Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Virol ; 90(20): 9096-113, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27489277

ABSTRACT

UNLABELLED: Under physiological conditions, the liver sinusoidal endothelial cells (LSECs) mediate hepatic immune tolerance toward self or foreign antigens through constitutive expression of anti-inflammatory mediators. However, upon viral infection or Toll-like receptor 2 (TLR2) activation, LSECs can achieve proinflammatory functions, but their role in hepatic inflammation during acute viral hepatitis is unknown. Using the highly virulent mouse hepatitis virus type 3 (MHV3) and the attenuated variants 51.6-MHV3 and YAC-MHV3, exhibiting lower tropism for LSECs, we investigated in vivo and in vitro the consequence of LSEC infection on their proinflammatory profiles and the aggravation of acute hepatitis process. In vivo infection with virulent MHV3, in comparison to attenuated strains, resulted in fulminant hepatitis associated with higher hepatic viral load, tissue necrosis, and levels of inflammatory mediators and earlier recruitment of inflammatory cells. Such hepatic inflammatory disorders correlated with disturbed production of interleukin-10 (IL-10) and vascular factors by LSECs. We next showed in vitro that infection of LSECs by the virulent MHV3 strain altered their production of anti-inflammatory cytokines and promoted higher release of proinflammatory and procoagulant factors and earlier cell damage than infection by attenuated strains. This higher replication and proinflammatory activation in LSECs by the virulent MHV3 strain was associated with a specific activation of TLR2 signaling by the virus. We provide evidence that TLR2 activation of LSCEs by MHV3 is an aggravating factor of hepatic inflammation and correlates with the severity of hepatitis. Taken together, these results indicate that preservation of the immunotolerant properties of LSECs during acute viral hepatitis is imperative in order to limit hepatic inflammation and damage. IMPORTANCE: Viral hepatitis B and C infections are serious health problems affecting over 350 million and 170 million people worldwide, respectively. It has been suggested that a balance between protection and liver damage mediated by the host's immune response during the acute phase of infection would be determinant in hepatitis outcome. Thus, it appears crucial to identify the factors that predispose in exacerbating liver inflammation to limit hepatocyte injury. Liver sinusoidal endothelial cells (LSECs) can express both anti- and proinflammatory functions, but their role in acute viral hepatitis has never been investigated. Using mouse hepatitis virus (MHV) infections as animal models of viral hepatitis, we report for the first time that in vitro and in vivo infection of LSECs by the pathogenic MHV3 serotype leads to a reversion of their intrinsic anti-inflammatory phenotype toward a proinflammatory profile as well to as disorders in vascular factors, correlating with the severity of hepatitis. These results highlight a new virus-promoted mechanism of exacerbation of liver inflammatory response during acute hepatitis.


Subject(s)
Endothelial Cells/pathology , Endothelial Cells/virology , Hepatitis, Viral, Animal/pathology , Liver/pathology , Liver/virology , Murine hepatitis virus/pathogenicity , Toll-Like Receptor 2/metabolism , Animals , Inflammation Mediators/analysis , Leukocytes/immunology , Mice , Necrosis/pathology , Viral Load
2.
Immunology ; 149(2): 204-24, 2016 10.
Article in English | MEDLINE | ID: mdl-27273587

ABSTRACT

Viral replication in the liver is generally detected by cellular endosomal Toll-like receptors (TLRs) and cytosolic helicase sensors that trigger antiviral inflammatory responses. Recent evidence suggests that surface TLR2 may also contribute to viral detection through recognition of viral coat proteins but its role in the outcome of acute viral infection remains elusive. In this study, we examined in vivo the role of TLR2 in acute infections induced by the highly hepatotrophic mouse hepatitis virus (MHV) type 3 and weakly hepatotrophic MHV-A59 serotype. To address this, C57BL/6 (wild-type; WT) and TLR2 knockout (KO) groups of mice were intraperitoneally infected with MHV3 or MHV-A59. MHV3 infection provoked a fulminant hepatitis in WT mice, characterized by early mortality and high alanine and aspartate transaminase levels, histopathological lesions and viral replication whereas infection of TLR2 KO mice was markedly less severe. MHV-A59 provoked a comparable mild and subclinical hepatitis in WT and TLR2 KO mice. MHV3-induced fulminant hepatitis in WT mice correlated with higher hepatic expression of interferon-ß, interleukin-6, tumour necrosis factor-α, CXCL1, CCL2, CXCL10 and alarmin (interleukin-33) than in MHV-A59-infected WT mice and in MHV3-infected TLR2 KO mice. Intrahepatic recruited neutrophils, natural killer cells, natural killer T cells or macrophages rapidly decreased in MHV3-infected WT mice whereas they were sustained in MHV-A59-infected WT mice and MHV3-infected TLR2 KO. MHV3 in vitro infection of macrophagic cells induced rapid and higher viral replication and/or interleukin-6 induction in comparison to MHV-A59, and depended on viral activation of TLR2 and p38 mitogen-activated protein kinase. Taken together, these results support a new aggravating inflammatory role for TLR2 in MHV3-induced acute fulminant hepatitis.


Subject(s)
Hepatitis, Viral, Animal/immunology , Murine hepatitis virus/physiology , Toll-Like Receptor 2/metabolism , Acute Disease , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Cells, Cultured , Cytokines/metabolism , Female , Humans , Inflammation Mediators/metabolism , Macrophages , Mice , Mice, Inbred C57BL , Mice, Knockout , Toll-Like Receptor 2/genetics , Virus Replication/genetics , p38 Mitogen-Activated Protein Kinases/administration & dosage , p38 Mitogen-Activated Protein Kinases/metabolism
3.
J Virol ; 89(19): 9896-908, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26202229

ABSTRACT

UNLABELLED: Coronaviruses (CoVs) have shown neuroinvasive properties in humans and animals secondary to replication in peripheral organs, but the mechanism of neuroinvasion is unknown. The major aim of our work was to evaluate the ability of CoVs to enter the central nervous system (CNS) through the blood-brain barrier (BBB). Using the highly hepatotropic mouse hepatitis virus type 3 (MHV3), its attenuated variant, 51.6-MHV3, which shows low tropism for endothelial cells, and the weakly hepatotropic MHV-A59 strain from the murine coronavirus group, we investigated the virus-induced dysfunctions of BBB in vivo and in brain microvascular endothelial cells (BMECs) in vitro. We report here a MHV strain-specific ability to cross the BBB during acute infection according to their virulence for liver. Brain invasion was observed only in MHV3-infected mice and correlated with enhanced BBB permeability associated with decreased expression of zona occludens protein 1 (ZO-1), VE-cadherin, and occludin, but not claudin-5, in the brain or in cultured BMECs. BBB breakdown in MHV3 infection was not related to production of barrier-dysregulating inflammatory cytokines or chemokines by infected BMECs but rather to a downregulation of barrier protective beta interferon (IFN-ß) production. Our findings highlight the importance of IFN-ß production by infected BMECs in preserving BBB function and preventing access of blood-borne infectious viruses to the brain. IMPORTANCE: Coronaviruses (CoVs) infect several mammals, including humans, and are associated with respiratory, gastrointestinal, and/or neurological diseases. There is some evidence that suggest that human respiratory CoVs may show neuroinvasive properties. Indeed, the severe acute respiratory syndrome coronavirus (SARS-CoV), causing severe acute respiratory syndrome, and the CoVs OC43 and 229E were found in the brains of SARS patients and multiple sclerosis patients, respectively. These findings suggest that hematogenously spread CoVs may gain access to the CNS at the BBB level. Herein we report for the first time that CoVs exhibit the ability to cross the BBB according to strain virulence. BBB invasion by CoVs correlates with virus-induced disruption of tight junctions on BMECs, leading to BBB dysfunction and enhanced permeability. We provide evidence that production of IFN-ß by BMECs during CoV infection may prevent BBB breakdown and brain viral invasion.


Subject(s)
Blood-Brain Barrier/virology , Brain/virology , Endothelial Cells/metabolism , Interferon-beta/metabolism , Microvessels/cytology , Murine hepatitis virus/physiology , Tight Junctions/virology , Animals , Cell Line , DNA Primers/genetics , Electric Impedance , Enzyme-Linked Immunosorbent Assay , Female , Immunohistochemistry , Liver/virology , Mice , Mice, Inbred C57BL , Murine hepatitis virus/pathogenicity , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Viral Tropism/physiology , Virulence
4.
Diabetes Metab Res Rev ; 31(6): 545-61, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25352002

ABSTRACT

Obesity is associated with a systemic chronic low-grade inflammation that contributes to the development of metabolic disorders such as cardiovascular diseases and type 2 diabetes. However, the etiology of this obesity-related pro-inflammatory process remains unclear. Most studies have focused on adipose tissue dysfunctions and/or insulin resistance in skeletal muscle cells as well as changes in adipokine profile and macrophage recruitment as potential sources of inflammation. However, low-grade systemic inflammation probably involves a complex network of signals interconnecting several organs. Recent evidences have suggested that disturbances in the composition of the gut microbial flora and alterations in levels of gut peptides following the ingestion of a high-fat diet may be a cause of low-grade systemic inflammation that may even precede and predispose to obesity, metabolic disorders or type 2 diabetes. This hypothesis is appealing because the gastrointestinal system is first exposed to nutrients and may thereby represent the first link in the chain of events leading to the development of obesity-associated systemic inflammation. Therefore, the present review will summarize the latest advances interconnecting intestinal mucosal bacteria-mediated inflammation, adipose tissue and skeletal muscle in a coordinated circuitry favouring the onset of a high-fat diet-related systemic low-grade inflammation preceding obesity and predisposing to metabolic disorders and/or type 2 diabetes. A particular emphasis will be given to high-fat diet-induced alterations of gut homeostasis as an early initiator event of mucosal inflammation and adverse consequences contributing to the promotion of extended systemic inflammation, especially in adipose and muscular tissues.


Subject(s)
Adipose Tissue, White/metabolism , Diabetes Mellitus, Type 2/etiology , Enteritis/physiopathology , Gastrointestinal Microbiome , Models, Biological , Muscle, Skeletal/metabolism , Obesity/etiology , Adipose Tissue, White/immunology , Animals , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/microbiology , Diet, High-Fat/adverse effects , Enteritis/etiology , Enteritis/immunology , Enteritis/microbiology , Gastrointestinal Hormones/metabolism , Humans , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Muscle, Skeletal/immunology , Myositis/etiology , Myositis/immunology , Myositis/microbiology , Myositis/physiopathology , Obesity/immunology , Obesity/metabolism , Obesity/microbiology , Panniculitis/etiology , Panniculitis/immunology , Panniculitis/microbiology , Panniculitis/physiopathology , Systemic Vasculitis/etiology , Systemic Vasculitis/immunology , Systemic Vasculitis/microbiology , Systemic Vasculitis/physiopathology
5.
PLoS One ; 8(9): e74278, 2013.
Article in English | MEDLINE | ID: mdl-24058536

ABSTRACT

The IL-33/ST2 axis is known to be involved in liver pathologies. Although, the IL-33 levels increased in sera of viral hepatitis patients in human, the cellular sources of IL-33 in viral hepatitis remained obscure. Therefore, we aimed to investigate the expression of IL-33 in murine fulminant hepatitis induced by a Toll like receptor (TLR3) viral mimetic, poly(I:C) or by pathogenic mouse hepatitis virus (L2-MHV3). The administration of poly(I:C) plus D-galactosamine (D-GalN) in mice led to acute liver injury associated with the induction of IL-33 expression in liver sinusoidal endothelial cells (LSEC) and vascular endothelial cells (VEC), while the administration of poly(I:C) alone led to hepatocyte specific IL-33 expression in addition to vascular IL-33 expression. The hepatocyte-specific IL-33 expression was down-regulated in NK-depleted poly(I:C) treated mice suggesting a partial regulation of IL-33 by NK cells. The CD1d KO (NKT deficient) mice showed hepatoprotection against poly(I:C)-induced hepatitis in association with increased number of IL-33 expressing hepatocytes in CD1d KO mice than WT controls. These results suggest that hepatocyte-specific IL-33 expression in poly(I:C) induced liver injury was partially dependent of NK cells and with limited role of NKT cells. In parallel, the L2-MHV3 infection in mice induced fulminant hepatitis associated with up-regulated IL-33 expression as well as pro-inflammatory cytokine microenvironment in liver. The LSEC and VEC expressed inducible expression of IL-33 following L2-MHV3 infection but the hepatocyte-specific IL-33 expression was only evident between 24 to 32h of post infection. In conclusion, the alarmin cytokine IL-33 was over-expressed during fulminant hepatitis in mice with LSEC, VEC and hepatocytes as potential sources of IL-33.


Subject(s)
Endothelial Cells/immunology , Hepatitis, Viral, Animal/immunology , Hepatitis/immunology , Interleukins/genetics , Liver/immunology , Murine hepatitis virus/immunology , Animals , Antigens, CD1d/genetics , Antigens, CD1d/immunology , Endothelial Cells/pathology , Endothelial Cells/virology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Endothelium, Vascular/virology , Galactosamine/administration & dosage , Gene Deletion , Gene Expression/immunology , Hepatitis/etiology , Hepatitis/genetics , Hepatitis/pathology , Hepatitis, Viral, Animal/genetics , Hepatitis, Viral, Animal/pathology , Hepatitis, Viral, Animal/virology , Interleukin-33 , Interleukins/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Liver/pathology , Liver/virology , Mice , Mice, Knockout , Murine hepatitis virus/pathogenicity , Natural Killer T-Cells/immunology , Natural Killer T-Cells/pathology , Natural Killer T-Cells/virology , Poly I-C/administration & dosage
6.
Avian Pathol ; 42(2): 88-99, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23581435

ABSTRACT

The use of attenuated vaccines or the occurrence of low virulent T-lymphotropic or B-lymphotropic viruses in flocks may alter the immune responses of young chicks in spite of the absence of clinical signs. Infections with a low virulent T-lymphotropic chicken infectious anaemia virus (lvCIAV) followed by infection with an intermediate B-lymphotropic infectious bursal disease virus (iIBDV) were conducted in specific pathogen free chicks. Thirty-six 1-day-old chicks were infected with the lvCIAV strain (CAV-VAC®) and a similar number of chicks were inoculated with phosphate-buffered saline. At 14 days after lvCIAV infection, one group of 18 lvCIAV-infected chicks and one group of 18 uninfected chicks were infected with an iIBDV strain. At 4, 7 and 14 days post infection with iIBDV, six chicks from each group were euthanized and lymphoid organs were collected. Detection of lvCIAV and iIBDV genomes was conducted by polymerase chain reaction and reverse transcriptase-polymerase chain reaction, respectively. Double-labelled lymphoid subsets from the thymus, spleen and bursa were studied by cytofluorometric analysis. The results reveal that previous infection with lvCIAV increases the occurrence of the lvCIAV and iIBDV genome in thymus and/or bursa without the occurrence of clinical signs in dually lvCIAV/iIBDV-infected chicks. However, the decreases of B cells in spleen and bursa and increases of T-cell subsets in bursa observed in chicks infected with iIBDV did not occur in chicks previously infected with lvCIAV. Taken together, these results suggest that previous infection of young chicks with lvCIAV decreases lymphoid disorders induced by iIBDV while subsequent iIBDV infection increases the lvCIAV genome in lymphoid organs.


Subject(s)
Birnaviridae Infections/veterinary , Chicken anemia virus , Chickens , Circoviridae Infections/veterinary , Coinfection/veterinary , Infectious bursal disease virus , Poultry Diseases/virology , Analysis of Variance , Animals , B-Lymphocytes/immunology , Bursa of Fabricius/virology , Coinfection/virology , DNA Primers/genetics , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Thymus Gland/virology
7.
Avian Pathol ; 40(4): 377-85, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21812717

ABSTRACT

The chicken infectious anaemia virus (CIAV) infection may induce immunosuppression and persistent infection. The use of vaccination in young chicks is still controversial due to its low immune efficiency. In order to verify the viral persistency of a vaccinal strain of CIAV and its associated-lymphoid cell disorders, 54 1-day-old specific pathogen free chicks were vaccinated (CIAV-VAC(®); Intervet, Millsboro, Delaware, USA) and haematologic examination, expression of viral VP3 gene, humoral response and phenotyping of lymphoid cells were studied in lymphoid organs at various times post vaccination (p.v.). No clinical signs were observed but light heteropaenia was detected in CIAV-vaccinated chicks. The VP3 gene of CIAV was detected by polymerase chain reaction in the thymus and spleen from day 7 until 28 days p.v. Thymic larger CD4(+)CD8(+) cells increased only at 7 days p.v. while smaller CD4(+)CD8(+) cells decreased after 14 and 28 days in CIAV-vaccinated birds. The CD4 expression, in contrast to that seen for CD8, decreased in thymocytes from the CIAV-vaccinated group. In the spleen and bursa, the percentage of CD8(+) cells increased at 7 and 28 days p.v. only, while CD4(+) cells decreased simultaneously. The vaccinated chicks also exhibited a higher number of splenic CD3(-)CD8(+) cells (natural killer cells). The anti-CIAV antibody responses, however, remained low in most vaccinated chicks and did not persist up to 18 days p.v. These results suggest that the vaccinal virus strain is clinically attenuated but persists in the thymus and spleen in some birds, inducing a low humoral immune response and altering thymopoiesis.


Subject(s)
Chicken anemia virus/immunology , Chickens/immunology , Lymphocytes/pathology , Poultry Diseases/immunology , Spleen/virology , Thymus Gland/virology , Animals , Chick Embryo , Chicken anemia virus/genetics , Delaware , Immunity, Humoral/immunology , Immunosuppression Therapy/veterinary , Lymphocytes/immunology , Lymphocytes/virology , Phenotype , Poultry Diseases/pathology , Poultry Diseases/virology , Specific Pathogen-Free Organisms , Thymocytes/virology , Time Factors , Vaccination/adverse effects , Vaccination/veterinary , Vaccines, Attenuated/adverse effects , Viral Vaccines/adverse effects
8.
Immunology ; 128(1 Suppl): e181-92, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19740307

ABSTRACT

A rapid antiviral immune response may be related to viral interaction with the host cell leading to activation of macrophages via pattern recognition receptors (PPRs) or specific viral receptors. Carcinoembryonic cell adhesion antigen 1a (CEACAM1a) is the specific receptor for the mouse hepatitis virus (MHV), a coronavirus known to induce acute viral hepatitis in mice. The objective of this study was to understand the mechanisms responsible for the secretion of high-pathogenic MHV3-induced inflammatory cytokines. We report that the induction of the pro-inflammatory cytokines interleukin (IL)-6 and tumour necrosis factor (TNF)-alpha in peritoneal macrophages does not depend on CEACAM1a, as demonstrated in cells isolated from Ceacam1a(-/-) mice. The induction of IL-6 and TNF-alpha production was related rather to the fixation of the spike (S) protein of MHV3 on Toll-like receptor 2 (TLR2) in regions enriched in heparan sulphate and did not rely on viral replication, as demonstrated with denatured S protein and UV-inactivated virus. High levels of IL-6 and TNF-alpha were produced in livers from infected C57BL/6 mice but not in livers from Tlr2(-/-) mice. The histopathological observations were correlated with the levels of those inflammatory cytokines. Depending on mouse strain, the viral fixation to heparan sulfate/TLR2 stimulated differently the p38 mitogen-activated protein kinase (MAPK) and nuclear factor (NF)-kappaB in the induction of IL-6 and TNF-alpha. These results suggest that TLR2 and heparan sulphate receptors can act as new viral PPRs involved in inflammatory responses.


Subject(s)
Carcinoembryonic Antigen/metabolism , Coronavirus Infections/complications , Hepatitis, Animal/immunology , Macrophages, Peritoneal/immunology , Membrane Glycoproteins/immunology , Toll-Like Receptor 2/metabolism , Tumor Necrosis Factor-alpha/metabolism , Viral Envelope Proteins/immunology , Animals , Butadienes/pharmacology , Carcinoembryonic Antigen/genetics , Cell Line , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/virology , Heparitin Sulfate/immunology , Heparitin Sulfate/metabolism , Hepatitis, Animal/virology , Imidazoles/pharmacology , Interleukin-6/agonists , Interleukin-6/biosynthesis , Liver/immunology , Liver/pathology , Liver/virology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/virology , Mice , Mice, Knockout , Murine hepatitis virus/immunology , Nitriles/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Pyridines/pharmacology , Spike Glycoprotein, Coronavirus , Toll-Like Receptor 2/genetics , Tumor Necrosis Factor-alpha/agonists , Virus Replication/immunology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism , NF-kappaB-Inducing Kinase
9.
Immunology ; 128(1 Suppl): e551-61, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19740316

ABSTRACT

The production of interferon-gamma (IFN-gamma) by infiltrating natural killer (NK) cells in liver is involved in the control of mouse hepatitis virus (MHV) infection. The objectives of this study were to identify the mechanisms used by MHV type 3 to modulate the production of IFN-gamma by NK cells during the acute hepatitis in susceptible C57BL/6 mice. Ex vivo and in vitro experiments revealed that NK cells, expressing carcinoembryonic antigen-related cell adhesion molecules (CEACAM) 1a (the MHV receptor), can produce a higher level of IFN-gamma in the presence of both L2-MHV3 and interleukin-12 (IL-12)/IL-18. The synergistic production of IFN-gamma by NK cells depends on viral replication rather than viral fixation only, because it is inhibited or not induced in cells infected with ultraviolet-inactivated viruses and in cells from Ceacam1a(-/-) mice infected with virulent viruses. The synergistic IFN-gamma production involves the p38 mitogen-activated protein kinase (MAPK) rather than the extracellular signal-regulated kinase-1/2 MAPK signalling pathway. However, the signal triggered through the engagement of CEACAM1a decreases the production of IFN-gamma, when these molecules are cross-linked using specific monoclonal antibodies. These results suggest that control of acute hepatitis by IFN-gamma-producing NK cells may depend on both production of IL-12 and IL-18 in the liver environment and viral infection of NK cells.


Subject(s)
Carcinoembryonic Antigen/metabolism , Hepatitis, Viral, Animal/immunology , Interferon-gamma/biosynthesis , Killer Cells, Natural/immunology , Murine hepatitis virus/immunology , Acute Disease , Animals , Butadienes/pharmacology , Carcinoembryonic Antigen/genetics , Cell Line , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/virology , Hepatitis, Viral, Animal/virology , Imidazoles/pharmacology , Interferon-gamma/agonists , Interleukin-12/pharmacology , Interleukin-18/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/virology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/immunology , Mitogen-Activated Protein Kinase Kinases/metabolism , Nitriles/pharmacology , Pyridines/pharmacology , Recombinant Proteins/pharmacology
10.
Can J Microbiol ; 53(8): 944-56, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17898851

ABSTRACT

Several bacterial and immunogenic factors are involved in the host response to probiotic strains of Lactobacillus. Here, we report the isolation of new intestinal lactobacilli from chicken, with different immunomodulating properties on lymphoid cells from SJL and C57BL/6 mice. Analysis of biochemical markers in the Lactobacillus acidophilus CBA4P, CBA3P, and TPA3P isolates reveal that these bacterial isolates belong to the type 2 prototype, although they differ from each other. The effect of conditioned media (CM) from SJL- and C57BL/6-derived peritoneal macrophages incubated with the 3 sonicated bacterial isolates from chicken, as well as with Lactobacillus rhamnosus 9595, Escherichia coli lipopolysaccharide, or Staphylococcus aureus peptidoglycan were compared. Our results show that the CM of macrophages from C57BL/6 and SJL mice treated with the CBA4P isolate stimulated syngeneic splenic lymphocytes at a level similar to the one induced with CM from peptidoglycan-stimulated macrophages. In contrast, the CM from TPA3P- and CBA3P-treated macrophages promoted low or no stimulation of lymphoid cells. Incubation of splenic cells with CM from macrophages treated with L. rhamnosus or TPA3P led to a relative decrease in the percentages of splenic CD4+ T cells, whereas the relative percentages of B cells increased. The CBA4P and CBA3P isolates induced higher levels of gamma interferon when compared with the TPA3P isolate. The effects of the lactobacilli isolates differed according to the mouse strain used but correlated with the production of macrophagic tumor necrosis factor alpha and interleukins 6, 10, and 12 and with the modulation of the p38 mitogen-activated protein kinase (MAPK). Taken together, these results indicate that the immunomodulating properties of the new L. acidophilus isolates depend on their capacity to induce production of interleukins 10 and 12 by macrophages, which is under genetic control and depends on the p38 MAPK pathway.


Subject(s)
Interleukin-10/metabolism , Interleukin-12/metabolism , Lactobacillus acidophilus/metabolism , Macrophages, Peritoneal/drug effects , Animals , Cell Proliferation/drug effects , Cells, Cultured , Chickens , Culture Media, Conditioned/pharmacology , Enzyme-Linked Immunosorbent Assay , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Interleukin-4/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Lymphocytes/cytology , Lymphocytes/drug effects , Lymphocytes/metabolism , Macrophages, Peritoneal/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Peptidoglycan/pharmacology , Probiotics/metabolism , Probiotics/pharmacology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...