Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
N Engl J Med ; 382(5): 416-426, 2020 01 30.
Article in English | MEDLINE | ID: mdl-31995687

ABSTRACT

BACKGROUND: Acute kidney injury is common, with a major effect on morbidity and health care utilization. Soluble urokinase plasminogen activator receptor (suPAR) is a signaling glycoprotein thought to be involved in the pathogenesis of kidney disease. We investigated whether a high level of suPAR predisposed patients to acute kidney injury in multiple clinical contexts, and we used experimental models to identify mechanisms by which suPAR acts and to assess it as a therapeutic target. METHODS: We measured plasma levels of suPAR preprocedurally in patients who underwent coronary angiography and patients who underwent cardiac surgery and at the time of admission to the intensive care unit in critically ill patients. We assessed the risk of acute kidney injury at 7 days as the primary outcome and acute kidney injury or death at 90 days as a secondary outcome, according to quartile of suPAR level. In experimental studies, we used a monoclonal antibody to urokinase plasminogen activator receptor (uPAR) as a therapeutic strategy to attenuate acute kidney injury in transgenic mice receiving contrast material. We also assessed cellular bioenergetics and generation of reactive oxygen species in human kidney proximal tubular (HK-2) cells that were exposed to recombinant suPAR. RESULTS: The suPAR level was assessed in 3827 patients who were undergoing coronary angiography, 250 who were undergoing cardiac surgery, and 692 who were critically ill. Acute kidney injury developed in 318 patients (8%) who had undergone coronary angiography. The highest suPAR quartile (vs. the lowest) had an adjusted odds ratio of 2.66 (95% confidence interval [CI], 1.77 to 3.99) for acute kidney injury and 2.29 (95% CI, 1.71 to 3.06) for acute kidney injury or death at 90 days. Findings were similar in the surgical and critically ill cohorts. The suPAR-overexpressing mice that were given contrast material had greater functional and histologic evidence of acute kidney injury than wild-type mice. The suPAR-treated HK-2 cells showed heightened energetic demand and mitochondrial superoxide generation. Pretreatment with a uPAR monoclonal antibody attenuated kidney injury in suPAR-overexpressing mice and normalized bioenergetic changes in HK-2 cells. CONCLUSIONS: High suPAR levels were associated with acute kidney injury in various clinical and experimental contexts. (Funded by the National Institutes of Health and others.).


Subject(s)
Acute Kidney Injury/blood , Cardiac Surgical Procedures/adverse effects , Coronary Angiography/adverse effects , Receptors, Urokinase Plasminogen Activator/blood , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Acute Kidney Injury/etiology , Acute Kidney Injury/prevention & control , Aged , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Biomarkers/blood , Critical Illness , Disease Models, Animal , Female , Humans , Intensive Care Units , Kidney Tubules/cytology , Kidney Tubules/drug effects , Kidney Tubules/pathology , Male , Mice , Mice, Transgenic , Middle Aged , Odds Ratio , Podocytes/drug effects , Podocytes/metabolism , Postoperative Complications/blood , Postoperative Complications/etiology , Risk Assessment/methods , Urokinase-Type Plasminogen Activator/pharmacology
2.
J Clin Invest ; 129(4): 1713-1726, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30747722

ABSTRACT

Soluble urokinase receptor (suPAR) is a circulatory molecule that activates αvß3 integrin on podocytes, causes foot process effacement, and contributes to proteinuric kidney disease. While active integrin can be targeted by antibodies and small molecules, endogenous inhibitors haven't been discovered yet. Here we report what we believe is a novel renoprotective role for the inducible costimulator ligand (ICOSL) in early kidney disease through its selective binding to podocyte αvß3 integrin. Contrary to ICOSL's immune-regulatory role, ICOSL in nonhematopoietic cells limited the activation of αvß3 integrin. Specifically, ICOSL contains the arginine-glycine-aspartate (RGD) motif, which allowed for a high-affinity and selective binding to αvß3 and modulation of podocyte adhesion. This binding was largely inhibited either by a synthetic RGD peptide or by a disrupted RGD sequence in ICOSL. ICOSL binding favored the active αvß3 rather than the inactive form and showed little affinity for other integrins. Consistent with the rapid induction of podocyte ICOSL by inflammatory stimuli, glomerular ICOSL expression was increased in biopsies of early-stage human proteinuric kidney diseases. Icosl deficiency in mice resulted in an increased susceptibility to proteinuria that was rescued by recombinant ICOSL. Our work identified a potentially novel role for ICOSL, which serves as an endogenous αvß3-selective antagonist to maintain glomerular filtration.


Subject(s)
Inducible T-Cell Co-Stimulator Ligand , Integrin alphaVbeta3 , Kidney Failure, Chronic , Podocytes , Proteinuria , Amino Acid Motifs , Animals , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/genetics , Glomerular Filtration Rate/immunology , Humans , Inducible T-Cell Co-Stimulator Ligand/genetics , Inducible T-Cell Co-Stimulator Ligand/immunology , Inducible T-Cell Co-Stimulator Ligand/pharmacology , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/immunology , Kidney Failure, Chronic/drug therapy , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/immunology , Kidney Failure, Chronic/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Podocytes/immunology , Podocytes/pathology , Proteinuria/drug therapy , Proteinuria/genetics , Proteinuria/immunology , Proteinuria/pathology
3.
J Clin Invest ; 129(5): 1946-1959, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30730305

ABSTRACT

Soluble urokinase plasminogen activator receptor (suPAR) is an immune-derived circulating signaling molecule that has been implicated in chronic kidney disease, such as focal segmental glomerulosclerosis (FSGS). Typically, native uPAR (isoform 1) translates to a 3-domain protein capable of binding and activating integrins, yet the function of additional isoforms generated by alternative splicing is unknown. Here, we characterized mouse uPAR isoform 2 (msuPAR2), encoding domain I and nearly one-half of domain II, as a dimer in solution, as revealed by 3D electron microscopy structural analysis. In vivo, msuPAR2 transgenic mice exhibited signs of severe renal disease characteristic of FSGS with proteinuria, loss of kidney function, and glomerulosclerosis. Sequencing of the glomerular RNAs from msuPAR2-Tg mice revealed a differentially expressed gene signature that includes upregulation of the suPAR receptor Itgb3, encoding ß3 integrin. Crossing msuPAR2-transgenic mice with 3 different integrin ß3 deficiency models rescued msuPAR2-mediated kidney function. Further analyses indicated a central role for ß3 integrin and c-Src in msuPAR2 signaling and in human FSGS kidney biopsies. Administration of Src inhibitors reduced proteinuria in msuPAR2-transgenic mice. In conclusion, msuPAR2 may play an important role in certain forms of scarring kidney disease.


Subject(s)
Kidney Diseases/genetics , Receptors, Urokinase Plasminogen Activator/chemistry , Receptors, Urokinase Plasminogen Activator/genetics , Adipocytes/cytology , Animals , Biopsy , Disease Models, Animal , HEK293 Cells , Humans , Mice , Mice, Transgenic , Microscopy, Electron , Podocytes/cytology , Protein Domains , Protein Isoforms , Protein Multimerization , Receptor, PAR-2/genetics , Retrospective Studies , Signal Transduction
4.
J Am Soc Nephrol ; 29(2): 409-415, 2018 02.
Article in English | MEDLINE | ID: mdl-29061651

ABSTRACT

Transient receptor potential channel 5 (TRPC5) is highly expressed in brain and kidney and mediates calcium influx and promotes cell migration. In the kidney, loss of TRPC5 function has been reported to benefit kidney filter dynamics by balancing podocyte cytoskeletal remodeling. However, in vivo gain-in-function studies of TRPC5 with respect to kidney function have not been reported. To address this gap, we developed two transgenic mouse models on the C57BL/6 background by overexpressing either wild-type TRPC5 or a TRPC5 ion-pore mutant. Compared with nontransgenic controls, neither transgenic model exhibited an increase in proteinuria at 8 months of age or a difference in LPS-induced albuminuria. Moreover, activation of TRPC5 by Englerin A did not stimulate proteinuria, and inhibition of TRPC5 by ML204 did not significantly lower the level of LPS-induced proteinuria in any group. Collectively, these data suggest that the overexpression or activation of the TRPC5 ion channel does not cause kidney barrier injury or aggravate such injury under pathologic conditions.


Subject(s)
Albuminuria/genetics , Kidney Diseases/genetics , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Albuminuria/chemically induced , Animals , Brain/metabolism , Female , Indoles/pharmacology , Kidney Diseases/chemically induced , Kidney Diseases/mortality , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Mice, Transgenic , Piperidines/pharmacology , Podocytes/ultrastructure , Sesquiterpenes, Guaiane/pharmacology , TRPC Cation Channels/agonists , TRPC Cation Channels/antagonists & inhibitors
6.
J Diabetes Res ; 2017: 3232848, 2017.
Article in English | MEDLINE | ID: mdl-28596971

ABSTRACT

Diabetic nephropathy (DN) is the leading cause of end-stage renal disease (ESRD) worldwide. DN typically manifests by glomerular hyperfiltration and microalbuminuria; then, the disease progresses to impaired glomerular filtration rate, which leads to ESRD. Treatment options for DN include the strict control of blood glucose levels and pressure (e.g., intraglomerular hypertension). However, the search for novel therapeutic strategies is ongoing. These include seeking specific molecules that contribute to the development and progression of DN to potentially interfere with these "molecular targets" as well as with the cellular targets within the kidney such as podocytes, which play a major role in the pathogenesis of DN. Recently, podocyte membrane protein urokinase receptor (uPAR) and its circulating form (suPAR) are found to be significantly induced in glomeruli and sera of DN patients, respectively, and elevated suPAR levels predicted diabetic kidney disease years before the occurrence of microalbuminuria. The intent of this review is to summarize the emerging evidence of uPAR and suPAR in the clinical manifestations of DN. The identification of specific pathways that govern DN will help us build a more comprehensive molecular model for the pathogenesis of the disease that can inform new opportunities for treatment.


Subject(s)
Diabetes Mellitus/metabolism , Kidney Failure, Chronic/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Albuminuria/metabolism , Animals , Diabetic Nephropathies/metabolism , Disease Progression , Glomerular Filtration Rate , Humans , Hypertension/complications , Integrins/metabolism , Kidney Glomerulus/physiopathology , Models, Biological , Podocytes/metabolism , Signal Transduction
7.
Nat Med ; 23(8): 945-953, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28650456

ABSTRACT

Soluble urokinase plasminogen activator receptor (suPAR) independently predicts chronic kidney disease (CKD) incidence and progression. Apolipoprotein L1 (APOL1) gene variants G1 and G2, but not the reference allele (G0), are associated with an increased risk of CKD in individuals of recent African ancestry. Here we show in two large, unrelated cohorts that decline in kidney function associated with APOL1 risk variants was dependent on plasma suPAR levels: APOL1-related risk was attenuated in patients with lower suPAR, and strengthened in those with higher suPAR levels. Mechanistically, surface plasmon resonance studies identified high-affinity interactions between suPAR, APOL1 and αvß3 integrin, whereby APOL1 protein variants G1 and G2 exhibited higher affinity for suPAR-activated avb3 integrin than APOL1 G0. APOL1 G1 or G2 augments αvß3 integrin activation and causes proteinuria in mice in a suPAR-dependent manner. The synergy of circulating factor suPAR and APOL1 G1 or G2 on αvß3 integrin activation is a mechanism for CKD.


Subject(s)
Apolipoproteins/genetics , Integrin alphaVbeta3/metabolism , Lipoproteins, HDL/genetics , Podocytes/metabolism , Proteinuria/genetics , Receptors, Urokinase Plasminogen Activator/metabolism , Renal Insufficiency, Chronic/genetics , Adolescent , Adult , Black or African American , Aged , Alleles , Animals , Apolipoprotein L1 , Apolipoproteins/metabolism , Cohort Studies , Female , Genetic Predisposition to Disease , Genotype , Humans , Lipoproteins, HDL/metabolism , Male , Mice , Middle Aged , Proteinuria/metabolism , Renal Insufficiency, Chronic/metabolism , Surface Plasmon Resonance , Young Adult
8.
Phys Chem Chem Phys ; 15(23): 8908-15, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23493863

ABSTRACT

A key factor in the development of type II diabetes is the loss of insulin-producing beta-cells. Human islet amyloid polypeptide protein (human-IAPP) is believed to play a crucial role in this process by forming small aggregates that exhibit toxicity by disrupting the cell membrane. The actual mechanism of membrane disruption is complex and appears to involve an early component before fiber formation and a later component associated with fiber formation on the membrane. By comparing the peptide-lipid interactions derived from solid-state NMR experiments of two IAPP fragments that cause membrane disordering to IAPP derived peptides known to cause significant early membrane permeabilization, we show here that membrane disordering is not likely to be sufficient by itself to cause the early membrane permeabilization observed by IAPP, and may play a lesser role in IAPP membrane disruption than expected.


Subject(s)
Amyloid/metabolism , Cell Membrane/metabolism , Diabetes Mellitus, Type 2/metabolism , Islet Amyloid Polypeptide/metabolism , Unilamellar Liposomes/metabolism , Amyloid/chemistry , Animals , Cell Membrane/chemistry , Cell Membrane/pathology , Cell Membrane Permeability , Diabetes Mellitus, Type 2/pathology , Humans , Islet Amyloid Polypeptide/chemistry , Rats , Unilamellar Liposomes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...