Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
J Microsc ; 268(2): 129-140, 2017 11.
Article in English | MEDLINE | ID: mdl-28806482

ABSTRACT

The actin cytoskeleton is a main component of cells and it is crucially involved in many physiological processes, e.g. cell motility. Changes in the actin organization can be effected by diseases or vice versa. Due to the nonuniform pattern, it is difficult to quantify reasonable features of the actin cytoskeleton for a significantly high cell number. Here, we present an approach capable to fully segment and analyse the actin cytoskeleton of 2D fluorescence microscopic images with a special focus on stress fibres. The extracted feature data include length, width, orientation and intensity distributions of all traced stress fibres. Our approach combines morphological image processing techniques and a trace algorithm in an iterative manner, classifying the segmentation result with respect to the width of the stress fibres and in nonfibre-like actin. This approach enables us to capture experimentally induced processes like the condensation or the collapse of the actin cytoskeleton. We successfully applied the algorithm to F-actin images of cells that were treated with the actin polymerization inhibitor latrunculin A. Furthermore, we verified the robustness of our algorithm by a sensitivity analysis of the parameters, and we benchmarked our algorithm against established methods. In summary, we present a new approach to segment actin stress fibres over time to monitor condensation or collapse processes.


Subject(s)
Actins/analysis , Automation, Laboratory/methods , Image Processing, Computer-Assisted/methods , Microscopy, Fluorescence/methods , Podocytes/chemistry , Podocytes/ultrastructure , Stress Fibers/ultrastructure , Algorithms , Animals , Cell Line , Mice
2.
Acta Physiol (Oxf) ; 220(1): 167-173, 2017 05.
Article in English | MEDLINE | ID: mdl-27414464

ABSTRACT

AIM: Zebrafish larvae with their simplified pronephros are an ideal model to study glomerular physiology. Although several groups use zebrafish larvae to assess glomerular barrier function, temporary or slight changes are still difficult to measure. The aim of this study was to investigate the potential of in vivo two-photon microscopy (2-PM) for long-term imaging of glomerular barrier function in zebrafish larvae. METHODS: As a proof of principle, we adapted the nitroreductase/metronidazole model of targeted podocyte ablation for 2-PM. Combination with a strain, which expresses eGFP-vitamin D-binding protein in the blood plasma, led to a strain that allowed induction of podocyte injury with parallel assessment of glomerular barrier function. We used four-dimensional (4D) 2-PM to assess eGFP fluorescence over 26 h in the vasculature and in tubules of multiple zebrafish larvae (5 days post-fertilization) simultaneously. RESULTS: By 4D 2-PM, we observed that, under physiological conditions, eGFP fluorescence was retained in the vasculature and rarely detected in proximal tubule cells. Application of metronidazole induced podocyte injury and cell death as shown by TUNEL staining. Induction of podocyte injury resulted in a dramatic decrease of eGFP fluorescence in the vasculature over time (about 50% and 90% after 2 and 12 h respectively). Loss of vascular eGFP fluorescence was paralleled by an endocytosis-mediated accumulation of eGFP fluorescence in proximal tubule cells, indicating proteinuria. CONCLUSION: We established a microscopy-based method to monitor the dynamics of glomerular barrier function during induction of podocyte injury in multiple zebrafish larvae simultaneously over 26 h.


Subject(s)
Kidney Glomerulus/physiopathology , Microscopy/methods , Optical Imaging/methods , Animals , Disease Models, Animal , Proteinuria , Zebrafish
3.
Am J Physiol Renal Physiol ; 308(6): F602-13, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25503725

ABSTRACT

Elevated glomerular pressure represents a high risk for the development of severe kidney diseases and causes an increase in mechanical load to podocytes. In this study, we investigated whether mechanical stress alters gene expression in cultured podocytes using gene arrays. We found that tetraspanin CD9 is significantly upregulated in cultured podocytes after mechanical stress. The differential expression of CD9 was confirmed by RT-PCR and Western blotting under stretched and unstretched conditions. Furthermore, mechanical stress resulted in a relocalization of CD9. To get an insight into the functional role of CD9, podocytes were transfected with pEGFP-CD9. The expression of CD9 induced the formation of substratum-attached thin arborized protrusions. Ca(2+) depletion revealed that podocytes overexpressing CD9 possess altered adhesive properties in contrast to the control transfected cells. Finally, elevated CD9 expression increased migration of podocytes in a wound assay. In summary, our results suggest that upregulation of CD9 may play an important role in podocyte morphology, adhesion, and migration.


Subject(s)
Podocytes/metabolism , Stress, Mechanical , Tetraspanin 29/metabolism , Actin Cytoskeleton/metabolism , Animals , Calcium/metabolism , Cell Line , Cell Movement , Fluorescent Antibody Technique , Mice , Podocytes/cytology , Up-Regulation
4.
Diabetologia ; 52(11): 2445-54, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19707743

ABSTRACT

AIMS/HYPOTHESIS: Chemokine (C-X-C motif) ligand 12 (CXCL12) (also known as stromal cell-derived factor-1 [SDF-1]-alpha) is a homeostatic chemokine with multiple roles in cell homing, tumour metastasis, angiogenesis and tissue regeneration after acute injuries. However, its role in chronic diseases remains poorly defined, e.g. in chronic glomerular diseases like diabetic glomerulosclerosis. We hypothesised that CXCL12 may have a functional role during the evolution of diabetic glomerulosclerosis, either by assisting glomerular repair or by supporting the maladaptive tissue remodelling in response to hyperglycaemia and glomerular hyperfiltration. METHODS: To define the functional role of CXCL12 in the progression of glomerular disease, we used the CXCL12-specific inhibitor NOX-A12, an L: -enantiomeric RNA oligonucleotide (Spiegelmer). A mouse model of type 2 diabetes (db/db mice) was used. Male db/db mice, uni-nephrectomised at 6 weeks of age, received subcutaneous injections with a PEGylated form of NOX-A12, non-functional control Spiegelmer or vehicle on alternate days from 4 to 6 months of age. RESULTS: Immunostaining localised renal CXCL12 production to glomerular podocytes in db/db mice with early or advanced diabetic nephropathy. CXCL12 inhibition significantly reduced the degree of glomerulosclerosis, increased the number of podocytes, prevented the onset of albuminuria and maintained the peritubular vasculature without affecting blood glucose levels, body weight or glomerular macrophage infiltration. CONCLUSIONS/INTERPRETATION: We conclude that podocytes produce CXCL12, which contributes to proteinuria and glomerulosclerosis in our mouse model of type 2 diabetes. This novel pathomechanism provides the first evidence that CXCL12 could be a therapeutic target in (diabetic) glomerulosclerosis.


Subject(s)
Chemokine CXCL12/biosynthesis , Diabetes Mellitus, Type 2/physiopathology , Diabetic Nephropathies/physiopathology , Podocytes/physiology , Albuminuria/epidemiology , Animals , Base Sequence , Chemokine CXCL12/genetics , Chemokine CXCL12/physiology , DNA Primers , Diabetic Nephropathies/pathology , Disease Models, Animal , Disease Progression , Inflammation/physiopathology , Kidney Glomerulus/pathology , Kidney Glomerulus/physiology , Male , Mice , Mice, Inbred C57BL , Nephrectomy , Podocytes/pathology , RNA/genetics , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction
5.
Br J Pharmacol ; 134(6): 1113-36, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11704631

ABSTRACT

The cloning of the so-called 'parathyroid hormone-related protein' (PTHrP) in 1987 was the result of a long quest for the factor which, by mimicking the actions of PTH in bone and kidney, is responsible for the hypercalcemic paraneoplastic syndrome, humoral calcemia of malignancy. PTHrP is distinct from PTH in a number of ways. First, PTHrP is the product of a separate gene. Second, with the exception of a short N-terminal region, the structure of PTHrP is not closely related to that of PTH. Third, in contrast to PTH, PTHrP is a paracrine factor expressed throughout the body. Finally, most of the functions of PTHrP have nothing in common with those of PTH. PTHrP is a poly-hormone which comprises a family of distinct peptide hormones arising from post-translational endoproteolytic cleavage of the initial PTHrP translation products. Mature N-terminal, mid-region and C-terminal secretory forms of PTHrP are thus generated, each of them having their own physiologic functions and probably their own receptors. The type 1 PTHrP receptor, binding both PTH(1-34) and PTHrP(1-36), is the only cloned receptor so far. PTHrP is a PTH-like calciotropic hormone, a myorelaxant, a growth factor and a developmental regulatory molecule. The present review reports recent aspects of PTHrP pharmacology and physiology, including: (a) the identification of new peptides and receptors of the PTH/PTHrP system; (b) the recently discovered nuclear functions of PTHrP and the role of PTHrP as an intracrine regulator of cell growth and cell death; (c) the physiological and developmental actions of PTHrP in the cardiovascular and the renal glomerulo-vascular systems; (d) the role of PTHrP as a regulator of pancreatic beta cell growth and functions, and, (e) the interactions of PTHrP and calcium-sensing receptors for the control of the growth of placental trophoblasts. These new advances have contributed to a better understanding of the pathophysiological role of PTHrP, and will help to identify its therapeutic potential in a number of diseases.


Subject(s)
Islets of Langerhans/metabolism , Parathyroid Hormone/physiology , Proteins/physiology , Receptors, Parathyroid Hormone/physiology , Animals , Apoptosis , Calcium-Binding Proteins/metabolism , Cardiovascular System/metabolism , Cell Nucleus/metabolism , Female , Humans , Kidney/metabolism , Mice , Nuclear Localization Signals , Parathyroid Hormone/genetics , Parathyroid Hormone-Related Protein , Placenta/metabolism , Pregnancy , Proteins/genetics , Rats , Receptors, Parathyroid Hormone/genetics , Trophoblasts/metabolism
6.
Exp Nephrol ; 9(6): 436-43, 2001.
Article in English | MEDLINE | ID: mdl-11702004

ABSTRACT

Podocyte function appears to be regulated by vasoactive factors. In vivo podocytes express parathyroid hormone-related protein (PTHrP), the N-terminal fragment of which has vasoactive properties. Since the signaling pathway(s) of PTHrP(1-36) are unknown in podocytes, differentiated cells of a conditionally immortalized mouse podocyte cell line were studied. Gene expression of PTHrP and the PTH/PTHrP receptor was investigated by RT-PCR; protein distribution of PTHrP was examined by immunofluorescence. Accumulation of cAMP was determined by an enzyme immunoassay; [Ca2+]i was measured by fura-2 ratio imaging. PTHrP and PTH/PTHrP receptor mRNA was detected in differentiated podocytes. Immunoreactive PTHrP exhibited a granular distribution in the cytoplasm of differentiated podocytes. With regard to the signaling pathway(s) of PTHrP(1-36), a concentration-dependent increase of cAMP levels with an EC50 value of 4 +/- 2 nM was found. PTHrP(1-36) (1 microM) increased cAMP levels 5.5 +/- 1.1-fold above baseline as compared with a 25.4 +/- 4.2-fold increase in response to forskolin (10 microM). The PTH/PTHrP receptor antagonist PTHrP(7-34) significantly diminished the PTHrP(1-36)-induced cAMP increase. While superfusion of podocytes with bradykinin (100 nM) increased [Ca2+]i, PTHrP(1-36) (100 nM) was without effect on [Ca2+]i. However, PTHrP(1-36) attenuated the bradykinin-induced increase in [Ca2+]i. Our results suggest that PTHrP is an autocrine hormone in podocytes, which selectively activates the cAMP pathway through the PTH/PTHrP receptor.


Subject(s)
Kidney Glomerulus/metabolism , Proteins/physiology , Signal Transduction , Animals , Calcium/metabolism , Cell Line, Transformed , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/metabolism , Fluorescent Antibody Technique , Immunoenzyme Techniques , Intracellular Membranes/metabolism , Kidney Glomerulus/cytology , Mice , Mice, Transgenic , Osmolar Concentration , Parathyroid Hormone-Related Protein , Peptide Fragments/pharmacology , Peptide Fragments/physiology , Proteins/metabolism , Proteins/pharmacology , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/metabolism , Tissue Distribution
7.
Am J Physiol Renal Physiol ; 281(4): F769-77, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11553524

ABSTRACT

Mice lacking the 80-kDa CD2-associated protein (CD2AP) develop progressive renal failure that starts soon after birth with proteinuria and foot process effacement by unknown mechanisms. CD2AP has been identified and cloned independently by virtue of its interaction with the T cell protein CD2 and with the docking protein p130Cas. In the present study we examined the localization of CD2AP and p130Cas in the mouse glomerulus and in cultured podocytes. In glomeruli, CD2AP and p130Cas immunofluorescence were observed in podocytes, where they colocalized with F-actin in foot processes. In addition, p130Cas was strongly expressed in mesangial cells. Immunoelectron microscopy demonstrated that CD2AP was present in podocyte foot processes without a prevailing localization. In cultured podocytes, p130Cas was enriched at sites of focal adhesions, where it colocalized like vinculin with F-actin at stress fiber ends. In contrast, CD2AP colocalized with F-actin at the leading edge of lamellipodia and in small spots, which were unevenly distributed in the cytoplasm. The spot-shaped F-actin structures were also stained by antibodies against the actin nucleation Arp2/3 complex and cortactin, both contributing to dynamic actin assembly. Moreover, CD2AP spots in cultured podocytes were in close spatial association with actinin-4, but not actinin-1. Our results suggest that CD2AP and p130Cas, which both colocalize with F-actin in podocytes in situ, possess different functions. Whereas p130Cas is found in focal adhesions, CD2AP seems to be involved in the regulation of highly dynamic F-actin structures in podocyte foot processes.


Subject(s)
Actins/analysis , Cytoskeletal Proteins , Cytoskeleton/chemistry , Kidney Glomerulus/chemistry , Kidney Glomerulus/cytology , Phosphoproteins/analysis , Proteins/analysis , Actin-Related Protein 2 , Actin-Related Protein 3 , Actinin/analysis , Adaptor Proteins, Signal Transducing , Animals , Cell Line, Transformed , Cortactin , Crk-Associated Substrate Protein , Focal Adhesions/chemistry , Mice , Microfilament Proteins/analysis , Retinoblastoma-Like Protein p130 , Stress Fibers/chemistry
8.
Curr Opin Nephrol Hypertens ; 10(3): 331-40, 2001 May.
Article in English | MEDLINE | ID: mdl-11342794

ABSTRACT

Knowledge of podocyte biology is growing rapidly. Podocytes are crucially involved in most hereditary diseases affecting the glomerulus, which all exhibit podocyte-specific defects, that is, foot process effacement and protein leakage. Efforts to understand molecular mechanisms causing these derangements are increasingly successful and will allow a better targeting of interventions to halt the progression of chronic renal disease.


Subject(s)
Kidney Diseases , Kidney Glomerulus , Epithelial Cells/pathology , Epithelial Cells/physiology , Humans , Kidney Diseases/etiology , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Kidney Glomerulus/pathology , Kidney Glomerulus/physiology
9.
FASEB J ; 15(3): 707-18, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11259389

ABSTRACT

In previous studies, added parathyroid hormone-related protein (PTHrP) inhibits whereas transfected PTHrP stimulates the proliferation of A10 aortic smooth muscle cells by nuclear translocation of the peptide. In the present studies, we asked whether these paradoxical trophic actions of PTHrP occur in smooth muscle cells (SMC) cultured from small intrarenal arteries of, and whether they are altered in, 12-wk-old spontaneously hypertensive rats (SHR) as compared to normotensive Wistar-Kyoto (WKY) rats. SHR cells grew faster than WKY cells. PTHrP transcript was increased in SHR-derived cells whereas PTH1 receptor (PTH1R) transcripts were similar in both cell lines. In both strains of cells, stable transfection with human PTHrP(1-139) cDNA did not further induce proliferation, suggesting maximal effect of endogenous PTHrP in wild cells. In contrast, transfection with antisense hPTHrP(1-139) cDNA, which abolished PTHrP mRNA, decreased WKY but increased SHR cell proliferation. Added PTHrP(1-36) (1-100 pM) decreased WKY and increased SHR cell proliferation. Additional studies indicated that the preferential coupling of PTH1-R to G-protein Gi was responsible for the proliferative effect of exogenous PTHrP in SHR cells. Moreover, PTHrP was detected in the nucleolus of a fraction of WKY and SHR renal SMC, in vitro as well as in situ, suggesting that the nucleolar translocation of PTHrP might be involved in the proliferative effects of endogenous PTHrP. In renovascular SMC, added PTHrP is antimitogenic, whereas endogenously produced PTHrP is mitogenic. These paradoxical effects of PTHrP on renovascular SMC proliferation appear to be reversed in the SHR model of genetic hypertension. A new concept emerges from these results, according to which a single molecule may have opposite effects on VSMC proliferation under physiological and pathophysiological conditions.


Subject(s)
Cell Division/drug effects , Hypertension/pathology , Kidney/blood supply , Muscle, Smooth, Vascular/drug effects , Proteins/pharmacology , Receptors, Parathyroid Hormone/metabolism , Animals , Arteries/anatomy & histology , Blotting, Western , Cells, Cultured , Cholera Toxin/pharmacology , Cloning, Molecular , Disease Models, Animal , Humans , Immunohistochemistry , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , Parathyroid Hormone-Related Protein , Proteins/genetics , Proteins/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Virulence Factors, Bordetella/pharmacology
10.
Kidney Int ; 57(6): 2468-75, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10844615

ABSTRACT

BACKGROUND: In contrast to arterioles, small arteries appear to be the preferential site of renal vascular smooth muscle cell (VSMC) proliferation under pathophysiological conditions. To date, techniques have been described to isolate renal arterioles and to culture VSMCs. The aim of the present study was to develop a method of culturing VSMCs from isolated small arteries of the rat kidney and to characterize their growth as compared with that of aortic VSMCs. METHODS: Renal vascular trees were isolated from kidneys of male Wistar rats by a sieving technique. VSMCs were grown from explants of collagenase-treated renal vascular trees and thoracic aorta. Growth curves and proliferation of renal and aortic VSMCs in response to fetal bovine serum (FBS) were compared by determination of cell number and DNA synthesis, measured as incorporation of 5-bromo-2'-deoxyuridine. RESULTS: Renal vascular trees consisted mainly of small arteries with a diameter of 80 to 400 microm (interlobar and arcuate arteries). As compared with total kidney or renal cortex, alkaline phosphatase activity was decreased by 81%, and vasopressin (10 micromol/L) was unable to stimulate adenylyl cyclase in renal vascular trees, indicating little tubular contamination. A homogenous population of spindle-shaped cells was cultured from renal vascular trees, which grew in a hill-and-valley pattern and stained positively for smooth muscle alpha-actin, according to the characteristics of VSMC phenotype. Renal VSMCs proliferated more slowly than aortic VSMCs and reached the plateau of growth at about half of the cell density of aortic VSMCs. Furthermore, proliferation of renal VSMCs depended more heavily on FBS concentration, since about threefold higher concentrations of FBS were needed for renal VSMCs to multiply at the same rate and to similarly stimulate DNA synthesis as compared with aortic VSMCs. CONCLUSIONS: We present a method to culture renal VSMCs from small arteries of the rat kidney, which possess distinct growth characteristics as compared with aortic VSMCs.


Subject(s)
Cytological Techniques , Muscle, Smooth, Vascular/cytology , Renal Artery/cytology , Actins/metabolism , Adenylyl Cyclases/metabolism , Alkaline Phosphatase/metabolism , Animals , Aorta/cytology , Arginine Vasopressin/pharmacology , Cattle/blood , Cattle/embryology , Cell Division , Cells, Cultured , Fetal Blood , Male , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Phenotype , Rats , Rats, Wistar , Renal Artery/enzymology , Renal Artery/metabolism , Renal Artery/physiology
11.
Kidney Int ; 57(6): 2476-84, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10844616

ABSTRACT

BACKGROUND: Diadenosine polyphosphates (APXA) are vasoactive nucleotides that elicit effects via purinoceptors. Recent data suggest differential effects of APXA on kidney vasculature. METHODS: The in vivo effects of AP3A, AP5A, and adenosine on renal microvessels and the role of purinoceptors were investigated by the application of agonists to the hydronephrotic rat kidney and preincubation with respective antagonists. RESULTS: The addition of the agonists (10-7 mol/L up to 10-4 mol/L) resulted in a concentration-dependent transient vasoconstriction [interlobular artery (ILOB): adenosine 30 +/- 7%, N = 7, AP3A 35 +/- 10%, N = 5; AP5A 66 +/- 19%, N = 5; 10-5 mol/L each] lasting up to one minute, followed by a concentration-dependent vasodilation (ILOB: adenosine 10 +/- 3%, N = 6; AP3A 19 +/- 4%, N = 5; AP5A 12 +/- 5%, N = 6; 10-5 mol/L each). In ILOB and in the afferent arteriole (AFF), the constrictory effects of AP5A were more pronounced than those of AP3A and adenosine. In the efferent arteriole (EFF), vascular tone was only slightly affected by all agonists. The dilatory potency was comparable for all agonists in ILOB and EFF. No significant vasodilation occurred in AFF. The application of the selective A1 receptor antagonist DPCPX (10-5 mol/L) completely abolished the adenosine-induced vasoconstriction, whereas the A2 receptor antagonist DMPX and the P2 purinoceptor antagonists PPADS and A3P5P (all 10-5 mol/L) did not affect adenosine-induced constriction. The AP3A-induced constriction was abolished by DPCPX and was partially inhibited by PPADS. The constriction induced by AP5A was less sensitive to DPCPX but more sensitive to PPADS. In ILOB and EFF, DMPX or A3P5P abolished dilation after the addition of the agonists. The dilation after AP5A was not significantly reduced. In AFF, no significant dilation was observed with these agonists alone, but it was clearly visible in the presence of DPCPX or PPADS. CONCLUSIONS: APXA evoke transient constrictions in vessels of the hydronephrotic rat kidney, which are mediated by A1 and P2 purinoceptors. The length of the phosphate chain determines the degree of vasoconstriction and the extent to which the substances exert effects on the P2 purinoceptor subtypes. ILOB and AFF are more potently affected by APXA than EFF. Afferent vasodilation is partially overridden by sustained vasoconstriction.


Subject(s)
Dinucleoside Phosphates/pharmacology , Renal Circulation/drug effects , Adenosine/pharmacology , Animals , Arterioles/drug effects , Blood Vessels/drug effects , Female , Hydronephrosis/physiopathology , Microcirculation/drug effects , Rats , Rats, Wistar , Receptors, Purinergic P1/physiology , Receptors, Purinergic P2/physiology , Renal Artery/drug effects , Vasoconstriction/physiology , Vasomotor System/drug effects
12.
J Am Soc Nephrol ; 10(12): 2510-7, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10589689

ABSTRACT

Leukocytes play a central role in the pathogenesis of anti-glomerular basement membrane glomerulonephritis (anti-GBM GN). Understanding the mechanisms underlying their recruitment in the glomerulus is of critical importance, because this may lead to more specific anti-inflammatory drug design. The requirement for integrins, especially from the beta2 group, and their Ig superfamily counter-receptors has been established, however, the role of selectins remains controversial. An intravital microscopy technique was developed to study concomitantly the glomerular and venular leukocyte kinetics and the hemodynamic alterations in a rat model of anti-GBM GN, induced by injection of 10 mg of nephrotoxic serum (NTS). Histologic studies of the kidney were performed in parallel and urinary protein excretion was measured. The animals received NTS alone or were pretreated with either a monoclonal antibody against the beta2 integrin CD11b (OX42, 4 mg/kg) or fucoidan F7 (FF7, 8 mg/kg), an oligosaccharide that blocks both L- and P-selectin function. Administration of NTS resulted in a time-dependent increase in the number of adherent leukocytes in the glomeruli and a parallel decrease of the perfused glomerular capillary area. Substantial proteinuria was observed. Pretreatment with OX42 significantly attenuated these changes. FF7 almost abolished the rolling of the leukocytes in the venules, thus demonstrating efficient anti-selectin activity. Nevertheless, FF7 had no influence on the glomerular events or on the development of proteinuria. These results confirm that glomerular leukocyte adhesion in anti-GBM GN is CD11b-dependent. However, selectin-mediated interaction between the leukocytes and the glomerular capillary endothelium does not appear to be a prerequisite for leukocyte adhesion in the glomerulus. These results therefore question the potential utility of anti-selectin therapy in the treatment of anti-GBM GN.


Subject(s)
Glomerulonephritis/etiology , Kidney Glomerulus/immunology , Leukocytes/immunology , Selectins/physiology , Animals , Antibodies, Monoclonal/pharmacology , Basement Membrane/immunology , Basement Membrane/pathology , CD18 Antigens/immunology , CD18 Antigens/physiology , Cell Adhesion , Cell Movement , Female , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Hemodynamics , Kidney Glomerulus/blood supply , Kidney Glomerulus/pathology , L-Selectin/immunology , L-Selectin/physiology , Leukocytes/pathology , P-Selectin/immunology , P-Selectin/physiology , Polysaccharides/antagonists & inhibitors , Polysaccharides/immunology , Polysaccharides/physiology , Rats , Rats, Wistar
13.
Br J Pharmacol ; 127(8): 1929-35, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10482926

ABSTRACT

1. Renal vasoconstriction in response to angiotensin II (ANGII) is known to be modulated by nitric oxide (NO). Since shear stress stimulates the release of a variety of vasoactive compounds from endothelial cells, we studied the impact of shear stress on the haemodynamic effect of ANGII in isolated perfused kidneys of rats under control conditions and during NO synthase inhibition with L-NAME (100 microM). 2. Kidneys were perfused in the presence of cyclo-oxygenase inhibitor (10 microM indomethacin) with Tyrode's solution of relative viscosity zeta=1 (low viscosity perfusate, LVP) or, in order to augment shear stress, with Tyrode's solution containing 7% Ficoll 70 of relative viscosity zeta=2 (high viscosity perfusate, HVP). 3. Vascular conductance was 3.5+/-0.4 fold larger in HVP as compared with LVP kidneys, associated with an augmentation of overall wall shear stress by 37+/-5%. During NO inhibition, vascular conductance was only 2.5+/-0.2 fold elevated in HVP vs LVP kidneys, demonstrating shear stress-induced vasodilatation by NO and non-NO/non-prostanoid compound(s). 4. ANGII (10 - 100 pM) constricted the vasculature in LVP kidneys, but was without effect in HVP kidneys. During NO inhibition, in contrast, ANGII vasoconstriction was potentiated in HVP as compared with LVP kidneys. 5. The potentiation of ANGII vasoconstriction during NO inhibition has been shown to be mediated by endothelium-derived P450 metabolites and to be sensitive to AT2 receptor blockade in our earlier studies. Accordingly, in HVP kidneys, increasing concentrations of the AT2 receptor antagonist PD123319 (5 and 500 nM) gradually abolished the potentiation of ANGII vasoconstriction during NO inhibition, but did not affect vasoconstriction in response to ANGII in LVP kidneys. 6. Our results demonstrate, that augmentation of shear stress by increasing perfusate viscosity induces vasodilatation in the rat kidney, which is partially mediated by NO. Elevated levels of shear stress attenuate renal ANGII vasoconstriction through enhanced NO production and are required for AT2 sensitive potentiation during NO inhibition.


Subject(s)
Angiotensin II/pharmacology , Kidney/drug effects , Nitric Oxide/metabolism , Vasoconstrictor Agents/pharmacology , Angiotensin Receptor Antagonists , Animals , Blood Viscosity , Enzyme Inhibitors/pharmacology , Hemorheology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Rats , Rats, Wistar , Stress, Physiological
14.
Endocrinology ; 140(9): 4342-50, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10465308

ABSTRACT

Although PTH-related protein-(1-36) [PTHrP-(1-36)] is known to be expressed in smooth muscle and to exert potent myorelaxant effects, its tonic effects on cavernosal smooth muscle has not yet been explored. Using the RT-PCR technique, the present study establishes that PTHrP messenger RNA is present in microdissected corpus cavernosa in the rat. In immunohistochemical studies using affinity-purified antibodies to middle regions of PTHrP, immunostaining was localized throughout the penile structures, including vessels, cavernosal smooth muscle, and trabecular fibroblasts. Strong immunostaining for PTHrP was also detected in the dorsal nerve bundles. In anesthetized rats, intracavernosally injected boluses of increasing doses of PTHrP-(1-36) (0.3-30 pmol in 100 microl saline) had little effect on intracavernosal pressure. However, they markedly potentiated the dilatory response to papaverine (8-800 nmol), increasing the papaverine-induced intracavernous pressure by 2.5-fold, close to the mean arterial pressure. In conclusion, the cavernosal expression of PTHrP messenger RNA, the distribution of immunoreactive PTHrP throughout the structuro-functional components of the erectile apparatus and its strong potentiating action on papaverine-induced cavernosal relaxation, collectively suggest that PTHrP participates in the control of cavernosal tone.


Subject(s)
Penis/metabolism , Proteins/metabolism , Animals , Immunohistochemistry , In Vitro Techniques , Male , Parathyroid Hormone-Related Protein , Penis/physiology , Peptide Fragments/pharmacology , Pressure , Proteins/genetics , Proteins/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution/physiology
15.
Kidney Int Suppl ; 67: S207-10, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9736292

ABSTRACT

Parathyroid hormone-related protein (PTHrP) is expressed throughout the renovascular system, and it dilates renal vessels, increases renal blood flow and glomerular filtration rate, and stimulates renin release. Mechanical forces and experimental hypertension have been shown to stimulate PTHrP expression in smooth muscles, suggesting a negative feedback control of vascular tone by PTHrP in hypertension. In this study, we compared the impact of a PTHrP receptor antagonist, PTHrP (7-34), and a PTHrP receptor agonist, PTHrP (1-36), on the vascular resistance of perfused kidneys isolated from spontaneously hypertensive rats (SHR) and Wistar Kyoto rats (WKY). Endogenous PTHrP appears not to act as a renal vasodilator in either WKY or SHR. However, the vasodilation following infused PTHrP (1-36) is blunted markedly in SHR, possibly due to desensitization or down-regulation of PTH/PTHrP receptors. Negative feedback control of vascular tone by PTHrP in SHR thus appears unlikely. The results raise the question of whether endogenous renovascular PTHrP behaves rather as a growth factor than as a vasodilator.


Subject(s)
Hypertension, Renal/physiopathology , Kidney/blood supply , Proteins/pharmacology , Renal Circulation/drug effects , Vasodilation/drug effects , Animals , Feedback/physiology , Growth Substances/physiology , Kidney/chemistry , Kidney/physiopathology , Male , Organ Culture Techniques , Parathyroid Hormone-Related Protein , Phenylephrine/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Parathyroid Hormone/physiology , Vascular Resistance , Vasoconstrictor Agents/pharmacology
16.
Kidney Int Suppl ; 67: S234-7, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9736302

ABSTRACT

Nitric oxide (NO) buffers the effect of vasoconstrictors currently active in the renovascular system. Enhancement of the angiotensin II (Ang II)-induced vasoconstriction during NO blockade comprises both AT2-sensitive potentiation, decreasing the half maximal vasoconstriction (EC50) value to the subnanomolar concentration range, and augmentation, increasing the maximal effect (Emax) value in the isolated perfused rat kidney. In this study, we examine whether constrictory prostanoids are involved in Ang II subtype receptor (AT2)-sensitive potentiation of the Ang II effect during NO blockade. Thus, Ang II-induced vasoconstriction (0.1 or 10 nM Ang II) was measured in six series of constant-flow perfused isolated rat kidneys in the presence of indomethacin under control conditions, during NO inhibition, and during combined inhibition of NO and all arachidonic pathways by eicosatetraynoic acid (ETYA), an analog of arachidonic acid. The vasoconstriction elicited by 10 nM Ang II, which is the maximal response, increased about threefold during NO inhibition compared with control. This augmentation was not affected by ETYA. In contrast, the vasoconstriction elicited by 0.1 nM Ang II increased about 20-fold during NO inhibition, reflecting mainly potentiation of the Ang II effect. This increase was abrogated by ETYA. We conclude that vasoconstrictor eicosanoids, which are suppressed by endogenous NO, mediate AT2-sensitive potentiation of the Ang II-induced vasoconstriction in the rat kidney.


Subject(s)
5,8,11,14-Eicosatetraynoic Acid/pharmacology , Angiotensin II/physiology , Nitric Oxide/antagonists & inhibitors , Vasoconstriction/physiology , Angiotensin II/antagonists & inhibitors , Animals , Eicosanoids/pharmacology , Enzyme Inhibitors/pharmacology , Kidney/blood supply , Kidney/chemistry , Kidney/physiology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Organ Culture Techniques , Prostaglandins/physiology , Rats , Rats, Wistar , Renal Circulation/physiology , Vascular Resistance
17.
Br J Pharmacol ; 124(5): 946-52, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9692780

ABSTRACT

1. We showed earlier that NO inhibition caused a left-shift and augmented Emax of the concentration-response curve of AT1-mediated (angiotensin II)-induced vasoconstrictions (AII-VC) in the rat kidney. The 0.01-0.1 nM AII-VC unmasked by the potentiating effect of NO inhibition, were sensitive not only to AT1 (L158809), but also to AT2 receptor (PD123319) antagonists. We now demonstrate the role of endothelium and eicosanoids in the NO-masked AT1/AT2-mediated component of the AII-VC in isolated indomethacin-perfused kidneys of the rat. 2. L-NAME increased 0.1 nM AII-VC 7.2 fold. Pretreatment of the kidneys with factor VIII antibody/complement or with the detergent CHAPS to damage endothelium, decreased carbachol-induced vasodilatation and blunted by 60 and 30% respectively, the enhancement of AII-VC during NO inhibition. 3. L-NAME also increased 3 microM noradrenaline (NA)-induced vasoconstriction (NA-VC) 8.1 fold. In contrast to AII-VC, endothelium damage was without effect on the enhancement of NA-VC by L-NAME, suggesting a dominant role of endothelium-derived NO in the enhancement of NA-VC. 4. During NO inhibition, ETYA (2 microM; an inhibitor of all arachidonic acid derived pathways) and alpha-naphtoflavone (10 microM; an inhibitor of the cytochrome P450 isozymes), decreased by 85% the 0.1 nM AII-VC. 5. In conclusion, during NO inhibition, the AT1-mediated constriction to low concentrations of AII, which is sensitive to AT2 antagonists, depends on intact endothelium, and can be blocked by inhibition of eicosanoid synthesis. The results suggest that the AII-mediated vasoconstriction through AT1 receptors is potentiated in the absence of NO, by the release of eicosanoids from the endothelium through AT2 receptors.


Subject(s)
Angiotensin II/physiology , Angiotensin Receptor Antagonists , Arachidonic Acid/physiology , Cytochrome P-450 Enzyme System/physiology , Endothelium, Vascular/physiology , Kidney/blood supply , Nitric Oxide/antagonists & inhibitors , Vasoconstriction/physiology , 5,8,11,14-Eicosatetraynoic Acid/pharmacology , Angiotensin II/pharmacology , Animals , Arachidonic Acid/metabolism , Benzoflavones/pharmacology , Bosentan , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Endothelin Receptor Antagonists , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Kidney/drug effects , Male , Norepinephrine/pharmacology , Perfusion , Rats , Rats, Wistar , Receptor, Angiotensin, Type 2 , Sulfonamides/pharmacology , Vascular Resistance/drug effects , Vasoconstriction/drug effects
18.
Kidney Int ; 53(4): 963-9, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9551405

ABSTRACT

Cyclosporine A (CsA), a widely used immunosuppressive agent, causes renal vasoconstriction and systemic hypertension. Recent data suggest that the renal effect of CsA is possibly mediated by endothelin (ET). We investigated the effects of CsA on renal microvessels and the efficacy of ETA or ETA/ETB receptor antagonists in ameliorating CsA effects in the hydronephrotic rat kidney. Infusion of CsA (30 mg.kg-1) induced a transient increase (20%) in mean arterial pressure (MAP) and a sustained reduction (85%) in glomerular blood flow (GBF) due to preferential constriction of the arcuate artery (39%) and the proximal segment of the interlobular artery (23%). Under basal conditions the ETA receptor antagonist BQ-123 had marginal effects consisting of reduction in MAP, rise in GBF and dilation of preglomerular vessels. The non-selective ETA/ETB receptor antagonist PD 145065 also reduced MAP, but tended to decrease GBF and constrict large preglomerular vessels. The difference in effects of the two antagonists indicated that under basal conditions ETB blockade constricts large preglomerular vessels and reduces GBF. After BQ-123 or PD 145065, the constriction of large preglomerular vessels and reduction in GBF induced by CsA was attenuated by about 50%, but the rise in MAP was not influenced. Our data indicate that a sizable part of renal vasoconstriction due to CsA is mediated via ET production in large preglomerular arteries and can be avoided by the blockade of ETA receptors. Additional blockade of ETB receptors does not attenuate the CsA effects further, possibly because ETB receptors mediate both vasoconstriction and dilation.


Subject(s)
Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Kidney/blood supply , Receptors, Endothelin/physiology , Vasoconstriction/drug effects , Animals , Blood Pressure/drug effects , Capillaries/chemistry , Capillaries/physiology , Drug Interactions , Endothelin Receptor Antagonists , Female , Peptides, Cyclic/pharmacology , Rats , Rats, Wistar , Vasoconstriction/physiology
19.
Kidney Int ; 52(1): 202-7, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9211363

ABSTRACT

Atrial natriuretic peptide (ANP) has unique effects on the renal vasculature, in that it dilates preglomerular vessels and constricts efferent arterioles. In the present study we aimed to characterize the natriuretic peptide receptor (NPR) subtypes, which mediate the renovascular effects of ANP, using in vivo microscopy in the split hydronephrotic kidney model of rats. ANP (10(-9) and 3.10(-9)), which binds to NPR-A and NPR-C, dilated preglomerular vessels and constricted efferent arterioles similarly to that found in previous studies. C-type natriuretic peptide (10(-9) to 10(-7)), which binds to NPR-B and NPR-C, dilated pre- and postglomerular vessels and profoundly increased glomerular blood flow. A specific ligand of NPR-C, C-ANP (des-[Gln18,Ser19,Gly20,Leu21,Gly22]ANP 4-23-NH2, 10(-9) to 10(-7)) was devoid of vascular effects. The ANP antagonist A71915 (10(-9) to 10(-6)) induced moderate dilation in renal vessels possibly due to some agonistic activity on NPR-B, ANP-induced preglomerular vasodilation was attenuated by A71915 (10(-6)) to 36 +/- 6% of the initial response, whereas efferent vasoconstriction was completely abolished (-4 +/- 4% of initial response). Our results indicate that ANP dilates preglomerular vessels and constricts efferent arterioles through NPR-A, both responses being antagonized by A71915 with different potencies. Furthermore, our data show that in the rat renal microcirculation stimulation of NPR-B results in vasodilation only, whereas NPR-C does not mediate vascular responses.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Kidney/drug effects , Microcirculation/drug effects , Receptors, Atrial Natriuretic Factor/physiology , Animals , Female , Kidney/blood supply , Natriuretic Peptide, C-Type , Peptide Fragments/pharmacology , Proteins/pharmacology , Rats , Rats, Wistar , Tetrahydroisoquinolines
20.
J Hypertens ; 15(6): 633-41, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9218183

ABSTRACT

OBJECTIVE: To assess directly the vasodilating effects of angiotensin converting enzyme (ACE) inhibition in different renal vessels and to determine the role of kinins and angiotensin II (ANGII) therein. METHODS: Lumen diameters of different vessels and glomerular blood flows were measured in cortical and juxtamedullary glomeruli by in-vivo microscopy in the split hydronephrotic kidney of anesthetized female Wistar rats. RESULTS: Injection of the ACE inhibitor quinapril at a dose of 0.9 mg/kg intravenously, which blocks conversion of locally applied angiotensin I (1 mumol/l), increased glomerular blood flows by 39 +/- 6 and 18 +/- 4% in cortical and juxtamedullary glomeruli, respectively, due to vasodilatation in all renal vessels. The most pronounced vasodilatation was observed in interlobular arteries (19 +/- 2%) and in cortical afferent arterioles (16 +/- 3%). Pretreatment of the hydronephrotic kidney by local application of 40 nmol/l Hoe140, a bradykinin B2 receptor antagonist, or 3 mumol/l valsartan, an ANGII type 1 receptor antagonist, attenuated the vasodilatation in response to quinapril. ANGII receptor blockade affected only weakly, whereas bradykinin receptor blockade blunted markedly, the quinapril-induced vasodilatation, suggesting that kinins play an important role in our experimental model. Administration of valsartan, which abrogated the renal vasoconstriction induced by 10 nmol/l ANGII completely, caused vasodilation of magnitude similar to that caused by administration of quinapril. Yet, the vasodilatation induced by the combination of valsartan and quinapril was significantly larger than that induced by administration of quinapril alone in interlobular arteries, afferent arterioles, and cortical efferent arterioles. CONCLUSIONS: Our results indicate that kinins and ANGII can contribute to the renal vasodilatation in response to ACE inhibitors, but ACE inhibitors appear to have only minor effects on ANGII levels in those renal vessels, which are the well-known sites of renin expression.


Subject(s)
Angiotensin II/physiology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Kidney/blood supply , Kidney/drug effects , Kinins/physiology , Tetrahydroisoquinolines , Vasodilation/drug effects , Vasodilation/physiology , Angiotensin II/administration & dosage , Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Animals , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Bradykinin Receptor Antagonists , Drug Synergism , Female , Isoquinolines/administration & dosage , Isoquinolines/pharmacology , Quinapril , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptor, Bradykinin B2 , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Tetrazoles/administration & dosage , Tetrazoles/pharmacology , Valine/administration & dosage , Valine/analogs & derivatives , Valine/pharmacology , Valsartan
SELECTION OF CITATIONS
SEARCH DETAIL
...