101
|
Lu L, Khan S, Lencer W, Walker WA. Endocytosis of cholera toxin by human enterocytes is developmentally regulated. Am J Physiol Gastrointest Liver Physiol 2005; 289:G332-41. [PMID: 15790757 DOI: 10.1152/ajpgi.00521.2004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Many secretory diarrheas including cholera are more prevalent and fulminant in young infants than in older children and adults. Cholera toxin (CT) elicits a cAMP-dependent chloride secretory response in intestinal epithelia, which accounts for the fundamental pathogenesis of this toxigenic diarrhea. We have previously reported that the action of this bacterial enterotoxin is excessive in immature enterocytes and under developmental regulation. In this study, we tested the hypothesis that enhanced endocytosis by immature human enterocytes may, in part, account for the excessive secretory response to CT noted in the immature intestine and that enterocyte endocytosis of CT is developmentally regulated. To test this hypothesis, we used specific inhibitors to define endocytic pathways in mature and immature cell lines. We showed that internalization of CT in adult enterocytes is less and occurs via the caveolae/raft-mediated pathway in contrast to an enhanced immature human enterocyte CT uptake that occurs via a clathrin pathway. We also present evidence that this clathrin pathway is developmentally regulated as demonstrated by its response to corticosteroids, a known maturation factor that causes a decreased CT endocytosis by this pathway.
Collapse
Affiliation(s)
- Lei Lu
- Developmental Gastroenterology Laboratory, Massachusetts General Hospital, 114 16th St. (114-3503), Charlestown, MA 02129-4404, USA
| | | | | | | |
Collapse
|
102
|
Kjolby MJ, Kompanowska-Jezierska E, Wamberg S, Bie P. Effects of sodium intake on plasma potassium and renin angiotensin aldosterone system in conscious dogs. ACTA ACUST UNITED AC 2005; 184:225-34. [PMID: 15954990 DOI: 10.1111/j.1365-201x.2005.01452.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIMS The operating range of the renin-angiotensin-aldosterone system is ill-defined. This study quantifies renin-angiotensin-aldosterone system activity as a function of sodium intake. METHODS Renin-angiotensin-aldosterone system variables were measured daily after a sudden reduction in sodium intake (3.0-0.5 mmol kg(-1) day(-1)) or at steady states generated by eight levels of sodium intake (0.5-8.0 mmol kg(-1) day(-1)). Potassium intake was 2.79 +/- 0.03 mmol kg(-1) day(-1). Arterial blood pressure was measured invasively. Hormone concentrations were determined by radioimmunoassays. Glomerular filtration rate and plasma volume were determined by standard methods. RESULTS Sudden sodium intake reduction doubled plasma renin activity and angiotensin II, and tripled aldosterone on day 1 with only small non-significant additional changes on the following days. Different levels of sodium intake did not affect arterial blood pressure, heart rate, and plasma concentrations of sodium, angiotensinogen, atrial natriuretic peptide, vasopressin, glomerular filtration rate and diuresis. With increasing sodium intake, plasma volume increased by 0.47 +/- 0.04 mL (kg body mass)(-1) (unit increase in Na intake)(-1) (P < 0.01), and plasma potassium decreased with the slope -0.038 mm [(mmol Na+ intake) (kg body mass)(-1) day(-1)](-1) (P = 0.001) while plasma renin-activity, angiotensin II, and aldosterone decreased systematically as expected. CONCLUSIONS A step reduction in sodium intake alters renin-angiotensin-aldosterone system activity on day 1 with little further change the subsequent 4 days. Week-long increases in sodium intake decreases renin-angiotensin-aldosterone system activity, increases plasma volume, and decreases plasma potassium. Isolated decreases in sodium intake increase aldosterone secretion via volume-mediated action on the renin-angiotensin system and via increases in plasma potassium.
Collapse
Affiliation(s)
- M J Kjolby
- Department of Physiology and Pharmacology, Institute of Medical Biology, University of Southern Denmark, Odense, Denmark
| | | | | | | |
Collapse
|
103
|
Cugno M, Agostoni P, Mari D, Meroni PL, Gregorini L, Bussotti M, Anguissola GB, Donatelli F, Nussberger J. Impaired bradykinin response to ischaemia and exercise in patients with mild congestive heart failure during angiotensin-converting enzyme treatment. Relationships with endothelial function, coagulation and inflammation. Br J Haematol 2005; 130:113-20. [PMID: 15982353 DOI: 10.1111/j.1365-2141.2005.05569.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Inflammation and endothelial dysfunction play important roles in the pathophysiology of congestive heart failure (CHF), and the peptide bradykinin, generated during inflammation, may act as a defence mechanism by inducing vasodilation. Plasma bradykinin levels are increased in experimental heart failure but low in patients with advanced chronic CHF despite treatment with angiotensin-converting enzyme (ACE) inhibitors. It is not currently known how bradykinin behaves in less severe phases of CHF controlled by long-term ACE inhibitor treatment. We studied 10 male patients with clinically stable chronic CHF [New York Heart Association (NYHA) class II] on long-term ACE inhibitor treatment and 10 normal sex- and age-matched control subjects. High performance liquid chromatography/radioimmunoassay methods were used to evaluate plasma levels of bradykinin in relation to an array of parameters of endothelial function, coagulation and inflammation before and after stimuli of forearm arterial occlusion and physical exercise. CHF patients had higher levels of bradykinin (P = 0.008), activated factor XII (P = 0.049), interleukin-6 (P = 0.050) and tumour necrosis factor receptor II (sTNFRII) (P = 0.026) than controls. Arterial occlusion and exercise significantly increased bradykinin and von Willebrand factor levels in controls but not in CHF patients. The increase in brachial artery diameter after arterial occlusion was less in CHF patients (P = 0.036) and inversely related to baseline plasma levels of bradykinin (r = -0.855, P = 0.002) and sTNFRII (r = -0.780, P = 0.008). NYHA class II CHF patients during long-term treatment with ACE inhibitors have increased bradykinin levels and signs of inflammation. They are unable to respond adequately to stimuli of ischaemia and physical exercise which both require vasodilation.
Collapse
Affiliation(s)
- Massimo Cugno
- Department of Internal Medicine, University of Milan, Milan, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Gioli-Pereira L, Nascimento EA, Santos EL, Bracht A, Juliano MA, Pesquero JB, Borges DR, Kouyoumdjian M. Fate of bradykinin on the rat liver when administered by the venous or arterial route. J Gastroenterol Hepatol 2005; 20:463-73. [PMID: 15740493 DOI: 10.1111/j.1440-1746.2005.03580.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND AIM Bradykinin (BK) infused into the portal vein elicits a hypertensive response via the B2 receptor (B2R) and is efficiently hydrolyzed by the liver. Our purpose was to characterize the mechanism of interaction between BK and the liver. METHOD BK, HOE-140 (a B2R antagonist), des-R(9)-BK (a B1R agonist) and enzyme inhibitors were used in monovascular or bivascular perfusions and in isolated liver cell assays. RESULTS Des-R(9)-BK did not elicit a portal hypertensive response (PHR); BK infused into the hepatic artery elicited a calcium-dependent PHR and a calcium-independent arterial hypertensive response (HAHR), with the latter being almost abolished by naproxen. BK has a predominant distribution in the extracellular space and an average hepatic extraction of 8% in the steady state. Hydrolysis products of infused BK (R(1)-F(5) and R(1)-P(7)) did not elicit PHR. Angiotensin converting enzyme (ACE) is concentrated in the perivenous region and B2R in the periportal region. Microphysiometry showed that BK (and not a B1 agonist) interacts with stellate cells and the endothelial sinusoidal/Kupffer cell fraction. This effect was inhibited by the B2R antagonist. CONCLUSIONS Events can be summarized as: the hypertensive action of BK on sinusoidal cells of the periportal region is followed by its hydrolysis by ACE which is primarily present in the perivenous region; there is no functional B1R in the normal liver; BK induces HAHR via eicosanoid release and PHR by a distinct pathway on the B2R. Our data suggest that BK may participate in the modulation of sinusoidal microvasculature tonus both in the portal and the arterial routes.
Collapse
MESH Headings
- Adrenergic beta-Antagonists/pharmacology
- Animals
- Bradykinin/administration & dosage
- Bradykinin/analogs & derivatives
- Bradykinin/pharmacokinetics
- Bradykinin/pharmacology
- Bradykinin B2 Receptor Antagonists
- Chromatography, High Pressure Liquid
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Extracellular Space/metabolism
- Fluorescent Antibody Technique
- Hepatic Artery
- Hydrolysis/drug effects
- Hypertension, Portal/drug therapy
- Hypertension, Portal/metabolism
- Hypertension, Portal/physiopathology
- Infusions, Intra-Arterial
- Infusions, Intravenous
- Kupffer Cells/cytology
- Kupffer Cells/drug effects
- Kupffer Cells/metabolism
- Liver/cytology
- Liver/drug effects
- Liver/metabolism
- Liver Circulation/drug effects
- Liver Circulation/physiology
- Male
- Mass Spectrometry
- Peptidyl-Dipeptidase A/metabolism
- Portal Pressure/drug effects
- Portal Pressure/physiology
- Portal Vein
- Rats
- Rats, Wistar
- Receptor, Bradykinin B2/metabolism
Collapse
|
105
|
Nascimento EA, Gioli-Pereira L, Carvalho LT, Santos EL, Pesquero JB, Kouyoumdjian M, Borges DR. Hemodynamic and metabolic effects of angiotensin II on the liver. Peptides 2005; 26:315-22. [PMID: 15629544 DOI: 10.1016/j.peptides.2004.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2004] [Revised: 09/28/2004] [Accepted: 09/28/2004] [Indexed: 10/26/2022]
Abstract
To ascertain the mechanism of interaction between angiotensins (AI and AII) and the liver, an angiotensin-converting enzyme inhibitor (captopril) and a receptor antagonist (losartan) were used. Monovascular or bivascular liver perfusion was used to assess both hemodynamic (portal and arterial hypertensive responses) and metabolic (glucose production and oxygen consumption) effects. Microphysiometry was used for isolated liver cell assays to assess AII or losartan membrane receptor-mediated interaction. Captopril abolishes portal hypertensive response (PHR) to AI but not the AII effect. AII infused via the portal pathway promotes calcium-dependent PHR but not a hypertensive response in the arterial pathway (AHR); when infused into the arterial pathway AII promotes calcium-dependent PHR and AHR. Losartan infused into the portal vein abolishes PHR to AII but not the metabolic response; when infused via both pathways it abolishes the hypertensive responses and inhibits the metabolic effects. Isolated liver cells specifically respond to AII. Sinusoidal cells, but not hepatocytes, respond to 10 nM losartan. We conclude that AI has to be converted to AII to produce PHR. Quiescent stellate cells interacts in vitro with AII and losartan. Hemodynamic responses to AII are losartan-dependent but metabolic responses are partially losartan-independent. AII hemodynamic actions are mainly presinusoidal.
Collapse
|
106
|
Barki-Harrington L. Oligomerisation of angiotensin receptors: novel aspects in disease and drug therapy. J Renin Angiotensin Aldosterone Syst 2004; 5:49-52. [PMID: 15295714 DOI: 10.3317/jraas.2004.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The concept that 7 transmembrane receptors (7TMRs) exist and function as independent monomers has facilitated a therapeutic approach of selective targeting of receptors. However, oligomerisation of 7TMRs is now being recognised as an important biological phenomenon that adds a level of complexity to their signalling. In vitro, many 7TMR heterodimers display altered binding, signalling, and trafficking properties compared to their constituent monomeric units. This review discusses an emerging concept regarding the role of 7TMR heterodimerisation in vivo, and its significance to drug therapy. Studies of angiotensin receptor signalling indicate a pivotal role for heterodimerisation in the pathogenesis of human disorders. Furthermore, the occurrence of angiotensin receptor heterodimers has a profound effect on the responsiveness to treatment with 7TMR blockers. Global assessment of the prevalence of different heterodimers during disease and their responsiveness to drug therapy is likely to optimise patient treatment and reduce side-effects associated with 7TMR blockers.
Collapse
Affiliation(s)
- Liza Barki-Harrington
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, 27710, USA.
| |
Collapse
|
107
|
Müller AM, Gruhn K, Lange S, Franke FE, Müller KM. [Angiotensin converting enzyme (ACE, CD143) in the regular pulmonary vasculature]. DER PATHOLOGE 2004; 25:141-6. [PMID: 15011000 DOI: 10.1007/s00292-004-0681-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Angiotensin I converting enzyme (ACE, CD143) is an endothelial transmembrane Zn2+-dipeptidylpeptidase. By formation of angiotensin II and degrading bradykinin it acts as a vasoconstrictor. We examined endothelial ACE expression in human pulmonary vessels in specimens from 20 female and 19 male patients (age: 34-76 years) by immunohistochemistry. In all specimens, capillary endothelial cells showed the strongest expression, followed by those in arterioles and arteries. Venules and veins showed next to no staining. The differences in staining intensities were significant ( P<0.001). Sex affected neither the expression intensity nor the expression pattern. Summarizing, we demonstrate the existence of a vessel-type specific ACE expression pattern for pulmonary vessels. The nearly exclusive endothelial ACE expression in capillaries and arterial vessels points to ACE as an immunohistochemical marker for these vessels in normal lung tissue.
Collapse
Affiliation(s)
- A M Müller
- Institut für Pathologie an den Berufsgenossenschaftlichen Kliniken Bergmannsheil-Universitätsklinik, Bergmannsheil der Ruhr-Universität Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany.
| | | | | | | | | |
Collapse
|
108
|
Tan Y, Hutchison FN, Jaffa AA. Mechanisms of angiotensin II-induced expression of B2 kinin receptors. Am J Physiol Heart Circ Physiol 2004; 286:H926-32. [PMID: 14766673 DOI: 10.1152/ajpheart.00757.2003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although the primary roles of the kallikreinkinin system and the renin-angiotensin system are quite divergent, they are often intertwined under pathophysiological conditions. We examined the effect of ANG II on regulation of B(2) kinin receptors (B2KR) in vascular cells. Vascular smooth muscle cells (VSMC) were treated with ANG II in a concentration (10(-9)-10(-6) M)- and time (0-24 h)-dependent manner, and B2KR protein and mRNA levels were measured by Western blots and PCR, respectively. A threefold increase in B2KR protein levels was observed as early as 6 h, with a peak response at 10(-7) M. ANG II (10(-7) M) also increased B2KR mRNA levels twofold 4 h after stimulation. Actinomycin D suppressed the increase in B2KR mRNA and protein levels induced by ANG II. To elucidate the receptor subtype involved in mediating this regulation, VSMC were pretreated with losartan (AT(1) receptor antagonist) and/or PD-123319 (AT(2) receptor antagonist) at 10 microM for 30 min, followed by ANG II (10(-7) M) stimulation. Losartan completely blocked the ANG II-induced B2KR increase, whereas PD-123319 had no effect. In addition, expression of B2KR mRNA levels was decreased in AT(1A) receptor knockout mice. Finally, to determine whether ANG II stimulates B2KR expression via activation of the MAPK pathway, VSMC were pretreated with an inhibitor of p42/p44(mapk) (PD-98059) and/or an inhibitor of p38(mapk) (SB-202190), followed by ANG II (10(-7) M) for 24 h. Selective inhibition of the p42/p44(mapk) pathway significantly blocked the ANG II-induced increase in B2KR expression. These findings demonstrate that ANG II regulates expression of B2KR in VSMC and provide a rationale for studying the interaction between ANG II and bradykinin in the pathogenesis of vascular dysfunction.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Aorta/cytology
- Cells, Cultured
- Extracellular Fluid/metabolism
- Gene Expression/drug effects
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Rats
- Rats, Sprague-Dawley
- Receptor, Bradykinin B2/genetics
- Receptor, Bradykinin B2/metabolism
- Signal Transduction/drug effects
- Vasoconstrictor Agents/pharmacology
Collapse
Affiliation(s)
- Yan Tan
- Dept. of Medicine, Endocrinology-Diabetes-Medical Genetics, Medical Univ. of South Carolina, 114 Doughty St., PO Box 250776, Charleston, SC 29425, USA
| | | | | |
Collapse
|
109
|
Shariat-Madar Z, Mahdi F, Schmaier AH. Recombinant prolylcarboxypeptidase activates plasma prekallikrein. Blood 2004; 103:4554-61. [PMID: 14996700 DOI: 10.1182/blood-2003-07-2510] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The serine protease prolylcarboxypeptidase (PRCP), isolated from human umbilical vein endothelial cells (HUVECs), is a plasma prekallikrein (PK) activator. PRCP cDNA was cloned in pMT/BIP/V5-HIS-C, transfected into Schneider insect (S2) cells, and purified from serum-free media. Full-length recombinant PRCP (rPRCP) activates PK when bound to high-molecular-weight kininogen (HK). Recombinant PRCP is inhibited by leupeptin, angiotensin II, bradykinin, anti-PRCP, diisopropyl-fluorophosphonate (DFP), phenylmethylsulfonyl fluoride (PMSF), and Z-Pro-Proaldehyde-dimethyl acetate, but not by 1 mM EDTA (ethylenediaminetetraacetic acid), bradykinin 1-5, or angiotensin 1-7. Corn trypsin inhibitor binds to prekallikrein to prevent rPRCP activation, but it does not directly inhibit the active site of either enzyme. Unlike factor XIIa, the ability of rPRCP to activate PK is blocked by angiotensin II, not by neutralizing antibody to factor XIIa. PRCP antigen is detected on HUVEC membranes using flow cytometry and laser scanning confocal microscopy. PRCP antigen does not colocalize with LAMP1 on nonpermeabilized HUVECs, but it partially colocalizes in permeabilized cells. PRCP colocalizes with all the HK receptors, gC1qR, uPAR, and cytokeratin 1 antigen, on nonpermeabilized HUVECs. PRCP activity and antigen expression on cultured HUVECs are blocked by a morpholino antisense oligonucleotide. These investigations indicate that rPRCP is functionally identical to isolated HUVEC PRCP and is a major HUVEC membrane-expressed, PK-activating enzyme detected in the intravascular compartment.
Collapse
Affiliation(s)
- Zia Shariat-Madar
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, 5301 MSRB III, 1150 W Medical Center Drive, Ann Arbor, MI 48109-0649, USA
| | | | | |
Collapse
|
110
|
Amin RP, Vickers AE, Sistare F, Thompson KL, Roman RJ, Lawton M, Kramer J, Hamadeh HK, Collins J, Grissom S, Bennett L, Tucker CJ, Wild S, Kind C, Oreffo V, Davis JW, Curtiss S, Naciff JM, Cunningham M, Tennant R, Stevens J, Car B, Bertram TA, Afshari CA. Identification of putative gene based markers of renal toxicity. ENVIRONMENTAL HEALTH PERSPECTIVES 2004; 112:465-79. [PMID: 15033597 PMCID: PMC1241901 DOI: 10.1289/ehp.6683] [Citation(s) in RCA: 172] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
This study, designed and conducted as part of the International Life Sciences Institute working group on the Application of Genomics and Proteomics, examined the changes in the expression profile of genes associated with the administration of three different nephrotoxicants--cisplatin, gentamicin, and puromycin--to assess the usefulness of microarrays in the understanding of mechanism(s) of nephrotoxicity. Male Sprague-Dawley rats were treated with daily doses of puromycin (5-20 mg/kg/day for 21 days), gentamicin (2-240 mg/kg/day for 7 days), or a single dose of cisplatin (0.1-5 mg/kg). Groups of rats were sacrificed at various times after administration of these compounds for standard clinical chemistry, urine analysis, and histological evaluation of the kidney. RNA was extracted from the kidney for microarray analysis. Principal component analysis and gene expression-based clustering of compound effects confirmed sample separation based on dose, time, and degree of renal toxicity. In addition, analysis of the profile components revealed some novel changes in the expression of genes that appeared to be associated with injury in specific portions of the nephron and reflected the mechanism of action of these various nephrotoxicants. For example, although puromycin is thought to specifically promote injury of the podocytes in the glomerulus, the changes in gene expression after chronic exposure of this compound suggested a pattern similar to the known proximal tubular nephrotoxicants cisplatin and gentamicin; this prediction was confirmed histologically. We conclude that renal gene expression profiling coupled with analysis of classical end points affords promising opportunities to reveal potential new mechanistic markers of renal toxicity.
Collapse
Affiliation(s)
- Rupesh P Amin
- National Institute of Environmental Health Sciences, National Institutes of Health/DHHS, Research Triangle Park, North Carolina, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Barros NMT, Puzer L, Tersariol ILS, Oliva MLV, Sampaio CAM, Carmona AK, Motta GD. Plasma prekallikrein/kallikrein processing by lysosomal cysteine proteases. Biol Chem 2004; 385:1087-91. [PMID: 15576330 DOI: 10.1515/bc.2004.141] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Plasma kallikrein plays a role in coagulation, fibrinolysis and inflammation. Cathepsins B and L participate in (patho)physiological processes such as peptide antigen processing, tissue remodeling events, protein turnover in cells, hormone processing and tumor invasion. The present work analyzes the processing of prekallikrein/kallikrein by lysosomal cathepsins. Prekallikrein is not hydrolyzed by catB, and catL generates an inactive fragment of prekallikrein. Both kallikrein chains are hydrolyzed by catL and the light chain is mainly hydrolyzed by catB; kallikrein activity is lower after incubation with catL compared to catB. Our data suggest that the plasma kallikrein/ kinin system can be controlled by cathepsins.
Collapse
Affiliation(s)
- Nilana M T Barros
- Departamento de Bioquímica, Universidade Federal de São Paulo/EPM, CEP 04044-020, São Paulo, SP, Brazil
| | | | | | | | | | | | | |
Collapse
|
112
|
Paterna S, Di Garbo V, Avellone G, Di Pasquale P, Cacia A, Tuttolomondo A, Follone G, Cardinale A, Maniscalchi T, Licata G. Effects of Losartan and Delapril on the Fibrinolytic System in Patients with Mild to Moderate Hypertension. Clin Drug Investig 2003; 23:717-24. [PMID: 17536885 DOI: 10.2165/00044011-200323110-00004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
BACKGROUND AND OBJECTIVES Angiotensin-converting enzyme (ACE) probably influences the fibrinolytic system at a central point by converting angiotensin I to angiotensin II, which increases plasminogen activator inhibitor-1 (PAI-1) activity. This effect appears to be mediated in humans via the angiotensin II type 1 (AT(1)) receptor. The objective of this study was to evaluate, in patients with mild to moderate hypertension, the change in tissue plasminogen activator (t-PA) and PAI-1 plasma levels after treatment with an AT(1)-receptor blocker (losartan 50 mg/day) or an ACE inhibitor (delapril 60 mg/day). PATIENTS AND METHODS 30 hypertensive patients and 15 controls were enrolled. Essential hypertension was established by a medical history, physical examination and the absence of clinical findings suggestive of a secondary form of hypertension. Preliminary investigations, routine biochemical tests (including clearance of creatinine and oral glucose tolerance test), chest x-ray, standard and 24-hour ECG monitoring, M- and B-mode echocardiography and fundus oculi examinations were performed. No patients had previously received ACE inhibitors or AT(1)-receptor blockers. After a 14-day run-in period with placebo, patients were randomised in a double-blind fashion into two groups: 15 patients were randomised to losartan 50 mg/day (group 1), 15 patients were randomised to delapril 60 mg/day (group 2), and 15 healthy subjects were used as controls (group 3). Plasma PAI-1 and t-PA antigen were determined by enzyme-linked immunosorbent assay and a photometric method at the end of the run-in period and after 6 months of treatment. RESULTS There were no significant differences among the three groups regarding age, sex, body mass index and smoking. After 6 months, both groups of patients showed a reduction in blood pressure values. The losartan group did not demonstrate significant changes in PAI-1 levels (96.52 +/- 23.73 and 99.89 +/- 22.18 mug/L, pre- and post-treatment, respectively) or in t-PA antigen levels (26.17 +/- 6.18 and 27.32 +/- 5.91 mug/L, pre- and post-treatment, respectively). The delapril group showed no significant changes in PAI-1 levels (97.73 +/- 25.75 and 86.12 +/- 13.12 mug/L, pre- and post-treatment, respectively), but did show a statistically significant difference (p < 0.005) in t-PA antigen levels (25.71 +/- 6.40 and 32.24 +/- 5.31 mug/L, pre- and post-treatment, respectively). The losartan group demonstrated significantly higher post-treatment PAI-1 values than the delapril group (p = 0.048). CONCLUSION The study showed that losartan does not affect fibrinolytic parameters, while delapril resulted in an insignificant reduction in PAI-1 and a significant increase in t-PA levels. Further studies are clearly required in order to establish whether these different effects on the fibrinolytic system between ACE inhibitors and AT(1)-receptor blockers may have clinical relevance.
Collapse
Affiliation(s)
- Salvatore Paterna
- Department of Emergency Medicine, University of Palermo, Palermo, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|