Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 9(1): 19338, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31853095

ABSTRACT

IFNγ enhances allograft immunogenicity and facilitates T-cell mediated rejection. This may cause interstitial fibrosis and tubular atrophy (IFTA), contributing to chronic allograft loss. We assessed if inhibition of T-cell activation by N-octanoyl dopamine (NOD) impairs adherence of activated T-cells to endothelial cells and the ability of activated T-cells to produce IFNγ. We also assessed if NOD affects IFNγ mediated gene expression in endothelial cells. The presence of NOD during T-cell activation significantly blunted their adhesion to unstimulated and cytokine stimulated HUVEC. Supernatants of these T-cells displayed significantly lower concentrations of TNFα and IFNγ and were less capable to facilitate T-cell adhesion. In the presence of NOD VLA-4 (CD49d/CD29) and LFA-1 (CD11a/CD18) expression on T-cells was reduced. NOD treatment of IFNγ stimulated HUVEC reduced the expression of MHC class II transactivator (CIITA), of MHC class II and its associated invariant chain CD74. Since IFTA is associated with T-cell mediated rejection and IFNγ to a large extent regulates immunogenicity of allografts, our current data suggest a potential clinical use of NOD in the treatment of transplant recipients. Further in vivo studies are warranted to confirm these in vitro findings and to assess the benefit of NOD on IFTA in clinically relevant models.


Subject(s)
Cell Adhesion Molecules/metabolism , Dopamine/analogs & derivatives , Histocompatibility Antigens Class II/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Interferon-gamma/pharmacology , Lymphocyte Activation/drug effects , T-Lymphocytes/immunology , Antigens, Differentiation, B-Lymphocyte/metabolism , Cell Adhesion/drug effects , Dopamine/pharmacology , Gene Expression Regulation/drug effects , HLA-DR Antigens/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Integrin alpha4beta1/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Nuclear Proteins/metabolism , Signal Transduction/drug effects , T-Lymphocytes/drug effects , Trans-Activators/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
2.
J Neurotrauma ; 36(2): 370-379, 2019 01 15.
Article in English | MEDLINE | ID: mdl-29768967

ABSTRACT

Important differences in the biology of focal and diffuse traumatic brain injury (TBI) subtypes may result in unique pathophysiological responses to shared molecular mechanisms. Interleukin-1 (IL-1) signaling has been tested as a potential therapeutic target in preclinical models of cerebral contusion and diffuse TBI, and in a phase II clinical trial, but no published studies have examined IL-1 signaling in an impact/acceleration closed head injury (CHI) model. We hypothesized that genetic deletion of IL-1 receptor-1 (IL-1R1 KO) would be beneficial in focal (contusion) and CHI in mice. Wild type and IL-1R1 KO mice were subjected to controlled cortical impact (CCI), or to CHI. CCI produced brain leukocyte infiltration, HMGB1 translocation and release, edema, cell death, and cognitive deficits. CHI induced peak rotational acceleration of 9.7 × 105 ± 8.1 × 104 rad/s2, delayed time to righting reflex, and robust Morris water maze deficits without deficits in tests of anxiety, locomotion, sensorimotor function, or depression. CHI produced no discernable acute plasmalemma damage or cell death, blood-brain barrier permeability to IgG, or brain edema and only a modest increase in brain leukocyte infiltration at 72 h. In both models, mature (17 kDa) interleukin-1 beta (IL-1ß) was induced by 24 h in CD31+ endothelial cells isolated from injured brain but was not induced in CD11b+ cells in either model. High mobility group box protein-1 was released from injured brain cells in CCI but not CHI. Surprisingly, cognitive outcome in mice with global deletion of IL-1R1 was improved in CHI, but worse after CCI without affecting lesion size, edema, or infiltration of CD11b+/CD45+ leukocytes in CCI. IL-1R1 may induce unique biological responses, beneficial or detrimental to cognitive outcome, after TBI depending on the pathoanatomical subtype. Brain endothelium is a hitherto unrecognized source of mature IL-1ß in both models.


Subject(s)
Brain Concussion/metabolism , Brain Concussion/pathology , Brain Contusion/metabolism , Brain Contusion/pathology , Receptors, Interleukin-1/metabolism , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-1/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...