Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Kidney Int ; 70(8): 1456-62, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16955103

ABSTRACT

Albuminuria in diabetic nephropathy is due to endothelial dysfunction, a loss of negative charges in the basement membrane, and changes a of the slit-membrane diaphragm composition. We have recently shown that protein kinase C alpha (PKCalpha)-deficient mice are protected against the development of albuminuria under diabetic conditions. We here tested the hypothesis that PKCalpha mediates the hyperglycemia-induced downregulation of the slit-diaphragm protein nephrin. After 8 weeks of streptozotocin (STZ)-induced hyperglycemia the expression of glomerular nephrin was significantly reduced. In contrast, other slit-diaphragm proteins such as podocin and CD2AP were unaltered in diabetic state. In PKCalpha-/- mice, hyperglycemia-induced downregulation of nephrin was prevented. Podocin and CD2AP remained unchanged. In addition, the nephrin messenger RNA expression was also reduced in hyperglycemic wild-type mice but remained unaltered in PKCalpha-/- mice. We postulate that the underlying mechanism of the hyperglycemia-induced regulation of various proteins of the glomerular filtration barrier is a PKCalpha-dependent regulation of the Wilms' Tumor Suppressor (WT1) which previously has been shown to act as a direct transcription factor on the nephrin promoter. Our data suggest that PKCalpha activation may be an important intracellular signaling pathway in the regulation of nephrin expression and glomerular albumin permeability in the diabetic state.


Subject(s)
Diabetic Nephropathies/metabolism , Membrane Proteins/metabolism , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Albuminuria/etiology , Albuminuria/metabolism , Animals , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/physiopathology , Gene Expression Regulation/physiology , Humans , Hyperglycemia , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Glomerulus/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism
2.
J Mol Med (Berl) ; 79(10): 601-8, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11692158

ABSTRACT

Angiotensin II is known to stimulate cardiac hypertrophy and contractility. Most angiotensin II effects are mediated via membrane bound AT1 receptors. However, the role of myocardial AT1 receptors in cardiac hypertrophy and contractility is still rarely defined. To address the hypothesis that increased myocardial AT1 receptor density causes cardiac hypertrophy apart from high blood pressure we developed a transgenic rat model which expresses the human AT1 receptor under the control of the alpha-myosin heavy-chain promoter specifically in the myocardium. Expression was identified and quantified by northern blot analysis and radioligand binding assays, demonstrating overexpression of angiotensin II receptors in the transgenic rats up to 46 times the amount seen in nontransgenic rats. Coupling of the human AT1 receptor to rat G proteins and signal transduction cascade was verified by sensitivity to GTP-gamma-S and increased sensitivity of intracellular Ca2+ [Ca2+]i to angiotensin II in fluo-3 loaded transgenic cardiomyocytes. Transgenic rats exhibited normal cardiac growth and function under baseline conditions. Pronounced hypertrophic growth and contractile responses to angiotensin II, however, were noted in transgenic rats challenged by volume and pressure overload. In summary, we generated a new transgenic rat model that exhibits an upregulated myocardial AT1 receptor density and demonstrates augmented cardiac hypertrophy and contractile response to angiotensin II after volume and pressure overload, but not under baseline conditions.


Subject(s)
Cardiomegaly/genetics , Myocardium/metabolism , Receptors, Angiotensin/genetics , Angiotensin II/pharmacology , Animals , Animals, Genetically Modified , Animals, Newborn , Binding, Competitive , Blood Pressure/drug effects , Calcium/metabolism , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Hemodynamics/drug effects , Humans , Membranes/metabolism , Myocardium/pathology , Organ Size/drug effects , Perfusion , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Time Factors , Transgenes/genetics
3.
Science ; 293(5539): 2449-52, 2001 Sep 28.
Article in English | MEDLINE | ID: mdl-11498544

ABSTRACT

Caveolae are plasma membrane invaginations that may play an important role in numerous cellular processes including transport, signaling, and tumor suppression. By targeted disruption of caveolin-1, the main protein component of caveolae, we generated mice that lacked caveolae. The absence of this organelle impaired nitric oxide and calcium signaling in the cardiovascular system, causing aberrations in endothelium-dependent relaxation, contractility, and maintenance of myogenic tone. In addition, the lungs of knockout animals displayed thickening of alveolar septa caused by uncontrolled endothelial cell proliferation and fibrosis, resulting in severe physical limitations in caveolin-1-disrupted mice. Thus, caveolin-1 and caveolae play a fundamental role in organizing multiple signaling pathways in the cell.


Subject(s)
Aorta/physiology , Caveolae/physiology , Caveolins/genetics , Caveolins/physiology , Endothelium, Vascular/physiology , Mice, Inbred C57BL , Muscle, Smooth, Vascular/physiology , Pulmonary Alveoli/pathology , Signal Transduction , Albumins/cerebrospinal fluid , Animals , Aorta/ultrastructure , Asthenia/etiology , Calcium Signaling , Caveolae/ultrastructure , Caveolin 1 , Caveolins/deficiency , Cell Division , Cells, Cultured , Cholesterol/metabolism , Endothelium/cytology , Endothelium, Vascular/cytology , Gene Targeting , In Vitro Techniques , Lipids/analysis , Lung/ultrastructure , Membrane Microdomains/chemistry , Membrane Microdomains/physiology , Mice , Mice, Knockout , Muscle Contraction , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/ultrastructure , Nitric Oxide/metabolism , Pulmonary Alveoli/cytology , Pulmonary Fibrosis/etiology
4.
Am J Hypertens ; 14(6 Pt 2): 55S-61S, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11411766

ABSTRACT

Angiotensin (Ang) II is capable of producing inflammatory changes by signals through its AT1 receptor. Reactive oxygen species production, adhesion molecule expression, chemokines, and other mediators are involved. Nuclear factor-kappaB (NK-kappaB) and activator protein 1 (AP-1) are two of the transcription factors activating the responsible genes. We have studied Ang II-independent modulating effects in a double transgenic rat model harboring the human renin and angiotensinogen genes. We have recently focused on the protective effects of HMG-CoA reductase inhibition and review these data here. We found that cerivastatin decreased mortality, lowered blood pressure, preserved renal function, decreased cardiac hypertrophy, and inhibited the entire chain of inflammatory events. Furthermore, NF-kappaB and AP-1 activation was sharply attenuated. We also observed that cerivastatin blocked ERK1/2 phosphorylation in vivo and in vitro. Cerivastatin also inhibited phorbol ester-transmitted events in vascular smooth muscle cells. Because Rho, a member of the Ras protein superfamily is important to Ang II-dependent and -independent vascular smooth muscle signaling events, we suggest that cerivastatin may act by inhibiting the prenylation, membrane anchoring, and subsequent activation of Ras proteins. These data may in part explain cholesterol-independent, HMG-CoA reductase-related, protective effects.


Subject(s)
Angiotensin II/physiology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Inflammation/etiology , Animals , Heart/drug effects , Humans , Inflammation/prevention & control , Kidney/drug effects , Pyridines/pharmacology
5.
Kidney Int ; 58(4): 1420-30, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11012877

ABSTRACT

BACKGROUND: Statins are effective in prevention of end-organ damage; however, the benefits cannot be fully explained on the basis of cholesterol reduction. We used an angiotensin II (Ang II)-dependent model to test the hypothesis that cerivastatin prevents leukocyte adhesion and infiltration, induction of inducible nitric oxide synthase (iNOS), and ameliorates end-organ damage. METHODS: We analyzed intracellular targets, such as mitogen-activated protein kinase and transcription factor (nuclear factor-kappaB and activator protein-1) activation. We used immunohistochemistry, immunocytochemistry, electrophoretic mobility shift assays, and enzyme-linked immunosorbent assay techniques. We treated rats transgenic for human renin and angiotensinogen (dTGR) chronically from week 4 to 7 with cerivastatin (0.5 mg/kg by gavage). RESULTS: Untreated dTGR developed hypertension, cardiac hypertrophy, and renal damage, with a 100-fold increased albuminuria and focal cortical necrosis. dTGR mortality at the age of seven weeks was 45%. Immunohistochemistry showed increased iNOS expression in the endothelium and media of small vessels, infiltrating cells, afferent arterioles, and glomeruli of dTGR, which was greater in cortex than medulla. Phosphorylated extracellular signal regulated kinase (p-ERK) was increased in dTGR; nuclear factor-kappaB and activator protein-1 were both activated. Cerivastatin decreased systolic blood pressure compared with untreated dTGR (147 +/- 14 vs. 201 +/- 6 mm Hg, P < 0.001). Albuminuria was reduced by 60% (P = 0.001), and creatinine was lowered (0.45 +/- 0.01 vs. 0.68 +/- 0.05 mg/dL, P = 0. 003); however, cholesterol was not reduced. Intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 expression was diminished, while neutrophil and monocyte infiltration in the kidney was markedly reduced. ERK phosphorylation and transcription factor activation were reduced. In addition, in vitro incubation of vascular smooth muscle cells with cerivastatin (0.5 micromol/L) almost completely prevented the Ang II-induced ERK phosphorylation. CONCLUSION: Cerivastatin reduced inflammation, cell proliferation, and iNOS induction, which led to a reduction in cellular damage. Our findings suggest that 3-hydroxy-3-methylglutaryl coenzyme (HMG-CoA) reductase inhibition ameliorates Ang II-induced end-organ damage. We suggest that these effects were independent of cholesterol.


Subject(s)
Angiotensin II/pharmacology , Blood Pressure/drug effects , Cholesterol/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Kidney Failure, Chronic/drug therapy , Pyridines/pharmacology , Vasoconstrictor Agents/pharmacology , Albuminuria/chemically induced , Albuminuria/drug therapy , Albuminuria/enzymology , Angiotensinogen/genetics , Animals , Animals, Genetically Modified , Cell Division/drug effects , Creatinine/blood , Disease Models, Animal , Humans , Intercellular Adhesion Molecule-1/analysis , Kidney/cytology , Kidney/drug effects , Kidney Failure, Chronic/chemically induced , Kidney Failure, Chronic/enzymology , Leukocytes/chemistry , Leukocytes/cytology , Male , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Organ Size/drug effects , Phosphorylation , Plasminogen Activators/metabolism , Rats , Rats, Sprague-Dawley , Renin/genetics , Thromboplastin/metabolism , Transcription Factor AP-1/metabolism , Urea/blood , Vascular Cell Adhesion Molecule-1/analysis
6.
FASEB J ; 14(11): 1653-63, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10929000

ABSTRACT

Protein kinase C (PKC) isoforms exert specific intracellular functions, but the different isoforms display little substrate specificity in vitro. Selective PKC isoform targeting may be a mechanism to achieve specificity. We used a green fluorescent fusion protein (GFP) to test the hypothesis that local changes in [Ca(2+)](i) regulate translocation of PKCalpha and that different modes of Ca(2+) and Ca(2+) release play a role in PKCalpha targeting. We constructed deletion mutants of PKCalpha to analyze the Ca(2+)-sensitive domains and their role in targeting. Confocal microscopy was used and [Ca(2+)](i) was measured by fluo-3. The fusion protein PKCalpha-GFP was expressed in vascular smooth muscle cells and showed a cytosolic distribution similar to the wild-type PKCalpha protein. The Ca(2+) ionophore ionomycin induced a speckled cytosolic PKCalpha-GFP distribution, followed by membrane translocation, while depolarization by KCl induced primarily membrane translocation. Selective voltage-operated Ca(2+) channel opening led to a localized accumulation of PKCalpha-GFP near the plasma membrane. Opening Ca(2+) stores with InsP(3), thapsigargin, or ryanodine induced a specific PKCalpha-GFP targeting to distinct intracellular areas. The G-protein-coupled receptor agonist thrombin induced a rapid translocation of the fusion protein to focal domains. The tyrosine kinase receptor agonist PDGF induced Ca(2+) influx and led to a linear PKCalpha-GFP membrane association. PKCalpha-GFP deletion mutants demonstrated that the C2 domain, but not the catalytic subunit, is necessary for Ca(2+)-induced PKCalpha targeting. Targeting was also abolished when the ATP binding site was deleted. We conclude that PKCalpha can rapidly be translocated to distinct intracellular or membrane domains by local increases in [Ca(2+)](i). The targeting mechanism is dependent on the C2 and ATP binding site of the enzyme. Localized [Ca(2+)](i) changes determine the spatial and temporal targeting of PKCalpha.


Subject(s)
Calcium/metabolism , Isoenzymes/metabolism , Muscle, Smooth, Vascular/metabolism , Protein Kinase C/metabolism , Adenosine Triphosphate/metabolism , Animals , Aorta , Binding Sites , Biological Transport/drug effects , Calcium Channel Blockers/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Cytoplasm/drug effects , Cytoplasm/metabolism , Inositol 1,4,5-Trisphosphate/pharmacology , Ion Channel Gating/drug effects , Ionomycin/pharmacology , Isoenzymes/chemistry , Isoenzymes/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Platelet-Derived Growth Factor/pharmacology , Potassium Chloride/pharmacology , Protein Kinase C/chemistry , Protein Kinase C/genetics , Protein Kinase C-alpha , Protein Structure, Tertiary , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ryanodine/pharmacology , Sequence Deletion/genetics , Substrate Specificity , Thapsigargin/pharmacology , Thrombin/pharmacology , Transfection
7.
Biochim Biophys Acta ; 1497(2): 253-60, 2000 Jul 21.
Article in English | MEDLINE | ID: mdl-10903430

ABSTRACT

Profilin I was identified, by mass spectrometric sequencing and immunoblotting, as a component of purified Golgi cisternae from HepG2 cells. Binding to the Golgi was verified by indirect immunofluorescence in MT-1 cells showing that a fraction of profilin I colocalizes with TGN38, a marker of the trans-Golgi network (TGN). Studying the formation of constitutive exocytic vesicles at the TGN in a cell-free system demonstrated that cytosolic profilin I has no effect, while incubation of Golgi cisternae with a profilin I-specific antibody reduced vesicle formation by about 50%. Notably, the antibody displaces a fraction of the Golgi-bound dynamin II indicating that profilin I may indirectly promote vesicle formation by supporting the binding of dynamin II to the Golgi membrane. The impact of dynamin II on vesicle formation is demonstrated by incubating the Golgi with the proline-rich domain of dynamin II which concomitantly displaces dynamin II and inhibits vesicle formation. The data provide evidence that profilin I attaches to the Golgi apparatus and is required for the formation of constitutive transport vesicles.


Subject(s)
Contractile Proteins , Glycoproteins , Golgi Apparatus/metabolism , Intracellular Membranes/metabolism , Membrane Proteins , Microfilament Proteins/metabolism , Dynamins , Fluorescent Antibody Technique, Indirect , GTP Phosphohydrolases/metabolism , Immunoblotting , Mass Spectrometry , Membrane Glycoproteins/analysis , Microfilament Proteins/analysis , Microscopy, Confocal , Profilins , Tumor Cells, Cultured
8.
FASEB J ; 13(15): 2161-72, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10593863

ABSTRACT

A novel calcium channel-associated protein of approximately 700 kDa has been identified in mammalian cardiomyocytes that undergoes substantial cAMP-dependent protein kinase (PKA) phosphorylation. It was therefore designated as phosphoprotein 700 (pp700). The pp700 interacts specifically with the beta(2) subunit of cardiac L-type calcium channels as revealed by coprecipitation experiments using affinity-purified antibodies against different calcium channel subunits. It is surprising that amino acid sequence analysis of pig pp700 revealed homology to AHNAK-encoded protein, which was originally identified in human cell lines of neural crest origin as 700-kDa phosphoprotein. Cardiac AHNAK expression was assessed on mRNA level by reverse transcriptase-polymerase chain reaction. Sequence-directed antibodies raised against human AHNAK recognized pp700 in immunoblotting and immunoprecipitation experiments, confirming the homology between both proteins. Anti-AHNAK antibodies labeled preferentially the plasma membrane of cardiomyocytes in cryosections of rat cardiac tissue and isolated cardiomyocytes. Sarcolemmal pp700/AHNAK localization was not influenced by stimulation of either the PKA or the protein kinase C pathway. In back-phosphorylation studies with cardiac biopsies, we identified distinct pp700 pools. The membrane-associated fraction of pp700 underwent substantial in vivo phosphorylation on beta-adrenergic receptor stimulation by isoproterenol, whereas the cytoplasmic fraction of pp700 was not accessible to endogenous PKA. It is important that in vivo phosphorylation occurred in that pp700 fraction which coprecipitated with the calcium channel beta subunit. We hypothesize that both phosphorylation of pp700 and its coupling to the beta subunit play a physiological role in cardiac beta-adrenergic signal transduction. Haase, H., Podzuweit, T., Lutsch, G., Hohaus, A., Kostka, S., Lindschau, C., Kott, M., Kraft, R., Morano, I. Signaling from beta-adrenoceptor to L-type calcium channel: identification of a novel cardiac protein kinase A target that has similarities to AHNAK.


Subject(s)
Calcium Channels, L-Type/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Membrane Proteins/chemistry , Myocardium/enzymology , Neoplasm Proteins/chemistry , Receptors, Adrenergic, beta/metabolism , Amino Acid Sequence , Animals , Gene Expression , Humans , In Vitro Techniques , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Sequence Data , Molecular Weight , Myocardium/cytology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Substrate Specificity , Swine
9.
Circulation ; 99(19): 2523-9, 1999 May 18.
Article in English | MEDLINE | ID: mdl-10330383

ABSTRACT

BACKGROUND: Dihydropyridines block calcium channels; however, they also influence endothelial cells, which do not express calcium channels. We tested the hypothesis that nifedipine can prevent ischemia-induced endothelial permeability increases by inhibiting protein kinase C (PKC) in cultured porcine endothelial cells. METHODS AND RESULTS: Ischemia was induced by potassium cyanide/deoxyglucose, and permeability was measured by albumin flux. Ion channels were characterized by patch clamp. [Ca2+]i was measured by fura 2. PKC activity was measured by substrate phosphorylation after cell fractionation. PKC isoforms were assessed by Western blot and confocal microscopy. Nifedipine prevented the ischemia-induced increase in permeability in a dose-dependent manner. Ischemia increased [Ca2+]i, which was not affected by nifedipine. Instead, ischemia-induced PKC translocation was prevented by nifedipine. Phorbol ester also increased endothelial cell permeability, which was dose dependently inhibited by nifedipine. The effects of non-calcium-channel-binding dihydropyridine derivatives were similar. Analysis of the PKC isoforms showed that nifedipine prevented ischemia-induced translocation of PKC-alpha and PKC-zeta. Specific inhibition of PKC isoforms with antisense oligodeoxynucleotides demonstrated a major role for PKC-alpha. CONCLUSIONS: Nifedipine exerts a direct effect on endothelial cell permeability that is independent of calcium channels. The inhibition of ischemia-induced permeability by nifedipine seems to be mediated primarily by PKC-alpha inhibition. Anti-ischemic effects of dihydropyridine calcium antagonists could be due in part to their effects on endothelial cell permeability.


Subject(s)
Calcium Channel Blockers/pharmacology , Capillary Permeability/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Ischemia , Nifedipine/pharmacology , Protein Kinase C/antagonists & inhibitors , Animals , Cells, Cultured , Endothelium, Vascular/drug effects , Poisons/toxicity , Potassium Cyanide/toxicity , Swine
10.
J Clin Invest ; 103(7): 945-52, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10194466

ABSTRACT

Immune mechanisms and the renin-angiotensin system are implicated in preeclampsia. We investigated 25 preeclamptic patients and compared them with 12 normotensive pregnant women and 10 pregnant patients with essential hypertension. Antibodies were detected by the chronotropic responses to AT1 receptor-mediated stimulation of cultured neonatal rat cardiomyocytes coupled with receptor-specific antagonists. Immunoglobulin from all preeclamptic patients stimulated the AT1 receptor, whereas immunoglobulin from controls had no effect. The increased autoimmune activity decreased after delivery. Affinity-column purification and anti-human IgG and IgM antibody exposure implicated an IgG antibody directed at the AT1 receptor. Peptides corresponding to sites on the AT1 receptor's second extracellular loop abolished the stimulatory effect. Western blotting with purified patient IgG and a commercially obtained AT1 receptor antibody produced bands of identical molecular weight. Furthermore, confocal microscopy of vascular smooth muscle cells showed colocalization of purified patient IgG and AT1 receptor antibody. The protein kinase C (PKC) inhibitor calphostin C prevented the stimulatory effect. Our results suggest that preeclamptic patients develop stimulatory autoantibodies against the second extracellular AT1 receptor loop. The effect appears to be PKC-mediated. These novel autoantibodies may participate in the angiotensin II-induced vascular lesions in these patients.


Subject(s)
Autoantibodies/immunology , Pre-Eclampsia/immunology , Receptors, Angiotensin/agonists , Amino Acid Sequence , Angiotensin II/pharmacology , Angiotensin Receptor Antagonists , Animals , Cells, Cultured , Female , Heart Ventricles/immunology , Humans , Immunoglobulin G/pharmacology , Immunoglobulin M/pharmacology , Molecular Sequence Data , Muscle, Smooth, Vascular/immunology , Myocardial Contraction/drug effects , Myocardial Contraction/immunology , Naphthalenes/pharmacology , Peptide Fragments/pharmacology , Postpartum Period , Pregnancy , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/immunology
11.
Arterioscler Thromb Vasc Biol ; 19(1): 178-85, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9888881

ABSTRACT

The heparin-binding protein vascular endothelial growth factor (VEGF) is a highly specific growth factor for endothelial cells. VEGF binds to specific tyrosine kinase receptors, which mediate intracellular signaling. We investigated 2 hypotheses: (1) VEGF affects intracellular calcium [Ca2+]i regulation and [Ca2+]i-dependent messenger systems; and (2) these mechanisms are important for VEGF's proliferative effects. [Ca2+]i was measured in human umbilical vein endothelial cells using fura-2 and fluo-3. Protein kinase C (PKC) activity was measured by histone-like pseudosubstrate phosphorylation. PKC isoform distribution was observed with confocal microscopy and Western blot. Inhibition of PKC isoforms was assessed by specific antisense oligonucleotides (ODN) for the PKC isoforms. VEGF (10 ng/mL) induced a transient increase in [Ca2+]i followed by a sustained elevation. The sustained [Ca2+]i plateau was abolished by EGTA. Pertussis toxin also abolished the plateau phase, whereas the initial peak was not affected. The PKC isoforms alpha, delta, epsilon, and zeta were identified in endothelial cells. VEGF induced a translocation of PKC-alpha and PKC-zeta toward the nucleus and the perinuclear area, whereas cellular distribution of PKC-delta and PKC-epsilon was not influenced. Cell exposure to TPA led to a down-regulation of PKC-alpha and reduced the proliferative effect of VEGF. VEGF-induced endothelial cell proliferation also was reduced by the PKC inhibitors staurosporine and calphostin C. Specific down-regulation of PKC-alpha and PKC-zeta with antisense ODN reduced the proliferative effect of VEGF significantly. Our data show that VEGF induces initial and sustained Ca2+ influx. VEGF leads to the translocation of the [Ca2+]i-sensitive PKC isoform alpha and the atypical PKC isoform zeta. Antisense ODN for these PKC isoforms block VEGF-induced proliferation. These findings suggest that PKC isoforms alpha and zeta are important for VEGF's angiogenic effects.


Subject(s)
Cell Division , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Isoenzymes/metabolism , Lymphokines/pharmacology , Protein Kinase C/metabolism , Calcium/metabolism , Calcium/pharmacology , Egtazic Acid/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , Microscopy, Confocal , Naphthalenes/pharmacology , Oligodeoxyribonucleotides, Antisense/pharmacology , Pertussis Toxin , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-alpha , Signal Transduction , Staurosporine/pharmacology , Umbilical Veins , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Virulence Factors, Bordetella/pharmacology
12.
J Am Soc Nephrol ; 10 Suppl 11: S75-83, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9892144

ABSTRACT

Angiotensin II (AngII) is present inside vascular smooth muscle cells (VSMC); however, its intracellular functions, if any, are unknown. AngII was administered by microinjection. AngII was identified in endosomes and in the nucleus. Microinjection of AngII (10(-10) M) led to a rapid increase in the intracellular Ca2+ concentration ([Ca2+]i) in the cytosol and in the nucleus. The [Ca2+]i increase was the result of an influx of extracellular Ca2+ ions. The intracellular AngII effect was totally inhibited by concomitant injection of the AngII type 1 receptor blocker candesartan. Desensitization of extracellular AngII receptors, on the other hand, did not influence the intracellular effects, and neither did extracellular candesartan. The increase in [Ca2+]i was observed not only in the microinjected cell but also in directly adjacent VSMC. In contrast to the microinjected cells, the [Ca2+]i increase in the adjacent cells was mostly the result of Ca2+ release from intracellular stores. Pretreatment with thapsigargin, which interferes with Ca2+ release from intracellular stores, abolished the AngII response in adjacent cells. Microinjection of inositol trisphosphate induced a [Ca2+]i response in adjacent cells that was similar to the AngII-induced effects. Preincubation of VSMC with uncoupling substances did not decrease the AngII response but prevented a [Ca2+]i surge in adjacent cells. Tyrosine phosphorylation was next examined. Phosphorylation was detected in the injected cells, primarily in the cytoskeleton. It can be concluded that intracellular AngII binds to intracellular AngII receptors and elicits increased [Ca2+]i in the injected cell and then in cells in the immediate neighborhood. Cell-cell contact is necessary for the AngII-mediated effects. These data suggest that intracellular AngII may stimulate a cluster of VSMC from a single cell, via the release of second messengers.


Subject(s)
Angiotensin II/metabolism , Muscle, Smooth, Vascular/metabolism , Angiotensin II/pharmacology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cell Nucleus/metabolism , Cells, Cultured , Cytoplasm/metabolism , Inositol Phosphates/metabolism , Inositol Phosphates/pharmacology , Microinjections , Microscopy, Confocal , Nitrendipine/pharmacology , Rats
13.
Lancet ; 351(9107): 945-9, 1998 Mar 28.
Article in English | MEDLINE | ID: mdl-9734941

ABSTRACT

BACKGROUND: Oedema and vascular leakage play a part in the pathogenesis of pre-eclampsia. We tested the hypothesis that serum from pre-eclamptic patients increases endothelial-cell permeability and examined possible signal-transduction pathways. METHODS: We studied eight patients with pre-eclampsia, eight normotensive pregnant women, eight non-pregnant women, five pregnant patients with pre-existing hypertension, and four hypertensive non-pregnant women. Cultured human umbilical-vein endothelial-cell monolayers were used and permeability was measured by albumin flux. The part played by protein kinase C (PKC) signalling was examined by down-regulation with phorbol ester and with the inhibitors Goe 6976 and staurosporine. PKC isoforms were assessed by western blot and confocal microscopy. Antisense oligodesoxynucleotides (ODN) were used to test for specific PKC isoforms. FINDINGS: Serum from pre-eclamptic women increased endothelial permeability significantly (by 100%, p<0.01). The change in permeability decreased rapidly after delivery. Serum from normotensive pregnant women and non-pregnant women had no effect. Permeability was not influenced by serum from patients with essential hypertension or pregnant patients with pre-existing hypertension. Serum from pre-eclamptic patients induced a translocation of PKC isoforms alpha and epsilon within the cells. Goe 6976 and staurosporine (10(-8) mol/L) inhibited the increase in permeability induced by serum from pre-eclamptic patients. Down-regulation of PKC alpha and, to a lesser extent, PKC epsilon by antisense ODN also inhibited the pre-eclampsia-induced permeability increase. INTERPRETATION: Serum from pre-eclamptic patients contains a factor or factors that increase endothelial-cell permeability. The effect of pre-eclamptic serum may be mediated by PKC alpha and epsilon.


Subject(s)
Cell Membrane Permeability , Endothelium, Vascular/physiopathology , Pre-Eclampsia/physiopathology , Protein Kinase C/metabolism , Adult , Female , Humans , Isoenzymes , Pre-Eclampsia/blood , Pregnancy , Signal Transduction/physiology
14.
FASEB J ; 12(7): 593-601, 1998 May.
Article in English | MEDLINE | ID: mdl-9576486

ABSTRACT

Despite intensive interest in understanding the differentiation of vascular smooth muscle cells (VSMC), no information is available about differential regulation of ion channels in these cells. Since expression of the L-type Ca2+ channel can be influenced by differentiation in other cell types, we tested the hypothesis that the L-type (C class) channel is a specific differentiation marker of VSMC and that expression of these channels depends on the state of cell differentiation. We used rat aortic (A7r5) VSMC, which express functional L-type Ca2+ channels, and induced dedifferentiation by cell culture in different media. Treatment with retinoic acid was used to redifferentiate the VSMC. We characterized the differentiated state of the cells by using immunohistochemistry and Western blot analysis for smooth muscle (SM) alpha-actin and SM-myosin heavy chain (MHC). The number of functional Ca2+ channels was significantly decreased in dedifferentiated VSMC and increased upon differentiation with retinoic acid. Ca2+ channel function was assessed by whole-cell voltage clamp techniques. Using Western blot and dihydropyridine binding analysis, we found that the expression of the Ca2+ channel alpha1 subunit, and to a lesser extent the beta2 subunit, was directly correlated with the expression of SM alpha-actin and SM-MHC. We conclude that expression of L-type Ca2+ channel alpha1 subunits, and thus a functional Ca2+ channel, is highly coordinated with expression of the SM-specific proteins required for specialized smooth muscle cell functions. Furthermore, our results demonstrate that the L-type Ca2+ channel is a novel marker for differentiation of VSMC. The data suggest that regulation of ion channel expression during differentiation may have physiological importance for normal smooth muscle function and may influence VSMC behavior under pathophysiological conditions.


Subject(s)
Calcium Channels/biosynthesis , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Actins/analysis , Animals , Aorta , Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Calcium Channels, L-Type , Cell Differentiation/drug effects , Cell Line , Culture Media , Dihydropyridines/metabolism , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Muscle, Smooth, Vascular/drug effects , Myosin Heavy Chains/analysis , Nimodipine/pharmacology , Patch-Clamp Techniques , Rats , Tretinoin/pharmacology
15.
Circ Res ; 82(2): 157-65, 1998 Feb 09.
Article in English | MEDLINE | ID: mdl-9468186

ABSTRACT

The extracellular matrix influences the cellular spreading of vascular smooth muscle cells (VSMCs) via integrin receptors. However, the intracellular signaling mechanisms are still incompletely understood. We investigated the hypothesis that VSMCs binding to fibronectin activates the protein kinase C (PKC) pathway, causes differential intracellular PKC isoform translocation, and mediates cell spreading. VSMCs binding to poly-L-lysine or preincubated with Arg-Gly-Asp (RGD) peptides were used as controls. Diacylglycerol (DAG) and phospholipase D (PLD) activity were measured by thin-layer chromatography. Intracellular distribution of PKC isoforms was assessed by confocal microscopy. VSMCs binding to fibronectin induced focal adhesions and cell spreading within 30 minutes. Fibronectin induced a rapid increase in DAG content, peaking at 10 minutes with a sustained response for <1 hour. In contrast, PLD activity was not influenced by specific binding to fibronectin. PKC isoforms alpha, delta, epsilon, and zeta were assessed by confocal microscopy. Fibronectin induced a PKC isoform translocation to the cell nucleus and to focal adhesions within minutes. The nuclear PKCalpha immunoreactivity was transiently increased. PKC isoforms a and epsilon were both translocated to focal adhesions. The intracellular distributions of other PKC isoforms were not influenced by fibronectin. The effects of fibronectin on DAG generation, the translocation of PKCalpha and PKCepsilon, and cell spreading were all abolished by the incubation with RGD peptides. Downregulation of PKC isoforms alpha and epsilon with specific antisense oligodinucleotides resulted in a significant inhibition of cell spreading. Our results show that integrins induce intracellular signaling in VSMCs via DAG and PKC. PKC isoform a is translocated to the nucleus, whereas PKC isoforms alpha and epsilon are translocated to focal adhesions. Both isoforms seem to play a role in inside-out integrin signaling and cell spreading.


Subject(s)
Integrins/physiology , Isoenzymes/metabolism , Muscle, Smooth, Vascular/cytology , Protein Kinase C/metabolism , Animals , Biological Transport/physiology , Cell Adhesion/physiology , Diglycerides/biosynthesis , Fibronectins/physiology , Isoenzymes/antagonists & inhibitors , Oligonucleotides, Antisense/pharmacology , Protein Kinase C/antagonists & inhibitors , Rats
16.
Am J Med Sci ; 315(1): 17-23, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9427570

ABSTRACT

The occurrence of autoimmune phenomena in 28 patients with thromboangiitis obliterans (Buerger's disease) was determined. The following were sought: antineutrophil cytoplasmic antibodies against proteinase 3 (cANCA) and myeloperoxidase (pANCA), antinuclear antibodies, anti-Ro antibodies, anticardiolipin antibodies, and antiendothelial cell antibodies (AECA). For the last, an enzyme-linked immunosorbent assay was developed which verified the presence of the AECA phenomenon with immunofluorescence and confocal microscopy. Seven patients with active disease had AECA titers of 1,857 +/- 450 arbitrary units (AU) compared with 126 +/- 15 AU in 30 normal control subjects (P < 0.001) and 461 +/- 41 AU in 21 patients in remission (P < 0.01). Antibodies from the sera of patients with active disease reacted not only with surface epitopes but also with sites within the cytoplasm of human endothelial cells. AECA may be useful in following disease activity and may play a role in the pathogenesis of thromboangiitis obliterans.


Subject(s)
Autoantibodies/blood , Endothelium, Vascular/immunology , Thromboangiitis Obliterans/immunology , Adult , Antibodies, Anticardiolipin/blood , Antibodies, Antineutrophil Cytoplasmic/blood , Azathioprine/therapeutic use , Biomarkers/blood , Enzyme-Linked Immunosorbent Assay , Humans , Immunosuppressive Agents/therapeutic use , Male , Myeloblastin , Peroxidase/immunology , Prednisolone/therapeutic use , Reference Values , Serine Endopeptidases/immunology , Smoking , Thromboangiitis Obliterans/blood , Thromboangiitis Obliterans/drug therapy
17.
Acta Physiol Scand ; 164(4): 599-609, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9887982

ABSTRACT

Protein kinase C is an important second messenger system, which is translocated from the cytosol to the cell membrane upon cell stimulation. We used confocal microscopy to study the spatial distribution of protein kinase C isoforms after stimulation of cultured vascular smooth muscle cells with different agonists. First, we analysed the effects of angiotensin II and platelet-derived growth factor (PDGF). Confocal microscopy showed a rapid assembly of PKC alpha along cytosolic fibres followed by a translocation towards the nucleus with angiotensin II. PDGF engendered a similar, but much slower response; however, a cytoskeletal distribution was not observed. We then investigated the effects of thrombin and bFGF on nuclear translocation. bFGF induced a rapid translocation of the isoform towards the perinuclear region and into the nucleus. bFGF had a similar effect on PKC epsilon. In contrast, thrombin had a smaller effect on nuclear translocation of PKC alpha and did not influence PKC epsilon, but instead induced a rapid nuclear translocation of PKC zeta. Thus, tyrosine kinase receptor activation via bFGF induces a rapid association of PKC alpha and epsilon within nuclear structures. Our results show that agonists cause, not only a translocation of protein kinase C isoforms into the cell membrane but also into the cell nucleus. Lastly, we analyzed the nuclear immunoreactivity of the PKC isoforms, alpha, delta, epsilon and zeta in vascular smooth muscle cells during the cell cycle. Resting cells were stimulated with foetal calf serum (FCS, 10%), which translocated PKC alpha and epsilon to the perinuclear region and into the nucleus, while PKC delta and zeta showed no increase in nuclear immunoreactivity. After 4 h of FCS, the nuclear immunoreactivity for PKC alpha and epsilon was reduced to or below control values. At 8 h, increased nuclear expression of isoforms alpha, epsilon and zeta was observed, while isoform delta was not affected. Our results demonstrate a complex spatial and temporal regulation of PKC isoforms in response to vasoactive hormones and growth factors. We suggest that protein kinase C may be important for nuclear signaling and demonstrate that nuclear translocation of PKC isoforms is differentially regulated during the cell cycle.


Subject(s)
Isoenzymes/physiology , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiology , Protein Kinase C/physiology , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Angiotensin II/physiology , Animals , Cell Division/physiology , Cell Nucleus/physiology , Cells, Cultured , Fibroblast Growth Factor 2/physiology , Immunohistochemistry , Microscopy, Confocal , Muscle, Smooth, Vascular/cytology , Platelet-Derived Growth Factor/physiology , Rats , Thrombin/physiology
18.
Circ Res ; 81(3): 363-71, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9285638

ABSTRACT

Endothelial cell permeability is impaired in diabetes mellitus and may be increased by high extracellular glucose concentrations. High glucose activates protein kinase C (PKC), a family of kinases vital to intracellular signaling. We tested the hypothesis that high glucose concentration activates PKC in endothelial cells and leads to an increase in endothelial cell permeability via distinct PKC isoforms. Porcine aortic endothelial cells were used, and the PKC isoforms alpha, delta, epsilon, zeta, and theta were identified in these cells. Glucose caused a rapid dose-dependent increase in endothelial cell permeability, with an EC50 of 17.5 mmol/L. Phorbol 12-myristate 13-acetate (TPA) induced an increase in permeability very similar to that elicited by glucose. The effect of glucose and TPA was totally reversed by preincubating the cells with the PKC inhibitors staurosporine (10(-8) mol/L) and Goe 6976 (10(-8) mol/L). Downregulation of PKC by preincubation with TPA for 24 hours also abolished the effect of glucose and TPA on endothelial cell permeability. High glucose (20 mmol/L) caused an increase in PKC activity at 2, 10, and 30 minutes. Cell fractionation and Western blot analysis showed a glucose-induced translocation of PKC alpha and PKC epsilon. Confocal microscopy confirmed the translocation and showed an association of PKC alpha and PKC epsilon with nuclear structures and the cell membrane. Specific antisense oligodesoxynucleotides (ODNs) against PKC alpha reduced the expression of the isoform, abolished the effects of glucose on endothelial cell permeability completely, and reduced the TPA effect significantly. In contrast, specific antisense ODNs against PKC epsilon had no effect on glucose-induced permeability and only a minor effect on the TPA-induced increase in permeability. We conclude that an increase in extracellular glucose leads to a rapid dose-dependent increase in endothelial cell permeability via the activiation of PKC and that this effect is mediated by the PKC isoform alpha.


Subject(s)
Cell Membrane Permeability/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Glucose/pharmacology , Isoenzymes/metabolism , Protein Kinase C/metabolism , Animals , Base Sequence , Cell Membrane Permeability/physiology , Cells, Cultured , Diabetic Angiopathies/enzymology , Diabetic Angiopathies/etiology , Diabetic Angiopathies/physiopathology , Enzyme Activation/drug effects , Humans , Immunohistochemistry , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Oligonucleotide Probes/genetics , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C-alpha , Protein Kinase C-epsilon , Swine , Tetradecanoylphorbol Acetate/pharmacology
19.
FASEB J ; 11(11): 905-15, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9285489

ABSTRACT

Vascular smooth muscle cell (VSMC) differentiation is important in understanding vascular disease; however, no in vitro model is available. Totipotent mouse embryonic stem (ES) cells were used to establish such a model. To test whether the ES cell-derived smooth muscle cells expressed VSMC-specific properties, the differentiated cells were characterized by 1) morphological analysis, 2) gene expression, 3) immunostaining for VSMC-specific proteins, 4) expression of characteristic VSMC ion channels, and 5) formation of [Ca2+]i transients in response to VSMC-specific agonists. Treatment of embryonic stem cell-derived embryoid bodies with retinoic acid and dibutyryl-cyclic adenosine monophosphate (db-cAMP) induced differentiation of spontaneously contracting cell clusters in 67% of embryoid bodies compared with 10% of untreated controls. The highest differentiation rate was observed when retinoic acid and db-cAMP were applied to the embryoid bodies between days 7 and 11 in combination with frequent changes of culture medium. Other protocols with retinoic acid and db-cAMP, as well as single or combined treatment with VEGF, ECGF, bFGF, aFGF, fibronectin, matrigel, or hypoxia did not influence the differentiation rate. Single-cell RT-PCR and sequencing of the PCR products identified myosin heavy chain (MHC) splice variants distinguishing between gut and VSMC isoforms. RT-PCR with VSMC-specific MHC primers and immunostaining confirmed the presence of VSMC transcripts and MHC protein. Furthermore, VSMC expressing MHC had typical ion channels and responded to specific agonists with an increased [Ca2+]i. Here we present a retinoic acid + db-cAMP-inducible embryonic stem cell model of in vitro vasculogenesis. ES cell-derived cells expressing VSMC-specific MHC and functional VSMC properties may be a suitable system to study mechanisms of VSMC differentiation.


Subject(s)
Bucladesine/pharmacology , Embryo, Mammalian/cytology , Muscle, Smooth, Vascular/cytology , Stem Cells/cytology , Tretinoin/pharmacology , Animals , Base Sequence , Calcium/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Mice , Molecular Sequence Data , Myosin Heavy Chains/analysis , Potassium Channels/physiology
20.
FEBS Lett ; 406(1-2): 61-5, 1997 Apr 07.
Article in English | MEDLINE | ID: mdl-9109386

ABSTRACT

Using activity measurements and Western blotting, we demonstrated that PKC alpha is constitutively present in nuclei of Neuro-2a neuroblastoma cells. Confocal microscopy revealed that PKC alpha is present in the nucleoplasm and that this localization does not change after stimulation with phorbol ester. However, as revealed by extraction experiments, phorbol ester leads to a firmer association of PKC alpha with nuclear components. Our findings suggest that PKC alpha not only associates with lipids but also with proteins inside the nucleus. The presence of active PKC alpha inside the nucleus allows the enzyme to phosphorylate not only proteins at the nuclear envelope but also proteins in the nucleoplasm.


Subject(s)
Cell Nucleus/enzymology , Isoenzymes/metabolism , Neuroblastoma/enzymology , Protein Kinase C/metabolism , Animals , Immunohistochemistry , Mice , Neuroblastoma/pathology , Protein Kinase C-alpha , Subcellular Fractions/enzymology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...