Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Kidney Int ; 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38431215

ABSTRACT

Despite the recent advances in our understanding of the role of lipids, metabolites, and related enzymes in mediating kidney injury, there is limited integrated multi-omics data identifying potential metabolic pathways driving impaired kidney function. The limited availability of kidney biopsies from living donors with acute kidney injury has remained a major constraint. Here, we validated the use of deceased transplant donor kidneys as a good model to study acute kidney injury in humans and characterized these kidneys using imaging and multi-omics approaches. We noted consistent changes in kidney injury and inflammatory markers in donors with reduced kidney function. Neighborhood and correlation analyses of imaging mass cytometry data showed that subsets of kidney cells (proximal tubular cells and fibroblasts) are associated with the expression profile of kidney immune cells, potentially linking these cells to kidney inflammation. Integrated transcriptomic and metabolomic analysis of human kidneys showed that kidney arachidonic acid metabolism and seven other metabolic pathways were upregulated following diminished kidney function. To validate the arachidonic acid pathway in impaired kidney function we demonstrated increased levels of cytosolic phospholipase A2 protein and related lipid mediators (prostaglandin E2) in the injured kidneys. Further, inhibition of cytosolic phospholipase A2 reduced injury and inflammation in human kidney proximal tubular epithelial cells in vitro. Thus, our study identified cell types and metabolic pathways that may be critical for controlling inflammation associated with impaired kidney function in humans.

2.
Kidney Int ; 94(6): 1111-1126, 2018 12.
Article in English | MEDLINE | ID: mdl-30385042

ABSTRACT

Insufficient podocyte regeneration after injury is a central pathomechanism of glomerulosclerosis and chronic kidney disease. Podocytes constitutively secrete the chemokine CXCL12, which is known to regulate homing and activation of stem cells; hence we hypothesized a similar effect of CXCL12 on podocyte progenitors. CXCL12 blockade increased podocyte numbers and attenuated proteinuria in mice with Adriamycin-induced nephropathy. Similar studies in lineage-tracing mice revealed enhanced de novo podocyte formation from parietal epithelial cells in the setting of CXCL12 blockade. Super-resolution microscopy documented full integration of these progenitor-derived podocytes into the glomerular filtration barrier, interdigitating with tertiary foot processes of neighboring podocytes. Quantitative 3D analysis revealed that conventional 2D analysis underestimated the numbers of progenitor-derived podocytes. The 3D analysis also demonstrated differences between juxtamedullary and cortical nephrons in both progenitor endowment and Adriamycin-induced podocyte loss, with more robust podocyte regeneration in cortical nephrons with CXCL12 blockade. Finally, we found that delayed CXCL12 inhibition still had protective effects. In vitro studies found that CXCL12 inhibition uncoupled Notch signaling in podocyte progenitors. These data suggest that CXCL12-driven podocyte-progenitor feedback maintains progenitor quiescence during homeostasis, but also limits their intrinsic capacity to regenerate lost podocytes, especially in cortical nephrons. CXCL12 inhibition could be an innovative therapeutic strategy in glomerular disorders.


Subject(s)
Aptamers, Nucleotide/pharmacology , Chemokine CXCL12/antagonists & inhibitors , Glomerulosclerosis, Focal Segmental/drug therapy , Regeneration/drug effects , Stem Cells/drug effects , Animals , Aptamers, Nucleotide/therapeutic use , Cell Differentiation/drug effects , Cells, Cultured , Chemokine CXCL12/metabolism , Disease Models, Animal , Doxorubicin/toxicity , Feedback, Physiological/drug effects , Glomerulosclerosis, Focal Segmental/chemically induced , Glomerulosclerosis, Focal Segmental/complications , Humans , Imaging, Three-Dimensional , Male , Mice , Mice, Transgenic , Microscopy, Confocal/methods , Podocytes/drug effects , Podocytes/pathology , Proteinuria/drug therapy , Proteinuria/etiology , Stem Cells/physiology , Treatment Outcome
3.
Biosci Rep ; 37(6)2017 12 22.
Article in English | MEDLINE | ID: mdl-29054964

ABSTRACT

Notch and interleukin-22 (IL-22) signaling are known to regulate tissue homeostasis and respond to injury in humans and mice, and the induction of endogenous aryl hydrocarbon receptor (Ahr) ligands through Notch links the two pathways in a hierarchical fashion. However in adults, the species-, organ- and injury-specific gene expression of the Notch-AhR-IL22 axis components is unknown. We therefore performed gene expression profiling of DLL1, DLL3, DLL4, DLK1, DLK2, JAG1, JAG2, Notch1, Notch2, Notch3, Notch4, ADAM17/TNF-α ADAM metalloprotease converting enzyme (TACE), PSEN1, basigin (BSG)/CD147, RBP-J, HES1, HES5, HEY1, HEYL, AHR, ARNT, ARNT2, CYP1A1, CYP24A1, IL-22, IL22RA1, IL22RA2, IL10RB, and STAT3 under homeostatic conditions in ten mature murine and human organs. Additionally, the expression of these genes was assessed in murine models of acute sterile inflammation and progressive fibrosis. We show that there are organ-specific gene expression profiles of the Notch-AhR-IL22 axis in humans and mice. Although there is an overall interspecies congruency, specific differences between human and murine expression signatures do exist. In murine tissues with AHR/ARNT expression CYP1A1 and IL-22 were correlated with HES5 and HEYL expression, while in human tissues no such correlation was found. Notch and AhR signaling are involved in renal inflammation and fibrosis with specific gene expression changes in each model. Despite the presence of all Notch pathway molecules in the kidney and a model-specific induction of Notch ligands, IL-22 was only up-regulated in acute inflammation, but rapidly down-regulated during regeneration. This implies that for targeting injury responses, e.g. via IL-22, species-specific differences, injury type and time points have to be considered.


Subject(s)
Acute Kidney Injury/genetics , Inflammation/genetics , Interleukins/genetics , Receptors, Aryl Hydrocarbon/genetics , Acute Kidney Injury/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/economics , Basic Helix-Loop-Helix Transcription Factors/genetics , Cytochrome P-450 CYP1A1/genetics , Gene Expression Profiling/methods , Gene Expression Regulation , Humans , Inflammation/physiopathology , Mice , Receptors, Notch/genetics , Repressor Proteins/economics , Signal Transduction/genetics , Interleukin-22
4.
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
5.
J Am Soc Nephrol ; 28(1): 76-83, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27151924

ABSTRACT

CKD associates with systemic inflammation, but the underlying cause is unknown. Here, we investigated the involvement of intestinal microbiota. We report that collagen type 4 α3-deficient mice with Alport syndrome-related progressive CKD displayed systemic inflammation, including increased plasma levels of pentraxin-2 and activated antigen-presenting cells, CD4 and CD8 T cells, and Th17- or IFNγ-producing T cells in the spleen as well as regulatory T cell suppression. CKD-related systemic inflammation in these mice associated with intestinal dysbiosis of proteobacterial blooms, translocation of living bacteria across the intestinal barrier into the liver, and increased serum levels of bacterial endotoxin. Uremia did not affect secretory IgA release into the ileum lumen or mucosal leukocyte subsets. To test for causation between dysbiosis and systemic inflammation in CKD, we eradicated facultative anaerobic microbiota with antibiotics. This eradication prevented bacterial translocation, significantly reduced serum endotoxin levels, and fully reversed all markers of systemic inflammation to the level of nonuremic controls. Therefore, we conclude that uremia associates with intestinal dysbiosis, intestinal barrier dysfunction, and bacterial translocation, which trigger the state of persistent systemic inflammation in CKD. Uremic dysbiosis and intestinal barrier dysfunction may be novel therapeutic targets for intervention to suppress CKD-related systemic inflammation and its consequences.


Subject(s)
Bacterial Translocation , Dysbiosis , Inflammation/etiology , Inflammation/microbiology , Intestines/microbiology , Renal Insufficiency, Chronic/complications , Animals , Mice
6.
JCI Insight ; 1(17): e87877, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27777973

ABSTRACT

To derive new insights in diabetic complications, we integrated publicly available human protein-protein interaction (PPI) networks with global metabolic networks using metabolomic data from patients with diabetic nephropathy. We focused on the participating proteins in the network that were computationally predicted to connect the urine metabolites. MDM2 had the highest significant number of PPI connections. As validation, significant downregulation of MDM2 gene expression was found in both glomerular and tubulointerstitial compartments of kidney biopsy tissue from 2 independent cohorts of patients with diabetic nephropathy. In diabetic mice, chemical inhibition of MDM2 with Nutlin-3a led to reduction in the number of podocytes, increased blood urea nitrogen, and increased mortality. Addition of Nutlin-3a decreased WT1+ cells in embryonic kidneys. Both podocyte- and tubule-specific MDM2-knockout mice exhibited severe glomerular and tubular dysfunction, respectively. Interestingly, the only 2 metabolites that were reduced in both podocyte and tubule-specific MDM2-knockout mice were 3-methylcrotonylglycine and uracil, both of which were also reduced in human diabetic kidney disease. Thus, our bioinformatics tool combined with multi-omics studies identified an important functional role for MDM2 in glomeruli and tubules of the diabetic nephropathic kidney and links MDM2 to a reduction in 2 key metabolite biomarkers.


Subject(s)
Diabetic Nephropathies/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Systems Biology , Albuminuria , Animals , Computational Biology , Diabetes Mellitus, Experimental/metabolism , Humans , Kidney Glomerulus/physiopathology , Kidney Tubules/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Podocytes
7.
Nephrol Dial Transplant ; 31(9): 1541-5, 2016 09.
Article in English | MEDLINE | ID: mdl-27325253

ABSTRACT

Often the cause of refractory lupus nephritis (RLN) remains unclear. We performed next-generation sequencing for podocyte genes in an RLN patient and identified compound heterozygosity for APOL1 risk alleles G1 and G2 and a novel homozygous c.[1049C>T]+[1049C>T] NPHS1 gene variant of unknown significance. To test for causality renal progenitor cells isolated from urine of this patient were differentiated into podocytes in vitro. Podocytes revealed aberrant nephrin trafficking, cytoskeletal structure and lysosomal leakage, and increased detachment as compared with podocytes isolated from controls. Thus, lupus podocytopathy can be confirmed as a cause of RLN by functional genetics on patient-derived podocytes.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Kidney/physiopathology , Lupus Nephritis/diagnosis , Lupus Nephritis/urine , Podocytes/metabolism , Stem Cells/metabolism , Adolescent , Female , Humans , Lupus Nephritis/etiology , Podocytes/pathology , Recurrence , Stem Cells/pathology
8.
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
9.
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
10.
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
11.
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
12.
Stem Cell Reports ; 5(2): 248-63, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26235895

ABSTRACT

Podocyte loss is a general mechanism of glomerular dysfunction that initiates and drives the progression of chronic kidney disease, which affects 10% of the world population. Here, we evaluate whether the regenerative response to podocyte injury influences chronic kidney disease outcome. In models of focal segmental glomerulosclerosis performed in inducible transgenic mice where podocytes are tagged, remission or progression of disease was determined by the amount of regenerated podocytes. When the same model was established in inducible transgenic mice where renal progenitors are tagged, the disease remitted if renal progenitors successfully differentiated into podocytes, while it persisted if differentiation was ineffective, resulting in glomerulosclerosis. Treatment with BIO, a GSK3s inhibitor, significantly increased disease remission by enhancing renal progenitor sensitivity to the differentiation effect of endogenous retinoic acid. These results establish renal progenitors as critical determinants of glomerular disease outcome and a pharmacological enhancement of their differentiation as a possible therapeutic strategy.


Subject(s)
Cell Differentiation , Podocytes/cytology , Regeneration , Renal Insufficiency, Chronic/pathology , Stem Cells/cytology , Animals , Cells, Cultured , Glycogen Synthase Kinase 3/antagonists & inhibitors , Indoles/pharmacology , Indoles/therapeutic use , Mice , Mice, Inbred C57BL , Oximes/pharmacology , Oximes/therapeutic use , Podocytes/drug effects , Podocytes/metabolism , Renal Insufficiency, Chronic/drug therapy , Stem Cells/drug effects , Stem Cells/metabolism
13.
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
14.
Lab Invest ; 95(3): 273-82, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25531564

ABSTRACT

The metabolic and hemodynamic alterations in diabetes activate podocytes to increase extracellular matrix (ECM) production, leading to thickening of the glomerular basement membrane (GBM). We hypothesized that diabetes would activate parietal epithelial cells (PECs) in a similar manner and cause thickening of Bowman's capsules. Periodic acid Schiff staining of human kidney biopsies of 30 patients with diabetic nephropathy (DN) revealed a significantly thicker Bowman's capsule as compared with 20 non-diabetic controls. The average thickness was 4.55±0.21 µm in the group of patients with DN compared with 2.92±0.21 µm in the group of non-diabetic controls (P<0.001). Transmission electron microscopy confirmed this finding. In vitro, short-term exposure of human PECs to hyperglycemic conditions (30 mM glucose) advanced glycation end products (100 µg/ml) or transforming growth factor-ß1 (TGF-ß1; 5 ng/ml) increased the mRNA expression of collagen type I α-1, collagen type IV (all six α-chains), bamacan, nidogen 1, laminin α-1, and perlecan. Western blot and colorimetric collagen assays confirmed these results for collagen type IV at the protein level. The production and secretion of TGF-ß1 as a possible positive feedback loop was excluded as a mechanism for the autocrine activation of human PECs. To validate these findings in vivo, activation of the PECs was assessed by immunohistochemical staining for CD44 of 12 human biopsy cases with DN. Thickening of the Bowman's capsule showed strong association with CD44-positive PECs. In summary, metabolic alterations in diabetes activate PECs to increase the expression and secretion of Bowman's capsule proteins. This process may contribute to the thickening of the Bowman's capsule, similar to the thickening of the GBM that is driven by activated podocytes. These data may also imply that activated PECs contribute to ECM production once they migrate to the glomerular tuft, a process resulting in glomerular scaring, for example, in diabetic glomerulosclerosis.


Subject(s)
Bowman Capsule/metabolism , Collagen/metabolism , Diabetic Nephropathies/metabolism , Epithelial Cells/metabolism , Kidney Glomerulus/metabolism , Adult , Blotting, Western , Bowman Capsule/pathology , Cells, Cultured , Collagen/genetics , Collagen Type I, alpha 1 Chain , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Epithelial Cells/pathology , Epithelial Cells/ultrastructure , Female , Gene Expression/drug effects , Glucose/pharmacology , Glycation End Products, Advanced/pharmacology , Humans , Hyaluronan Receptors/metabolism , Immunohistochemistry , Kidney Glomerulus/pathology , Male , Microscopy, Electron, Transmission , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology
15.
J Am Soc Nephrol ; 26(7): 1513-23, 2015 07.
Article in English | MEDLINE | ID: mdl-25349197

ABSTRACT

Murine double minute-2 (MDM2), an E3 ligase that regulates the cell cycle and inflammation, is highly expressed in podocytes. In podocyte injury, MDM2 drives podocyte loss by mitotic catastrophe, but the function of MDM2 in resting podocytes has not been explored. Here, we investigated the effects of podocyte MDM2 deletion in vitro and in vivo. In vitro, MDM2 knockdown by siRNA caused increased expression of p53 and podocyte death, which was completely rescued by coknockdown of p53. Apoptosis, pyroptosis, pyronecrosis, necroptosis, ferroptosis, and parthanatos were excluded as modes of occurrence for this p53-overactivation-related cell death (here referred to as podoptosis). Podoptosis was associated with cytoplasmic vacuolization, endoplasmic reticulum stress, and dysregulated autophagy (previously described as paraptosis). MDM2 knockdown caused podocyte loss and proteinuria in a zebrafish model, which was consistent with the phenotype of podocyte-specific MDM2-knockout mice that also showed the aforementioned ultrastructual podocyte abnormalities before and during progressive glomerulosclerosis. The phenotype of both animal models was entirely rescued by codeletion of p53. We conclude that MDM2 maintains homeostasis and long-term survival in podocytes by preventing podoptosis, a p53-regulated form of cell death with unspecific features previously classified as paraptosis.


Subject(s)
Autophagy/genetics , Cell Death/genetics , Genes, p53/physiology , Glomerulosclerosis, Focal Segmental/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Transcriptional Activation/genetics , Analysis of Variance , Animals , Blotting, Western , Cell Survival/genetics , Cells, Cultured , Disease Models, Animal , Genes, p53/genetics , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/physiopathology , Homeostasis/genetics , Immunohistochemistry , Kidney Function Tests , Mice , Mice, Knockout , Microscopy, Confocal , Podocytes/cytology , Podocytes/physiology , Real-Time Polymerase Chain Reaction/methods , Sensitivity and Specificity , Transcriptional Activation/physiology , Zebrafish
16.
Methods Mol Biol ; 1169: 77-86, 2014.
Article in English | MEDLINE | ID: mdl-24957231

ABSTRACT

Foreign nucleic acids are recognized by germ-line-encoded receptors expressed in immune and nonimmune cells. Activation of the nucleic acid-specific pattern recognition receptors by foreign nucleic acid promotes production of inflammatory cytokines (mostly type I IFNs) and at the later stage leads to cell death. Here, we describe reliable and simple methods to quantify cell death caused by nucleic acid recognition. Additionally, we report two different methods to discriminate between two cell death modalities: apoptosis and necrosis.


Subject(s)
Nucleic Acids/pharmacology , Animals , Apoptosis/drug effects , Cell Death/drug effects , Cells, Cultured , Flow Cytometry , In Vitro Techniques , Mice , Receptors, Pattern Recognition/metabolism
17.
J Am Soc Nephrol ; 25(5): 978-89, 2014 May.
Article in English | MEDLINE | ID: mdl-24459235

ABSTRACT

AKI involves early Toll-like receptor (TLR)-driven immunopathology, and resolution of inflammation is needed for rapid regeneration of injured tubule cells. Notably, activation of TLRs also has been implicated in epithelial repair. We hypothesized that TLR signaling drives tubule regeneration after acute injury through the induction of certain ILs. Systematic screening in vitro identified IL-22 as a candidate proregeneratory factor in primary tubular cell recovery, and IL-22 deficiency or IL-22 blockade impaired post-ischemic tubular recovery after AKI in mice. Interstitial mononuclear cells, such as dendritic cells and macrophages, were the predominant source of IL-22 secretion, whereas IL-22 receptor was expressed by tubular epithelial cells exclusively. Depleting IL-22-producing cells during the healing phase impaired epithelial recovery, which could be rescued entirely by reconstituting mice with IL-22. In vitro, necrotic tubular cells and oxidative stress induced IL-22 secretion selectively through TLR4. Although TLR4 blockade during the early injury phase prevented tubular necrosis and AKI, TLR4 blockade during the healing phase suppressed IL-22 production and impaired kidney regeneration. Taken together, these results suggest that necrotic cell-derived TLR4 agonists activate intrarenal mononuclear cells to secrete IL-22, which accelerates tubular regeneration and recovery in AKI.


Subject(s)
Acute Kidney Injury/therapy , Interleukins/biosynthesis , Kidney Tubules/pathology , Regeneration/physiology , Reperfusion Injury/therapy , Toll-Like Receptor 4/physiology , Urothelium/pathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukins/metabolism , Janus Kinases/metabolism , Kidney Tubules/cytology , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Mononuclear Phagocyte System/metabolism , Mononuclear Phagocyte System/pathology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , STAT3 Transcription Factor/metabolism , Urothelium/cytology , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL
...