Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Type of study
Language
Publication year range
1.
J Am Soc Nephrol ; 28(4): 1145-1161, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27799486

ABSTRACT

Maladaptive repair after AKI may lead to progressive fibrosis and decline in kidney function. Sphingosine 1-phosphate has an important role in kidney injury and pleiotropic effects in fibrosis. We investigated the involvement of sphingosine kinase 1 and 2 (SphK1 and SphK2), which phosphorylate sphingosine to produce sphingosine 1-phosphate, in kidney fibrosis induced by folic acid (FA) or unilateral ischemia-reperfusion injury. Analysis of Masson trichrome staining and fibrotic marker protein and mRNA expression 14 days after AKI revealed that wild-type (WT) and Sphk1-/- mice exhibited more kidney fibrosis than Sphk2-/- mice. Furthermore, kidneys of FA-treated WT and Sphk1-/- mice had greater immune cell infiltration and expression of fibrotic and inflammatory markers than kidneys of FA-treated Sphk2-/- mice. In contrast, kidneys of Sphk2-/- mice exhibited greater expression of Ifng and IFN-γ-responsive genes (Cxcl9 and Cxcl10) than kidneys of WT or Sphk1-/- mice did at this time point. Splenic T cells from untreated Sphk2-/- mice were hyperproliferative and produced more IFN-γ than did those of WT or Sphk1-/- mice. IFN-γ blocking antibody administered to Sphk2-/- mice or deletion of Ifng (Sphk2-/-Ifng-/- mice) blocked the protective effect of SphK2 deficiency in fibrosis. Moreover, adoptive transfer of Sphk2-/- (but not Sphk2-/-Ifng-/- ) CD4 T cells into WT mice blocked FA-induced fibrosis. Finally, a selective SphK2 inhibitor blocked FA-induced kidney fibrosis in WT mice. These studies demonstrate that SphK2 inhibition may serve as a novel therapeutic approach for attenuating kidney fibrosis.


Subject(s)
Interferon-gamma/physiology , Kidney Diseases/enzymology , Kidney/enzymology , Kidney/pathology , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Animals , Fibrosis/enzymology , Fibrosis/etiology , Fibrosis/prevention & control , Kidney Diseases/prevention & control , Mice , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors
2.
J Am Soc Nephrol ; 26(4): 908-25, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25145931

ABSTRACT

Sphingosine 1-phosphate (S1P), the natural sphingolipid ligand for a family of five G protein- coupled receptors (S1P1-S1P5Rs), regulates cell survival and lymphocyte circulation. We have shown that the pan-S1PR agonist, FTY720, attenuates kidney ischemia-reperfusion injury by directly activating S1P1 on proximal tubule (PT) cells, independent of the canonical lymphopenic effects of S1P1 activation on B and T cells. FTY720 also reduces cisplatin-induced AKI. Therefore, in this study, we used conditional PT-S1P1-null (PepckCreS1pr1(fl/fl)) and control (PepckCreS1pr1(w/wt)) mice to determine whether the protective effect of FTY720 in AKI is mediated by PT-S1P1. Cisplatin induced more renal injury in PT-S1P1-null mice than in controls. Although FTY720 produced lymphopenia in both control and PT-S1P1-null mice, it reduced injury only in control mice. Furthermore, the increase in proinflammatory cytokine (CXCL1, MCP-1, TNF-α, and IL-6) expression and infiltration of neutrophils and macrophages induced by cisplatin treatment was attenuated by FTY720 in control mice but not in PT-S1P1-null mice. Similarly, S1P1 deletion rendered cultured PT cells more susceptible to cisplatin-induced injury, whereas S1P1 overexpression protected PT cells from injury and preserved mitochondrial function. We conclude that S1P1 may have an important role in stabilizing mitochondrial function and that FTY720 administration represents a novel strategy in the prevention of cisplatin-induced AKI.


Subject(s)
Acute Kidney Injury/prevention & control , Kidney Tubules, Proximal/drug effects , Mitochondria/drug effects , Propylene Glycols/therapeutic use , Receptors, Lysosphingolipid/agonists , Sphingosine/analogs & derivatives , Acute Kidney Injury/chemically induced , Animals , Apoptosis/drug effects , Cell Respiration , Cisplatin , Drug Evaluation, Preclinical , Epithelial Cells/drug effects , Fingolimod Hydrochloride , Male , Mice, Inbred C57BL , Mitochondrial Dynamics , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/metabolism , Sphingosine/pharmacology , Sphingosine/therapeutic use
3.
J Immunol ; 189(5): 2584-96, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22855711

ABSTRACT

Dendritic cells (DCs) are central to innate and adaptive immunity of early kidney ischemia-reperfusion injury (IRI), and strategies to alter DC function may provide new therapeutic opportunities. Sphingosine 1-phosphate (S1P) modulates immunity through binding to its receptors (S1P1-5), and protection from kidney IRI occurs in S1P3-deficient mice. Through a series of experiments we determined that this protective effect was owing in part to differences between S1P3-sufficient and -deficient DCs. Mice lacking S1P3 on bone marrow cells were protected from IRI, and S1P3-deficient DCs displayed an immature phenotype. Wild-type (WT) but not S1P3-deficient DCs injected into mice depleted of DCs prior to kidney IR reconstituted injury. Adoptive transfer (i.e., i.v. injection) of glycolipid (Ag)-loaded WT but not S1P3-deficient DCs into WT mice exacerbated IRI, suggesting that WT but not S1P3-deficient DCs activated NKT cells. Whereas WT DC transfers activated the Th1/IFN-γ pathway, S1P3-deficient DCs activated the Th2/IL-4 pathway, and an IL-4-blocking Ab reversed protection from IRI, supporting the concept that IL-4 mediates the protective effect of S1P3-deficient DCs. Administration of S1P3-deficient DCs 7 d prior to or 3 h after IRI protected mice from IRI and suggests their potential use in cell-based therapy. We conclude that absence of DC S1P3 prevents DC maturation and promotes a Th2/IL-4 response. These findings highlight the importance of DC S1P3 in modulating NKT cell function and IRI and support development of selective S1P3 antagonists for tolerizing DCs for cell-based therapy or for systemic administration for the prevention and treatment of IRI and autoimmune diseases.


Subject(s)
Cell Polarity/immunology , Dendritic Cells/immunology , Ischemia/pathology , Kidney/pathology , Receptors, Lysosphingolipid/physiology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Dendritic Cells/metabolism , Ischemia/immunology , Kidney/blood supply , Kidney/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology , Neutrophil Infiltration/immunology , Receptors, Lysosphingolipid/deficiency , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Sphingosine-1-Phosphate Receptors , Th1 Cells/pathology , Th2 Cells/pathology
4.
J Am Soc Nephrol ; 21(6): 955-65, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20338995

ABSTRACT

Agonists of the sphingosine-1-phosphate receptor (S1PR) attenuate kidney ischemia-reperfusion injury (IRI). Previous studies suggested that S1P1R-induced lymphopenia mediates this protective effect, but lymphocyte-independent mechanisms could also contribute. Here, we investigated the effects of S1PR agonists on kidney IRI in mice that lack T and B lymphocytes (Rag-1 knockout mice). Administration of the nonselective S1PR agonist FTY720 or the selective S1P1R agonist SEW2871 reduced injury in both Rag-1 knockout and wild-type mice. In vitro, SEW2871 significantly attenuated LPS- or hypoxia/reoxygenation-induced apoptosis in cultured mouse proximal tubule epithelial cells, supporting a direct protective effect of S1P1R agonists via mitogen-activated protein kinase and/or Akt pathways. S1P1Rs in the proximal tubule mediated IRI in vivo as well: Mice deficient in proximal tubule S1P1Rs experienced a greater decline in renal function after IRI than control mice and their kidneys were no longer protected by SEW2871 administration. In summary, S1PRs in the proximal tubule are necessary for stress-induced cell survival, and S1P1R agonists are renoprotective via direct effects on the tubule cells. Selective agonists of S1P1Rs may hold therapeutic potential for the prevention and treatment of acute kidney injury.


Subject(s)
Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Receptors, Lysosphingolipid/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Acute Kidney Injury/pathology , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Disease Models, Animal , Epithelial Cells/pathology , Fingolimod Hydrochloride , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Kidney Tubules, Proximal/pathology , Leukocytes/drug effects , Leukocytes/pathology , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/metabolism , Oxadiazoles/pharmacology , Propylene Glycols/pharmacology , RNA, Messenger/metabolism , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/genetics , Reperfusion Injury/pathology , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Thiophenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL