101
|
Elkins KL, Cooper A, Colombini SM, Cowley SC, Kieffer TL. In vivo clearance of an intracellular bacterium, Francisella tularensis LVS, is dependent on the p40 subunit of interleukin-12 (IL-12) but not on IL-12 p70. Infect Immun 2002; 70:1936-48. [PMID: 11895957 PMCID: PMC127864 DOI: 10.1128/iai.70.4.1936-1948.2002] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To determine the role of interleukin-12 (IL-12) in primary and secondary immunity to a model intracellular bacterium, we have comprehensively evaluated infection with Francisella tularensis LVS in three murine models of IL-12 deficiency. Mice lacking the p40 protein of IL-12 (p40 knockout [KO] mice) and mice treated in vivo with neutralizing anti-IL-12 antibodies survived large doses of primary and secondary LVS infection but never cleared bacteria and exhibited a chronic infection. In dramatic contrast, mice lacking the p35 protein (p35 KO mice) of heterodimeric IL-12 readily survived large doses of primary sublethal LVS infection as well as maximal secondary lethal challenge, with only a slight delay in clearance of bacteria. LVS-immune wild-type (WT) lymphocytes produced large amounts of gamma interferon (IFN-gamma), but p35 KO and p40 KO lymphocytes produced much less; nonetheless, similar amounts of NO were found in all cultures containing immune lymphocytes, and all immune lymphocytes were equally capable of controlling intracellular growth of LVS in vitro. Purified CD4(+) and CD8(+) T cells from both WT and p40 KO mice controlled intracellular growth, even though T cells from WT mice produced much more IFN-gamma than those from p40 KO mice, and p40 KO T cells did not adopt a Th2 phenotype. Thus, while IL-12 p70 stimulation of IFN-gamma production may be important for bacteriostasis, IL-12 p70 is not necessary for appropriate development of LVS-immune T cells that are capable of controlling intracellular bacterial growth and for clearance of primary or secondary LVS infection. Instead, an additional mechanism dependent on the IL-12 p40 protein, either alone or in another complex such as the newly discovered heterodimer IL-23, appears to be responsible for actual clearance of this intracellular bacterium.
Collapse
Affiliation(s)
- Karen L Elkins
- Laboratory of Mycobacteria, Division of Bacterial, Parasitic, and Allergenic Products, CBER/FDA, Rockville, Maryland 20852, USA.
| | | | | | | | | |
Collapse
|
102
|
Conlan JW, KuoLee R, Shen H, Webb A. Different host defences are required to protect mice from primary systemic vs pulmonary infection with the facultative intracellular bacterial pathogen, Francisella tularensis LVS. Microb Pathog 2002; 32:127-34. [PMID: 11855943 DOI: 10.1006/mpat.2001.0489] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Francisella tularensis is a zoonotic, facultative intracellular bacterial pathogen capable of initiating infection, tularemia, via multiple routes including dermal micro-abrasions and inhalation. Mouse models of systemically-initiated infection with F. tularensis LVS have been used extensively to reveal potential host defence mechanisms against the pathogen. Such studies have demonstrated the critical need for neutrophils and interferon-gamma (IFN-gamma) to combat the early stages of primary experimental tularaemia initiated by systemic routes. Surprisingly, however, the present study shows that these defences appear not to combat early pulmonary tularaemia initiated by inhalation of the pathogen into the lower airways. The results imply that the effectiveness of particular anti-bacterial host defences vary with invasion site. Thus, it is impossible to predict effective host defence mechanisms against inhalation-initiated tularaemia from current knowledge of anti- Francisella defences that have been shown to combat systemically-initiated infection.
Collapse
Affiliation(s)
- J Wayne Conlan
- National Research Council Canada, Institute for Biological Sciences, Ottawa, Ontario, K1A OR6, Canada.
| | | | | | | |
Collapse
|
103
|
Fulop M, Mastroeni P, Green M, Titball RW. Role of antibody to lipopolysaccharide in protection against low- and high-virulence strains of Francisella tularensis. Vaccine 2001; 19:4465-72. [PMID: 11483272 DOI: 10.1016/s0264-410x(01)00189-x] [Citation(s) in RCA: 139] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mice immunised with lipopolysaccharide (LPS) from Francisella tularensis were protected against challenge with the live vaccine strain (LVS). However, when similarly immunised mice were challenged using the fully virulent F. tularensis strain Schu4, only an increase in the time to death was observed. Passive transfer of serum from LPS-immunised mice to naive mice afforded protection against F. tularensis LVS. LPS-immunised mice depleted of either CD4+ or CD8+ T-cells survived a F. tularensis LVS challenge although the rate of clearance of bacteria from the spleen was significantly reduced in the CD8+ depleted group. LPS-immunised mice boosted with F. tularensis LVS were re-challenged with F. tularensis Schu4. This cohort was significantly protected (LD(50) increased from <1 to >1000 CFU). However, passive transfer of serum did not confer protection and mice depleted of CD4+ or CD8+ T-cells did not survive.
Collapse
Affiliation(s)
- M Fulop
- Defence Evaluation and Research Agency, CBD, Porton Down, Wiltshire SP4 0JQ, Salisbury, UK
| | | | | | | |
Collapse
|
104
|
Bosio CM, Elkins KL. Susceptibility to secondary Francisella tularensis live vaccine strain infection in B-cell-deficient mice is associated with neutrophilia but not with defects in specific T-cell-mediated immunity. Infect Immun 2001; 69:194-203. [PMID: 11119506 PMCID: PMC97872 DOI: 10.1128/iai.69.1.194-203.2001] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previous studies have demonstrated a role for B cells, not associated with antibody production, in protection against lethal secondary infection of mice with Francisella tularensis live vaccine strain (LVS). However, the mechanism by which B cells contribute to this protection is not known. To study the specific role of B cells during secondary LVS infection, we developed an in vitro culture system that mimics many of the same characteristics of in vivo infection. Using this culture system, we showed that B cells do not directly control LVS infection but that control of LVS growth is mediated primarily by LVS-primed T cells. Importantly, B cells were not required for the generation of effective memory T cells since LVS-primed, B-cell-deficient (BKO) mice generated CD4(+) and CD8(+) T cells that controlled LVS infection similarly to LVS-primed CD4(+) and CD8(+) T cells from wild-type mice. The control of LVS growth appeared to depend primarily on gamma interferon and nitric oxide and was similar in wild-type and BKO mice. Rather, the inability of BKO mice to survive secondary LVS infection was associated with marked neutrophil influx into the spleen very early after challenge. The neutrophilia was directly associated with B cells, since BKO mice reconstituted with naive B cells prior to a secondary challenge with LVS had decreased bacterial loads and neutrophils in the spleen and survived.
Collapse
Affiliation(s)
- C M Bosio
- Laboratory of Mycobacteria, Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics, Evaluation, and Research, Food and Drug Administration, Rockville, Maryland 20852, USA
| | | |
Collapse
|
105
|
Masihi KN. Immunomodulators in infectious diseases: panoply of possibilites. INTERNATIONAL JOURNAL OF IMMUNOPHARMACOLOGY 2000; 22:1083-91. [PMID: 11137615 DOI: 10.1016/s0192-0561(00)00074-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Infections which caused ravages in the past centuries are again resurgent and newly emerging pathogens capable of human diseases continue to surface. Multidrug antibiotic resistance has turned into a major medical problem. Judicious concepts for combating infections in the 21st century have acquired a new poignancy. Immunomodulators of natural, synthetic, and recombinant origin can stimulate host defense mechanisms for the prophylaxis and treatment of diverse viral, bacterial, parasitic and fungal diseases. Some immunomodulator preparations are already licensed for use in patients and numerous others are being extensively investigated in preclinical and clinical studies. Immunomodulators offer a novel adjunct to established antimicrobial therapies.
Collapse
Affiliation(s)
- K N Masihi
- Robert Koch Institute, Nordufer 20, D-13353, Berlin, Germany.
| |
Collapse
|
106
|
Dreisbach VC, Cowley S, Elkins KL. Purified lipopolysaccharide from Francisella tularensis live vaccine strain (LVS) induces protective immunity against LVS infection that requires B cells and gamma interferon. Infect Immun 2000; 68:1988-96. [PMID: 10722593 PMCID: PMC97377 DOI: 10.1128/iai.68.4.1988-1996.2000] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previous results have demonstrated that nonspecific protective immunity against lethal Francisella tularensis live vaccine strain (LVS) or Listeria monocytogenes infection can be stimulated either by sublethal infection with bacteria or by treatment with bacterial DNA given 3 days before lethal challenge. Here we characterize the ability of purified lipopolysaccharide (LPS) from F. tularensis LVS to stimulate similar early protective immunity. Treatment of mice with surprisingly small amounts of LVS LPS resulted in very strong and long-lived protection against lethal LVS challenge within 2 to 3 days. Despite this strong protective response, LPS purified from F. tularensis LVS did not activate murine B cells for proliferation or polyclonal immunoglobulin secretion, nor did it activate murine splenocytes for secretion of interleukin-4 (IL-4), IL-6, IL-12, or gamma interferon (IFN-gamma). Immunization of mice with purified LVS LPS induced a weak specific anti-LPS immunoglobulin M (IgM) response and very little IgG; however, infection of mice with LVS bacteria resulted in vigorous IgM and IgG, particularly IgG2a, anti-LPS antibody responses. Studies using various immunodeficient mouse strains, including LPS-hyporesponsive C3H/HeJ mice, muMT(-) (B-cell-deficient) knockout mice, and IFN-gamma-deficient mice, demonstrated that the mechanism of protection does not involve recognition through the Lps(n) gene product; nonetheless, protection was dependent on B cells as well as IFN-gamma.
Collapse
Affiliation(s)
- V C Dreisbach
- Laboratory of Mycobacteria, Division of Bacterial Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, Maryland 20852, USA
| | | | | |
Collapse
|
107
|
Abstract
The advent of the antibiotic era ushered in a shift towards non-pathogen-specific therapy of infectious diseases. This led to an overt emphasis on targeting microbial pathogens while strategies directed towards enhancing host immunity were neglected. In an effort to decrease sole reliance on antimicrobials, the time has come for a critical reappraisal of nonantibiotic, albeit immune response-enhancing substances. The diverse array of natural, synthetic, and recombinant immunomodulators discussed in this review succinctly demonstrate the potential of these agents to stimulate host defense mechanisms for prophylaxis and treatment of various microbial infections.
Collapse
Affiliation(s)
- K N Masihi
- Robert Koch Institute, Nordufer 20, D-13353, Berlin, Germany.
| |
Collapse
|
108
|
Elkins KL, Bosio CM, Rhinehart-Jones TR. Importance of B cells, but not specific antibodies, in primary and secondary protective immunity to the intracellular bacterium Francisella tularensis live vaccine strain. Infect Immun 1999; 67:6002-7. [PMID: 10531260 PMCID: PMC96986 DOI: 10.1128/iai.67.11.6002-6007.1999] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although there appears to be little if any role for specific antibodies in protection against intracellular bacteria, such as the model pathogen F. tularensis live vaccine strain (LVS), the role of B cells themselves in primary and secondary infection with such bacteria has not been examined directly. We show here that mice deficient in mature B cells and antibodies (B-cell knockout mice) are marginally compromised in controlling primary sublethal infection but are 100-fold less well protected against secondary lethal challenge than are their normal counterparts. This defect in optimal specific protective immunity was readily reconstituted by the transfer of primed, and to a lesser degree, unprimed B cells, but not by the transfer of specific antibodies. The results indicate a previously unappreciated role for B cells in secondary immunity to intracellular pathogens through a function other than antibody production.
Collapse
Affiliation(s)
- K L Elkins
- Laboratory of Mycobacteria, Division of Bacterial Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, Maryland 20852, USA.
| | | | | |
Collapse
|
109
|
Stenmark S, Sunnemark D, Bucht A, Sjöstedt A. Rapid local expression of interleukin-12, tumor necrosis factor alpha, and gamma interferon after cutaneous Francisella tularensis infection in tularemia-immune mice. Infect Immun 1999; 67:1789-97. [PMID: 10085019 PMCID: PMC96529 DOI: 10.1128/iai.67.4.1789-1797.1999] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Francisella tularensis LVS is an effective live vaccine strain used for cutaneous vaccination against tularemia in man. In mice, injection of LVS causes invasive disease and subsequent development of immunity that is characterized by effective control of otherwise lethal doses of the organism. In the present investigation, it is shown that LVS-immune mice controlled an intradermal infection much more effectively than did naive mice; bacterial counts in skin samples were 1.5 to 2.0 log10 lower 24 h after injection and 6 log10 lower 72 h after injection in immune mice. Moreover, in contrast to naive mice, no bacteria were demonstrated in samples from livers and spleens of immune mice. By immunohistochemistry, skin samples from immune mice showed an intense staining for interleukin-12 (IL-12) and a moderate staining for tumor necrosis factor alpha (TNF-alpha) at 24 h postinoculation, after which staining for both cytokines faded. In naive mice, the staining for IL-12 was weak at all time points and no staining for TNF-alpha was observed. No staining for gamma interferon (IFN-gamma) was observed in any group before 72 h. At that time point, skin samples from immune mice showed moderate staining and skin samples from naive mice showed weak staining. Reverse transcriptase PCR showed an induction of mRNA of the three cytokines in the skin within the first day after injection. A quantitative analysis demonstrated higher IFN-gamma and TNF-alpha mRNA levels in immune mice at 24 h postinoculation. In conclusion, immunization with F. tularensis LVS conferred a capability to respond to cutaneous reinfection, with rapid local expression of IL-12, TNF-alpha, and IFN-gamma, and this expression was paralleled by containment and mitigation of the infection. The cytokine response may be part of a local barrier function of the skin, important to host protection against tularemia.
Collapse
Affiliation(s)
- S Stenmark
- Department of Infectious Diseases, University of Umeâ, Stockholm, Sweden
| | | | | | | |
Collapse
|
110
|
Scharton-Kersten T, Contursi C, Masumi A, Sher A, Ozato K. Interferon consensus sequence binding protein-deficient mice display impaired resistance to intracellular infection due to a primary defect in interleukin 12 p40 induction. J Exp Med 1997; 186:1523-34. [PMID: 9348310 PMCID: PMC2199126 DOI: 10.1084/jem.186.9.1523] [Citation(s) in RCA: 175] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/1997] [Revised: 08/15/1997] [Indexed: 02/05/2023] Open
Abstract
Mice lacking the transcription factor interferon consensus sequence binding protein (ICSBP), a member of the interferon regulatory factor family of transcription proteins, were infected with the intracellular protozoan, Toxoplasma gondii. ICSBP-deficient mice exhibited unchecked parasite replication in vivo and rapidly succumbed within 14 d after inoculation with an avirulent Toxoplasma strain. In contrast, few intracellular parasites were observed in wild-type littermates and these animals survived for at least 60 d after infection. Analysis of cytokine synthesis in vitro and in vivo revealed a major deficiency in the expression of both interferon (IFN)-gamma and interleukin (IL)-12 p40 in the T. gondii exposed ICSBP-/- animals. In related experiments, macrophages from uninfected ICSBP-/- mice were shown to display a selective impairment in the mRNA expression of IL-12 p40 but not IL-1alpha, IL-1beta, IL-1Ra, IL-6, IL-10, or TNF-alpha in response to live parasites, parasite antigen, lipopolysaccharide, or Staphylococcus aureus. This selective defect in IL-12 p40 production was observed regardless of whether the macrophages had been primed with IFN-gamma. We hypothesize that the impaired synthesis of IL-12 p40 in ICSBP-/- animals is the primary lesion responsible for the loss in resistance to T. gondii because IFN-gamma-induced parasite killing was unimpaired in vitro and, more importantly, administration of exogenous IL-12 in vivo significantly prolonged survival of the infected mice. Together these findings implicate ICSBP as a major transcription factor which directly or indirectly regulates IL-12 p40 gene activation and, as a consequence, IFN-gamma-dependent host resistance.
Collapse
Affiliation(s)
- T Scharton-Kersten
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892-2753, USA
| | | | | | | | | |
Collapse
|
111
|
Abstract
Interleukin-12 (IL-12) is a potent immunoregulatory cytokine that is crucially involved in a wide range of infectious diseases. In several experimental models of bacterial, parasitic, viral, and fungal infection, endogenous IL-12 is required for early control of infection and for generation and perhaps maintenance of acquired protective immunity, directed by T helper type 1 (Th1) cells and mediated by phagocytes. Although the relative roles of IL-12 and gamma interferon in Th1-cell priming may be to a significant extent pathogen dependent, common to most infections is that IL-12 regulates the magnitude of the gamma interferon response at the initiation of infection, thus potentiating natural resistance, favoring Th1-cell development; and inhibiting Th2 responses. Treatment of animals with IL-12, either alone or as a vaccine adjuvant, has been shown to prevent disease by many of the same infectious agents, by stimulating innate resistance or promoting specific reactivity. Although IL-12 may enhance protective memory responses in vaccination or in combination with antimicrobial chemotherapy, it is yet unclear whether exogenous IL-12 can alter established responses in humans. Continued investigation into the possible application of IL-12 therapy to human infections is warranted by the role of the cytokine in inflammation, immunopathology, and autoimmunity.
Collapse
Affiliation(s)
- L Romani
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Italy.
| | | | | |
Collapse
|
112
|
Hernychova L, Kovarova H, Macela A, Kroca M, Krocova Z, Stulik J. Early consequences of macrophage-Francisella tularensis interaction under the influence of different genetic background in mice. Immunol Lett 1997; 57:75-81. [PMID: 9232429 DOI: 10.1016/s0165-2478(97)00063-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The induction, regulation and expression of protective immunity against Francisella tularensis LVS infection is dependent on the results of primary interaction between the cells of host's immunoregulatory system and the microbe. The early events, at least on the side of macrophages, are under the genetic control. To determine the impact of genes that might be involved in the control of resistance to Francisella tularensis LVS infection, we have used three different inbred strains of mice with increasing resistance to this infection in order C3H/HeJ (Lpsd), C3H/HeN (Lpsn"), and C57B1/10N (Lpsn"). The controlled production of IL-10, IFN-gamma, and TNF-alpha coupled with increased production of reactive oxygen metabolites during early phase of infection distinguished less susceptible C3H/HeN mice from their more susceptible cogenic C3H/HeJ counterparts. The enhancement of oxidative metabolism that appeared on day 5 after the infection of both C3H/HeN and C57B1/10N mice closely correlated with increasing resistance of these two strains of mice to Francisella tularensis LVS infection. These mice were also capable to reach the highest level of TNF-alpha on day 5 after the infection. At the same time interval, only C57B1/10N mice produced significantly enhanced level of nitric oxide. Overall, these parameters may suggest their possible biological role in early-phase resistance to Francisella tularensis LVS infection and their subsequent consequences for ultimate control of infection and its clearance.
Collapse
Affiliation(s)
- L Hernychova
- Institute for Immunology, Purkyne Military Medical Academy, Hradec Kralove, Czech Republic
| | | | | | | | | | | |
Collapse
|
113
|
Elkins KL, Rhinehart-Jones TR, Culkin SJ, Yee D, Winegar RK. Minimal requirements for murine resistance to infection with Francisella tularensis LVS. Infect Immun 1996; 64:3288-93. [PMID: 8757866 PMCID: PMC174220 DOI: 10.1128/iai.64.8.3288-3293.1996] [Citation(s) in RCA: 142] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Intraperitoneal or intravenous infection of mice with Francisella tularensis LVS is lethal, with an intraperitoneal 50% lethal dose (LD50) approaching a single bacterium. Intradermal (i.d.) LVS infection has a much higher LD50, about 10(6) bacteria in BALB/cByJ mice, and survival of i.d. infection leads to solid generation of immunity against lethal challenge. To define the minimal requirements for both initial and long-term survival of i.d. infection, we characterized the nature of i.d. LVS infection in lymphocyte-deficient BALB/cByJ.scid (scid) mice. scid mice infected i.d. with strain LVS survived for about 20 days and then died from overwhelming disseminated infection. However, scid mice treated with monoclonal antibodies to gamma interferon, tumor necrosis factor alpha, or neutrophils-granulocytes all died within 1 week of infection, indicating that these were essential for early control of infection. Studies using GKO (gamma interferon knockout) mice emphasized that gamma interferon is absolutely required for initial survival of i.d. LVS infection. scid mice could be reconstituted for long-term survival of i.d. LVS infection and clearance of bacteria by intravenous transfer of splenic lymphocytes or purified B220-/T+ lymphocytes but not nu/nu lymphocytes. T cells are therefore required for long-term clearance and survival of i.d. LVS infection; efforts to determine whether CD4+ T cells, CD8+ T cells, or both are involved are ongoing.
Collapse
Affiliation(s)
- K L Elkins
- Laboratory of Enteric and Sexually Transmitted Diseases, Division of Bacterial Products, Center for Biologics Evaluation and Research, Rockville, Maryland 20852, USA
| | | | | | | | | |
Collapse
|
114
|
Sjöstedt A, North RJ, Conlan JW. The requirement of tumour necrosis factor-alpha and interferon-gamma for the expression of protective immunity to secondary murine tularaemia depends on the size of the challenge inoculum. MICROBIOLOGY (READING, ENGLAND) 1996; 142 ( Pt 6):1369-1374. [PMID: 8704976 DOI: 10.1099/13500872-142-6-1369] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The present study was conducted to determine the extent to which the cytokines tumour necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma) are required to protect against primary or secondary murine tularaemia caused by the live vaccine strain of the facultative intracellular bacterium Francisella tularensis. It is shown that non-immune mice treated with neutralizing monoclonal antibodies (mAbs) against TNF-alpha and IFN-gamma are rendered defenceless against otherwise sublethal intravenous inocula of the bacterium. Treatment with either of the anti-cytokine mAbs resulted in even a very small inoculum of 500 c.f.u. of the pathogen multiplying unrestrictedly in the livers, spleens and lungs of non-immune mice to rapidly reach lethal numbers. By contrast, Francisella-immune mice treated with either of the mAbs remained capable of resolving secondary infection with 50-fold larger inocula. However, the need for TNF-alpha and IFN-gamma for controlling secondary tularaemia became critical when challenge inocula exceeded 10(6) c.f.u. Overall, the results imply that different defence mechanisms operate to control primary versus secondary murine tularaemia. Additionally, they show that the need for TNF-alpha and IFN-gamma to combat secondary infection depends on the size of the challenge inoculum.
Collapse
Affiliation(s)
- Anders Sjöstedt
- Department of Infectious Diseases, University of Umeå, S-901 87 Umeå, Sweden
- Department of Microbiology, National Defence Research Establishment, S-901 82 Umeå, Sweden
- Trudeau Institute, Inc., Saranac Lake, NY 12983, USA
| | | | | |
Collapse
|
115
|
Mokhtarian F, Wesselingh SL, Choi S, Maeda A, Griffin DE, Sobel RA, Grob D. Production and role of cytokines in the CNS of mice with acute viral encephalomyelitis. J Neuroimmunol 1996; 66:11-22. [PMID: 8964904 DOI: 10.1016/0165-5728(95)00181-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Semliki Forest Virus (SFV) causes a more severe acute encephalomyelitis in B6 than in SJL mice despite similar T cell proliferation and antibody responses in these two strains. To determine the immunological mechanisms that may contribute to this difference, CNS tissues from SFV-infected B6 and SJL mice were analyzed for viral replication, inflammatory responses and cytokine production, by semiquantitative reverse transcriptase-PCR and immunohistochemistry. Although initially similar on day 2 p.i., SFV replicated to higher viral titers in B6 than SJL mice on days 4 and 7 p.i. Infectious virus was cleared from both strains by day 10 p.i. There were no differences in numbers of CD4+, CD8+ or MHC class I and II+ inflammatory cells at any time point. Higher levels of IL-4 mRNA, lower levels of TNF-alpha, IL-6, IL-1 beta and IL-2 mRNAs and lower IL-2+ and IFN-gamma+ cells were found in B6. These findings suggest that despite comparable immune responses, different patterns of cytokine production correlated with higher levels of virus in the brains and more severe clinical disease in B6, and more efficient clearance of virus and less severe disease in SJL mice.
Collapse
Affiliation(s)
- F Mokhtarian
- Department of Medicine, SUNY Health Science Center/Maimonides Medical Center, Brooklyn, 11219 USA.
| | | | | | | | | | | | | |
Collapse
|
116
|
Sjöstedt A, Tärnvik A, Sandström G. Francisella tularensis: host-parasite interaction. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 1996; 13:181-4. [PMID: 8861025 DOI: 10.1111/j.1574-695x.1996.tb00233.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- A Sjöstedt
- Department of Microbiology, National Defence Research Establishment, Umeå, Sweden
| | | | | |
Collapse
|
117
|
Fulop M, Manchee R, Titball R. Role of lipopolysaccharide and a major outer membrane protein from Francisella tularensis in the induction of immunity against tularemia. Vaccine 1995; 13:1220-5. [PMID: 8578807 DOI: 10.1016/0264-410x(95)00062-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
A crude outer membrane preparation from Francisella tularensis Live Vaccine Strain (LVS) was used to immunize mice. Immunized mice were completely protected from a F. tularensis challenge. We evaluated the role of two major outer membrane antigens in the induction of protective immunity, namely lipopolysaccharide and an outer membrane protein FopA. We presented FopA to the immune system using an aromatic amino acid-dependent Salmonella typhimurium as a vector. Although mice mounted an immune response to cloned FopA no significant protection was induced. However, LPS immunized mice were completely protected. We conclude that LPS is a major protective antigen whereas FopA has a limited or no role in the induction of protective immunity.
Collapse
Affiliation(s)
- M Fulop
- Chemical and Biological Defence Establishment, Porton Down, Salisbury, Wiltshire, UK
| | | | | |
Collapse
|
118
|
Krakauer T. Levels of interleukin 6 and tumor necrosis factor in serum from humans vaccinated with live, attenuated Francisella tularensis. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 1995; 2:487-8. [PMID: 7583930 PMCID: PMC170185 DOI: 10.1128/cdli.2.4.487-488.1995] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The levels of interleukin 6 (IL-6) and tumor necrosis factor alpha in serum did not change significantly during the course of immunization with the live vaccine strain of Francisella tularensis. Higher levels of circulating IL-6 were found in the sera of vaccinees with good antibody responses than in the sera from nonresponders. Preimmunization levels of IL-6 in serum were also higher in responders than in nonresponders.
Collapse
Affiliation(s)
- T Krakauer
- Applied Research Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland 21702, USA
| |
Collapse
|
119
|
Nakane A, Okamoto M, Asano M, Kohanawa M, Minagawa T. Endogenous gamma interferon, tumor necrosis factor, and interleukin-6 in Staphylococcus aureus infection in mice. Infect Immun 1995; 63:1165-72. [PMID: 7890367 PMCID: PMC173129 DOI: 10.1128/iai.63.4.1165-1172.1995] [Citation(s) in RCA: 98] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The production and roles of endogenous gamma interferon (IFN-gamma), tumor necrosis factor (TNF), and interleukin-6 (IL-6) in both lethal and nonlethal infections of Staphylococcus aureus were investigated in mice. In the case of nonlethal infection, although no bacteria were detected in the bloodstreams, bacteria that colonized and proliferated persistently for 3 weeks were found in the kidneys. All mice given lethal injections died within 7 days, and large numbers of bacteria were detected in the bloodstreams, spleens, and kidneys. The first peaks of IFN-gamma, TNF, and IL-6 were observed in the bloodstreams and spleens of the mice with nonlethal and lethal infections within 24 h. Thereafter, in the nonlethal cases, IFN-gamma, TNF, and IL-6 peaked again in the spleens and kidneys during the period of maximum growth of bacteria in the kidneys, although only IL-6 was detected in the sera. In contrast, in the case of lethal infection, the titers of IFN-gamma and IL-6 in the sera and TNF in the kidneys peaked before death. Effects of in vivo administration of monoclonal antibodies (MAbs) against IFN-gamma and TNF on the fates of S. aureus-infected mice were studied. In the nonlethal infections, anti-TNF alpha (anti-TNF-alpha) MAb-treated mice, but not anti-IFN-gamma MAb-treated mice, died as a result of worsening infection, suggesting that endogenous TNF plays a protective role in host resistance to S. aureus infection. In the mice that received lethal doses, injection of anti-TNF-alpha MAb accelerated death. However, although injection of anti-IFN-gamma MAb inhibited host resistance of the infected mice early in infection, most of the animals survived the lethal infection by injection of anti-IFN-gamma MAb, suggesting that endogenous IFN-gamma plays a detrimental role in S. aureus infection. Thus, this study demonstrated that IFN-gamma and TNF play different roles in S. aureus infection.
Collapse
Affiliation(s)
- A Nakane
- Department of Bacteriology, Hirosaki University School of Medicine, Japan
| | | | | | | | | |
Collapse
|
120
|
Golovliov I, Sandström G, Ericsson M, Sjöstedt A, Tärnvik A. Cytokine expression in the liver during the early phase of murine tularemia. Infect Immun 1995; 63:534-8. [PMID: 7822019 PMCID: PMC173028 DOI: 10.1128/iai.63.2.534-538.1995] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Cytokine expression was determined in the livers of mice inoculated subcutaneously with Francisella tularensis LVS. During the first 48 h of infection, there was a logarithmic increase of bacteria in the liver, with a doubling time of 2.5 h. Within 48 h, tumor necrosis factor alpha (TNF-alpha), interleukin 10 (IL-10), IL-12, and gamma interferon (IFN-gamma) mRNAs were expressed, and production of TNF-alpha and IFN-gamma was demonstrated. There was no expression within 96 h of mRNA from IL-2, IL-3, or IL-4. After subcutaneous inoculation of heat-killed LVS, no expression of any of the cytokine mRNAs and no increase in the levels of TNF-alpha or IFN-gamma occurred. The expression of TNF-alpha, IL-12, and IFN-gamma is held to be important to evoke an early T-cell-independent host defense against F. tularensis as well as to drive the expansion of a protective Th1 cell response.
Collapse
Affiliation(s)
- I Golovliov
- Department of Microbiology, National Defence Research Establishment, Umeå, Sweden
| | | | | | | | | |
Collapse
|
121
|
Conlan JW, Sjöstedt A, North RJ. CD4+ and CD8+ T-cell-dependent and -independent host defense mechanisms can operate to control and resolve primary and secondary Francisella tularensis LVS infection in mice. Infect Immun 1994; 62:5603-7. [PMID: 7960142 PMCID: PMC303308 DOI: 10.1128/iai.62.12.5603-5607.1994] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Immunity to experimental infection with the facultative intracellular bacterium Francisella tularensis is generally considered an example of T-cell-mediated, macrophage-expressed immunity. However, the results of the present study indicate that T-cell-independent mechanisms are also important in anti-Francisella defense. They show that mice selectively depleted of CD4+, CD8+, or both T-cell populations by treatment with T-cell subset-specific monoclonal antibodies remained capable of controlling and partly resolving a primary sublethal Francisella infection. Similarly, it was found that Francisella-immune mice depleted of either or both subsets of T cells retain a high degree of acquired immunity to reinfection. Together, these findings imply that resistance to primary and secondary tularemia can be mediated by cells other than CD4+ and CD8+ T cells.
Collapse
Affiliation(s)
- J W Conlan
- Trudeau Institute, Inc., Saranac Lake, New York 12983
| | | | | |
Collapse
|
122
|
Anthony LS, Cowley SC, Mdluli KE, Nano FE. Isolation of a Francisella tularensis mutant that is sensitive to serum and oxidative killing and is avirulent in mice: Correlation with the loss of MinD homologue expression. FEMS Microbiol Lett 1994. [DOI: 10.1111/j.1574-6968.1994.tb07278.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
123
|
Shirahata T, Yamashita T, Ohta C, Goto H, Nakane A. CD8+ T lymphocytes are the major cell population involved in the early gamma interferon response and resistance to acute primary Toxoplasma gondii infection in mice. Microbiol Immunol 1994; 38:789-96. [PMID: 7869956 DOI: 10.1111/j.1348-0421.1994.tb01858.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Gamma interferon (IFN-gamma) is known to be a major mediator influencing host defense against Toxoplasma (T.) gondii. To evaluate lymphocyte populations involved in this cytokine-mediated early resistance to T. gondii, the effects of in vivo administration of monoclonal antibodies (MAbs) against T-cell subsets and anti-asialo GM1 antibody on the course of infection and IFN-gamma response were investigated in mice infected acutely with this parasitic protozoan. A single injection of anti-CD8 MAb on day -1 or day 4 severely exacerbated the infection, in accordance with a marked suppression of endogenous IFN-gamma production. Moreover, the administration of anti-IFN-gamma MAb on day 0 but not later than day 4 resulted in a total abrogation of resistance to T. gondii, suggesting that endogenous IFN-gamma produced during the first several days of infection is critical for the generation of antitoxoplasmal resistance in mice. In contrast, no significant increase in mortality was observed when injected with either anti-CD4 MAb or anti-asialo GM1 antibody on day -1, while these antibodies reduced significantly the ability of mice to produce IFN-gamma. Indeed, simultaneous depletion of CD4+ and CD8+ cells had no greater suppressive effect on host defense and endogenous IFN-gamma production than depletion of CD8+ cells alone. Together, these results suggest that CD8+ T cells play a central role for resolution of acute toxoplasmosis by participating in endogenous IFN-gamma production. The possible role of early produced IFN-gamma in the development of protective immune response to T. gondii is also discussed.
Collapse
Affiliation(s)
- T Shirahata
- Department of Veterinary Microbiology, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| | | | | | | | | |
Collapse
|
124
|
Yamamoto Y, Okubo S, Klein TW, Onozaki K, Saito T, Friedman H. Binding of Legionella pneumophila to macrophages increases cellular cytokine mRNA. Infect Immun 1994; 62:3947-56. [PMID: 8063412 PMCID: PMC303052 DOI: 10.1128/iai.62.9.3947-3956.1994] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Infection of macrophages with Legionella pneumophila induces formation of interleukin 1 beta (IL-1 beta), but the molecular basis of this is not understood. Binding of bacteria to macrophage surfaces is the first step in an infection process. Therefore, we examined whether this step was sufficient to increase the cellular level of mRNAs for IL-1 beta and other cytokines. To assess the effect of binding of L. pneumophila on the steady-state levels of cytokine mRNAs, cultures of thioglycolate-elicited macrophages from L. pneumophila-susceptible A/J mice were treated with cytochalasin D and infected with L. pneumophila and the total RNA was extracted for analysis by reverse transcription-PCR with primers for IL-1 alpha, IL-1 beta, IL-6, tumor necrosis factor alpha, granulocyte macrophage colony-stimulating factor, and beta interferon (IFN-beta). L. pneumophila treatment increased the cellular steady-state mRNA levels of all cytokines except IFN-beta. To determine the specificity of this effect, macrophage cultures were treated with cytochalasin D and either bacterial lipopolysaccharide, bovine serum albumin-sensitized latex, Salmonella typhimurium, or Escherichia coli. Lipopolysaccharide treatment increased all mRNAs, bovine serum albumin-sensitized latex had no significant effect, and treatment with S. typhimurium or E. coli increased all mRNAs except that of IFN-beta. These results suggested that the binding of gram-negative bacteria to the macrophage surface was sufficient to induce a unique pattern of cytokine mRNAs. Additional studies that examined the characteristics of the bacterial ligands involved indicated involvement of both heat-labile and heat-stable surface ligands.
Collapse
Affiliation(s)
- Y Yamamoto
- Department of Medical Microbiology and Immunology, University of South Florida College of Medicine, Tampa 33612
| | | | | | | | | | | |
Collapse
|
125
|
Rhinehart-Jones TR, Fortier AH, Elkins KL. Transfer of immunity against lethal murine Francisella infection by specific antibody depends on host gamma interferon and T cells. Infect Immun 1994; 62:3129-37. [PMID: 8039881 PMCID: PMC302937 DOI: 10.1128/iai.62.8.3129-3137.1994] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Both serum and spleen cells from mice immune to Francisella tularensis transfer protection to naive recipients. Here we characterize the mechanism of protection induced by transfer of immune mouse serum (IMS). IMS obtained 4 weeks after intradermal infection with 10(3) bacteria of the live vaccine strain (LVS) contained high levels of immunoglobulin G2 (IgG2a) and IgM (end point titers, 1:16,600 and 1:7,200, respectively) and little IgG1, IgG2b, or IgG3. LVS-specific antibodies were detected 5 days after intradermal infection, and reached peak levels by 2 weeks postinfection. Only sera obtained 10 days or more after sublethal infection, when IgG titers peaked, transferred protection against a challenge of 100 50% lethal doses (LD50s). Purified high-titer IgG anti-LVS antibody but not IgM anti-LVS antibody was responsible for transfer of protection against an intraperitoneal challenge of up to 3,000 LD50s. IMS had no direct toxic effects on LVS and did not affect uptake or growth of bacteria in association with peritoneal cells. One day after LVS infection, liver, spleen, and lung tissue from mice treated with IMS contained 1 to 2 log units fewer bacteria than did tissue from mice treated with normal mouse serum or phosphate-buffered saline. Between 2 and 4 days after infection, however, bacterial growth rates in tissues were similar in both serum-protected mice and unprotected mice. Bacterial burdens in IMS-treated, LVS-infected mice declined in infected tissues after day 5, whereas control animals died. This lag phase suggested that development of a host response was involved in complete bacterial clearance. In fact, transfer of IMS into normal recipients that were simultaneously treated with anti-gamma interferon and challenged with LVS did not protect mice from death. Further, transfer of IMS into athymic nu/nu mice did not protect against LVS challenge; protection was, however, reconstituted by transfer of normal T cells into nu/nu mice. Thus, "passive" transfer of protection against LVS with specific antibody is not passive but depends on a host T-cell response to promote clearance of systemic infection and protection against lethal disease.
Collapse
Affiliation(s)
- T R Rhinehart-Jones
- Department of Cellular Immunology, Walter Reed Army Institute of Research, Rockville, Maryland 20850
| | | | | |
Collapse
|
126
|
Kogut MH, McGruder ED, Hargis BM, Corrier DE, DeLoach JR. Dynamics of avian inflammatory response to Salmonella-immune lymphokines. Changes in avian blood leukocyte populations. Inflammation 1994; 18:373-88. [PMID: 7982728 DOI: 10.1007/bf01534435] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Investigations in our laboratories have indicated that an increased resistance to SE organ infectivity in chicks was conferred by the immunoprophylactic administration of SE-immune lymphokines (SE-ILK). This resistance was associated with an increase in the lamina propria thickness due to a marked infiltration of inflammatory polymorphonuclear cells (PMNs). In the present study, we determined whether the hematological profile of SE-ILK-treated chicks might reflect changes that are associated with the protection against organ invasion by SE. As protection has been observed in previous studies within 24 h of SE-ILK administration, we evaluated alterations in the circulating leukocyte profile in 1-day-old Leghorn chicks during this time period. We also determined whether the alterations in the peripheral blood leukocytes correlated with the increased protection against SE organ invasion induced by the SE-ILK. Within 4 h after an intraperitoneal injection of SE-ILK and challenge with SE, the number of circulating leukocytes increased significantly (P < 0.05) from all of the other treatment groups. The number of circulating PMNs was found to account for more than 80% of the increase in the number of circulating leukocytes. Using correlation analysis, we found a strong association between the number of circulating PMNs and the protection induced by SE-ILK against SE organ invasion. These studies associate the expansion of the available pool of circulating PMNs and the expression of innate resistance to organ invasion by SE.
Collapse
Affiliation(s)
- M H Kogut
- United States Department of Agriculture, Agricultural Research Service,Food Animal Protection Research Laboratory, Texas 77845
| | | | | | | | | |
Collapse
|
127
|
Sjöstedt A, Conlan JW, North RJ. Neutrophils are critical for host defense against primary infection with the facultative intracellular bacterium Francisella tularensis in mice and participate in defense against reinfection. Infect Immun 1994; 62:2779-83. [PMID: 8005668 PMCID: PMC302881 DOI: 10.1128/iai.62.7.2779-2783.1994] [Citation(s) in RCA: 112] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
It is generally believed that immunity to experimental infection with the facultative intracellular bacterium Francisella tularensis is an example of T-cell-mediated immunity that is expressed by activated macrophages and mediated by Francisella-specific T cells. According to the results presented herein, neutrophils are also essential for defense against primary infection with this organism. It is shown that mice depleted of neutrophils by treatment with the granulocyte-specific monoclonal antibody RB6-8C5 are rendered defenseless against otherwise sublethal doses of F. tularensis LVS inoculated intravenously or intradermally. In neutrophil-depleted mice, the organism grew progressively in the livers, spleens, and lungs to reach lethal numbers, whereas infection was resolved in normal mice. Although neutrophils were found to resistance to reinfection, their participation was less important. The results suggest that neutrophils are needed for defense against primary infection because they serve to restrict the growth of F. tularensis before it reaches numbers capable of overwhelming a developing specific immune response. The exact way that neutrophils achieve this is not clear at this time, although it is probable that they contribute in ways other than by ingesting and killing the bacterium.
Collapse
Affiliation(s)
- A Sjöstedt
- Trudeau Institute, Inc., Saranac Lake, New York 12983
| | | | | |
Collapse
|
128
|
Autenrieth IB, Beer M, Bohn E, Kaufmann SH, Heesemann J. Immune responses to Yersinia enterocolitica in susceptible BALB/c and resistant C57BL/6 mice: an essential role for gamma interferon. Infect Immun 1994; 62:2590-9. [PMID: 8188382 PMCID: PMC186549 DOI: 10.1128/iai.62.6.2590-2599.1994] [Citation(s) in RCA: 149] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Susceptibility of mice to infection with Yersinia enterocolitica has been shown to be related to neither the Ity locus encoding for resistance to Salmonella typhimurium and other pathogens nor the H-2 locus. Recent studies in our laboratory have demonstrated that T-cell-mediated immune responses are required for overcoming primary Yersinia infection. In the present study, we investigated the course of infection with Y. enterocolitica and the resulting immune responses in Yersinia-susceptible BALB/c and Yersinia-resistant C57BL/6 mice. In the early phase of infection, the clearance of the pathogen was comparable in both strains of mice, suggesting similar mechanisms of innate resistance. Splenic T cells from Yersinia-infected C57BL/6 mice exhibited marked proliferative responses and produced gamma interferon (IFN-gamma) upon exposure to heat-killed yersiniae. By contrast, the Yersinia-specific T-cell response in BALB/c mice was weak, and IFN-gamma production could not be detected before day 21 postinfection. T cells isolated from C57BL/6 mice 7 days after infection mediated immunity to Y. enterocolitica but those from BALB/c mice did not, while at 21 days postinfection T cells from both strains mediated protection. Neutralization of IFN-gamma abrogated resistance to yersiniae in C57BL/6 mice but to a far smaller extent in BALB/c mice. Administration of recombinant IFN-gamma or anti-interleukin-4 antibodies rendered BALB/c mice resistant to yersiniae, whereas this treatment did not significantly affect the course of the infection in C57BL/6 mice. These results indicate that the cellular immune response, in particular the production of IFN-gamma by Yersinia-specific T cells, is associated with resistance of mice to Y. enterocolitica.
Collapse
Affiliation(s)
- I B Autenrieth
- Institute for Hygiene and Microbiology, University of Würzburg, Federal Republic of Germany
| | | | | | | | | |
Collapse
|
129
|
Denich K, Börlin P, O'Hanley PD, Howard M, Heath AW. Expression of the murine interleukin-4 gene in an attenuated aroA strain of Salmonella typhimurium: persistence and immune response in BALB/c mice and susceptibility to macrophage killing. Infect Immun 1993; 61:4818-27. [PMID: 8406883 PMCID: PMC281239 DOI: 10.1128/iai.61.11.4818-4827.1993] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Cytokines are potentially useful in vaccination as adjuvants or modulators of the type of response induced. The work below describes the expression of a cloned cytokine gene for murine interleukin-4 (mIL-4) by a live vaccine vector, an attenuated aroA strain (SL7207) of Salmonella typhimurium, in a murine model system. SL7207 was used as a carrier for two different high-level expression vectors. Both resulting strains, designated SL7207(pOmpAmIL-4) and SL7207(pKKmIL-4), expressed the cloned gene product as monitored by both immunological and biological assays. However, SL7207(pOmpAmIL-4) produced mIL-4 at higher levels and was more stable in vitro than SL7207(pKKmIL-4). When SL7207(pOmpAmIL-4) was used as a live vaccine in BALB/c mice, this strain grew and survived at higher levels than the parental attenuated strain or empty plasmid-carrying strain in spleens, livers, and intestines. This difference in growth and survival did not appear to be caused by alterations in specific lymphocyte-mediated anti-Salmonella immune responses such as delayed-type hypersensitivity or serum antibody as measured by enzyme-linked immunosorbent assay; such alterations have been induced by IL-4 administration in other in vivo systems, and the lack of effect here may reflect the fact that IL-4 is not secreted from the bacteria in large quantities, most of the cytokine being in the cytoplasmic-membrane-bound fraction. Conversely, the ability of mouse macrophages to kill the bacteria in vitro was inhibited by bacterial production of mIL-4. This reduction in macrophage killing activity suggests that bacterial production of mIL-4 may be detrimental to host defense against Salmonella infection and may explain the enhanced bacterial growth and survival in vivo.
Collapse
Affiliation(s)
- K Denich
- Department of Medicine, Stanford University, California 94305-5402
| | | | | | | | | |
Collapse
|
130
|
Havell EA, Rogerson BJ. Endotoxin-induced tumor necrosis factor alpha synthesis in murine embryo fibroblasts. Infect Immun 1993; 61:1630-5. [PMID: 8478050 PMCID: PMC280744 DOI: 10.1128/iai.61.5.1630-1635.1993] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Murine embryo fibroblasts (MEF) were found to secrete tumor necrosis factor (TNF) in response to stimulation with endotoxin. Endotoxin-induced TNF production by MEF was inhibited by cycloheximide. However, reversal of the effect of this inhibitor on protein synthesis results in TNF being secreted in amounts equivalent to those produced by endotoxin-induced MEF not treated with cycloheximide. Actinomycin D treatment of MEF blocked the production of endotoxin-induced TNF. Maximal production of TNF required MEF gene transcription during the first 6 h of incubation with endotoxin. To determine whether endotoxin-induced TNF alpha (TNF-alpha) and/or TNF beta were produced by MEF, cDNA was synthesized from the total RNA isolated from endotoxin-induced MEF and amplified by the polymerase chain reaction in the presence of oligonucleotide primers specific for each cytokine. On the basis of the polymerase chain reaction analysis, it was determined that TNF-alpha mRNA levels were increased in endotoxin-induced MEF. Thus, production of TNF-alpha by fibroblasts in response to the endotoxin component of bacterial cell walls is likely to contribute to the expression of TNF-mediated effects occurring in fibroblast-rich tissues infected with gram-negative bacteria.
Collapse
Affiliation(s)
- E A Havell
- Trudeau Institute, Inc., Saranac Lake, New York 12983
| | | |
Collapse
|
131
|
Elkins KL, Rhinehart-Jones T, Nacy CA, Winegar RK, Fortier AH. T-cell-independent resistance to infection and generation of immunity to Francisella tularensis. Infect Immun 1993; 61:823-9. [PMID: 8432603 PMCID: PMC302807 DOI: 10.1128/iai.61.3.823-829.1993] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The intraperitoneal 50% lethal dose (LD50) for Francisella tularensis LVS in both normal control heterozygote BALB/c nu/+ mice and BALB/c nu/nu mice was 2 x 10(0). Both nu/+ and nu/nu mice given 10(7) LVS bacteria or more intradermally (i.d.) died, with a mean time to death of about 7 to 8 days. On the other hand, nu/+ mice given 10(6) LVS bacteria or less survived for more than 60 days and cleared systemic bacteria, while nu/nu mice given 10(6) LVS bacteria or less survived for more than 10 days but died between days 25 and 30. Thus, the short-term (i.e., < 10-day) i.d. LD50 of both nu/nu and nu/+ mice was 3 x 10(6), but the long-term (i.e., > 10-day) i.d. LD50 of nu/nu mice was less than 7 x 10(0). The short-term survival of i.d. infection was dependent on tumor necrosis factor and gamma interferon: treatment of nu/nu mice with anti-tumor necrosis factor or anti-gamma interferon at the time of i.d. infection resulted in death from infection 7 to 8 days later, whereas control infected nu/nu mice survived for 26 days. nu/nu mice infected with LVS i.d. generated LVS-specific serum antibodies, which were predominantly immunoglobulin M: titers peaked 7 days after i.d. infection but declined sharply by day 21, after which mice died. Surprisingly, nu/nu mice given 10(3) LVS bacteria i.d. became resistant to a lethal challenge (5,000 LD50s) of LVS intraperitoneally within 2 days after i.d. infection; nu/nu mice similarly infected with LVS i.d. and challenged with Salmonella typhimurium (10 LD50s) were not protected. nu/nu mice given nu/+ spleen cells intravenously as a source of mature T cells survived i.d. infection for more than 60 days and cleared bacteria. Taken together, these studies demonstrate that i.d. infection of nu/nu mice with LVS rapidly generates T-cell-independent, short-term, specific protective immunity against lethal challenge, but T lymphocytes are essential for long-term survival.
Collapse
Affiliation(s)
- K L Elkins
- Department of Cellular Immunology, Walter Reed Army Institute of Research, Rockville, Maryland 20850
| | | | | | | | | |
Collapse
|
132
|
Harty JT, Schreiber RD, Bevan MJ. CD8 T cells can protect against an intracellular bacterium in an interferon gamma-independent fashion. Proc Natl Acad Sci U S A 1993; 89:11612-6. [PMID: 1360672 PMCID: PMC50603 DOI: 10.1073/pnas.89.23.11612] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Specific T-cell immunity to Listeria monocytogenes is thought to occur through the action of lymphokines which activate phagocytes to ingest and kill microorganisms. Interferon gamma (IFN-gamma) has been shown to be an effective mediator of this type of macrophage activation in vivo and in vitro. The monoclonal antibody H22.1 efficiently neutralizes endogenous IFN-gamma, exacerbates disease in a mouse model of L. monocytogenes infection, and inhibits the in vivo protective activity of a Listeria antigen-specific CD4 T-cell line. In contrast, in vivo protection by Listeria-immune CD8 T cells is not inhibited by the neutralizing anti-IFN-gamma monoclonal antibody. These results suggest that CD8 T cells can protect against an intracellular pathogen in an IFN-gamma-independent manner.
Collapse
Affiliation(s)
- J T Harty
- Howard Hughes Medical Institute, Seattle, WA
| | | | | |
Collapse
|
133
|
Abstract
Since its initial description as an antiviral, it has become clear that Interferon-gamma (IFN-gamma) has potent immunoregulatory and cell growth regulatory activities. As a result of these additional activities, it is now apparent that IFN-gamma plays a major role in regulation of bacterial infections. IFN-gamma can be both induced by bacteria and bacterial products; endogenous IFN-gamma production has been shown to play a protective role in the natural host response to several bacterial infections; and administration of exogenous IFN-gamma is effective in the prevention and treatment of bacterial infections in numerous animal model systems. Although it is now clear that IFN-gamma plays a role in regulation of bacterial infections, the mechanisms of its anti-bacterial effects in vivo remain to be established due to the pleiotropic nature of IFN-gamma activity.
Collapse
|
134
|
Nakajima R, Brubaker RR. Association between virulence of Yersinia pestis and suppression of gamma interferon and tumor necrosis factor alpha. Infect Immun 1993; 61:23-31. [PMID: 8418045 PMCID: PMC302683 DOI: 10.1128/iai.61.1.23-31.1993] [Citation(s) in RCA: 196] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
It is established that Yersinia pestis, the causative agent of bubonic plague, and enteropathogenic Yersinia pseudotuberculosis and Yersinia enterocolitica share a ca. 70-kb low-calcium response or Lcr plasmid (Lcr+). The latter is known to encode regulatory functions that restrict growth at 37 degrees C in Ca(2+)-deficient medium and virulence factors that are expressed only in vitro within this environment (e.g., certain Yops and V antigen). In this study, gamma interferon (IFN-gamma) was never detected in mice infected with 10 minimum lethal doses (MLD) of Lcr+ cells of Y. pestis, and significant levels of tumor necrosis factor alpha (TNF-alpha) arose only prior to death. Prompt and marked synthesis of these cytokines was observed upon infection with avirulent Lcr- mutants. Treatment of mice with exogenous IFN-gamma plus TNF-alpha inhibited multiplication of Lcr+ yersiniae in vivo, thereby providing protection against challenge with 10 MLD. Administration of both cytokines was required for absolute survival, suggesting a synergistic rather than cumulative interaction. This protective effect entailed cytokine priming as judged by subsequent detection of substantial levels of endogenous IFN-gamma and TNF-alpha. Monospecific anti-V-antigen, known to provide passive immunity against 10 MLD of Lcr+ Y. pestis, permitted significant synthesis of endogenous IFN-gamma and TNF-alpha. These findings demonstrate that Lcr+ yersiniae suppress synthesis of cytokines and suggest that this effect is mediated by one or more Lcr plasmid-encoded virulence factors.
Collapse
Affiliation(s)
- R Nakajima
- Department of Microbiology, Michigan State University, East Lansing 48824-1101
| | | |
Collapse
|
135
|
Conlan JW, North RJ. Early pathogenesis of infection in the liver with the facultative intracellular bacteria Listeria monocytogenes, Francisella tularensis, and Salmonella typhimurium involves lysis of infected hepatocytes by leukocytes. Infect Immun 1992; 60:5164-71. [PMID: 1452350 PMCID: PMC258293 DOI: 10.1128/iai.60.12.5164-5171.1992] [Citation(s) in RCA: 180] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The results show that Listeria monocytogenes, Francisella tularensis, and Salmonella typhimurium are facultative intracellular bacteria with a capacity to invade and grow in nonphagocytic cells in vivo. In the liver, all of these pathogens were seen to invade and to multiply extensively in hepatocytes. In all three cases, inflammatory phagocytes were rapidly marshalled to foci of infection where they appeared to cause the destruction of infected hepatocytes, thereby releasing bacteria into the extracellular space, in which presumably they could be ingested and destroyed by the phagocytes. If phagocytic cells were prevented from accumulating at foci of liver infection by treatment of the mice with a monoclonal antibody (NIMP-R10) directed against the type 3 complement receptor of myelomonocytic cells, then lysis of hepatocytes failed to occur and bacteria proliferated unrestrictedly within them. Under these circumstances, otherwise sublethal infections became rapidly lethal. These findings strongly suggest that lysis of infected hepatocytes by phagocytic cells is an important general early-defense strategy against liver infection with at least three different intracellular bacteria.
Collapse
Affiliation(s)
- J W Conlan
- Trudeau Institute, Inc., Saranac Lake, New York 12983
| | | |
Collapse
|
136
|
Elkins KL, Winegar RK, Nacy CA, Fortier AH. Introduction of Francisella tularensis at skin sites induces resistance to infection and generation of protective immunity. Microb Pathog 1992; 13:417-21. [PMID: 1297917 DOI: 10.1016/0882-4010(92)90085-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Mice are susceptible to systemic infection with Francisella tularensis strain LVS; thus, the intraperitoneal (i.p.) lethal dose at 50% (LD50) in C3H/HeN and C57BI/6J mice is only a single bacterium, while the intradermal (i.d.) LD50 is more than 10(4). Here we show that the LD50 when LVS is introduced via the skin, either i.d. or subcutaneously (s.c.), ranges from 7 x 10(4) to 2 x 10(6). Sublethal i.d. or s.c. infection (priming) invariably leads to the generation of systemic and specific protective immunity: primed mice survive lethal i.p., intravenous (i.v.), or i.d. challenges of LVS but not Salmonella typhimurium W118 or Escherichia coli 018:K1:H7 strain BORT.
Collapse
Affiliation(s)
- K L Elkins
- Department of Cellular Immunology, Walter Reed Army Institute of Research, Rockville, MD 20850
| | | | | | | |
Collapse
|
137
|
Elkins KL, Leiby DA, Winegar RK, Nacy CA, Fortier AH. Rapid generation of specific protective immunity to Francisella tularensis. Infect Immun 1992; 60:4571-7. [PMID: 1398969 PMCID: PMC258204 DOI: 10.1128/iai.60.11.4571-4577.1992] [Citation(s) in RCA: 29] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Mice inoculated either subcutaneously (s.c.) or intradermally (i.d.) with a sublethal dose of Francisella tularensis LVS are immune to a lethal intraperitoneal (i.p.) or intravenous (i.v.) challenge of LVS. Here, we show that this immunity developed quite rapidly: mice given a sublethal dose of live LVS s.c. or i.d. (but not i.v.) withstood lethal i.p., i.v., or i.d. challenge as early as 2 days after the initial inoculation, despite the presence of bacterial burdens already in tissues. The magnitude of this early protection was quite impressive. The i.p. 50% lethal dose (LD50) in naive C3H/HeN mice was only 2 bacteria, while the i.p. LD50 in mice given 10(4) LVS i.d. 3 days previously was 3 x 10(6) bacteria. Similarly, the i.v. LD50 in C3H/HeN mice shifted from 3 x 10(2) in naive mice to 5 x 10(6) in primed mice within 3 days after i.d. LVS infection. Comparable changes in the i.p. and i.v. LD50 were observed in C57BL/6J mice. This rapid generation of protective immunity was specific for LVS, in that mice given a sublethal i.d. inoculation of LVS did not survive a lethal challenge with either Salmonella typhimurium W118 or Escherichia coli O118 BORT at any time, nor could mice given sublethal doses of S. typhimurium, E. coli, or Mycobacterium bovis BCG survive lethal doses of LVS. Although an increase in the mean time to death from S. typhimurium infection was noted when mice were given a sublethal i.d. dose of LVS 4 to 14 days earlier, no overall increase in protection or change in the S. typhimurium LD50 was observed. Thus, sublethal infection with LVS at skin sites induced rapid and specific protective immunity.
Collapse
Affiliation(s)
- K L Elkins
- Department of Cellular Immunology, Walter Reed Army Institute of Research, Rockville, Maryland 20850
| | | | | | | | | |
Collapse
|
138
|
Nagano T, Kita T, Tanaka N. The immunocytochemical localization of tumour necrosis factor and leukotriene in the rat liver after treatment with lipopolysaccharide. Int J Exp Pathol 1992; 73:675-83. [PMID: 1419781 PMCID: PMC2002017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
After administration of bacterial lipopolysaccharide, there is an increase in the number of leucocytes which adhere to the endothelial cell surface of the hepatic vessels and pass through the endothelial layer by comparison with controls. There is also marked endothelial cell damage including intracytoplasmic oedema, increased numbers of autophagic vacuoles and dilatation of the intercellular junction in LPS-treated samples. The presence of immunocytochemical products of leukotriene (LTR) and tumour necrosis factor (TNF) was examined using in both LPS-treated and control samples. Immunoreactions of LTR which were seen in specific granules of neutrophils and monocytes attached to the endothelial cell surface may indicate the onset of endothelial cell damage. Positive immunoreactions of TNF on the endothelial cell surface, seen only in LPS-treated samples, indicate that TNF may enhance the passage of blood cells through the endothelia and also increase the endocytotic activity of the liver parenchymal cells, as revealed by the present marker experiment using horseradish peroxidase. Positive reactions of TNF in lysosomes of the endothelial cells suggest that they are able to produce TNF and transport it to the cell surface.
Collapse
Affiliation(s)
- T Nagano
- Department of Legal Medicine, Kanazawa Medical University, Japan
| | | | | |
Collapse
|
139
|
Sjöstedt A, Kuoppa K, Johansson T, Sandström G. The 17 kDa lipoprotein and encoding gene of Francisella tularensis LVS are conserved in strains of Francisella tularensis. Microb Pathog 1992; 13:243-9. [PMID: 1291846 DOI: 10.1016/0882-4010(92)90025-j] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A T-cell-stimulating 17 kDa protein of the vaccine strain Francisella tularensis LVS has previously been cloned, sequenced and shown to be a lipoprotein. In the present study, it was investigated whether the protein, denoted TUL4, and its gene are present in various strains of the genus Francisella. By Western blot analysis, it was demonstrated that a TUL4-specific monoclonal antibody bound to a protein present in each of the Francisella strains. The immunoreactive proteins had an M(r) of 17 kDa in all F. tularensis strains and in the strain Francisella novicida, whereas the M(r) in strains of Francisella philomiragia was 20 kDa. When genomic preparations were probed with a radioactive DNA fragment of F. tularensis LVS encoding TUL4, hybridization was demonstrated in all strains of Francisella, although the F. philomiragia strains did not hybridize under conditions of high stringency. The hybridizing chromosomal DNA fragment of the F. philomiragia strains was larger than that of the other Francisella strains. No hybridization or Western blot reactivity was seen when various other Gram-negative and Gram-positive bacteria were probed. In summary, the 17 kDa lipoprotein of F. tularensis LVS appears to be Francisella-specific and present in the species F. tularensis and F. novicida, whereas an immunologically related protein is present in F. philomiragia.
Collapse
Affiliation(s)
- A Sjöstedt
- Department of Microbiology, National Defence Research Establishment, Umeå, Sweden
| | | | | | | |
Collapse
|
140
|
Urban JF, Madden KB, Svetić A, Cheever A, Trotta PP, Gause WC, Katona IM, Finkelman FD. The Importance of Th2 Cytokines in Protective Immunity to Nematodes. Immunol Rev 1992; 127:205-20. [PMID: 1354652 DOI: 10.1111/j.1600-065x.1992.tb01415.x] [Citation(s) in RCA: 275] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- J F Urban
- Helminthic Diseases Laboratory, U.S. Department of Agriculture, Beltsville, MD 20705
| | | | | | | | | | | | | | | |
Collapse
|