Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 8: 398, 2020.
Article in English | MEDLINE | ID: mdl-32582696

ABSTRACT

Alloantigen presentation is an essential process in acute allorejection. In this context, we speculated on a pathogenic role of cathepsin S (Cat-S), a cysteine protease known to promote antigenic peptide loading into MHC class II and to activate protease-activated receptor (PAR)-2 on intrarenal microvascular endothelial and tubular epithelial cells. Single-cell RNA sequencing and immunostaining of human kidney allografts confirmed Cat-S expression in intrarenal mononuclear phagocytes. In vitro, Cat-S inhibition suppressed CD4 + T cell lymphocyte activation in a mixed lymphocyte assay. In vivo, we employed a mouse model of kidney transplantation that showed preemptive Cat-S inhibition significantly protected allografts from tubulitis and intimal arteritis. To determine the contribution of PAR-2 activation, first, Balb/c donor kidneys were transplanted into Balb/c recipient mice without signs of rejection at day 10. In contrast, kidneys from C57BL/6J donor mice revealed severe intimal arteritis, tubulitis, interstitial inflammation, and glomerulitis. Kidneys from Par2-deficient C57BL/6J mice revealed partial protection from tubulitis and lower intrarenal expression levels for Fasl, Tnfa, Ccl5, and Ccr5. Together, we conclude that Cat-S and PAR-2 contribute to immune dysregulation and kidney allograft rejection, possibly involving Cat-S-mediated activation of PAR-2 on recipient parenchymal cells in the allograft.

2.
Nat Cell Biol ; 22(1): 60-73, 2020 01.
Article in English | MEDLINE | ID: mdl-31907413

ABSTRACT

Defining the ontogeny of the human adaptive immune system during embryogenesis has implications for understanding childhood diseases including leukaemias and autoimmune conditions. Using RAG1:GFP human pluripotent stem cell reporter lines, we examined human T-cell genesis from pluripotent-stem-cell-derived haematopoietic organoids. Under conditions favouring T-cell development, RAG1+ cells progressively upregulated a cohort of recognized T-cell-associated genes, arresting development at the CD4+CD8+ stage. Sort and re-culture experiments showed that early RAG1+ cells also possessed B-cell, myeloid and erythroid potential. Flow cytometry and single-cell-RNA-sequencing data showed that early RAG1+ cells co-expressed the endothelial/haematopoietic progenitor markers CD34, VECAD and CD90, whereas imaging studies identified RAG1+ cells within CD31+ endothelial structures that co-expressed SOX17+ or the endothelial marker CAV1. Collectively, these observations provide evidence for a wave of human T-cell development that originates directly from haemogenic endothelium via a RAG1+ intermediate with multilineage potential.


Subject(s)
Endothelium/cytology , Hemangioblasts/cytology , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Pluripotent Stem Cells/cytology , Cell Differentiation/physiology , Cell Line , Embryonic Development/physiology , Hematopoietic Stem Cell Transplantation/methods , Humans , Organoids/cytology
3.
Nephrol Dial Transplant ; 35(1): 86-97, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31081025

ABSTRACT

BACKGROUND: Dysbiosis, bacterial translocation and systemic inflammation have been found to be associated with human and experimental forms of chronic kidney disease (CKD), but the functional contribution of the intestinal microbiota to CKD-related intestinal barrier dysfunction and CKD progression is unknown, especially in CKD secondary to hyperoxaluria and nephrocalcinosis. METHODS: C57BL/6N mice fed an oxalate-rich diet for either 10 or 20 days developed reversible or progressive kidney disease, respectively. RESULTS: Oxalate-induced CKD manifested as azotaemia, renal anaemia and hyperkalaemia. CKD was associated with persistent dysbiosis and intestinal barrier dysfunction. Local as well as systemic inflammation was evident and partially persisted despite better renal function after returning to an oxalate-free diet, indicating some innate immune memory. Eradication of the microbiota with a combination of antibiotics improved intestinal barrier function but had no effect on renal function, nephrocalcinosis, kidney remodelling and atrophy compared with control mice not receiving antibiotics. CONCLUSIONS: Together, in chronic oxalate nephropathy, the intestinal microbiota contributes to the CKD-related dysfunction of the intestinal barrier but not to the progression of nephrocalcinosis itself, as well to its related kidney atrophy and excretory dysfunction.


Subject(s)
Dysbiosis/etiology , Gastrointestinal Microbiome , Hyperoxaluria/complications , Inflammation/etiology , Nephrocalcinosis/complications , Renal Insufficiency, Chronic/etiology , Animals , Disease Progression , Dysbiosis/pathology , Humans , Inflammation/pathology , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Renal Insufficiency, Chronic/pathology
4.
Front Immunol ; 10: 1223, 2019.
Article in English | MEDLINE | ID: mdl-31191559

ABSTRACT

Inflammasome-driven release of interleukin(IL)-1ß is a central element of many forms of sterile inflammation and has been evident to promote the onset and progression of diabetic kidney disease. We microdissected glomerular and tubulointerstitial samples from kidney biopsies of patients with diabetic kidney disease and found expression of IL-1ß mRNA. Immunostaining of such kidney biopsies across a broad spectrum of diabetic kidney disease stages revealed IL-1ß positivity in a small subset of infiltrating immune cell. Thus, we speculated on a potential of IL-1ß as a therapeutic target and neutralizing the biological effects of murine IL-1ß with a novel monoclonal antibody in uninephrectomized diabetic db/db mice with progressive type 2 diabetes- and obesity-related single nephron hyperfiltration, podocyte loss, proteinuria, and progressive decline of total glomerular filtration rate (GFR). At 18 weeks albuminuric mice were randomized to intraperitoneal injections with either anti-IL-1ß or control IgG once weekly for 8 weeks. During this period, anti-IL-1ß IgG had no effect on food or fluid intake, body weight, and fasting glucose levels. At week 26, anti-IL-1ß IgG had reduced renal mRNA expression of kidney injury markers (Ngal) and fibrosis (Col1, a-Sma), significantly attenuated the progressive decline of GFR in hyperfiltrating diabetic mice, and preserved podocyte number without affecting albuminuria or indicators of single nephron hyperfiltration. No adverse effect were observed. Thus, IL-1ß contributes to the progression of chronic kidney disease in type 2 diabetes and might therefore be a valuable therapeutic target, potentially in combination with drugs with different mechanisms-of-action such as RAS and SGLT2 inhibitors.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/therapy , Interleukin-1beta/physiology , Renal Insufficiency, Chronic/therapy , Actins/biosynthesis , Actins/genetics , Animals , Antibodies, Monoclonal/immunology , Collagen/biosynthesis , Collagen/genetics , Diabetes Mellitus, Type 2/genetics , Disease Progression , Glomerular Filtration Rate , Immunoglobulin G/immunology , Immunoglobulin G/therapeutic use , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/immunology , Lipocalin-2/biosynthesis , Lipocalin-2/genetics , Mice , Mice, Obese , Nephrectomy , Podocytes/pathology , Proteinuria/etiology , RNA, Messenger/biosynthesis , Random Allocation
5.
Development ; 146(5)2019 03 07.
Article in English | MEDLINE | ID: mdl-30846463

ABSTRACT

Kidney organoids have potential uses in disease modelling, drug screening and regenerative medicine. However, novel cost-effective techniques are needed to enable scaled-up production of kidney cell types in vitro We describe here a modified suspension culture method for the generation of kidney micro-organoids from human pluripotent stem cells. Optimisation of differentiation conditions allowed the formation of micro-organoids, each containing six to ten nephrons that were surrounded by endothelial and stromal populations. Single cell transcriptional profiling confirmed the presence and transcriptional equivalence of all anticipated renal cell types consistent with a previous organoid culture method. This suspension culture micro-organoid methodology resulted in a three- to fourfold increase in final cell yield compared with static culture, thereby representing an economical approach to the production of kidney cells for various biological applications.


Subject(s)
Cell Culture Techniques , Gene Expression Regulation, Developmental , Kidney/cytology , Pluripotent Stem Cells/cytology , Albumins/metabolism , Cell Differentiation , Cells, Cultured , Doxorubicin/pharmacology , Humans , Nephrons/metabolism , Organoids , Signal Transduction , Transcription, Genetic , Wnt Proteins/metabolism
6.
Annu Rev Physiol ; 81: 335-357, 2019 02 10.
Article in English | MEDLINE | ID: mdl-30742789

ABSTRACT

Human kidney tissue can now be generated via the directed differentiation of human pluripotent stem cells. This advance is anticipated to facilitate the modeling of human kidney diseases, provide platforms for nephrotoxicity screening, enable cellular therapy, and potentially generate tissue for renal replacement. All such applications will rely upon the accuracy and reliability of the model and the capacity for stem cell-derived kidney tissue to recapitulate both normal and diseased states. In this review, we discuss the models available, how well they recapitulate the human kidney, and how far we are from application of these cells for use in cellular therapies.


Subject(s)
Kidney/cytology , Stem Cells/cytology , Animals , Cell Differentiation/physiology , Humans , Kidney Diseases/physiopathology
7.
Semin Cell Dev Biol ; 91: 153-168, 2019 07.
Article in English | MEDLINE | ID: mdl-30184476

ABSTRACT

Decades of research into the molecular and cellular regulation of kidney morphogenesis in rodent models, particularly the mouse, has provided both an atlas of the mammalian kidney and a roadmap for recreating kidney cell types with potential applications for the treatment of kidney disease. With advances in both our capacity to maintain nephron progenitors in culture, reprogram to kidney cell types and direct the differentiation of human pluripotent stem cells to kidney endpoints, renal regeneration via cellular therapy or tissue engineering may be possible. Human kidney models also have potential for disease modelling and drug screening. Such applications will rely upon the accuracy of the model at the cellular level and the capacity for stem-cell derived kidney tissue to recapitulate both normal and diseased kidney tissue. In this review, we will discuss the available cell sources, how well they model the human kidney and how far we are from application either as models or for tissue engineering.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Kidney/physiology , Nephrons/physiology , Regeneration , Renal Insufficiency, Chronic/therapy , Animals , Cell Differentiation , Humans , Kidney/cytology , Nephrons/cytology , Pluripotent Stem Cells/cytology , Renal Insufficiency, Chronic/physiopathology , Tissue Engineering/methods
8.
J Immunol ; 199(4): 1440-1452, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28701510

ABSTRACT

Activation of various innate immune receptors results in IL-1 receptor-associated kinase (IRAK)-1/IRAK-4-mediated signaling and secretion of proinflammatory cytokines such as IL-12, IL-6, or TNF-α, all of which are implicated in tissue injury and elevated during tissue remodeling processes. IRAK-M, also known as IRAK-3, is an inhibitor of proinflammatory cytokine and chemokine expression in intrarenal macrophages. Innate immune activation contributes to both acute kidney injury and tissue remodeling that is associated with chronic kidney disease (CKD). Our study assessed the contribution of macrophages in CKD and the role of IRAK-M in modulating disease progression. To evaluate the effect of IRAK-M in chronic renal injury in vivo, a mouse model of unilateral ureteral obstruction (UUO) was employed. The expression of IRAK-M increased within 2 d after UUO in obstructed compared with unobstructed kidneys. Mice deficient in IRAK-M were protected from fibrosis and displayed a diminished number of alternatively activated macrophages. Compared to wild-type mice, IRAK-M-deficient mice showed reduced tubular injury, leukocyte infiltration, and inflammation following renal injury as determined by light microscopy, immunohistochemistry, and intrarenal mRNA expression of proinflammatory and profibrotic mediators. Taken together, these results strongly support a role for IRAK-M in renal injury and identify IRAK-M as a possible modulator in driving an alternatively activated profibrotic macrophage phenotype in UUO-induced CKD.


Subject(s)
Fibrosis/immunology , Interleukin-1 Receptor-Associated Kinases/immunology , Kidney/pathology , Macrophage Activation , Macrophages/cytology , Macrophages/immunology , Renal Insufficiency, Chronic/immunology , Animals , Cytokines/immunology , Disease Models, Animal , Disease Progression , Fibrosis/pathology , Humans , Immunomodulation , Inflammation/pathology , Interleukin-1 Receptor-Associated Kinases/deficiency , Interleukin-1 Receptor-Associated Kinases/genetics , Kidney/immunology , Mice , Mice, Inbred C57BL , Signal Transduction , Tumor Necrosis Factor-alpha/immunology , Ureteral Obstruction/pathology
9.
Sci Rep ; 7(1): 2775, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28584258

ABSTRACT

Cathepsin(Cat)-S processing of the invariant chain-MHC-II complex inside antigen presenting cells is a central pathomechanism of autoimmune-diseases. Additionally, Cat-S is released by activated-myeloid cells and was recently described to activate protease-activated-receptor-(PAR)-2 in extracellular compartments. We hypothesized that Cat-S blockade targets both mechanisms and elicits synergistic therapeutic effects on autoimmune tissue injury. MRL-(Fas)lpr mice with spontaneous autoimmune tissue injury were treated with different doses of Cat-S inhibitor RO5459072, mycophenolate mofetil or vehicle. Further, female MRL-(Fas)lpr mice were injected with recombinant Cat-S with/without concomitant Cat-S or PAR-2 blockade. Cat-S blockade dose-dependently reversed aberrant systemic autoimmunity, e.g. plasma cytokines, activation of myeloid cells and hypergammaglobulinemia. Especially IgG autoantibody production was suppressed. Of note (MHC-II-independent) IgM were unaffected by Cat-S blockade while they were suppressed by MMF. Cat-S blockade dose-dependently suppressed immune-complex glomerulonephritis together with a profound and early effect on proteinuria, which was not shared by MMF. In fact, intravenous Cat-S injection induced severe glomerular endothelial injury and albuminuria, which was entirely prevented by Cat-S or PAR-2 blockade. In-vitro studies confirm that Cat-S induces endothelial activation and injury via PAR-2. Therapeutic Cat-S blockade suppresses systemic and peripheral pathomechanisms of autoimmune tissue injury, hence, Cat-S is a promising therapeutic target in lupus nephritis.


Subject(s)
Autoimmune Diseases/etiology , Autoimmune Diseases/pathology , Autoimmunity/drug effects , Cathepsins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Animals , Autoimmune Diseases/drug therapy , Cathepsins/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacokinetics , Female , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lupus Nephritis/drug therapy , Lupus Nephritis/etiology , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Mice , Mice, Inbred MRL lpr , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Receptor, PAR-2/genetics , Receptor, PAR-2/metabolism
10.
Sci Rep ; 7: 43538, 2017 02 27.
Article in English | MEDLINE | ID: mdl-28240259

ABSTRACT

Cardiovascular complications determine morbidity/mortality in chronic kidney disease (CKD). We hypothesized that progressive CKD drives the release of cathepsin-S (Cat-S), a cysteine protease that promotes endothelial dysfunction and cardiovascular complications. Therefore, Cat-S, soluble tumor-necrosis-factor receptor (sTNFR) 1/2 and glomerular filtration rate (GFR) were measured in a CKD mouse model, a German CKD-cohort (MCKD, n = 421) and two Swedish community-based cohorts (ULSAM, n = 764 and PIVUS, n = 804). Association between Cat-S and sTNFR1/2/GFR was assessed using multivariable linear regression. In the mouse model, Cat-S and sTNFR1/2 concentrations were increased following the progressive decline of GFR, showing a strong correlation between Cat-S and GFR (r = -0.746, p < 0.001) and Cat-S and sTNFR1/sTNFR2 (r = 0.837/0.916, p < 0.001, respectively). In the human cohorts, an increase of one standard deviation of estimated GFR was associated with a decrease of 1.008 ng/ml (95%-confidence interval (95%-CI) -1.576-(-0.439), p < 0.001) in Cat-S levels in MCKD; in ULSAM and PIVUS, results were similar. In all three cohorts, Cat-S and sTNFR1/sTNFR2 levels were associated in multivariable linear regression (p < 0.001). In conclusion, as GFR declines Cat-S and markers of inflammation-related endothelial dysfunction increase. The present data indicating that Cat-S activity increases with CKD progression suggest that Cat-S might be a therapeutic target to prevent cardiovascular complications in CKD.


Subject(s)
Cathepsins/blood , Guanine Nucleotide Exchange Factors/blood , Receptors, Tumor Necrosis Factor, Type II/blood , Receptors, Tumor Necrosis Factor, Type I/blood , Adult , Aged , Animals , Biomarkers , Disease Models, Animal , Female , Germany , Glomerular Filtration Rate , Humans , Kidney Function Tests , Male , Mice , Middle Aged , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/epidemiology , Risk Factors , Sweden
11.
J Am Soc Nephrol ; 28(6): 1753-1768, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28073931

ABSTRACT

Severe AKI is often associated with multiorgan dysfunction, but the mechanisms of this remote tissue injury are unknown. We hypothesized that renal necroinflammation releases cytotoxic molecules that may cause remote organ damage. In hypoxia-induced tubular epithelial cell necrosis in vitro, histone secretion from ischemic tubular cells primed neutrophils to form neutrophil extracellular traps. These traps induced tubular epithelial cell death and stimulated neutrophil extracellular trap formation in fresh neutrophils. In vivo, ischemia-reperfusion injury in the mouse kidney induced tubular necrosis, which preceded the expansion of localized and circulating neutrophil extracellular traps and the increased expression of inflammatory and injury-related genes. Pretreatment with inhibitors of neutrophil extracellular trap formation reduced kidney injury. Dual inhibition of neutrophil trap formation and tubular cell necrosis had an additive protective effect. Moreover, pretreatment with antihistone IgG suppressed ischemia-induced neutrophil extracellular trap formation and renal injury. Renal ischemic injury also increased the levels of circulating histones, and we detected neutrophil infiltration and TUNEL-positive cells in the lungs, liver, brain, and heart along with neutrophil extracellular trap accumulation in the lungs. Inhibition of neutrophil extracellular trap formation or of circulating histones reduced these effects as well. These data suggest that tubular necrosis and neutrophil extracellular trap formation accelerate kidney damage and remote organ dysfunction through cytokine and histone release and identify novel molecular targets to limit renal necroinflammation and multiorgan failure.


Subject(s)
Acute Kidney Injury/complications , Extracellular Traps/physiology , Ischemia/complications , Kidney Cortex Necrosis/etiology , Kidney/blood supply , Neutrophils , Animals , Cells, Cultured , Histones/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Severity of Illness Index
12.
J Am Soc Nephrol ; 28(3): 761-768, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27612997

ABSTRACT

Intrarenal crystals trigger inflammation and renal cell necroptosis, processes that involve TNF receptor (TNFR) signaling. Here, we tested the hypothesis that TNFRs also have a direct role in tubular crystal deposition and progression of hyperoxaluria-related CKD. Immunohistochemical analysis revealed upregulated tubular expression of TNFR1 and TNFR2 in human and murine kidneys with calcium oxalate (CaOx) nephrocalcinosis-related CKD compared with controls. Western blot and mRNA expression analyses in mice yielded consistent data. When fed an oxalate-rich diet, wild-type mice developed progressive CKD, whereas Tnfr1-, Tnfr2-, and Tnfr1/2-deficient mice did not. Despite identical levels of hyperoxaluria, Tnfr1-, Tnfr2-, and Tnfr1/2-deficient mice also lacked the intrarenal CaOx deposition and tubular damage observed in wild-type mice. Inhibition of TNFR signaling prevented the induced expression of the crystal adhesion molecules, CD44 and annexin II, in tubular epithelial cells in vitro and in vivo, and treatment with the small molecule TNFR inhibitor R-7050 partially protected hyperoxaluric mice from nephrocalcinosis and CKD. We conclude that TNFR signaling is essential for CaOx crystal adhesion to the luminal membrane of renal tubules as a fundamental initiating mechanism of oxalate nephropathy. Furthermore, therapeutic blockade of TNFR might delay progressive forms of nephrocalcinosis in oxalate nephropathy, such as primary hyperoxaluria.


Subject(s)
Hyperoxaluria/complications , Kidney Calculi/etiology , Receptors, Tumor Necrosis Factor, Type II/physiology , Receptors, Tumor Necrosis Factor, Type I/physiology , Animals , Crystallization , Humans , Hyperoxaluria/metabolism , Mice , Mice, Inbred C57BL
13.
Semin Nephrol ; 36(3): 162-73, 2016 05.
Article in English | MEDLINE | ID: mdl-27339382

ABSTRACT

The nephrons of the kidney are independent functional units harboring cells of a low turnover during homeostasis. As such, physiological renal cell death is a rather rare event and dead cells are flushed away rapidly with the urinary flow. Renal cell necrosis occurs in acute kidney injuries such as thrombotic microangiopathies, necrotizing glomerulonephritis, or tubular necrosis. All of these are associated with intense intrarenal inflammation, which contributes to further renal cell loss, an autoamplifying process referred to as necroinflammation. But how does renal cell necrosis trigger inflammation? Here, we discuss the role of danger-associated molecular patterns (DAMPs), mitochondrial (mito)-DAMPs, and alarmins, as well as their respective pattern recognition receptors. The capacity of DAMPs and alarmins to trigger cytokine and chemokine release initiates the recruitment of leukocytes into the kidney that further amplify necroinflammation. Infiltrating neutrophils often undergo neutrophil extracellular trap formation associated with neutrophil death or necroptosis, which implies a release of histones, which act not only as DAMPs but also elicit direct cytotoxic effects on renal cells, namely endothelial cells. Proinflammatory macrophages and eventually cytotoxic T cells further drive kidney cell death and inflammation. Dissecting the molecular mechanisms of necroinflammation may help to identify the best therapeutic targets to limit nephron loss in kidney injury.


Subject(s)
Acute Kidney Injury/immunology , Apoptosis/immunology , Cytokines/immunology , Extracellular Traps/immunology , Inflammation/immunology , Necrosis/immunology , Alarmins/immunology , Cell Death , Histones , Humans , Kidney/immunology , Macrophages/immunology , Neutrophils/immunology , T-Lymphocytes, Cytotoxic/immunology
14.
Am J Pathol ; 186(6): 1442-53, 2016 06.
Article in English | MEDLINE | ID: mdl-27102769

ABSTRACT

Rapidly progressive glomerulonephritis is characterized by glomerular necroinflammation and crescent formation. Its treatment includes unspecific and toxic agents; therefore, the identification of novel therapeutic targets is required. The E3-ubiquitin ligase murine double minute (MDM)-2 is a nonredundant element of NF-κB signaling and the negative regulator of tumor suppressor gene TP53-mediated cell cycle arrest and cell death. We hypothesized that the MDM2 would drive crescentic glomerulonephritis by NF-κB-dependent glomerular inflammation and by p53-dependent parietal epithelial cell hyperproliferation. Indeed, the pre-emptive MDM2 blockade by nutlin-3a ameliorated all aspects of crescentic glomerulonephritis. MDM2 inhibition had identical protective effects in Trp53-deficient mice, with the exception of crescent formation, which was not influenced by nutlin-3a treatment. In vitro experiments confirmed the contribution of MDM2 for induction of NF-κB-dependent cytokines in murine glomerular endothelial cells and for p53-dependent parietal epithelial cell proliferation. To evaluate MDM2 blockade as a potential therapeutic intervention in rapidly progressive glomerulonephritis, we treated mice with established glomerulonephritis with nutlin-3a. Delayed onset of nutlin-3a treatment was equally protective as the pre-emptive treatment in abrogating crescentic glomerulonephritis. Together, the pathogenic effects of MDM2 are twofold, that is, p53-independent NF-κB activation increasing intraglomerular inflammation and p53-dependent parietal epithelial cell hyperplasia and crescent formation. We therefore propose MDM2 blockade as a potential novel therapeutic strategy in rapidly progressive glomerulonephritis.


Subject(s)
Glomerulonephritis/pathology , Imidazoles/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Animals , Blotting, Western , Disease Models, Animal , Glomerulonephritis/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism
15.
Semin Nephrol ; 36(1): 17-30, 2016 Jan.
Article in English | MEDLINE | ID: mdl-27085732

ABSTRACT

The clinical category of acute kidney injury includes a wide range of completely different disorders, many with their own pathomechanisms and treatment targets. In this review we focus on the role of inflammation in the pathogenesis of acute tubular necrosis (ATN). We approach this topic by first discussing the role of the immune system in the different phases of ATN (ie, early and late injury phase, recovery phase, and the long-term outcome phase of an ATN episode). A more detailed discussion focuses on putative therapeutic targets among the following mechanisms and mediators: oxidative stress and reactive oxygen species-related necroinflammation, regulated cell death-related necroinflammation, immunoregulatory lipid mediators, cytokines and cytokine signaling, chemokines and chemokine signaling, neutrophils and neutrophils extracellular traps (NETs) associated neutrophil cell death, called NETosis, extracellular histones, proinflammatory mononuclear phagocytes, humoral mediators such as complement, pentraxins, and natural antibodies. Any prioritization of these targets has to take into account the intrinsic differences between rodent models and human ATN, the current acute kidney injury definitions, and the timing of clinical decision making. Several conceptual problems need to be solved before anti-inflammatory drugs that are efficacious in rodent ATN may become useful therapeutics for human ATN.


Subject(s)
Acute Kidney Injury/therapy , Cytokines/metabolism , Disease Management , Inflammation , Acute Kidney Injury/complications , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/therapy
16.
Nat Commun ; 7: 10274, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26817517

ABSTRACT

Crystals cause injury in numerous disorders, and induce inflammation via the NLRP3 inflammasome, however, it remains unclear how crystals induce cell death. Here we report that crystals of calcium oxalate, monosodium urate, calcium pyrophosphate dihydrate and cystine trigger caspase-independent cell death in five different cell types, which is blocked by necrostatin-1. RNA interference for receptor-interacting protein kinase 3 (RIPK3) or mixed lineage kinase domain like (MLKL), two core proteins of the necroptosis pathway, blocks crystal cytotoxicity. Consistent with this, deficiency of RIPK3 or MLKL prevents oxalate crystal-induced acute kidney injury. The related tissue inflammation drives TNF-α-related necroptosis. Also in human oxalate crystal-related acute kidney injury, dying tubular cells stain positive for phosphorylated MLKL. Furthermore, necrostatin-1 and necrosulfonamide, an inhibitor for human MLKL suppress crystal-induced cell death in human renal progenitor cells. Together, TNF-α/TNFR1, RIPK1, RIPK3 and MLKL are molecular targets to limit crystal-induced cytotoxicity, tissue injury and organ failure.


Subject(s)
Apoptosis , Calcium Oxalate/toxicity , Calcium Pyrophosphate/toxicity , Kidney Diseases/physiopathology , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Uric Acid/toxicity , Animals , Calcium Oxalate/chemistry , Calcium Oxalate/metabolism , Calcium Pyrophosphate/chemistry , Calcium Pyrophosphate/metabolism , Humans , Kidney Diseases/genetics , Kidney Diseases/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Necrosis , Phosphorylation , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Uric Acid/chemistry , Uric Acid/metabolism
17.
Am J Physiol Renal Physiol ; 310(8): F785-F795, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26764204

ABSTRACT

Chronic kidney disease (CKD) research is limited by the lack of convenient inducible models mimicking human CKD and its complications in experimental animals. We demonstrate that a soluble oxalate-rich diet induces stable stages of CKD in male and female C57BL/6 mice. Renal histology is characterized by tubular damage, remnant atubular glomeruli, interstitial inflammation, and fibrosis, with the extent of tissue involvement depending on the duration of oxalate feeding. Expression profiling of markers and magnetic resonance imaging findings established to reflect inflammation and fibrosis parallel the histological changes. Within 3 wk, the mice reproducibly develop normochromic anemia, metabolic acidosis, hyperkalemia, FGF23 activation, hyperphosphatemia, and hyperparathyroidism. In addition, the model is characterized by profound arterial hypertension as well as cardiac fibrosis that persist following the switch to a control diet. Together, this new model of inducible CKD overcomes a number of previous experimental limitations and should serve useful in research related to CKD and its complications.


Subject(s)
Disease Models, Animal , Hypertension/etiology , Oxalic Acid , Renal Insufficiency, Chronic/complications , Uremia/etiology , Animals , Fibroblast Growth Factor-23 , Fibrosis , Hypertension/pathology , Mice , Mice, Inbred C57BL , Myocardium/pathology , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/pathology , Uremia/pathology
18.
Eur J Immunol ; 46(1): 223-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26531064

ABSTRACT

Neutrophil extracellular trap (NET) formation contributes to gout, autoimmune vasculitis, thrombosis, and atherosclerosis. The outside-in signaling pathway triggering NET formation is unknown. Here, we show that the receptor-interacting protein kinase (RIPK)-1-stabilizers necrostatin-1 or necrostatin-1s and the mixed lineage kinase domain-like (MLKL)-inhibitor necrosulfonamide prevent monosodium urate (MSU) crystal- or PMA-induced NET formation in human and mouse neutrophils. These compounds do not affect PMA- or urate crystal-induced production of ROS. Moreover, neutrophils of chronic granulomatous disease patients are shown to lack PMA-induced MLKL phosphorylation. Genetic deficiency of RIPK3 in mice prevents MSU crystal-induced NET formation in vitro and in vivo. Thus, neutrophil death and NET formation may involve the signaling pathway defining necroptosis downstream of ROS production. These data imply that RIPK1, RIPK3, and MLKL could represent molecular targets in gout or other crystallopathies.


Subject(s)
Extracellular Traps/metabolism , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction/immunology , Animals , Blotting, Western , Extracellular Traps/immunology , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Neutrophils/immunology , Neutrophils/metabolism , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/toxicity , Polymethacrylic Acids/toxicity , Protein Kinases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Uric Acid/toxicity
19.
J Am Soc Nephrol ; 26(10): 2399-413, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25644111

ABSTRACT

Severe GN involves local neutrophil extracellular trap (NET) formation. We hypothesized a local cytotoxic effect of NET-related histone release in necrotizing GN. In vitro, histones from calf thymus or histones released by neutrophils undergoing NETosis killed glomerular endothelial cells, podocytes, and parietal epithelial cells in a dose-dependent manner. Histone-neutralizing agents such as antihistone IgG, activated protein C, or heparin prevented this effect. Histone toxicity on glomeruli ex vivo was Toll-like receptor 2/4 dependent, and lack of TLR2/4 attenuated histone-induced renal thrombotic microangiopathy and glomerular necrosis in mice. Anti-glomerular basement membrane GN involved NET formation and vascular necrosis, whereas blocking NET formation by peptidylarginine inhibition or preemptive anti-histone IgG injection significantly reduced all aspects of GN (i.e., vascular necrosis, podocyte loss, albuminuria, cytokine induction, recruitment or activation of glomerular leukocytes, and glomerular crescent formation). To evaluate histones as a therapeutic target, mice with established GN were treated with three different histone-neutralizing agents. Anti-histone IgG, recombinant activated protein C, and heparin were equally effective in abrogating severe GN, whereas combination therapy had no additive effects. Together, these results indicate that NET-related histone release during GN elicits cytotoxic and immunostimulatory effects. Furthermore, neutralizing extracellular histones is still therapeutic when initiated in established GN.


Subject(s)
Blood Vessels/pathology , Extracellular Traps/physiology , Glomerulonephritis/complications , Histones/physiology , Animals , Cattle , Extracellular Traps/drug effects , Glomerulonephritis/drug therapy , Glomerulonephritis/etiology , Histones/drug effects , Mice , Necrosis/etiology , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL
...