TNF-alpha during infection, while pDC made neither of these cytokines. Instead other CD11c(int) cells produced these cytokines. Using five-colour flow cytometry and double intracellular cytokine staining, we detected phenotypically similar CD11c(int)CD11b(+)Gr1(+) cells with distinct capacities to produce TNF-alpha/IL-12 or TNF-alpha/iNOS (inducible nitric oxide synthase) in Listeria-infected tissues. IL-12p70 was also produced by sorted CD11c(hi) and CD11c(int)CD11b(+)Gr1(+) cells. Furthermore, production of TNF-alpha, iNOS and IL-12 was differentially dependent on cellular localization of the bacteria. Cytosol-restricted bacteria induced TNF-alpha and iNOS-producing cells, albeit at lower frequency than wild-type bacteria. In contrast, IL-12 was induced only with wild-type bacteria. These data provide new insight into the relative abundance and function of distinct CD11c-expressing populations during the early stage of Listeria infection.
Collapse
Affiliation(s)
- Miguel A Tam
- Department of Clinical Immunology, Göteborg University, Göteborg, Sweden
| | | |
Collapse
|
43
|
Ahmed M, Brzoza KL, Hiltbold EM. Matrix protein mutant of vesicular stomatitis virus stimulates maturation of myeloid dendritic cells. J Virol 2006; 80:2194-205. [PMID: 16474127 PMCID: PMC1395366 DOI: 10.1128/jvi.80.5.2194-2205.2006] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2005] [Accepted: 12/02/2005] [Indexed: 12/29/2022] Open
Abstract
Matrix (M) protein mutants of vesicular stomatitis virus have recently been used as oncolytic viruses for tumor therapies and are being developed as vaccine vectors for heterologous antigens. Because dendritic cell (DC) maturation is an important correlate of tumor immunosurveillance and vaccine efficacy, we sought to determine the ability of a recombinant M protein mutant virus (rM51R-M virus) to mature DC in vitro. We have previously shown that rM51R-M virus is defective at inhibiting host gene expression in several cell lines compared to its recombinant wild-type counterpart, rwt virus. Therefore, rM51R-M virus allows the expression of genes involved in antiviral responses, such as the type I interferon (IFN) gene. Our results demonstrate that, in contrast to the rwt virus, rM51R-M virus induced the maturation of myeloid DC (mDC) populations, as indicated by an increase in the surface expression of CD40, CD80, and CD86 as well as the secretion of interleukin-12 (IL-12), IL-6, and type I IFN. In addition, mDC infected with rM51R-M virus effectively activated naïve T cells in vitro, whereas rwt virus-infected mDC were defective in antigen presentation. The inability of rwt virus to induce mDC maturation was correlated with the inhibition of host gene expression in rwt virus-infected cells. Our studies also indicated that the production of costimulatory molecules on mDC by rM51R-M virus was dependent on the type I IFN receptor, while maturation induced by this virus was largely independent of MyD88. These data indicate that rM51R-M virus effectively stimulates the maturation of mDC and has the potential to promote effective T-cell responses to vector-expressed antigens, activate DC at tumor sites during therapy, and aid in tumor immunosurveillance and destruction.
Collapse
Affiliation(s)
- Maryam Ahmed
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | | | | |
Collapse
|
44
|
Zammit DJ, Lefrançois L. Dendritic cell-T cell interactions in the generation and maintenance of CD8 T cell memory. Microbes Infect 2006; 8:1108-15. [PMID: 16549379 DOI: 10.1016/j.micinf.2005.12.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Accepted: 11/19/2005] [Indexed: 02/04/2023]
Abstract
Dendritic cells (DCs) are the critical antigen-presenting cells involved in initiating CD8 T cell responses to microbial and viral pathogens. Hence the generation of memory T cells from naïve T cells is intricately intertwined with DCs at every level. This review broadly addresses DC-CD8 T cell interactions that result in the generation and maintenance of CD8 memory T cells.
Collapse
Affiliation(s)
- David J Zammit
- Department of Immunology, University of Connecticut Health Center, M/C 1319, 263 Farmington Avenue, Farmington, CT 06030, USA
| | | |
Collapse
|
45
|
Kupfahl C, Geginat G, Hof H. Lentinan has a stimulatory effect on innate and adaptive immunity against murine Listeria monocytogenes infection. Int Immunopharmacol 2005; 6:686-96. [PMID: 16504933 DOI: 10.1016/j.intimp.2005.10.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2005] [Revised: 08/22/2005] [Accepted: 10/18/2005] [Indexed: 10/25/2022]
Abstract
Lentinan, a (1-3)-beta glucan from Lentinus edodes, is licensed as an immunostimulatory drug. We tested the effect of lentinan in the well-established model system of the murine Listeria monocytogenes infection. Pre-treatment of bone marrow macrophages and dendritic cells with lentinan resulted in increased production of TNF-alpha and IL-12 after L. monocytogenes infection in vitro. After lentinan treatment bone marrow macrophages showed increased NO-production and enhanced cytotoxic activity against L. monocytogenes. Pre-treatment of mice with lentinan resulted in increased concentrations of TNF-alpha, IL-12 and IFN-gamma and also an increased number of L. monocytogenes specific CD8 T cells in the spleen. The bacterial burden in spleen and liver of mice was significantly reduced during primary and secondary Listeria infection after lentinan pre-treatment of mice. In summary these results show that lentinan enhances the protective CD8 T-cell response against L. monocytogenes probably by a mechanism that involves the IL-12-mediated augmentation of the specific antilisterial CD8 T-cell response.
Collapse
Affiliation(s)
- C Kupfahl
- Institut für Medizinische Mikrobiologie und Hygiene, Fakultät für klinische Medizin Mannheim der Universität Heidelberg, Germany
| | | | | |
Collapse
|
46
|
Feng H, Zhang D, Palliser D, Zhu P, Cai S, Schlesinger A, Maliszewski L, Lieberman J. Listeria-infected myeloid dendritic cells produce IFN-beta, priming T cell activation. THE JOURNAL OF IMMUNOLOGY 2005; 175:421-32. [PMID: 15972676 DOI: 10.4049/jimmunol.175.1.421] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The intracellular bacterium Listeria monocytogenes infects dendritic cells (DC) and other APCs and induces potent cell-mediated protective immunity. However, heat-killed bacteria fail to do so. This study explored whether DC differentially respond to live and killed Listeria and how this affects T cell activation. To control for bacterial number, a replication-deficient strain, Lmdd, defective in D-alanine biosynthesis, was used. We found that DC internalize both live and heat-killed Lmdd and similarly up-regulate the expression of costimulatory molecules, a necessary step for T cell activation. However, only live Lmdd-infected DC stimulate T cells to express the early activation marker CD69 and enhance T cell activation upon TCR engagement. Infection with live, but not heat-killed, Lmdd induces myeloid DC to secrete copious amounts of IFN-beta, which requires bacterial cytosolic invasion. Exposure to high concentrations of IFN-beta sensitizes naive T cells for Ag-dependent activation.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- CD3 Complex/immunology
- Dendritic Cells/immunology
- Hot Temperature
- In Vitro Techniques
- Interferon-beta/biosynthesis
- Interferon-beta/genetics
- Lectins, C-Type
- Listeria monocytogenes/genetics
- Listeria monocytogenes/immunology
- Listeria monocytogenes/pathogenicity
- Lymphocyte Activation
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mutation
- Myeloid Cells/immunology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Hanping Feng
- CBR Institute for Biomedical Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Decker T, Müller M, Stockinger S. The Yin and Yang of type I interferon activity in bacterial infection. Nat Rev Immunol 2005; 5:675-87. [PMID: 16110316 DOI: 10.1038/nri1684] [Citation(s) in RCA: 359] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Interferons (IFNs) are cytokines that are important for immune responses, particularly to intracellular pathogens. They are divided into two structurally and functionally distinct types that interact with different cell-surface receptors. Classically, type I IFNs are potent antiviral immunoregulators, whereas the type II IFN enhances antibacterial immunity. However, as outlined here, type I IFNs are also produced in response to infection with other pathogens, and an increasing body of work shows that type I IFNs have an important role in the host response to bacterial infection. Strikingly, their activity can be either favourable or detrimental, and can influence various immune effector mechanisms.
Collapse
Affiliation(s)
- Thomas Decker
- Max F. Perutz Laboratories, University Departments at the Vienna Biocenter, Department of Microbiology and Immunobiology, University of Vienna, Vienna, Austria.
| | | | | |
Collapse
|
48
|
Abstract
Listeria monocytogenes is a Gram-positive bacterium that is often used to study the mammalian immune response to infection because it is easy to culture, is relatively safe to work with and causes a highly predictable infection in laboratory mice. The broad application of this mouse model has resulted in a torrent of studies characterizing the contributions of different cytokines, receptors, adaptors and effector molecules to resistance against infection with Listeria monocytogenes. These studies, which are yielding one of the most comprehensive pictures of the 'battle' between host and microorganism, are reviewed here.
Collapse
Affiliation(s)
- Eric G Pamer
- Infectious Disease Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, Immunology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10021, USA.
| |
Collapse
|