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.
Am J Pathol ; 189(2): 231-247, 2019 02.
Article in English | MEDLINE | ID: mdl-30448408

ABSTRACT

Following renal ischemia-reperfusion injury (IRI), resolution of inflammation allows tubular regeneration, whereas ongoing inflammatory injury mediated by infiltrating leukocytes leads to nephron loss and renal fibrosis, typical hallmarks of chronic kidney disease. Atypical chemokine receptor 2 (ACKR2) is a chemokine decoy receptor that binds and scavenges inflammatory CC chemokines and reduces local leukocyte accumulation. We hypothesized that ACKR2 limits leukocyte infiltration, inflammation, and fibrotic tissue remodeling after renal IRI, thus preventing progression to chronic kidney disease. Compared with wild type, Ackr2 deficiency increases CC chemokine ligand 2 levels in tumor necrosis factor-stimulated tubulointerstitial tissue in vitro. In Ackr2-deficient mice with early IRI 1 or 5 days after transient renal pedicle clamping, tubular injury was similar to wild type, although accumulation of mononuclear phagocytes increased in postischemic Ackr2-/- kidneys. Regarding long-term outcomes, Ackr2-/- kidneys displayed more tubular injury 5 weeks after IRI, which was associated with persistently increased renal infiltrates of mononuclear phagocytes, T cells, Ly6Chigh inflammatory macrophages, and inflammation. Moreover, Ackr2 deficiency caused substantially aggravated renal fibrosis in Ackr2-/- kidneys 5 weeks after IRI, shown by increased expression of matrix molecules, renal accumulation of α-smooth muscle actin-positive myofibroblasts, and bone marrow-derived fibrocytes. ACKR2 is important in limiting persistent inflammation, tubular loss, and renal fibrosis after ischemic acute kidney injury and, thus, can prevent progression to chronic renal disease.


Subject(s)
Acute Kidney Injury/metabolism , Kidney/metabolism , Receptors, Chemokine/metabolism , Reperfusion Injury/metabolism , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Fibrosis , Kidney/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Myofibroblasts/metabolism , Myofibroblasts/pathology , Receptors, Chemokine/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
2.
Kidney Int ; 95(1): 75-93, 2019 01.
Article in English | MEDLINE | ID: mdl-30389199

ABSTRACT

Tumor necrosis factor-α (TNF) is a cytokine mediating inflammatory kidney diseases such as immune complex glomerulonephritis. Its two receptors, TNFR1 and TNFR2, play distinct roles in this process, with TNFR2 strongly required for induction of disease. In contrast to soluble TNF (sTNF), transmembrane TNF robustly activates TNFR2. Thus, we examined the functional role of transmembrane TNF by inducing heterologous nephrotoxic serum nephritis in wild-type and transgenic TNFΔ1-9,K11E knock-in mice expressing transmembrane TNF but no sTNF (memTNF mice). Compared to wild-type, nephritis was exacerbated in memTNF mice on day 5, indicated by increased albuminuria, higher serum urea levels, and more pronounced glomerular deposits, together with higher numbers of dying and proliferating glomerular cells. This was associated with greater loss of glomerular endothelial cells, increased podocyte stress, and signs of augmented necroptosis in memTNF kidneys. Aggravation of nephritis was dependent on transmembrane TNF expression in parenchymal cells, but not leukocytes. Surprisingly, increased kidney injury was associated with reduced renal leukocyte infiltration in memTNF mice, which correlated with decreased renal mRNA expression of pro-inflammatory mediators. This effect was also present in isolated memTNF glomeruli stimulated with interleukin-1ß in vitro. Thus, uncleaved transmembrane TNF is an important mediator of renal tissue damage characterized by increased renal cell death and loss of glomerular endothelial cells in murine glomerulonephritis. In contrast, sTNF predominantly mediates renal leukocyte recruitment and inflammation. These findings highlight the importance of transmembrane TNF in inflammatory kidney disease as a possible therapeutic target.


Subject(s)
Cell Membrane/metabolism , Glomerulonephritis/pathology , Kidney Glomerulus/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis , Biopsy , Cell Line , Datasets as Topic , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/pathology , Gene Knock-In Techniques , Glomerulonephritis/immunology , Humans , Interleukin-1beta/immunology , Kidney Glomerulus/cytology , Kidney Glomerulus/immunology , Leukocytes/immunology , Leukocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Tumor Necrosis Factor, Type II/metabolism , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...