TY - JOUR A1 - von Websky, Karoline A1 - Reichetzeder, Christoph A1 - Hocher, Berthold T1 - Physiology and pathophysiology of incretins in the kidney JF - Current opinion in nephrology and hypertension : reviews of all advances, evaluations of key references, comprehensive listing of papers N2 - Purpose of reviewIncretin-based therapy with glucagon-like peptide-1 receptor (GLP-1R) agonists and dipeptidyl peptidase-4 (DPP-4) inhibitors is considered a promising therapeutic option for type 2 diabetes mellitus. Cumulative evidence, mainly from preclinical animal studies, reveals that incretin-based therapies also may elicit beneficial effects on kidney function. This review gives an overview of the physiology, pathophysiology, and pharmacology of the renal incretin system.Recent findingsActivation of GLP-1R in the kidney leads to diuretic and natriuretic effects, possibly through direct actions on renal tubular cells and sodium transporters. Moreover, there is evidence that incretin-based therapy reduces albuminuria, glomerulosclerosis, oxidative stress, and fibrosis in the kidney, partially through GLP-1R-independent pathways. Molecular mechanisms by which incretins exert their renal effects are understood incompletely, thus further studies are needed.SummaryThe GLP-1R and DPP-4 are expressed in the kidney in various species. The kidney plays an important role in the excretion of incretin metabolites and most GLP-1R agonists and DPP-4 inhibitors, thus special attention is required when applying incretin-based therapy in renal impairment. Preclinical observations suggest direct renoprotective effects of incretin-based therapies in the setting of hypertension and other disorders of sodium retention, as well as in diabetic and nondiabetic nephropathy. Clinical studies are needed in order to confirm translational relevance from preclinical findings for treatment options of renal diseases. KW - DDP-4 inhibition KW - diabetes KW - diabetic nephropathy KW - GLP-1 receptor KW - hypertension KW - incretins KW - kidney KW - renal impairment Y1 - 2014 U6 - https://doi.org/10.1097/01.mnh.0000437542.77175.a0 SN - 1062-4821 SN - 1473-6543 VL - 23 IS - 1 SP - 54 EP - 60 PB - Lippincott Williams & Wilkins CY - Philadelphia ER - TY - JOUR A1 - Raila, Jens A1 - Schweigert, Florian J. A1 - Kohn, Barbara T1 - Relationship between urinary Tamm-Horsfall protein excretion and renal function in dogs with naturally occurring renal disease JF - Veterinary clinical pathology N2 - Background Tamm-Horsfall protein (THP) is physiologically excreted in urine, but little is known about the role of THP in the diagnosis of renal disease in dogs. Objective The aim of this study was to evaluate to which extent naturally occurring renal disease affects the urinary excretion of THP. Methods Dogs were divided into 5 groups according to plasma creatinine concentration, urinary protein-to-creatinine ratio (UP/UC), and exogenous plasma creatinine clearance (P-ClCr) rates: Group A (healthy control dogs; n=8), nonazotemic and nonproteinuric dogs, with P-ClCr rates > 90mL/min/m2; group B (n=25), nonazotemic and nonproteinuric dogs with reduced P-ClCr rates (51-89mL/min/m2); group C (n=7), nonazotemic but proteinuric dogs with P-ClCr rates 53-98mL/min/m2; group D (n=8), azotemic and borderline proteinuric dogs (P-ClCr rates: 22-45mL/min/m2); and group E (n=15), azotemic and proteinuric dogs (not tested for P-ClCr). THP was measured by quantitative Western blot analysis, and the ratio of THP-to-urinary creatinine (THP/UC) was calculated. Results The THP/UC concentrations were not different among dogs of groups A-D, but were reduced in dogs of group E (P<.001). THP/UC correlated negatively with serum creatinine (P<.01) and UP/UC (P<.01), but was not significantly associated with P-ClCr. Conclusions Decreased levels of THP/UC were present in moderately to severely azotemic and proteinuric dogs. This suggests tubular injury in these dogs and that THP might be useful as urinary marker to study the pathogenesis of renal disease. KW - Distal tubules KW - dog KW - kidney KW - lower nephron KW - proteinuria KW - urine Y1 - 2014 U6 - https://doi.org/10.1111/vcp.12143 SN - 0275-6382 SN - 1939-165X VL - 43 IS - 2 SP - 261 EP - 265 PB - Wiley-Blackwell CY - Hoboken ER - TY - JOUR A1 - Nair, Anil V. A1 - Hocher, Berthold A1 - Verkaart, Sjoerd A1 - van Zeeland, Femke A1 - Pfab, Thiemo A1 - Slowinski, Torsten A1 - Chen, You-Peng A1 - Schlingmann, Karl Peter A1 - Schaller, Andre A1 - Gallati, Sabina A1 - Bindels, Rene J. A1 - Konrad, Martin A1 - Hönderop, Joost G. T1 - Loss of insulin-induced activation of TRPM6 magnesium channels results in impaired glucose tolerance during pregnancy JF - Proceedings of the National Academy of Sciences of the United States of America N2 - Hypomagnesemia affects insulin resistance and is a risk factor for diabetes mellitus type 2 (DM2) and gestational diabetes mellitus (GDM). Two single nucleotide polymorphisms (SNPs) in the epithelial magnesium channel TRPM6 ((VI)-I-1393, (KE)-E-1584) were predicted to confer susceptibility for DM2. Here, we show using patch clamp analysis and total internal reflection fluorescence microscopy, that insulin stimulates TRPM6 activity via a phosphoinositide 3-kinase and Rac1-mediated elevation of cell surface expression of TRPM6. Interestingly, insulin failed to activate the genetic variants TRPM6 ((VI)-I-1393) and TRPM6((KE)-E-1584), which is likely due to the inability of the insulin signaling pathway to phosphorylate TRPM6(T-1391) and TRPM6(S-1583). Moreover, by measuring total glycosylated hemoglobin (TGH) in 997 pregnant women as a measure of glucose control, we demonstrate that TRPM6((VI)-I-1393) and TRPM6((KE)-E-1584) are associated with higher TGH and confer a higher likelihood of developing GDM. The impaired response of TRPM6((VI)-I-1393) and TRPM6((KE)-E-1584) to insulin represents a unique molecular pathway leading to GDM where the defect is located in TRPM6. KW - kidney KW - distal convoluted tubule KW - transient receptor potential KW - vesicular trafficking Y1 - 2012 U6 - https://doi.org/10.1073/pnas.1113811109 SN - 0027-8424 VL - 109 IS - 28 SP - 11324 EP - 11329 PB - National Acad. of Sciences CY - Washington ER - TY - THES A1 - Borschewski, Aljona T1 - Bedeutung der Interaktion von Calcineurin und SORLA für die Regulation des Na⁺,K⁺,2Cl⁻-Kotransporters (NKCC2) in der Niere T1 - Importance of the interaction between calcineurin and SORLA for the regulation of the renal Na⁺-K⁺-2Cl⁻−cotransporter (NKCC2) N2 - Der Na⁺-K⁺-2Cl⁻-Kotransporter (NKCC2) wird im distalen Nephron der Niere exprimiert. Seine Verteilung umfasst die Epithelien der medullären und kortikalen Teile der dicken aufsteigenden Henle-Schleife (Thick ascending limb, TAL) und die Macula densa. Resorptiver NaCl-Transport über den NKCC2 dient dem renalen Konzentrierungsmechanismus und reguliert systemisch auch Volumenstatus und Blutdruck. Die Aktivität des NKCC2 ist mit der Phosphorylierung seiner N-terminalen Aminosäurereste Serin 126 und Threonin 96/101 verbunden. Vermittelt wird diese durch die homologen Kinasen SPAK (SPS-related proline/alanine-rich kinase) und OSR1 (Oxidative stress responsive kinase 1), die hierzu ihrerseits phosphoryliert werden müssen. Der regulatorische Kontext dieser Kinasen ist mittlerweile gut charakterisiert. Über Mechanismen und Produkte, die den NKCC2 deaktivieren, war hingegen weniger bekannt. Ziel der Arbeit war daher zu untersuchen, welche Wege zur Deaktivierung des Transporters führen. Der intrazelluläre Sortierungsrezeptor SORLA (Sorting-protein-related receptor with A-type repeats) war zuvor in seiner Bedeutung für das Nephron charakterisiert worden. Ein SORLA-defizientes Mausmodell weist unter anderem eine stark verringerte NKCC2-Phosphorylierung auf. Unter osmotischem Stress können SORLA-defiziente Mäuse ihren Urin weniger effizient konzentrieren. Meine Resultate zeigen mit hochauflösender Technik, dass SORLA apikal im TAL lokalisiert ist und dass mit NKCC2 eine anteilige Kolokalisation besteht. Unter SORLA Defizienz war die für die NKCC2 Aktivität maßgebliche SPAK/OSR1-Phosphorylierung gegenüber dem Wildtyp nicht verändert. Jedoch war die ebenfalls im TAL exprimierte Phosphatase Calcineurin Aβ (CnAβ) per Western blot um das zweifache gesteigert. Parallel hierzu wurde immunhistochemisch die Kolokalisation von verstärktem CnAβ-Signal und NKCC2 bestätigt. Beide Befunde geben zusammen den Hinweis auf einen Bezug zwischen der reduzierten NKCC2-Phosphorylierung und der gesteigerten Präsenz von CnAβ bei SORLA Defizienz. Die parallel induzierte Überexpression von SORLA in HEK-Zellen zeigte entsprechend eine Halbierung der CnAβ Proteinmenge. SORLA steuert demzufolge sowohl die Abundanz als auch die zelluläre Verteilung der Phosphatase. Weiterhin ließ sich die Interaktion zwischen CnAβ und SORLA (intrazelluläre Domäne) mittels Co-Immunpräzipitation bzw. GST-pulldown assay nachweisen. Auch die Interaktion zwischen CnAβ und NKCC2 wurde auf diesem Weg belegt. Da allerdings weder SORLA noch NKCC2 ein spezifisches Bindungsmuster für CnAβ aufweisen, sind vermutlich intermediäre Adapterproteine bei ihrer Bindung involviert. Die pharmakologische Inhibition von CnAβ mittels Cyclosporin A (CsA; 1 h) führte bei SORLA Defizienz zur Normalisierung der NKCC2-Phosphorylierung. Entsprechend führte in vitro die Gabe von CsA bei TAL Zellen zu einer 7-fach gesteigerten NKCC2-Phosphorylierung. Zusammenfassend zeigen die Ergebnisse, dass die Phosphatase CnAβ über ihre Assoziation mit NKCC2 diesen im adluminalen Zellkompartiment deaktivieren kann. Gesteuert wird dieser Vorgang durch die Eigenschaft von SORLA, CnAβ apikal zu reduzieren und damit die adluminale Phosphorylierung und Aktivität von NKCC2 zu unterstützen. Da Calcineurin-Inhibitoren derzeit die Grundlage der immunsupprimierenden Therapie darstellen, haben die Ergebnisse eine klinische Relevanz. Angesichts der Co-Expression von SORLA und CnAβ in verschiedenen anderen Organen können die Ergebnisse auch über die Niere hinaus Bedeutung erlangen. N2 - The Na⁺-K⁺-2Cl⁻-cotransporter (NKCC2) of the thick ascending limb (TAL) is critical for renal salt handling. Activity of the cotransporter is facilitated by its phosphorylation at conserved N-terminal threonine and serine residues provided by homologous SPAK (SPS-related proline/alanine-rich kinase) and OSR1 (oxidative stress responsive kinase 1) kinases. The identification of factors which modulate the phosphorylation and hence, the activity of NKCC2 has received recent interest. SORLA (sorting-protein-related receptor with A-type repeats) is co-expressed with NKCC2 in TAL epithelium. Genetically engineered mice lacking SORLA show near-complete absence of NKCC2 phosphorylation at the SPAK/OSR1-dependent phosphoacceptors, indicating that SORLA acts as a mediator between these reaction partners, possibly by a cellular trafficking step involving additional molecules. The present study addresses molecular pathways modulating NKCC2 activity by phosphorylation or dephosphorylation reactions with a special focus on SORLA. Comparative evaluation of SORLA-deficient and wild-type mouse kidneys revealed similar levels of phosphorylated, catalytically active SPAK and OSR1 kinases, whereas abundance of the phosphatase calcineurin Aβ (CnAβ) was increased by approximately two-fold in the renal medulla of SORLA-deficient mice likely reflecting changes in TAL. In line with this, confocal microscopy revealed accumulation of CnAβ in the apical compartment of TAL in SORLA-deficient kidneys. In contrast, overexpression of SORLA in HEK cells resulted in approximately two-fold decreased CnAβ levels suggesting that SORLA modulates both the cellular abundance and distribution of the phosphatase. This data was further corroborated by co-immunoprecipitation and GST pull down assays which showed an interaction between the cytoplasmic tail of SORLA and CnAβ. Acute administration of the calcineurin inhibitor, cyclosporin A (CsA), for 1 h rapidly normalized NKCC2 phosphorylation in SORLA-deficient mice which demonstrates that the decrease in phospho-NKCC2 was functionally related with CnAβ. In sum, our data elucidate the role of calcineurin in the regulation of NKCC2 and establish SORLA as an endogenous regulator of the phosphatase. KW - Salztransport KW - Niere KW - Henlesche Schleife KW - Calcineurin KW - Phosphatase KW - SORLA KW - Calcineurin-Inhibitoren KW - Transporteraktivierung KW - Phosphorylierung KW - kidney KW - phosphorylation KW - thick ascending limb KW - epithelial salt transport KW - calcineurin inhibitors KW - cell signaling Y1 - 2015 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:kobv:517-opus4-89205 ER -