Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Trials ; 21(1): 1005, 2020 Dec 09.
Article in English | MEDLINE | ID: mdl-33298149

ABSTRACT

BACKGROUND: The peak of the global COVID-19 pandemic has not yet been reached, and many countries face the prospect of a second wave of infections before effective vaccinations will be available. After an initial phase of viral replication, some patients develop a second illness phase in which the host thrombotic and inflammatory responses seem to drive complications. Severe COVID-19 disease is linked to high mortality, hyperinflammation, and a remarkably high incidence of thrombotic events. We hypothesize a crucial pathophysiological role for the contact pathway of coagulation and the kallikrein-bradykinin pathway. Therefore, drugs that modulate this excessive thromboinflammatory response should be investigated in severe COVID-19. METHODS: In this adaptive, open-label multicenter randomized clinical trial, we compare low molecular weight heparins at 50 IU anti-Xa/kg twice daily-or 75 IU anti-Xa twice daily for intensive care (ICU) patients-in combination with aprotinin to standard thromboprophylaxis in hospitalized COVID-19 patients. In the case of hyperinflammation, the interleukin-1 receptor antagonist anakinra will be added on top of the drugs in the interventional arm. In a pilot phase, the effect of the intervention on thrombotic markers (D-dimer) will be assessed. In the full trial, the primary outcome is defined as the effect of the interventional drugs on clinical status as defined by the WHO ordinal scale for clinical improvement. DISCUSSION: In this trial, we target the thromboinflammatory response at multiple levels. We intensify the dose of low molecular weight heparins to reduce thrombotic complications. Aprotinin is a potent kallikrein pathway inhibitor that reduces fibrinolysis, activation of the contact pathway of coagulation, and local inflammatory response. Additionally, aprotinin has shown in vitro inhibitory effects on SARS-CoV-2 cellular entry. Because the excessive thromboinflammatory response is one of the most adverse prognostic factors in COVID-19, we will add anakinra, a recombinant interleukin-1 receptor antagonist, to the regimen in case of severely increased inflammatory parameters. This way, we hope to modulate the systemic response to SARS-CoV-2 and avoid disease progressions with a potentially fatal outcome. TRIAL REGISTRATION: The EU Clinical Trials Register 2020-001739-28 . Registered on April 10, 2020.


Subject(s)
COVID-19/complications , Inflammation/etiology , SARS-CoV-2/genetics , Venous Thromboembolism/etiology , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/therapeutic use , Aprotinin/administration & dosage , Aprotinin/therapeutic use , Belgium/epidemiology , Bradykinin/drug effects , Bradykinin/metabolism , COVID-19/epidemiology , COVID-19/virology , Critical Care/statistics & numerical data , Drug Therapy, Combination , Female , Heparin, Low-Molecular-Weight/administration & dosage , Heparin, Low-Molecular-Weight/therapeutic use , Humans , Incidence , Inflammation/epidemiology , Inflammation/metabolism , Inflammation/prevention & control , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Kallikreins/drug effects , Kallikreins/metabolism , Male , Outcome Assessment, Health Care , SARS-CoV-2/drug effects , Severity of Illness Index , Venous Thromboembolism/epidemiology , Venous Thromboembolism/metabolism , Venous Thromboembolism/prevention & control
2.
J Pharm Pract ; 27(5): 461-5, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25124378

ABSTRACT

Angiotensin inhibitors have been extensively evaluated in clinical trials and have demonstrated significant reductions in morbidity and mortality following myocardial infarction and stroke, as well as in patients with heart failure or who are at risk of cardiovascular disease. Further, both angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) are frequently prescribed for the treatment of hypertension and to preserve renal function in patients with diabetes mellitus and chronic kidney disease. Angioedema is a known, but rare, adverse effect of ACEIs and ARBs. Therefore, it is important for clinicians to have a thorough understanding of risks and benefits of prescribing these medications, particularly in patients with a history of angioedema. This review describes the literature evaluating the incidence and cross-reactivity of angioedema with ACEIs and ARBs in order to provide guidance for clinical decision making.


Subject(s)
Angioedema/chemically induced , Angioedema/physiopathology , Angiotensin Receptor Antagonists/adverse effects , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Angioedema/immunology , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Bradykinin/drug effects , Humans , Hypertension/drug therapy , Incidence , Renin-Angiotensin System/physiology
3.
J Clin Neurosci ; 21(11): 2004-6, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24974191

ABSTRACT

We present a patient with asymmetric oro-facial angioedema following thrombolysis for acute ischaemic stroke with serial photographs of this phenomenon. We discuss the mechanism for the development of asymmetric oro-facial oedema following thrombolysis and suggest a management plan.


Subject(s)
Angioedema/chemically induced , Brain Ischemia/complications , Face/pathology , Fibrinolytic Agents/adverse effects , Stroke/drug therapy , Tissue Plasminogen Activator/adverse effects , Aged, 80 and over , Angioedema/therapy , Bradykinin/drug effects , Bradykinin/metabolism , Fibrinolytic Agents/administration & dosage , Humans , Male , Stroke/etiology , Tissue Plasminogen Activator/administration & dosage
5.
Am J Physiol Heart Circ Physiol ; 302(10): H1929-35, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22427519

ABSTRACT

To evaluate the hypothesis that sphingosine-1-phosphate (S1P) and cAMP attenuate increased permeability of individually perfused mesenteric microvessels through a common Rac1-dependent pathway, we measured the attenuation of the peak hydraulic conductivity (L(p)) in response to the inflammatory agent bradykinin (BK) by either S1P or cAMP. We varied the extent of exposure to each agent (test) and measured the ratio L(p)(test)/L(p)(BK alone) for each vessel (anesthetized rats). S1P (1 µM) added at the same time as BK (concurrent, no pretreatment) was as effective to attenuate the response to BK (L(p) ratio: 0.14 ± 0.05; n = 5) as concurrent plus pretreatment with S1P for 30 min (L(p) ratio: 0.26 ± 0.06; n = 11). The same pretreatment with S1P, but with no concurrent S1P, caused no inhibition of the BK response (L(p) ratio 1.07 ± 0.11; n = 8). The rapid on and off action of S1P demonstrated by these results was in contrast to cAMP-dependent changes induced by rolipram and forskolin (RF), which developed more slowly, lasted longer, and resulted in partial inhibition when given either as pretreatment or concurrent with BK. In cultured endothelium, there was no Rac activation or peripheral cortactin localization at 1 min with RF, but cortactin localization and Rac activation were maximal at 1 min with S1P. When S1P was removed, Rac activation returned to control within 2 min. Because of such differing time courses, S1P and cAMP are unlikely to act through fully common effector mechanisms.


Subject(s)
Bradykinin/pharmacology , Capillary Permeability/drug effects , Lysophospholipids/pharmacology , Microvessels/drug effects , Sphingosine/analogs & derivatives , Vasodilator Agents/pharmacology , Animals , Bradykinin/drug effects , Capillary Permeability/physiology , Colforsin/pharmacology , Cyclic AMP/pharmacology , Male , Microvessels/physiology , Models, Animal , Rats , Rolipram/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Sphingosine/pharmacology , Time Factors , rac1 GTP-Binding Protein/metabolism
6.
Ther Adv Cardiovasc Dis ; 5(6): 281-95, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22032921

ABSTRACT

BACKGROUND: The bradykinin potentiating peptides (BPPs) are oligopeptides found in different animal venoms. BPPs isolated from Bothrops jararaca venom were the first natural inhibitors described for somatic angiotensin I-converting enzyme (ACE). They were used in the structural modeling for captopril development, a classical ACE inhibitor widely used to treat human hypertension. METHODS: We evaluated the effect of BPP-5a on cardiovascular parameters of conscious Wistar (WTs) and spontaneously hypertensive rats (SHRs). RESULTS: In SHR, BPP-5a showed potent cardiovascular effects, at doses ranging from 0.47 to 710 nmol/kg. The maximal changes in mean arterial pressure (MAP) and heart rate (HR) were found at the dose of 2.37 nmol/kg (Δ MAP: -38 ± 4 mmHg, p < 0.01; Δ HR: -71 ± 17 bpm, p < 0.05). Reductions in MAP and HR occurred throughout 6 hours of post-injection period. In contrast to active site-directed ACE inhibitors, no ACE inhibition, evaluated by the Ang I pressor effect, or bradykinin potentiation was observed during the antihypertensive effect of the pentapeptide. In vitro assays showed no effects of BPP-5a upon argininosuccinate synthetase and B(1), B(2), AT(1), AT(2) or Mas receptors. Ex vivo assays showed that BPP-5a induced endothelium-dependent vasorelaxation in isolated aortic rings of SHRs and WTs. CONCLUSIONS: Although the BPP-5a is considered an ACE inhibitor, our results indicate that its antihypertensive effect is exerted via a unique target, a nitric-oxide-dependent mechanism.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Bradykinin/metabolism , Hypertension/drug therapy , Oligopeptides/pharmacology , Viper Venoms/pharmacology , Amino Acid Motifs , Animals , Blood Pressure/drug effects , Bradykinin/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Heart Rate/drug effects , Hypertension/physiopathology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Protein Subunits/metabolism , Rats , Rats, Inbred SHR , Vasodilation/drug effects , Venoms/chemistry
7.
Med Princ Pract ; 20(2): 142-6, 2011.
Article in English | MEDLINE | ID: mdl-21252569

ABSTRACT

OBJECTIVE: The present study was conducted to assess the anti-inflammatory effect of a crude aqueous extract of Bixa orellana leaves (AEBO) and to examine the possible involvement of nitric oxide (NO) in its anti-inflammatory mechanism. MATERIALS AND METHODS: The air-dried, powdered leaves were soaked in distilled water (1:20 w/v) at 50°C for 24 h and the supernatant obtained was freeze-dried (yield 8.5% w/w). The dosage was recorded as the mass of extract per kg b.w. of rats in all inflammatory assays (bradykinin-induced paw edema, peritoneal vascular permeability and NO assay). RESULTS: Pretreatment with AEBO for 4 consecutive days exhibited significant inhibitory activity against inflammatory models, the bradykinin-induced hind paw edema model and bradykinin-induced increased peritoneal vascular permeability at both doses in dose-dependent manner. In addition, AEBO was also found to significantly suppress the production of NO at doses of 50 and 150 mg/kg. CONCLUSION: This study provides scientific data to support the traditional use of B. orellana leaves in treating inflammation. Results from this study suggest that AEBO exerts anti-inflammatory effects. Part of this anti-inflammatory effect may be associated with its antibradykinin activity and may be related to a reduction of the NO production.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Bixaceae/chemistry , Bradykinin/antagonists & inhibitors , Inflammation/drug therapy , Nitric Oxide/antagonists & inhibitors , Plant Extracts/therapeutic use , Analysis of Variance , Animals , Biological Assay , Bradykinin/drug effects , Capillary Permeability/drug effects , Disease Models, Animal , Edema/drug therapy , Male , Nitric Oxide/biosynthesis , Rats , Rats, Sprague-Dawley
8.
Cardiovasc Drugs Ther ; 24(4): 331-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20577898

ABSTRACT

Angiotensin-converting enzyme (ACE) inhibitors improve outcomes in patients with coronary artery disease (CAD), heart failure, and hypertension. This short review examines clinical evidence for such effects and the underlying mechanism of action. One potential mode of action for ACE inhibitors in CAD is blood pressure reduction. However, recent data suggest that the effects of ACE inhibitors on the endothelium may also be relevant in attenuating the progression of atherosclerosis. In CAD, chronic overexpression of tissue ACE disrupts the angiotensin II/bradykinin balance with a net result of endothelial dysfunction, mainly due to an increased rate of apoptosis. An imbalance between endothelial apoptosis (death) and its renewal from the bone marrow (life) causes discontinuity of the endothelial layer, favoring the initiation and progression of a biochemical sequence that leads to atherosclerosis, plaque rupture, and eventually acute coronary syndromes. There is clinical and experimental evidence that ACE inhibition improves the life and death cycle of the endothelium. By restoring the bradykinin/angiotensin II balance, ACE inhibition reduces the rate of endothelial apoptosis and experimental results suggest that ACE inhibition can also improve the production and mobilization of endothelial progenitor cells from bone marrow. We report our experience in this context with perindopril.


Subject(s)
Acute Coronary Syndrome/prevention & control , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Coronary Artery Disease/therapy , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Heart Failure/therapy , Hypertension/therapy , Secondary Prevention , Angiotensin II/drug effects , Angiotensin II/metabolism , Apoptosis/drug effects , Atherosclerosis/complications , Atherosclerosis/metabolism , Bradykinin/drug effects , Bradykinin/metabolism , Cardiotonic Agents/therapeutic use , Coronary Artery Disease/etiology , Coronary Artery Disease/physiopathology , Disease Progression , Endothelial Cells/pathology , Endothelial Cells/physiology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiology , Endothelium, Vascular/physiopathology , Heart Failure/etiology , Heart Failure/physiopathology , Humans , Hypertension/etiology , Hypertension/physiopathology , Peptidyl-Dipeptidase A/drug effects , Peptidyl-Dipeptidase A/metabolism , Perindopril/therapeutic use , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/physiopathology , Plaque, Atherosclerotic/prevention & control , Randomized Controlled Trials as Topic , Stem Cells/physiology
9.
Cardiovasc Pathol ; 19(4): e91-8, 2010.
Article in English | MEDLINE | ID: mdl-19733484

ABSTRACT

BACKGROUND: The objective of this study was to investigate whether reduction of cyclic circumferential stretch will impair endothelial function and elevate basal levels of oxidative stress, both known risk factors linked to cardiovascular disease. METHODS: Ex vivo and in vitro models were used to perfuse porcine carotid arteries and porcine endothelial cells, respectively, for 24 h. In both cases, one group was allowed to stretch naturally when exposed to a pulse shear stress (6+/-3 dynes/cm(2)) combined with a pulse pressure of 80+/-10 mmHg, yielding a physiological cyclic stretch of 4-5%. This group was compared to a reduced stretch group, achieved by wrapping the arterial segment with a silicon band or by seeding the endothelial cells inside less compliant tubes, decreasing cyclic stretch to 1%. RESULTS: The experimentally reduced compliance caused a significant decrease in bradykinin-dependent vascular relaxation. Reduced compliance significantly decreased the phosphorylation of serine 1177 (Ser1177) on eNOS, suggesting the activity of eNOS was decreased. Overall production of reactive oxygen species was increased by reducing compliance, as visualized with DHE. Finally, p22-phox and p47-phox, key players in the superoxide-generating NAD(P)H oxidase, were also up-regulated by reduced compliance. CONCLUSIONS: These findings point out how reduced arterial compliance increases the risk of arterial disease by creating a less functional endothelium, interrupting the eNOS activation pathway, and increasing the vascular levels of oxidative stress.


Subject(s)
Carotid Artery, Internal/physiology , Endothelium, Vascular/physiology , Mechanotransduction, Cellular/physiology , Oxidative Stress , Adaptation, Physiological , Animals , Bradykinin/drug effects , Carotid Artery, Internal/drug effects , Cells, Cultured , Compliance/drug effects , Compliance/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Mechanotransduction, Cellular/drug effects , Models, Animal , NADPH Oxidases/biosynthesis , Nitric Oxide Synthase Type III/metabolism , Pulsatile Flow , Reactive Oxygen Species/metabolism , Shear Strength/drug effects , Shear Strength/physiology , Stress, Mechanical , Swine , Tensile Strength/drug effects , Tensile Strength/physiology , Up-Regulation , Vasodilation/drug effects , Vasodilation/physiology
10.
Article in English | MEDLINE | ID: mdl-19076006

ABSTRACT

Hereditary angioedema (HAE) is a rare familial disease characterized by recurrent self-limiting episodes of soft tissue swelling affecting different parts of the body. Acute HAE attacks range from benign, but disfiguring skin edema, to painful abdominal, and even life-threatening laryngeal attacks. The disease is caused by an aberrant C1 esterase inhibitor (C1-INH), which regulates complement, fibrinolytic, and contact pathways. Elevated serum level of bradykinin as a result of contact pathway activation is thought to be the major mediator of pain and edema formation in HAE. Current therapy of acute HAE attacks is limited and mainly offers symptom control. In the United States only fresh frozen plasma provides some reconstitution of C1-INH, but the efficacy and safety of this treatment is controversial. In some European countries two human derived C1-INH concentrates have been used successfully. Prophylactic therapy for patients with frequent HAE attacks is confined to attenuated androgens and in some countries anti-fibrinolytics and C1-INH are also used. To satisfy the unmet needs, investigation of one recombinant C1-INH, two drugs working on bradykinin pathway and two human derived C1-INH concentrates are underway. This review article also discusses some recent patents related to the filed.


Subject(s)
Angioedema/therapy , Bradykinin/drug effects , Complement C1 Inactivator Proteins/therapeutic use , Androgens/therapeutic use , Angioedema/genetics , Angioedema/physiopathology , Animals , Antifibrinolytic Agents/therapeutic use , Bradykinin/blood , Complement C1 Inactivator Proteins/metabolism , Europe/epidemiology , Humans , Patents as Topic , Plasma , Recombinant Proteins , United States/epidemiology
11.
Naunyn Schmiedebergs Arch Pharmacol ; 378(1): 33-41, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18458878

ABSTRACT

The roles of kinin and protease-activated receptors (PAR) in endothelium-dependent relaxations to the serine protease, trypsin, were examined in rings of bovine left anterior descending coronary artery (LAD). Trypsin (0.01-30 U/ml) caused biphasic, endothelium-dependent relaxations-a high potency (0.01-0.3 U/ml), low efficacy relaxation [maximum relaxation (R (max)), 9.0 +/- 5.1%] followed by a lower potency (1-30 U/ml) but high efficacy (R (max), 90.4 +/- 5.5%) relaxation, which was abolished by aprotinin. Captopril (10 microM) caused an approximately 10-fold leftward shift of the second phase response such that the first phase was masked. The second phase relaxation to trypsin was inhibited in a concentration-dependent, non-surmountable manner by the B2 antagonist, HOE-140. At 3 nM HOE-140, the second phase response to trypsin was abolished unmasking the first phase. Kallikrein (0.0003-0.3 U/ml) caused monophasic, endothelium-dependent relaxations (R (max), 33.7 +/- 14.6%), which were potentiated by captopril (R (max), 94.2 +/- 1.0%) and abolished by HOE-140. In the presence of captopril, the second phase relaxation to trypsin was only minimally inhibited by either N(G)-nitro-L: -arginine (100 microM) or 67 mM [K(+)](o) alone but markedly reduced when these two treatments were combined (R (max), 26.1 +/- 11.6% versus 98.6 +/- 2.9% in controls). The PAR1-activating peptide, SFLLRN (0.1-30 microM), but not the PAR2-activating peptide, SLIGRL, caused concentration-dependent relaxations (pEC(50), 5.9 +/- 0.0%; R (max), 43.3 +/- 8.3%). In conclusion, trypsin causes endothelium-dependent relaxations in the bovine LAD predominantly via release of endogenous BK, which in turn activates endothelial B2 receptors. Only a minor role for PAR1-like receptors was evident in this tissue.


Subject(s)
Bradykinin/drug effects , Endothelium, Vascular/drug effects , Receptor, Bradykinin B2/drug effects , Trypsin/pharmacology , Animals , Bradykinin/administration & dosage , Bradykinin/analogs & derivatives , Bradykinin/metabolism , Bradykinin/pharmacology , Captopril/pharmacology , Cattle , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Kallikreins/administration & dosage , Kallikreins/pharmacology , Receptor, Bradykinin B2/metabolism , Receptor, PAR-1/drug effects , Receptor, PAR-1/metabolism , Trypsin/administration & dosage , Vasodilation/drug effects
12.
Neurosci Lett ; 434(3): 310-4, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18329172

ABSTRACT

Regulation of the kallikrein-kinin system in cerebral inflammation is still unclear. Here, we used reverse-transcription polymerase chain reaction (RT-PCR) techniques to show that lipopolysaccharide (LPS) activates the kallikrein-kinin system by enhancing liberation of bradykinin (BK), and alters mRNA levels of kallikrein-kinin system components, including high molecular weight (H-) and low molecular weight (L-) kininogens, in ECPC4 cells, a cell line of mouse choroid plexus epithelium. LPS treatment increased liberation of immunoreactive bradykinin in the supernatant of ECPC4 cells, and addition of LPS (500 ng/ml) to cultures resulted in elevation of H- and L-kininogen mRNA levels in ECPC4 cells within 24-48 h. Furthermore, LPS treatment elevated bradykinin type 2 and type 1 receptor mRNA levels within 4h, but did not change tissue kallikrein or plasma kallikrein mRNA levels. On the other hand, expression of pro-inflammatory mediators interleukin-1beta (IL-1beta), tumor necrosis factor-alpha (TNF-alpha), and cyclooxygenase-2 mRNA increased within 4-8h after addition of LPS to ECPC4 cells. The addition of IL-1beta and TNF-alpha to investigate the major mediator for kininogen expression in ECPC4 cells remarkably induced expression of H- and L-kininogen mRNAs in ECPC4 cells. These results suggest that LPS activates the kallikrein-kinin system in the choroid plexus via autocrine induction of IL-1beta and TNF-alpha.


Subject(s)
Choroid Plexus/metabolism , Cytokines/genetics , Encephalitis/cerebrospinal fluid , Inflammation Mediators/metabolism , Kallikreins/cerebrospinal fluid , Kinins/cerebrospinal fluid , Animals , Bradykinin/cerebrospinal fluid , Bradykinin/drug effects , Bradykinin/genetics , Cell Line , Cerebrospinal Fluid/metabolism , Choroid Plexus/drug effects , Choroid Plexus/physiopathology , Cyclooxygenase 2/genetics , Encephalitis/chemically induced , Encephalitis/physiopathology , Interleukin-1beta/genetics , Interleukin-1beta/pharmacology , Kallikreins/drug effects , Kallikreins/genetics , Kinins/drug effects , Kinins/genetics , Lipopolysaccharides , Mice , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Bradykinin/drug effects , Receptors, Bradykinin/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Up-Regulation/physiology
13.
Br J Pharmacol ; 153(5): 947-55, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18084312

ABSTRACT

BACKGROUND AND PURPOSE: Inhibition of bradykinin metabolizing enzymes (BMEs) can cause acute angioedema, as demonstrated in a recent clinical trial in patients administered the antihypertensive, omapatrilat. However, the relative contribution of specific BMEs to this effect is unclear and confounded by the lack of a predictive pre-clinical model of angioedema. EXPERIMENTAL APPROACH: Rats were instrumented to record blood pressure and heart rate; inhibitors were infused for 35 min and bradykinin was infused during the last 5 min to elicit hypotension, as a functional marker of circulating bradykinin and relative angioedema risk. KEY RESULTS: In the presence of omapatrilat bradykinin produced dose-dependent hypotension, an effect abolished by B(2) blockade. In the presence of lisinopril (ACE inhibitor), but not candoxatril (NEP inhibitor) or apstatin (APP inhibitor), bradykinin also elicited hypotension. Lisinopril-mediated hypotension was unchanged with concomitant blockade of NEP or NEP/DPPIV (candoxatril+A-899301). However, hypotension was enhanced upon concomitant blockade of APP and further intensified in the presence of NEP inhibition to values not different from omapatrilat alone. CONCLUSIONS AND IMPLICATIONS: We demonstrated that bradykinin is degraded in vivo with an enzyme rank-efficacy of ACE>APP>>NEP or DPPIV. These results suggest the effects of omapatrilat are mediated by inhibition of three BMEs, ACE/APP/NEP. However, dual inhibition of ACE/NEP or ACE/NEP/DPPIV elicits no increased risk of angioedema compared to ACE inhibition alone. Thus, novel BME inhibitors must display no activity against APP to avoid angioedema risk due to high prevalence of ACE inhibitor therapy in patients with diabetes and cardiovascular disease.


Subject(s)
Angioedema/etiology , Bradykinin/metabolism , Enzyme Inhibitors/pharmacology , Hypotension/etiology , Aminopeptidases/antagonists & inhibitors , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Bradykinin/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Indans/pharmacology , Lisinopril/pharmacology , Male , Neprilysin/antagonists & inhibitors , Peptides/pharmacology , Propionates/pharmacology , Pyridines/administration & dosage , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Thiazepines/administration & dosage , Thiazepines/pharmacology
14.
Eur J Pharmacol ; 579(1-3): 350-6, 2008 Jan 28.
Article in English | MEDLINE | ID: mdl-18036587

ABSTRACT

Kinins have been suggested to be involved in human airway diseases such as asthma and rhinitis. MEN16132 is a non-peptide kinin B(2) receptor antagonist able to inhibit the responses produced by intravenous bradykinin into the airways, as bronchoconstriction and microvascular leakage; we tested the effect of MEN16132 on endogenously generated bradykinin through the dextran sulfate-induced contact activation of kinin-kallikrein cascade in guinea-pigs. After dextran sulfate administration (1.5 mg/kg i.v.), the pulmonary insufflation pressure was monitored and the microvascular leakage of upper and lower airways was assessed using Evans blue as tracer of plasma protein extravasation. Our results demonstrated that topical MEN16132 strongly inhibited the dextran sulfate-induced bronchoconstriction (0.3 mM solution aerosol for 5 min) and plasma protein extravasation in both lower airways (3-10 microM solution aerosol for 5 min) and nasal mucosa (0.3 nmol/nostril); Icatibant, the peptide antagonist of kinin B(2) receptor, exerted a 3-30-fold less potent inhibitory effect than MEN16132. We conclude that local application of MEN16132 into the airways abolishes the responses produced by the endogenous generation of bradykinin and it can be useful as new pharmacological tool to check the role of kinins in human diseases.


Subject(s)
Bradykinin B2 Receptor Antagonists , Bradykinin/drug effects , Bronchoconstriction/drug effects , Ornithine/analogs & derivatives , Sulfonamides/pharmacology , Administration, Intranasal , Aerosols , Animals , Blood Proteins/metabolism , Bradykinin/administration & dosage , Bradykinin/analogs & derivatives , Bradykinin/metabolism , Bradykinin/pharmacology , Dextran Sulfate , Dose-Response Relationship, Drug , Guinea Pigs , Kallikrein-Kinin System/drug effects , Male , Nasal Mucosa/drug effects , Nasal Mucosa/metabolism , Ornithine/administration & dosage , Ornithine/pharmacology , Sulfonamides/administration & dosage
15.
J Cardiovasc Med (Hagerstown) ; 8(7): 473-82, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17568278

ABSTRACT

The prevalence of type 2 diabetes is increasing worldwide, and prevention of the disease is a key objective. Several clinical trials reported a consistent reduction in the incidence of newly diagnosed diabetes in high-risk patients treated with renin-angiotensin system-inhibiting drugs. In all those trials, however, diabetes reduction was either a post-hoc analysis result or a secondary endpoint. Therefore, we need the results of ongoing specific prospectively designed trials, with new-onset diabetes as the principal endpoint.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Cardiovascular Diseases/prevention & control , Diabetes Mellitus, Type 2/prevention & control , Adipose Tissue/drug effects , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Bradykinin/blood , Bradykinin/drug effects , Cardiovascular Diseases/mortality , Clinical Trials as Topic , Diabetes Mellitus, Type 2/epidemiology , Humans , Insulin Resistance , Pancreas/blood supply , Potassium/blood , Renin-Angiotensin System/drug effects
17.
J Cardiovasc Electrophysiol ; 18(1): 93-9, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17229306

ABSTRACT

OBJECTIVE: Brief reversible ischemic episodes (ischemic preconditioning, IPC) protect the heart against arrhythmias during a subsequent prolonged low-flow ischemia. We have recently shown that this protection involves release of bradykinin, activation of bradykinin B2 receptors followed by opening of sarcolemmal, but not mitochondrial ATP-sensitive K+ channels. The goal of this study was to clarify a trigger and/or mediator role of bradykinin in the antiarrhythmic effects of IPC during low-flow ischemia. METHODS: Isolated perfused rat hearts underwent 60 minutes of low-flow ischemia induced by reducing perfusion pressure followed by 60 minutes of reperfusion. Preconditioning was induced by 2 x 5 minutes episodes of zero-flow ischemia. In yet other groups, preconditioned or non-preconditioned hearts were treated either with bradykinin (10 nmol/L) or with HOE 140 (bradykinin B2 receptor antagonist, 100 nmol/L). RESULTS: IPC reduced the number of ventricular premature beats, as well as the incidence of ventricular tachycardia and of ventricular fibrillation during low-flow ischemia. In addition, this protection was abolished by HOE 140 given during low-flow ischemia. Pharmacological preconditioning using short bradykinin perfusion instead of IPC did not show antiarrhythmic effects. However, bradykinin administered during low-flow ischemia and reperfusion reduced the number of ventricular premature beats and the incidence of ventricular tachycardia and of ventricular fibrillation during low-flow ischemia. CONCLUSION: Bradykinin is a mediator, but unlikely a trigger, of antiarrhythmic effects of IPC during low-flow ischemia.


Subject(s)
Bradykinin/metabolism , Heart Ventricles/metabolism , Ischemic Preconditioning, Myocardial/methods , Tachycardia, Ventricular/etiology , Ventricular Fibrillation/etiology , Animals , Bradykinin/analogs & derivatives , Bradykinin/drug effects , Bradykinin/pharmacology , Bradykinin Receptor Antagonists , Disease Models, Animal , Disease Progression , Electrocardiography , Heart Rate/physiology , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Male , Pilot Projects , Prognosis , Rats , Rats, Sprague-Dawley , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/prevention & control , Ventricular Fibrillation/metabolism , Ventricular Fibrillation/prevention & control
18.
Eur J Heart Fail ; 9(2): 191-6, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16859993

ABSTRACT

BACKGROUND: The beneficial effects of angiotensin converting enzyme (ACE)-inhibitors are in part mediated through the inhibition of the degradation of the vasodilator bradykinin. The bradykinin effect is counteracted by cyclooxygenase-inhibitors. Angiotensin receptor blockers (ARBs) do not affect bradykinin. AIMS: To test the hypothesis that renal counteraction from a cyclooxygenase-inhibitor, diclofenac, is different in subjects treated with an ACE-inhibitor, enalapril compared with an ARB, losartan. METHODS: Twelve elderly, healthy, slightly over-hydrated subjects received diclofenac orally after pre-treatment with a diuretic, bendroflumethiazide, and enalapril or bendroflumethiazide and losartan, in a double-blind cross-over fashion, with a wash-out period of at least 1 week. RESULTS: Diclofenac reduced GFR significantly from 81(64-98) ml/min at first observations after dose for enalapril to 29(16-42) and from 76(64-88) after losartan to 35(24-46). There was no significant difference between enalapril and losartan in GFR. Diclofenac induced decreases in urine flow, excretion rates and clearances of sodium, osmolality clearance and free water clearance, irrespective of treatment with enalapril or losartan. However, serum potassium and handling of potassium were significantly lower after losartan-treatment. CONCLUSION: The negative renal effects of diclofenac administration in subjects with activation of the renin-angiotensin system and enalapril treatment are the same in subjects with activation of the renin-angiotensin system and losartan treatment.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Bradykinin/drug effects , Cyclooxygenase 2 Inhibitors/adverse effects , Diclofenac/adverse effects , Enalapril/pharmacology , Heart Failure/physiopathology , Losartan/pharmacology , Aged , Aged, 80 and over , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Diuretics/pharmacology , Diuretics/therapeutic use , Enalapril/therapeutic use , Female , Glomerular Filtration Rate/drug effects , Humans , Kidney/physiopathology , Losartan/therapeutic use , Male , Middle Aged , Renin-Angiotensin System/drug effects , Time Factors
19.
Immunol Allergy Clin North Am ; 26(4): 691-708, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17085285

ABSTRACT

Advances in our understanding of the molecular mechanisms underlying hereditary angioedema (HAE) have led to the development of new treatment modalities. Five new drugs for the treatment of HAE are currently undergoing clinical testing in the United States. These novel therapeutics can be divided into two groups: drugs that replace C1 inhibitor (C1INH) functional activity and drugs that abrogate the bradykinin-mediated increase in vascular permeability associated with HAE attacks. The first group includes two plasma-derived C1INH concentrates as well as a recombinant transgenic human C1INH protein, and the second group includes an engineered plasma kallikrein inhibitor as well as a B2 bradykinin receptor antagonist. This article reviews the rationale, development, and potential use of these novel therapeutics.


Subject(s)
Angioedema/drug therapy , Animals , Bradykinin/antagonists & inhibitors , Bradykinin/drug effects , Clinical Trials as Topic , Complement C1 Inhibitor Protein/metabolism , Complement C1 Inhibitor Protein/therapeutic use , Humans , Kallikreins/antagonists & inhibitors , Kallikreins/drug effects , Recombinant Proteins/therapeutic use
20.
J Neurotrauma ; 22(12): 1444-55, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16379582

ABSTRACT

Traumatic brain injury (TBI) mortality and morbidity remains a public health challenge. Because experimental studies support an important role of bradykinin (BK) in the neurological deterioration that follows TBI, a double-blind, randomized, placebo-controlled study of Anatibant (LF16- 0687Ms), a selective and potent antagonist of the BK B(2) receptor, was conducted in severe (Glasgow Coma Scale [GCS] < 8) TBI patients (n = 25) at six sites in the United States. At 8-12 h after injury (9.9 +/- 2.8 h), patients received a single subcutaneous injection of Anatibant (3.75 mg or 22.5 mg, n = 10 each) or placebo (n = 5). The primary objective was to investigate the pharmacokinetics of Anatibant; general safety, local tolerability, levels of the bradykinin metabolite BK1-5 in plasma and cerebrospinal fluid (CSF), intracranial pressure (ICP), and cerebral perfusion pressure were also assessed. We observed a dose-proportionality of the pharmacokinetics, Cmax, and AUC of Anatibant. V(d)/F, Cl/F, and t(1/2) were independent on the dose and protein binding was >97.7%. Anatibant, administered as single subcutaneous injections of 3.75 g and 22.5 mg, was well tolerated in severe TBI patients with no unexpected clinical adverse events or biological abnormalities observed. Interestingly, plasma and CSF levels of BK1-5 were significantly and markedly increased after trauma (e.g., 34,700 +/- 35,300 fmol/mL in plasma vs. 34.9 +/- 5.6 fmol/mL previously reported for normal volunteers), supporting the use of Anatibant as a treatment of secondary brain damage. To address this issue, a dose-response trial that would investigate the effects of Anatibant on the incidence of raised ICP and on functional outcome in severe TBI patients is needed.


Subject(s)
Bradykinin B2 Receptor Antagonists , Brain Injuries/drug therapy , Quinolines/adverse effects , Quinolines/pharmacokinetics , Adolescent , Adult , Aged , Area Under Curve , Bradykinin/blood , Bradykinin/cerebrospinal fluid , Bradykinin/drug effects , Double-Blind Method , Female , Glasgow Outcome Scale , Half-Life , Humans , Male , Middle Aged , Peptide Fragments/blood , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/drug effects , Pilot Projects
SELECTION OF CITATIONS
SEARCH DETAIL
...