Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
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
2.
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
3.
Clin Immunol ; 185: 119-127, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29111236

ABSTRACT

Induction therapy of proliferative lupus nephritis still requires the use of unselective immunosuppressive drugs with significant toxicities. In search of more specific drugs with equal efficacy but fewer side effects we considered blocking pro-inflammatory chemokine monocyte chemoattractant protein-1 (MCP-1/CCL2) and homeostatic chemokine stromal cell-derived factor-1 (SDF-1/CXCL12), which both contribute to the onset and progression of proliferative lupus nephritis yet through different mechanisms. We hypothesized that dual antagonism could be as potent on lupus nephritis as the unselective immunosuppressant cyclophosphamide (CYC). We estimated serum levels of CCL2 and CXCL12 in patients with SLE (n=99) and compared the results with healthy individuals (n=21). In order to prove our hypothesis we used l-enantiomeric RNA Spiegelmer® chemokine antagonists, i.e. the CCL2-specific mNOX-E36 and the CXCL12-specific NOX-A12 to treat female MRL/lpr mice from week 12 to 20 of age with either anti-CXCL12 or anti-CCL2 alone or both. SLE patients showed elevated serum levels of CCL2 but not of CXCL12. Female MRL/lpr mice treated with dual blockade showed significantly more effective than either monotherapy in preventing proteinuria, immune complex glomerulonephritis, and renal excretory failure and the results are at par with CYC treatment. Dual blockade reduced leukocyte counts and renal IL-6, IL-12p40, CCL-5, CCL-2 and CCR-2 mRNA expression. Dual blockade of CCL2 and CXCL12 can be as potent as CYC to suppress the progression of proliferative lupus nephritis probably because the respective chemokine targets mediate different disease pathomechanisms, i.e. systemic autoimmunity and peripheral tissue inflammation.

4.
J Am Soc Nephrol ; 27(6): 1635-49, 2016 06.
Article in English | MEDLINE | ID: mdl-26567242

ABSTRACT

Endothelial dysfunction is a central pathomechanism in diabetes-associated complications. We hypothesized a pathogenic role in this dysfunction of cathepsin S (Cat-S), a cysteine protease that degrades elastic fibers and activates the protease-activated receptor-2 (PAR2) on endothelial cells. We found that injection of mice with recombinant Cat-S induced albuminuria and glomerular endothelial cell injury in a PAR2-dependent manner. In vivo microscopy confirmed a role for intrinsic Cat-S/PAR2 in ischemia-induced microvascular permeability. In vitro transcriptome analysis and experiments using siRNA or specific Cat-S and PAR2 antagonists revealed that Cat-S specifically impaired the integrity and barrier function of glomerular endothelial cells selectively through PAR2. In human and mouse type 2 diabetic nephropathy, only CD68(+) intrarenal monocytes expressed Cat-S mRNA, whereas Cat-S protein was present along endothelial cells and inside proximal tubular epithelial cells also. In contrast, the cysteine protease inhibitor cystatin C was expressed only in tubules. Delayed treatment of type 2 diabetic db/db mice with Cat-S or PAR2 inhibitors attenuated albuminuria and glomerulosclerosis (indicators of diabetic nephropathy) and attenuated albumin leakage into the retina and other structural markers of diabetic retinopathy. These data identify Cat-S as a monocyte/macrophage-derived circulating PAR2 agonist and mediator of endothelial dysfunction-related microvascular diabetes complications. Thus, Cat-S or PAR2 inhibition might be a novel strategy to prevent microvascular disease in diabetes and other diseases.


Subject(s)
Cathepsins/physiology , Diabetic Angiopathies/etiology , Endothelial Cells/enzymology , Receptor, PAR-2/metabolism , Animals , Cathepsins/antagonists & inhibitors , Cells, Cultured , Kidney Glomerulus/cytology , Male , Mice , Microvessels , Proline/analogs & derivatives , Proline/pharmacology , Urothelium/cytology
5.
Kidney Int ; 90(5): 933-935, 2016 11.
Article in English | MEDLINE | ID: mdl-27742196

ABSTRACT

Cathepsins are a class of proteases with diverse biological activities inside and, in part, also outside cells. Recent studies suggest that cathepsins are involved in the pathogenesis of diabetic nephropathy and in disease activity of autoimmune immune complex glomerulonephritis. Targeting specific cathepsins with suitable antagonists may hold promises for therapeutic interventions.


Subject(s)
Cathepsins/antagonists & inhibitors , Diabetic Nephropathies , Cathepsin L , Humans
6.
Clin Immunol ; 169: 139-147, 2016 08.
Article in English | MEDLINE | ID: mdl-27392463

ABSTRACT

Induction therapy of proliferative lupus nephritis still requires the use of unselective immunosuppressive drugs with significant toxicities. In search of more specific drugs with equal efficacy but fewer side effects we considered blocking pro-inflammatory chemokine monocyte chemoattractant protein-1 (MCP-1/CCL2) and homeostatic chemokine stromal cell-derived factor-1 (SDF-1/CXCL12), which both contribute to the onset and progression of proliferative lupus nephritis yet through different mechanisms. We hypothesized that dual antagonism could be as potent on lupus nephritis as the unselective immunosuppressant cyclophosphamide (CYC). We estimated serum levels of CCL2 and CXCL12 in patients with SLE (n=99) and compared the results with healthy individuals (n=21). In order to prove our hypothesis we used l-enantiomeric RNA Spiegelmer® chemokine antagonists, i.e. the CCL2-specific mNOX-E36 and the CXCL12-specific NOX-A12 to treat female MRL/lpr mice from week 12 to 20 of age with either anti-CXCL12 or anti-CCL2 alone or both. SLE patients showed elevated serum levels of CCL2 but not of CXCL12. Female MRL/lpr mice treated with dual blockade showed significantly more effective than either monotherapy in preventing proteinuria, immune complex glomerulonephritis, and renal excretory failure and the results are at par with CYC treatment. Dual blockade reduced leukocyte counts and renal IL-6, IL-12p40, CCL-5, CCL-2 and CCR-2 mRNA expression. Dual blockade of CCL2 and CXCL12 can be as potent as CYC to suppress the progression of proliferative lupus nephritis probably because the respective chemokine targets mediate different disease pathomechanisms, i.e. systemic autoimmunity and peripheral tissue inflammation.


Subject(s)
Chemokine CCL2/antagonists & inhibitors , Chemokine CXCL12/antagonists & inhibitors , Cyclophosphamide/pharmacology , Lupus Nephritis/drug therapy , Oligoribonucleotides/pharmacology , Adult , Animals , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression/genetics , Glomerulonephritis/prevention & control , Homeostasis/genetics , Humans , Immunosuppressive Agents/pharmacology , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Lupus Erythematosus, Systemic/blood , Lupus Nephritis/genetics , Lupus Nephritis/metabolism , Male , Mice, Inbred MRL lpr , Middle Aged , Oligoribonucleotides/genetics , Oligoribonucleotides/metabolism , Proteinuria/prevention & control , Reverse Transcriptase Polymerase Chain Reaction , Treatment Outcome
7.
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