Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Immunity ; 41(6): 960-72, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25526309

ABSTRACT

Granzyme B (GzmB) is a protease with a well-characterized intracellular role in targeted destruction of compromised cells by cytotoxic lymphocytes. However, GzmB also cleaves extracellular matrix components, suggesting that it influences the interplay between cytotoxic lymphocytes and their environment. Here, we show that GzmB-null effector T cells and natural killer (NK) cells exhibited a cell-autonomous homing deficit in mouse models of inflammation and Ectromelia virus infection. Intravital imaging of effector T cells in inflamed cremaster muscle venules revealed that GzmB-null cells adhered normally to the vessel wall and could extend lamellipodia through it but did not cross it efficiently. In vitro migration assays showed that active GzmB was released from migrating cytotoxic lymphocytes and enabled chemokine-driven movement through basement membranes. Finally, proteomic analysis demonstrated that GzmB cleaved basement membrane constituents. Our results highlight an important role for GzmB in expediting cytotoxic lymphocyte diapedesis via basement membrane remodeling.


Subject(s)
Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Granzymes/metabolism , Killer Cells, Natural/physiology , T-Lymphocytes, Cytotoxic/physiology , Animals , Basement Membrane/metabolism , Cell Movement/genetics , Cells, Cultured , Chemokines/metabolism , Extracellular Matrix Proteins/metabolism , Granzymes/genetics , Killer Cells, Natural/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteolysis , T-Lymphocytes, Cytotoxic/virology , Transendothelial and Transepithelial Migration/genetics
2.
Immunol Cell Biol ; 95(10): 884-894, 2017 11.
Article in English | MEDLINE | ID: mdl-28722018

ABSTRACT

Intracellular serpins are proposed to inactivate proteases released from lysosome-related organelles into the host cell interior, preventing cell death. Serpinb9 opposes the immune cytotoxic protease, granzyme B, and in a number of settings protects cells against granzyme B-mediated cell death. Using a knockout mouse line engineered to express green fluorescent protein under the serpbinb9 promoter, we demonstrate that serpinb9 is vital for host survival during Ectromelia virus infection by maintaining both mature natural killer NK) cells, and activated CD8+ T cells. Serpinb9 expression parallels granzyme B expression within both populations during infection. Maturing serpinb9-null NK cells exhibit higher levels of granzyme B-mediated apoptosis during infection; hence there are fewer mature NK cells, and these cells also have lower cytotoxic potential. Thus the serpinb9-granzyme B axis is important for homeostasis of both major cytotoxic effector cell populations.


Subject(s)
Granzymes/antagonists & inhibitors , Killer Cells, Natural/immunology , Membrane Proteins/pharmacology , Poxviridae Infections/immunology , Poxviridae/immunology , Serpins/pharmacology , Animals , Cell Death , Cell Survival , Homeostasis , Humans , Intracellular Space , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
Eur J Immunol ; 43(7): 1789-98, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23568450

ABSTRACT

Japanese encephalitis, caused by infection with the neurotropic flavivirus, Japanese encephalitis virus (JEV), is among the most important viral encephalitides in Asia. While previous studies established an essential role of Ab and type I IFN, it is still unclear if the cell-mediated immune responses, through their direct antiviral effector functions, contribute to protection against the fatal disease. We report here that mice defective in both the granule exocytosis and death receptor pathways of cytotoxicity display increased susceptibility to JEV. The two cell contact-dependent cytotoxic effector mechanisms act redundantly within the CNS to reduce disease severity. We also demonstrate that IFN-γ is critical in recovery from primary infection with JEV by a mechanism involving suppression of virus growth in the CNS, and that T cells are the main source of the cytokine that promotes viral clearance from the brain. Finally, we show by in vivo depletion of NK cells that this innate immune cell population is dispensable for control of JEV infection in the periphery and in the CNS. Accordingly, cell contact-dependent cytolytic and IFN-γ-dependent noncytolytic clearance of virus mediated by T cells trafficking into the CNS help in recovery from lethal infection in a mouse model of Japanese encephalitis.


Subject(s)
Cytotoxicity, Immunologic/immunology , Encephalitis, Japanese/immunology , Interferon-gamma/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Disease Models, Animal , Immunity, Cellular/immunology , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Stud Health Technol Inform ; 305: 240-243, 2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37387007

ABSTRACT

A quantitative approach in the form of an online questionnaire was used to identify challenges and desires related to the Care Records Transmission Process and Care Transition Records (CTR). The questionnaire was sent to nurses, nursing assistants, and trainees working in ambulatory, acute inpatient, or long-term care settings. The survey revealed that creating CTRs is time-consuming, and the lack of standardization of CTRs makes the process even more cumbersome. In addition, most facilities transmit the CTR by physically handing it over to the patient or resident, resulting in little or no preparation time for the individual(s) receiving care. The key findings also suggest that most respondents are only partially satisfied with the completeness of the CTRs and that they must conduct additional interviews to obtain missing information. However, most respondents hoped that digital transmission of CTRs would lead to less administrative burden and that standardization of CTRs would be encouraged.


Subject(s)
Hospitals , Patient Transfer , Humans , Germany , Hand , Inpatients
5.
Eur J Immunol ; 41(7): 1948-57, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21590764

ABSTRACT

Cytotoxic T (Tc) cells play a key role in the defense against virus infections. Tc cells recognize infected cells via the T-cell receptor (TCR) and subsequently kill the target cells by one or more cytotoxic mechanisms. Induction of the cytotoxic mechanisms is finely tuned by the activation signals from the TCR. To determine whether TCR down-regulation affects the cytotoxicity of Tc cells, we studied TCR down-regulation-deficient CD3γLLAA mice. We found that Tc cells from CD3γLLAA mice have reduced cytotoxicity due to a specific deficiency in exocytosis of lytic granules. To determine whether this defect was reflected in an increased susceptibility to virus infections, we studied the course of ectromelia virus (ECTV) infection. We found that the susceptibility to ECTV infection was significantly increased in CD3γLLAA mice with a mortality rate almost as high as in granzyme B knock-out mice. Finally, we found that TCR signaling in CD3γLLAA Tc cells caused highly increased tyrosine phosphorylation and activation of the c-Cbl ubiquitin ligase, and that the impaired exocytosis of lytic granules could be rescued by the knockdown of c-Cbl. Thus, our work demonstrates that TCR down-regulation critically increases Tc cell cytotoxicity and protection against poxvirus infection.


Subject(s)
Cytotoxicity, Immunologic , Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/immunology , Animals , Blotting, Western , CD3 Complex/genetics , CD3 Complex/immunology , Cell Line , Exocytosis , Granzymes/metabolism , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Perforin/biosynthesis , Perforin/genetics , Phosphorylation , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/metabolism , RNA Interference , RNA, Small Interfering , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Cytotoxic/metabolism , Ubiquitin-Protein Ligases
6.
J Virol ; 85(11): 5446-55, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21450826

ABSTRACT

The immunological correlates for recovery from primary Japanese encephalitis virus (JEV) infection in humans and experimental animals remain poorly defined. To investigate the relative importance of the adaptive immune responses, we have established a mouse model for Japanese encephalitis in which a low-dose virus inoculum was administered into the footpads of adult C57BL/6 mice. In this model, ~60% of the mice developed a fatal encephalitis and a virus burden in the central nervous system (CNS). Using mice lacking B cells (µMT(-/-) mice) and immune B cell transfer to wild-type mice, we show a critically important role for humoral immunity in preventing virus spread to the CNS. T cell help played an essential part in the maintenance of an effective antibody response necessary to combat the infection, since mice lacking major histocompatibility complex class II showed truncated IgM and blunted IgG responses and uniformly high lethality. JEV infection resulted in extensive CD8(+) T cell activation, judged by upregulation of surface markers CD69 and CD25 and cytokine production after stimulation with a JEV NS4B protein-derived H-2D(b)-binding peptide and trafficking of virus-immune CD8(+) T cells into the CNS. However, no significant effect of CD8(+) T cells on the survival phenotype was found, which was corroborated in knockout mice lacking key effector molecules (Fas receptor, perforin, or granzymes) of cytolytic pathways triggered by T lymphocytes. Accordingly, CD8(+) T cells are mostly dispensable for recovery from infection with JEV. This finding highlights the conflicting role that CD8(+) T cells play in the pathogenesis of JEV and closely related encephalitic flaviviruses such as West Nile virus.


Subject(s)
Antibodies, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Encephalitis, Japanese/immunology , Animals , Central Nervous System/virology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Encephalitis, Japanese/mortality , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Rodent Diseases/immunology , Rodent Diseases/mortality , Survival Analysis
7.
J Virol ; 85(21): 11170-82, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21849445

ABSTRACT

Ectromelia virus (ECTV) is a natural pathogen of mice that causes mousepox, and many of its genes have been implicated in the modulation of host immune responses. Serine protease inhibitor 2 (SPI-2) is one of these putative ECTV host response modifier proteins. SPI-2 is conserved across orthopoxviruses, but results defining its mechanism of action and in vivo function are lacking or contradictory. We studied the role of SPI-2 in mousepox by deleting the SPI-2 gene or its serine protease inhibitor reactive site. We found that SPI-2 does not affect viral replication or cell-intrinsic apoptosis pathways, since mutant viruses replicate in vitro as efficiently as wild-type virus. However, in the absence of SPI-2 protein, ECTV is attenuated in mousepox-susceptible mice, resulting in lower viral loads in the liver, decreased spleen pathology, and substantially improved host survival. This attenuation correlates with more effective immune responses in the absence of SPI-2, including an earlier serum gamma interferon (IFN-γ) response, raised serum interleukin 18 (IL-18), increased numbers of granzyme B(+) CD8(+) T cells, and, most notably, increased numbers and activation of NK cells. Both virus attenuation and the improved immune responses associated with SPI-2 deletion from ECTV are lost when mice are depleted of NK cells. Consequently, SPI-2 renders mousepox lethal in susceptible strains by preventing protective NK cell defenses.


Subject(s)
Ectromelia virus/pathogenicity , Ectromelia, Infectious/mortality , Host-Pathogen Interactions , Killer Cells, Natural/immunology , Serpins/metabolism , Viral Proteins/metabolism , Virulence Factors/metabolism , Animals , Ectromelia virus/genetics , Ectromelia virus/immunology , Ectromelia, Infectious/virology , Gene Deletion , Interferon-gamma/metabolism , Interleukin-18/metabolism , Liver/virology , Lymphocyte Subsets/chemistry , Lymphocyte Subsets/immunology , Mice , Serpins/genetics , Spleen/pathology , Survival Analysis , Viral Load , Viral Proteins/genetics , Virus Replication
8.
Immunol Cell Biol ; 89(6): 706-15, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21173782

ABSTRACT

Using Toll-like receptor (TLR) and MyD88 gene knock-out (GKO) mice the effect of TLRs and MyD88 on virus replication, interferon (IFN)-ß production, natural killer (NK) cell and CD8T cell responses were assessed following ectromelia virus (ECTV) and recombinant vaccinia virus (rVV) infection. The capacity for rVVs encoding cytokines to restore immune function in MyD88(-/-) mice was clearly demonstrated. Results showed that TLR2(-/-), TLR4(-/-)and TLR7(-/-) mice survived ECTV infection whereas MyD88(-/-) and TLR9(-/-)mice, in contrast, were highly susceptible. Next, following infection with rVV, MyD88(-/-) mice elicited reduced serum IFN-ß, NK cell and CD8T cell responses compared with wild-type mice, whereas TLR9(-/-) mice showed elevated CD8T cell responses. When MyD88(-/-)mice were infected with rVV co-expressing IFN-ß these mice were able to restore IFN-ß levels and CD8T cell responses but not NK cell activation. Interestingly, even though rVV co-expressing interleukin (IL)-2 enhanced NK cell activation in MyD88(-/-) mice, this was not associated with an antiviral effect, as observed in normal mice. Surprisingly, co-infection with rVV IL-2/rVV IL-12, but not rVV IL-2/rVV IFN-ß, restored the attenuated phenotype of rVV IL-2 in MyD88(-/-) mice indicating that the IL-2/IL-12 combination promotes antiviral responses. Our results clearly show that the CD8T cell defect observed in MyD88(-/-) mice to vaccinia virus infection can be restored by rVV-encoding IFN-ß demonstrating the critical role of this cytokine in T cell mediated immunity and illustrates that the model can provide an effective platform for the elucidation of cytokine immunobiology.


Subject(s)
Cytokines/genetics , Myeloid Differentiation Factor 88/genetics , Toll-Like Receptors/genetics , Vaccinia virus/genetics , Vaccinia virus/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Chlorocebus aethiops , Cytokines/metabolism , Ectromelia virus/physiology , Ectromelia, Infectious/immunology , Ectromelia, Infectious/prevention & control , Female , Gene Expression Regulation, Viral , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Interleukin-12/metabolism , Interleukin-2/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Virus Replication/immunology
9.
J Virol ; 84(9): 4212-21, 2010 May.
Article in English | MEDLINE | ID: mdl-20164231

ABSTRACT

We previously demonstrated that a single dose of nonadjuvanted intranasal gamma-irradiated influenza A virus can provide robust protection in mice against both homologous and heterosubtypic challenges, including challenge with an H5N1 avian virus strain. We investigated the mechanism behind the observed cross-protection to define which arms of the adaptive immune response are involved in mediating this protection. Studies with gene knockout mice showed the cross-protective immunity to be mediated mainly by T cells and to be dependent on the cytolytic effector molecule perforin. Adoptive transfer of memory T cells from immunized mice, but not of memory B cells, protected naïve recipients against lethal heterosubtypic influenza virus challenge. Furthermore, gamma-irradiated influenza viruses induced cross-reactive Tc-cell responses but not cross-neutralizing or cross-protective antibodies. In addition, histological analysis showed reduced lung inflammation in vaccinated mice compared to that in unvaccinated controls following heterosubtypic challenge. This reduced inflammation was associated with enhanced early recruitment of T cells, both CD4(+) and CD8(+), and with early influenza virus-specific cytotoxic T-cell responses. Therefore, cross-protective immunity induced by vaccination with gamma-irradiated influenza A virus is mediated mainly by Tc-cell responses.


Subject(s)
Cross Protection , Gamma Rays , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , T-Lymphocytes, Cytotoxic/immunology , Adoptive Transfer , Animals , Antibodies, Viral/blood , Body Weight , Influenza A Virus, H1N1 Subtype/radiation effects , Influenza A Virus, H3N2 Subtype/radiation effects , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Survival Analysis , Vaccines, Inactivated/immunology
10.
Int Immunol ; 22(9): 757-67, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20682547

ABSTRACT

Type-I IFN (IFN-I) are highly pleiotropic cytokines known to modulate immune responses and play an early central role in mediating antiviral defenses. We have shown that IFN-I mediate transient up-regulation of a distinct subset of lymphocyte surface activation markers on both B and T cells in vivo independent of cognate antigen: a state referred to as 'partial lymphocyte activation'. Here we investigated in vitro the possibility that partial lymphocyte activation may serve to lower the antigen-specific activation thresholds for T cells. We found that the kinetics of Ca(2+) flux in T cells responding to TCR cross-linking was not enhanced in partially activated T cells. Furthermore, following TCR stimulation with anti-cluster of differentiation (CD) 3 epsilon, a lower proportion of partially activated than naive T cells proliferated. In contrast, the proliferation of partially activated and naive ovalbumin peptide (OVAp, SIINFEKL) specific CD8(+) T cells (OT-I CD8(+) T cells) was similar when stimulated with OVAp. Surprisingly, using an enzyme-linked immunospot (ELISPOT) assay for IFN-gamma secretion, we found that a higher number of partially activated OT-I CD8(+) T cells expressed effector functions than did naive OT-I CD8(+) T cells. This is most readily explained by an increased survival of activated antigen-specific CD8(+) T cells from a pool of partially activated T cells than naive T cells. Overall, when examining the effects of early (Ca(2+) flux), intermediate (proliferation) or late events (IFN-gamma secretion) of T-cell activation, we found that partial activation promotes the survival but does not alter the antigen-specific activation thresholds of CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Interferon Type I/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antigens/immunology , CD3 Complex/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Calcium Signaling/drug effects , Calcium Signaling/immunology , Cell Proliferation/drug effects , Cells, Cultured , Interferon Type I/immunology , Interferon-gamma/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology , Receptors, Antigen, T-Cell/agonists , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology
11.
J Immunol ; 183(1): 37-40, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19525394

ABSTRACT

Cytotoxic T (Tc) cells lyse target cells via exocytosis of granules containing perforin (perf) and granzymes (gzm). In vitro, gzm delivery into the target cell cytosol results in apoptosis, and in the absence of gzm A and B the induction of apoptosis is severely impaired. However, using in vivo Tc cell killing assays, we find that virus-immune, gzm A x B-deficient (gzmAxB(-/-)) mice are competent to eliminate adoptively transferred target cells pulsed with an immunodominant Tc cell determinant as rapidly and completely as their wild-type counterparts. Specific target cell elimination occurred with similar kinetics in both spleen and lymph nodes. Thus, neither gzmA nor gzmB are required for rapid and efficient in vivo cytotoxicity by Tc cells.


Subject(s)
Cytotoxicity, Immunologic , Granzymes/physiology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Death/genetics , Cell Death/immunology , Cytotoxicity, Immunologic/genetics , Ectromelia virus/immunology , Ectromelia, Infectious/enzymology , Ectromelia, Infectious/immunology , Ectromelia, Infectious/pathology , Granzymes/deficiency , Granzymes/genetics , Influenza A virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/enzymology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Cytotoxic/virology , Time Factors
12.
J Gen Virol ; 91(Pt 6): 1450-60, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20147516

ABSTRACT

We have recently shown that intranasal (i.n.) administration of gamma-irradiated A/PR/8 [A/Puerto Rico/8/34 (H1N1)] protects mice against lethal avian influenza A/Vietnam/1203/2004 (H5N1) and other heterosubtypic influenza A infections. Here, we used gamma-irradiated, formalin- and UV-inactivated A/PC [A/Port Chalmers/1/73 (H3N2)] virus preparations and compared their ability to induce both homologous and heterosubtypic protective immunity. Our data show that, in contrast to i.n. vaccination with formalin- or UV-inactivated virus, or the present commercially available trivalent influenza vaccine, a single dose of gamma-ray-inactivated A/PC (gamma-A/PC) conferred significant protection in mice against both homologous and heterosubtypic virus challenges. A multiple immunization regime was required for formalin-inactivated virus preparations to induce protective immunity against a homotypic virus challenge, but did not induce influenza A strain cross-protective immunity. The highly immunogenic gamma-A/PC, but not formalin- or UV-inactivated A/PC, nor the currently available subvirion vaccine, elicited cytotoxic T-cell responses that are most likely responsible for the cross-protective and long-lasting immunity against highly lethal influenza A infections in mice. Finally, freeze-drying of gamma-A/PC did not affect the ability to induce cross-protective immunity.


Subject(s)
Cross Protection , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Virus Inactivation , Animals , Body Weight , Female , Formaldehyde/toxicity , Gamma Rays , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/radiation effects , Lung/virology , Mice , Mice, Inbred BALB C , Survival Analysis , T-Lymphocytes, Cytotoxic/immunology , Ultraviolet Rays , Vaccines, Inactivated/immunology , Viral Load
13.
Virus Res ; 228: 61-65, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27865865

ABSTRACT

Mousepox is caused by the orthopoxvirus ectromelia virus (ECTV), and is thought to be transmitted via skin abrasions. We studied the ECTV virulence factor N1 following subcutaneous infection of mousepox-susceptible BALB/c mice. In this model, ECTV lacking N1L gene was attenuated more than 1000-fold compared with wild-type virus and replication was profoundly reduced as early as four days after infection. However, in contrast to data from an intranasal model, N1 protein was not required for virus dissemination. Further, neither T cell nor cytokine responses were enhanced in the absence of N1. Together with the early timing of reduced virus titres, this suggests that in a cutaneous model, N1 exerts its function at the level of infected cells or in the inhibition of the very earliest effectors of innate immunity.


Subject(s)
Ectromelia virus/physiology , Ectromelia, Infectious/virology , Viral Proteins/genetics , Animals , Host-Pathogen Interactions , Mice , Viral Load , Viral Proteins/metabolism , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism , Virus Replication
14.
FEMS Immunol Med Microbiol ; 43(2): 269-76, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15681158

ABSTRACT

A successful vaccine against human RSV (HRSV) is likely to induce a Th1 or a balanced Th1/TH2 cytokine response. We tested a panel of HRSV immunostimulating complexes (ISCOMs) containing different Quillaja saponin fractions (QH-A, QH-C, and 703: a mixture of 70% QH-A and 30% QH-C) with different immunological properties for their capacity of inducing innate and acquired immune responses. The HRSV 703 ISCOMs induced the strongest innate and acquired immune responses, followed by RSV QH-C and QH-A ISCOMs. All three formulations induced various degrees of Th1 bias response with prominent production of IFN-gamma being 10-50 times higher than that of IL-4 and IL-5. The HRSV specific IgG isotype profile correlated with the predominant secretion of Th1 cytokines, with strong induction of IgG2a antibodies. The 703 ISCOMs induced the most pronounced Th1 profile followed by QH-C and QH-A ISCOMs. The high incorporation of F protein in these ISCOMs compared to G protein combined with the Th1 biased nature of ISCOM are likely to be the causes to promote a Th1 type of profile. The prospect to formulate an RSV ISCOM formulation with an optimal Th1/Th2 balance is in reach particularly in view of the versatile properties of the ISCOM concept.


Subject(s)
Adjuvants, Immunologic , ISCOMs/immunology , Quillaja/chemistry , Quillaja/immunology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Animals , Antibodies, Viral/blood , Female , Immunoglobulin G/blood , Interferon-gamma/analysis , Interleukin-4/analysis , Interleukin-5/analysis , Mice , Respiratory Syncytial Virus Infections/prevention & control , Vaccination/methods , Viral Proteins/immunology
15.
Microbes Infect ; 6(7): 666-75, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15158774

ABSTRACT

To protect against human respiratory syncytial virus (hRSV)-induced bronchiolitis in early infancy, vaccines need to be designed which are effective in the neonatal period. To test the safety and efficacy of adjuvants in neonatal mice, we injected hRSV surface proteins combined with immune-stimulating complexes (ISCOMs) prepared from fractions A, C or A + C of Quillaja saponins. All were well tolerated in adults, but A + C ISCOMS proved lethal in neonates; A or C fractions alone were well tolerated by neonates up to the adult dose. hRSV-ISCOM A induced antibody responses similar to combined fractions, and potent in vitro cytotoxic T cell responses. Adult-like in vitro cytotoxicity against hRSV-infected targets and precursor cytotoxic T cell frequencies were observed within one week of neonatal priming and hRSV-ISCOM A-primed neonates showed virtually complete protection against subsequent viral challenge. hRSV challenge was associated with some pulmonary eosinophilia in both age groups, with higher IL-4 production by lung CD4+ T cells in mice primed as neonates. This was, however, accompanied by only minor (approximately 10%) and transient illness and weight loss. Thus, the identification of hRSV antigen delivery systems with an age-appropriate adjuvanticity/reactogenicity balance may be feasible even in the vulnerable early-life period.


Subject(s)
Adjuvants, Immunologic , Bronchiolitis, Viral/prevention & control , ISCOMs , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human/immunology , Saponins , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/therapeutic use , Animals , Animals, Newborn , Antibodies, Viral/blood , Bronchiolitis, Viral/virology , Humans , ISCOMs/administration & dosage , ISCOMs/adverse effects , ISCOMs/therapeutic use , Immunization , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus Vaccines/therapeutic use , Saponins/administration & dosage , Saponins/chemistry , Saponins/therapeutic use , T-Lymphocytes/immunology
16.
PLoS One ; 7(9): e44834, 2012.
Article in English | MEDLINE | ID: mdl-23028638

ABSTRACT

Japanese encephalitis is a severe central nervous system (CNS) inflammatory disease caused by the mosquito-borne flavivirus, Japanese encephalitis virus (JEV). In the current study we have investigated the immune responses against JEV in mice lacking expression of the chemokine receptor CCR5, which functions in activation and chemotaxis of leukocytes during infection. We show that CCR5 serves as a host antiviral factor against Japanese encephalitis, with CCR5 deficiency markedly increasing mortality, and viral burden in the CNS. Humoral immune responses, which are essential in recovery from JEV infection, were of similar magnitude in CCR5 sufficient and deficient mice. However, absence of CCR5 resulted in a multifaceted deficiency of cellular immune responses characterized by reduced natural killer and CD8⁺ T cell activity, low splenic cellularity, and impaired trafficking of leukocytes to the brain. Interestingly, adoptive transfer of immune spleen cells, depleted of B lymphocytes, increased resistance of CCR5-deficient recipient mice against JEV regardless of whether the cells were obtained from CCR5-deficient or wild-type donor mice, and only when transferred at one but not at three days post-challenge. This result is consistent with a mechanism by which CCR5 expression enhances lymphocyte activation and thereby promotes host survival in Japanese encephalitis.


Subject(s)
CCR5 Receptor Antagonists , Encephalitis, Japanese/immunology , Encephalitis, Japanese/metabolism , HIV Infections/drug therapy , Molecular Targeted Therapy , Receptors, CCR5/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Movement/drug effects , Cell Proliferation/drug effects , Chlorocebus aethiops , Disease Models, Animal , Female , Immunity, Humoral/drug effects , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Mice , Receptors, CCR5/deficiency , Spleen/cytology , Survival Analysis , Vero Cells
18.
PLoS One ; 6(8): e23252, 2011.
Article in English | MEDLINE | ID: mdl-21853094

ABSTRACT

NK cells kill target cells mainly via exocytosis of granules containing perforin (perf) and granzymes (gzm). In vitro, gzm delivery into the target cell cytosol results in apoptosis, and induction of apoptosis is severely impaired in the absence of gzm A and B. However, their importance for in vivo cytotoxicity by cytotoxic T cells has been questioned. We used an in vivo NK cytotoxicity assay, in which splenocytes from wild-type and ß(2)microglobulin-deficient (MHC-I(neg)) mice are co-injected into recipients whose NK cells were activated by virus infection or synthetic Toll-like receptor ligands. Elimination of adoptively transferred MHC-I(neg) splenocytes was unimpaired in the absence of gzmA and gzmB, but dependent on perforin. This target cell rejection was NK cell dependent, since NK cell depletion abrogated it. Furthermore, target cell elimination in vivo was equally rapid in both wild-type and gzmAxB-deficient recipients, with the majority of specific target cells lost from lymphoid tissue within less than one to two hours after transfer. Thus, similar to T cell cytotoxicity, the contribution of gzmA and B to in vivo target cell elimination remains unresolved.


Subject(s)
Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Animals , Cytotoxicity, Immunologic/genetics , Female , Gene Expression Regulation , Granzymes/deficiency , Granzymes/metabolism , Killer Cells, Natural/virology , Mice , Mice, Inbred C57BL , Perforin/metabolism , Semliki forest virus/physiology
19.
PLoS One ; 6(10): e25765, 2011.
Article in English | MEDLINE | ID: mdl-21998693

ABSTRACT

BACKGROUND: We have shown previously in mice, that infection with live viruses, including influenza/A and Semliki Forest virus (SFV), induces systemic partial activation of lymphocytes, characterized by cell surface expression of CD69 and CD86, but not CD25. This partial lymphocytes activation is mediated by type-I interferons (IFN-I). Importantly, we have shown that γ-irradiated SFV does not induce IFN-I and the associated lymphocyte activation. PRINCIPAL FINDINGS: Here we report that, in contrast to SFV, γ-irradiated influenza A virus elicits partial lymphocyte activation in vivo. Furthermore, we show that when using influenza viruses inactivated by a variety of methods (UV, ionising radiation and formalin treatment), as well as commercially available influenza vaccines, only γ-irradiated influenza virus is able to trigger IFN-I-dependent partial lymphocyte activation in the absence of the TLR7/MyD88 signalling pathways. CONCLUSIONS: Our data suggest an important mechanism for the recognition of γ-irradiated influenza vaccine by cytosolic receptors, which correspond with the ability of γ-irradiated influenza virus to induce cross-reactive and cross-protective cytotoxic T cell responses.


Subject(s)
Gamma Rays , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H1N1 Subtype/radiation effects , Influenza A Virus, H3N2 Subtype/physiology , Influenza A Virus, H3N2 Subtype/radiation effects , Interferon Type I/metabolism , Lymphocytes/virology , Animals , Cell Line , Cricetinae , Dogs , Female , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H3N2 Subtype/metabolism , Interferon Type I/biosynthesis , Lymphocytes/cytology , Lymphocytes/metabolism , Membrane Glycoproteins/metabolism , Mice , Myeloid Differentiation Factor 88/metabolism , Neuraminidase/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/virology , Toll-Like Receptor 7/metabolism , Virus Activation/radiation effects
20.
PLoS One ; 4(10): e7512, 2009 Oct 19.
Article in English | MEDLINE | ID: mdl-19838298

ABSTRACT

BACKGROUND: Ectromelia virus is a natural mouse pathogen, causing mousepox. The cytotoxic T (Tc) cell granule serine-protease, granzyme B, is important for its control, but the underlying mechanism is unknown. Using ex vivo virus immune Tc cells, we have previously shown that granzyme B is able to activate several independent pro-apoptotic pathways, including those mediated by Bid/Bak/Bax and caspases-3/-7, in target cells pulsed with Tc cell determinants. METHODS AND FINDINGS: Here we analysed the physiological relevance of those pro-apoptotic pathways in ectromelia infection, by incubating ectromelia-immune ex vivo Tc cells from granzyme A deficient (GzmB(+) Tc cells) or granzyme A and granzyme B deficient (GzmAxB(-/-) Tc cell) mice with ectromelia-infected target cells. We found that gzmB-induced apoptosis was totally blocked in ectromelia infected or peptide pulsed cells lacking caspases-3/-7. However ectromelia inhibited only partially apoptosis in cells deficient for Bid/Bak/Bax and not at all when both pathways were operative suggesting that the virus is able to interfere with apoptosis induced by gzmB in case not all pathways are activated. Importantly, inhibition of viral replication in vitro, as seen with wild type cells, was not affected by the lack of Bid/Bak/Bax but was significantly reduced in caspase-3/-7-deficient cells. Both caspase dependent processes were strictly dependent on gzmB, since Tc cells, lacking both gzms, neither induced apoptosis nor reduced viral titers. SIGNIFICANCE: Out findings present the first evidence on the biological importance of the independent gzmB-inducible pro-apoptotic pathways in a physiological relevant virus infection model.


Subject(s)
Caspases/metabolism , Ectromelia virus/genetics , Ectromelia, Infectious/virology , Fibroblasts/metabolism , Granzymes/metabolism , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/virology , Animals , Apoptosis , Calreticulin/metabolism , Caspase 3/metabolism , Caspase 7/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL