Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 183
Filter
1.
Molecules ; 26(14)2021 Jul 07.
Article in English | MEDLINE | ID: mdl-34299400

ABSTRACT

The goal of this study was to assess the pharmacological effects of black tea (Camellia sinensis var. assamica) water extract on human kinin-forming enzymes in vitro. Tea is a highly consumed beverage in the world. Factor XII (FXII, Hageman factor)-independent- and -dependent activation of prekallikrein to kallikrein leads to the liberation of bradykinin (BK) from high-molecular-weight kininogen (HK). The excessive BK production causes vascular endothelial and nonvascular smooth muscle cell permeability, leading to angioedema. The prevalence of angiotensin-converting enzyme inhibitor (ACEI)-induced angioedema appears to be through BK. Both histamine and BK are potent inflammatory mediators. However, the treatments for histamine-mediated angioedema are unsuitable for BK-mediated angioedema. We hypothesized that long-term consumption of tea would reduce bradykinin-dependent processes within the systemic and pulmonary vasculature, independent of the anti-inflammatory actions of polyphenols. A purified fraction of the black tea water extract inhibited both kallikrein and activated FXII. The black tea water extracts inhibited factor XII-induced cell migration and inhibited the production of kallikrein on the endothelial cell line. We compared the inhibitory effects of the black tea water extract and twenty-three well-known anti-inflammatory medicinal herbs, in inhibiting both kallikrein and FXII. Surprisingly, arjunglucoside II specifically inhibited the activated factor XII (FXIIa), but not the kallikrein and the activated factor XI. Taken together, the black tea water extract exerts its anti-inflammatory effects, in part, by inhibiting kallikrein and activated FXII, which are part of the plasma kallikrein-kinin system (KKS), and by decreasing BK production. The inhibition of kallikrein and activated FXII represents a unique polyphenol-independent anti-inflammatory mechanism of action for the black tea.


Subject(s)
Bradykinin/metabolism , Camellia/chemistry , Endothelium, Vascular/drug effects , Factor XII/antagonists & inhibitors , Kallikrein-Kinin System/drug effects , Plant Extracts/pharmacology , Pulmonary Artery/drug effects , Cell Proliferation , Cells, Cultured , Endothelium, Vascular/metabolism , Humans , Pulmonary Artery/metabolism
2.
Viruses ; 13(2)2021 02 16.
Article in English | MEDLINE | ID: mdl-33669276

ABSTRACT

Background: Coronavirus disease 19 (COVID-19) can develop into a severe respiratory syndrome that results in up to 40% mortality. Acute lung inflammatory edema is a major pathological finding in autopsies explaining O2 diffusion failure and hypoxemia. Only dexamethasone has been shown to reduce mortality in severe cases, further supporting a role for inflammation in disease severity. SARS-CoV-2 enters cells employing angiotensin-converting enzyme 2 (ACE2) as a receptor, which is highly expressed in lung alveolar cells. ACE2 is one of the components of the cellular machinery that inactivates the potent inflammatory agent bradykinin, and SARS-CoV-2 infection could interfere with the catalytic activity of ACE2, leading to the accumulation of bradykinin. Methods: In this case control study, we tested two pharmacological inhibitors of the kinin-kallikrein system that are currently approved for the treatment of hereditary angioedema, icatibant, and inhibitor of C1 esterase/kallikrein, in a group of 30 patients with severe COVID-19. Results: Neither icatibant nor inhibitor of C1 esterase/kallikrein resulted in changes in time to clinical improvement. However, both compounds were safe and promoted the significant improvement of lung computed tomography scores and increased blood eosinophils, which are indicators of disease recovery. Conclusions: In this small cohort, we found evidence for safety and a beneficial role of pharmacological inhibition of the kinin-kallikrein system in two markers that indicate improved disease recovery.


Subject(s)
Bradykinin/analogs & derivatives , COVID-19 Drug Treatment , Complement C1 Inhibitor Protein/therapeutic use , Kallikrein-Kinin System/drug effects , Kallikreins/antagonists & inhibitors , Adult , Aged , Bradykinin/therapeutic use , Case-Control Studies , Drug Repositioning , Female , Humans , Lung/drug effects , Lung/pathology , Male , Middle Aged
3.
Front Immunol ; 11: 2072, 2020.
Article in English | MEDLINE | ID: mdl-32922409

ABSTRACT

A dysregulated immune response with hyperinflammation is observed in patients with severe coronavirus disease 2019 (COVID-19). The aim of the present study was to assess the safety and potential benefits of human recombinant C1 esterase inhibitor (conestat alfa), a complement, contact activation and kallikrein-kinin system regulator, in severe COVID-19. Patients with evidence of progressive disease after 24 h including an oxygen saturation <93% at rest in ambient air were included at the University Hospital Basel, Switzerland in April 2020. Conestat alfa was administered by intravenous injections of 8400 IU followed by 3 additional doses of 4200 IU in 12-h intervals. Five patients (age range, 53-85 years; one woman) with severe COVID-19 pneumonia (11-39% lung involvement on computed tomography scan of the chest) were treated a median of 1 day (range 1-7 days) after admission. Treatment was well-tolerated. Immediate defervescence occurred, and inflammatory markers and oxygen supplementation decreased or stabilized in 4 patients (e.g., median C-reactive protein 203 (range 31-235) mg/L before vs. 32 (12-72) mg/L on day 5). Only one patient required mechanical ventilation. All patients recovered. C1INH concentrations were elevated before conestat alfa treatment. Levels of complement activation products declined after treatment. Viral loads in nasopharyngeal swabs declined in 4 patients. In this uncontrolled case series, targeting multiple inflammatory cascades by conestat alfa was safe and associated with clinical improvements in the majority of severe COVID-19 patients. Controlled clinical trials are needed to assess its safety and efficacy in preventing disease progression.


Subject(s)
Betacoronavirus/drug effects , Complement C1 Inhibitor Protein/therapeutic use , Complement C1/antagonists & inhibitors , Coronavirus Infections/drug therapy , Cytokine Release Syndrome/drug therapy , Kallikrein-Kinin System/drug effects , Pneumonia, Viral/drug therapy , Aged , Aged, 80 and over , COVID-19 , Complement C1 Inhibitor Protein/analysis , Factor XIa/antagonists & inhibitors , Female , Humans , Kallikreins/antagonists & inhibitors , Male , Middle Aged , Pandemics , Recombinant Proteins/therapeutic use , SARS-CoV-2 , Viral Load/drug effects
4.
Sci Rep ; 10(1): 14160, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32843685

ABSTRACT

Immediate hypersensitivity reaction (IHR) can be divided into allergic- and non-allergic-mediated, while "anaphylaxis" is reserved for severe IHR. Clinically, true penicillin allergy is rare and most reported penicillin allergy is "spurious". Penicillin-initiated anaphylaxis is possible to occur in skin test- and specific IgE-negative patients. The contact system is a plasma protease cascade initiated by activation of factor XII (FXII). Many agents with negative ion surface can activate FXII to drive contact system. Our data showed that penicillin significantly induced hypothermia in propranolol- or pertussis toxin-pretreated mice. It also caused a rapid and reversible drop in rat blood pressure, which did not overlap with IgE-mediated hypotension. These effects could be countered by a bradykinin-B2 receptor antagonist icatibant, and consistently, penicillin indeed increased rat plasma bradykinin. Moreover, penicillin not only directly activated contact system FXII-dependently, but also promoted bradykinin release in plasma incubated-human umbilical vein endothelial cells. In fact, besides penicillin, other beta-lactams also activated the contact system in vitro. Since the autoactivation of FXII can be affected by multiple-factors, plasma from different healthy individuals showed vastly different amidolytic activity in response to penicillin, suggesting the necessity of determining the potency of penicillin to induce individual plasma FXII activation. These results clarify that penicillin-initiated non-allergic anaphylaxis is attributed to contact system activation, which might bring more effective diagnosis options for predicting penicillin-induced fatal risk and avoiding costly and inappropriate treatment clinically.


Subject(s)
Anaphylaxis/chemically induced , Blood Coagulation/drug effects , Factor XIIa/metabolism , Kallikrein-Kinin System/drug effects , Penicillin G/toxicity , Anaphylaxis/immunology , Animals , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Bradykinin/physiology , Bradykinin Receptor Antagonists/pharmacology , Capillary Permeability/drug effects , Enzyme Activation/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Hypothermia/chemically induced , Male , Mice , Mice, Inbred BALB C , Penicillin G/adverse effects , Pertussis Toxin/toxicity , Propranolol/toxicity , Rats , Rats, Sprague-Dawley , Receptor, Bradykinin B2/drug effects , Receptor, Bradykinin B2/physiology , beta-Lactams/toxicity
5.
Nat Immunol ; 21(10): 1146-1151, 2020 10.
Article in English | MEDLINE | ID: mdl-32855555
6.
EBioMedicine ; 58: 102907, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32771682

ABSTRACT

BACKGROUND: SARS-CoV-2 enters cells by binding of its spike protein to angiotensin-converting enzyme 2 (ACE2). Angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin II receptor blockers (ARBs) have been reported to increase ACE2 expression in animal models, and worse outcomes are reported in patients with co-morbidities commonly treated with these agents, leading to controversy during the COVID-19 pandemic over whether these drugs might be helpful or harmful. METHODS: Animal, in vitro and clinical data relevant to the biology of the renin-angiotensin system (RAS), its interaction with the kallikrein-kinin system (KKS) and SARS-CoV-2, and clinical studies were reviewed. FINDINGS AND INTERPRETATION: SARS-CoV-2 hijacks ACE2to invade and damage cells, downregulating ACE2, reducing its protective effects and exacerbating injurious Ang II effects. However, retrospective observational studies do not show higher risk of infection with ACEI or ARB use. Nevertheless, study of the RAS and KKS in the setting of coronaviral infection may yield therapeutic targets.


Subject(s)
Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Coronavirus Infections/drug therapy , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/drug therapy , Angiotensin Receptor Antagonists/pharmacology , Angiotensin-Converting Enzyme 2 , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Betacoronavirus/pathogenicity , COVID-19 , Coronavirus Infections/metabolism , Coronavirus Infections/pathology , Coronavirus Infections/virology , Humans , Kallikrein-Kinin System/drug effects , Pandemics , Peptidyl-Dipeptidase A/genetics , Pneumonia, Viral/metabolism , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Renin-Angiotensin System/drug effects , SARS-CoV-2
7.
Pharmacol Res ; 160: 105096, 2020 10.
Article in English | MEDLINE | ID: mdl-32712319

ABSTRACT

The risk of thrombosis, a globally growing challenge and a major cause of death, is influenced by various factors in the intravascular coagulation, vessel wall, and cellular systems. Among the contributors to thrombosis, the contact activation system and the kallikrein/kinin system, two overlapping plasma proteolytic systems that are often considered as synonymous, regulate thrombosis from different aspects. On one hand, components of the contact activation system such as factor XII initiates activation of the coagulation proteins promoting thrombus formation on artificial surfaces through factor XI- and possibly prekallikrein-mediated intrinsic coagulation. On the other hand, physiological activation of plasma prekallikrein in the kallikrein/kinin system on endothelial cells liberates bradykinin from associated high-molecular-weight kininogen to stimulate the constitutive bradykinin B2 receptor to generate nitric oxide and prostacyclin to induce vasodilation and counterbalance angiotensin II signaling from the renin-angiotensin system which stimulates vasoconstriction. In addition to vascular tone regulation, this interaction between the kallikrein/kinin and renin-angiotensin systems has a thrombo-regulatory role independent of the contact pathway. At the level of the G-protein coupled receptors of these systems, defective bradykinin signaling due to attenuated bradykinin formation and/or decreased B2 receptor expression, as seen in murine prekallikrein and B2 receptor null mice, respectively, leads to compensatory overexpressed Mas, the receptor for angiotensin-(1-7) of the renin-angiotensin system. Mas stimulation and/or its increased expression contributes to maintaining a healthy vascular homeostasis by generating graded elevation of plasma prostacyclin which reduces thrombosis through two independent pathways: (1) increasing the vasoprotective transcription factor Sirtuin 1 to suppress tissue factor expression, and (2) inhibiting platelet activation. This review will summarize the recent advances in this field that support these understandings. Appreciating these subtle mechanisms help to develop novel anti-thrombotic strategies by targeting the vascular receptors in the renin-angiotensin and the kallikrein/kinin systems to maintain healthy vascular homeostasis.


Subject(s)
Blood Coagulation , Kallikrein-Kinin System/drug effects , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Renin-Angiotensin System , Thrombosis/blood , Animals , Epoprostenol/metabolism , Humans , Prekallikrein/metabolism , Proto-Oncogene Mas , Receptor, Bradykinin B2/metabolism , Signal Transduction , Sirtuin 1/metabolism
8.
Med Hypotheses ; 143: 109886, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32504925

ABSTRACT

Coronavirus disease 2019 (COVID-19) is an infectious disease with fast spreading all over the world caused by the SARS-CoV-2 virus which can culminate in a severe acute respiratory syndrome by the injury caused in the lungs. However, other organs can be also damaged. SARS-CoV-2 enter into the host cells using the angiotensin-converting enzyme 2 (ACE2) as receptor, like its ancestor SARS-CoV. ACE2 is then downregulated in lung tissues with augmented serum levels of ACE2 in SARS-CoV-2 patients. Interestingly, ACE2+ organs reveal the symptomatic repercussions, which are signals of the infection such as dry cough, shortness of breath, heart failure, liver and kidney damage, anosmia or hyposmia, and diarrhea. ACE2 exerts a chief role in the renin-angiotensin system (RAS) by converting angiotensin II to angiotensin-(1-7) that activates Mas receptor, inhibits ACE1, and modulates bradykinin (BK) receptor sensitivity, especially the BK type 2 receptor (BKB2R). ACE2 also hydrolizes des-Arg9-bradykinin (DABK), an active BK metabolite, agonist at BK type 1 receptors (BKB1R), which is upregulated by inflammation. In this opinion article, we conjecture a dialogue by the figure of Sérgio Ferreira which brought together basic science of classical pharmacology and clinical repercussions in COVID-19, then we propose that in the course of SARS-CoV-2 infection: i) downregulation of ACE2 impairs the angiotensin II and DABK inactivation; ii) BK and its metabolite DABK seems to be in elevated levels in tissues by interferences in kallikrein/kinin system; iii) BK1 receptor contributes to the outbreak and maintenance of the inflammatory response; iv) kallikrein/kinin system crosstalks to RAS and coagulation system, linking inflammation to thrombosis and organ injury. We hypothesize that targeting the kallikrein/kinin system and BKB1R pathway may be beneficial in SARS-CoV-2 infection, especially on early stages. This route of inference should be experimentally verified by SARS-CoV-2 infected mice.


Subject(s)
Betacoronavirus , Coronavirus Infections/drug therapy , Coronavirus Infections/physiopathology , Kallikrein-Kinin System/physiology , Models, Biological , Pneumonia, Viral/drug therapy , Pneumonia, Viral/physiopathology , Angiotensin-Converting Enzyme 2 , Animals , COVID-19 , Coronavirus Infections/etiology , Humans , Kallikrein-Kinin System/drug effects , Mice , Pandemics , Peptidyl-Dipeptidase A/physiology , Pneumonia, Viral/etiology , Receptors, Virus/physiology , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , SARS-CoV-2 , Translational Research, Biomedical , Virus Internalization/drug effects , COVID-19 Drug Treatment
9.
Br J Pharmacol ; 177(21): 4866-4872, 2020 11.
Article in English | MEDLINE | ID: mdl-32497257

ABSTRACT

The newly identified coronavirus SARS-CoV-2 that spread from China is causing the pandemic COVID-19 with a fatality rate from 5-15%. It causes fever, cough, myalgia, fatigue up to dyspnoea, responsible for hospitalization and artificial oxygenation. SARS-CoV-2 infects human cells using ACE2, the transmembrane protease serine 2 (TMPRSS2) and the SARS-CoV-2 main protease (Mpro ). Once bound to ACE2 and the other two proteases in concert they allow the virus replication and spread throughout the body. Our attention has been focused on the role of ACE2 as its binding to by the virus increases bradykinin and its metabolites, which facilitate inflammation in the lung (causing cough and fever), coagulation and the complement system. These three systems are involved in angioedema, cardiovascular dysfunction and sepsis, pathologies which occur in COVID-19 patients. Thus, we propose that blocking the kallikrein-kinin system with lanadelumab, approved for hereditary angioedema, will prevent facilitation of these 3 systems. LINKED ARTICLES: This article is part of a themed issue on The Pharmacology of COVID-19. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.21/issuetoc.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Antibodies, Monoclonal, Humanized/pharmacology , Betacoronavirus/isolation & purification , COVID-19 , Coronavirus Infections/virology , Humans , Kallikrein-Kinin System/drug effects , Kininogen, High-Molecular-Weight/metabolism , Pandemics , Pneumonia, Viral/virology , SARS-CoV-2 , COVID-19 Drug Treatment
10.
Acta Pharmacol Sin ; 41(12): 1597-1608, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32300244

ABSTRACT

Tissue kallikrein has protective function against various types of injury. In this study, we investigated whether exogenous pancreatic kininogenase (PK) conferred renoprotection in a rat model of unilateral ureteral obstruction (UUO) and H2O2-treated HK-2 cells in vitro. SD rats were subjected to UUO surgery, then PK (7.2 U/g per day, ip) was administered for 7 or 14 days. After the treatment, rats were euthanized; the obstructed kidneys were harvested for further examination. We found that PK administration significantly attenuated interstitial inflammation and fibrosis, and downregulated the expression of proinflammatory (MCP-1, TLR-2, and OPN) and profibrotic (TGF-ß1 and CTGF) cytokines in obstructed kidney. UUO-induced oxidative stress, closely associated with excessive apoptotic cell death and autophagy via PI3K/AKT/FoxO1a signaling, which were abolished by PK administration. We further showed that PK administration increased the expression of bradykinin receptors 1 and 2 (B1R and B2R) mRNA and the production of NO and cAMP in kidney tissues. Coadministration with either B1R antagonist (des-Arg9-[Leu8]-bradykinin) or B2R antagonist (icatibant) abrogated the renoprotective effects of PK, and reduced the levels of NO and cAMP in obstructed kidney. In H2O2-treated HK-2 cells, addition of PK (6 pg/mL) significantly decreased ROS production, regulated the expression of oxidant and antioxidant enzymes, suppressed the expression of TGF-ß1 and MCP-1, and inhibited cell apoptosis. Our data demonstrate that PK treatment protects against the progression of renal fibrosis in obstructed kidneys.


Subject(s)
Fibrosis/prevention & control , Kallikreins/therapeutic use , Kidney/metabolism , Pancreas/enzymology , Protective Agents/therapeutic use , Ureteral Obstruction/complications , Animals , Cell Death/drug effects , Cell Line , Fibrosis/etiology , Fibrosis/pathology , Humans , Inflammation/drug therapy , Inflammation/etiology , Inflammation/pathology , Kallikrein-Kinin System/drug effects , Kidney/pathology , Male , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Signal Transduction/drug effects , Ureteral Obstruction/pathology
11.
Sci Rep ; 9(1): 19437, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31857655

ABSTRACT

Pharmacological research in mice and human genetic analyses suggest that the kallikrein-kinin system (KKS) may regulate anxiety. We examined the role of the KKS in anxiety and stress in both species. In human genetic association analysis, variants in genes for the bradykinin precursor (KNG1) and the bradykinin receptors (BDKRB1 and BDKRB2) were associated with anxiety disorders (p < 0.05). In mice, however, neither acute nor chronic stress affected B1 receptor gene or protein expression, and B1 receptor antagonists had no effect on anxiety tests measuring approach-avoidance conflict. We thus focused on the B2 receptor and found that mice injected with the B2 antagonist WIN 64338 had lowered levels of a physiological anxiety measure, the stress-induced hyperthermia (SIH), vs controls. In the brown adipose tissue, a major thermoregulator, WIN 64338 increased expression of the mitochondrial regulator Pgc1a and the bradykinin precursor gene Kng2 was upregulated after cold stress. Our data suggests that the bradykinin system modulates a variety of stress responses through B2 receptor-mediated effects, but systemic antagonists of the B2 receptor were not anxiolytic in mice. Genetic variants in the bradykinin receptor genes may predispose to anxiety disorders in humans by affecting their function.


Subject(s)
Anxiety Disorders/metabolism , Bradykinin/metabolism , Kallikrein-Kinin System/physiology , Stress, Psychological/metabolism , Adult , Animals , Anxiety Disorders/drug therapy , Anxiety Disorders/genetics , Anxiety Disorders/pathology , Bradykinin B1 Receptor Antagonists/administration & dosage , Bradykinin B2 Receptor Antagonists/administration & dosage , Brain/pathology , Disease Models, Animal , Female , Humans , Kallikrein-Kinin System/drug effects , Kininogens/genetics , Kininogens/metabolism , Male , Mice , Naphthalenes/administration & dosage , Organophosphorus Compounds/administration & dosage , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Polymorphism, Single Nucleotide , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B1/metabolism , Receptor, Bradykinin B2/genetics , Receptor, Bradykinin B2/metabolism , Species Specificity , Stress, Psychological/drug therapy , Stress, Psychological/pathology , Up-Regulation
12.
EBioMedicine ; 47: 319-328, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31444145

ABSTRACT

BACKGROUND: The complement and kallikrein-kinin systems (KKS) are activated during vascular inflammation. The aim of this study was to investigate if blockade of the KKS can affect complement activation on the endothelium during inflammation. METHODS: Complement deposition on endothelial microvesicles was assayed in vasculitis patient plasma samples and controls. Plasma was perfused over glomerular endothelial cells and complement deposition assayed by flow cytometry. The effect of the kinin system was assessed using kinin receptor antagonists and C1-inhibitor. The in vivo effect was assessed in kidney sections from mice with nephrotoxic serum-induced glomerulonephritis treated with a kinin receptor antagonist. FINDINGS: Vasculitis patient plasma had significantly more C3- and C9-positive endothelial microvesicles than controls. Perfusion of patient acute-phase plasma samples over glomerular endothelial cells induced the release of significantly more complement-positive microvesicles, in comparison to remission or control plasma. Complement activation on endothelial microvesicles was reduced by kinin B1- and B2-receptor antagonists or by C1-inhibitor (the main inhibitor of the classical pathway and the KKS). Likewise, perfusion of glomerular endothelial cells with C1-inhibitor-depleted plasma induced the release of complement-positive microvesicles, which was significantly reduced by kinin-receptor antagonists or C1-inhibitor. Mice with nephrotoxic serum-induced glomerulonephritis exhibited significantly reduced glomerular C3 deposition when treated with a B1-receptor antagonist. INTERPRETATION: Excessive complement deposition on the endothelium will promote endothelial injury and the release of endothelial microvesicles. This study demonstrates that blockade of the KKS can reduce complement activation and thereby the inflammatory response on the endothelium. FUNDING: Full details are provided in the Acknowledgements/Funding section.


Subject(s)
Complement Activation/immunology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Kallikrein-Kinin System/drug effects , Vasculitis/etiology , Vasculitis/metabolism , Adult , Aged , Animals , Biological Transport , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Cell-Derived Microparticles/metabolism , Cells, Cultured , Complement C1 Inhibitor Protein/metabolism , Complement System Proteins/immunology , Complement System Proteins/metabolism , Disease Models, Animal , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , Immunoglobulin G/immunology , Male , Mice , Middle Aged , Protein Binding , Vasculitis/pathology
13.
J Cardiovasc Pharmacol ; 72(6): 291-295, 2018 12.
Article in English | MEDLINE | ID: mdl-30422889

ABSTRACT

Antihypertensive pharmacological treatments focus on the use of angiotensin-converting enzyme (ACE) inhibitors, AT1 receptor antagonists, and beta-blockers as single and combined treatments. The effect of single treatments on the mRNA expression of some components of the renin-angiotensin system has been studied, but not the effect of combined treatments. This study determined the expression of the AT1, AT2, B1, and B2 receptors and of the enzymes ACE and ACE2 in hypertensive rats treated with captopril-propranolol or losartan-propranolol. Methods: The mRNA expression of the receptors and enzymes was determined by reverse transcription-quantitative polymerase chain reaction in the aorta of spontaneously hypertensive rats under different treatments. Results: Rats under combined treatments showed a decrease in the expression of AT1 and ACE, and an increase in the expression of the B1 receptor (captopril + propranolol group: 0.43 ± 0.046, 2.243 ± 0.269, 3.356 ± 0.418; Group: losartan + propranolol: 0.727 ± 0.071, 0.852 ± 0.102, 1.277 ± 0.131 compared to the spontaneously hypertensive group: 1 ± 0.212, 1 ± 0.192, 1 ± 0.214). This decrease in the expression of ACE and AT1 suggests a reduction in the expression of Ang II that could be related to a lower response to this vasoconstrictor. An increase in the expression of B1 would improve vasodilation, which would be a beneficial effect of combined therapies for hypertension.


Subject(s)
Antihypertensive Agents/pharmacology , Aorta, Thoracic/drug effects , Hypertension/drug therapy , Kallikrein-Kinin System/drug effects , Renin-Angiotensin System/drug effects , Adrenergic beta-Antagonists/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiopathology , Blood Pressure/drug effects , Captopril/pharmacology , Disease Models, Animal , Gene Expression Regulation , Hypertension/genetics , Hypertension/metabolism , Hypertension/physiopathology , Kallikrein-Kinin System/genetics , Losartan/pharmacology , Male , Propranolol/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/genetics , Receptor, Angiotensin, Type 2/metabolism , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B1/metabolism , Receptor, Bradykinin B2/genetics , Receptor, Bradykinin B2/metabolism , Renin-Angiotensin System/genetics
14.
Nephron ; 139(4): 299-304, 2018.
Article in English | MEDLINE | ID: mdl-29772567

ABSTRACT

BACKGROUND/AIMS: Temporary proteinuria post-exercise is common and is caused predominantly by renal haemodynamic alterations. One reason is up-regulation of angiotensin II (Ang II) due to the reducing effect of angiotensin-converting enzyme (ACE) inhibitors. However, another, ignored, reason could be the kininase effect of ACE inhibition. This study investigated how ACE inhibition reduces post-exercise proteinuria: by either Ang II up-regulation inhibition or bradykinin elevation due to kininase activity inhibition. METHODS: Our study included 10 volunteers, who completed 3 high-intensity exercise protocols involving cycling at 1-week intervals. The first protocol was a control arm, the second evaluated the effect of ACE inhibition and the third examined the effect of angiotensin type 1 receptor blockade. Upon application, both agents reduced systolic and diastolic blood pressure; however, there were no statistically significant -differences. In addition, total protein, microalbumin and -ß2-microglobulin excretion levels in urine specimens were analysed before, 30 min after and 120 min after the exercise protocols. RESULTS: Total protein levels in urine samples were elevated in all 3 protocols after 30 min of high-intensity exercise, compared to baseline levels. However, both ACE inhibition and angiotensin type 1 receptor blockade suppressed total protein in the 30th min. In each protocol, total protein levels returned to the baseline after 120 min. Urinary microalbumin and ß2-microglobulin levels during the control protocol were significantly higher 30 min post-exercise; however, only angiotensin type 1 receptor blockade suppressed microalbumin levels. CONCLUSION: The results indicated Ang II up-regulation, not bradykinin elevation, plays a role in post-exercise proteinuria.


Subject(s)
Exercise , Kallikrein-Kinin System/drug effects , Proteinuria/prevention & control , Proteinuria/urine , Renin-Angiotensin System/drug effects , Albuminuria/prevention & control , Albuminuria/urine , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Bicycling , Bradykinin/urine , Healthy Volunteers , Humans , Male , Young Adult , beta 2-Microglobulin/urine
15.
JCI Insight ; 3(9)2018 05 03.
Article in English | MEDLINE | ID: mdl-29720566

ABSTRACT

Kidney injury is a frequent outcome in patients with disseminated Candida albicans fungal infections. IL-17 receptor (IL-17R) signaling is critical for renal protection against disseminated candidiasis, but the identity and function of IL-17-responsive cells in mediating renal defense remains an active area of debate. Using BM chimeras, we found that IL-17R signaling is required only in nonhematopoietic cells for immunity to systemic C. albicans infection. Since renal tubular epithelial cells (RTEC) are highly responsive to IL-17 in vitro, we hypothesized that RTEC might be the dominant target of IL-17 activity in the infected kidney. We generated mice with a conditional deletion of IL-17 receptor A (Il17ra) in RTEC (Il17raΔRTEC). Strikingly, Il17raΔRTEC mice showed enhanced kidney damage and early mortality following systemic infection, very similar to Il17ra-/- animals. Increased susceptibility to candidiasis in Il17raΔRTEC mice was associated with diminished activation of the renal protective Kallikrein-kinin system (KKS), resulting in reduced apoptosis of kidney-resident cells during hyphal invasion. Moreover, protection was restored by treatment with bradykinin, the major end-product of KKS activation, which was mediated dominantly via bradykinin receptor b1. These data show that IL-17R signaling in RTEC is necessary and likely sufficient for IL-17-mediated renal defense against fatal systemic C. albicans infection.


Subject(s)
Acute Kidney Injury/immunology , Candidemia/immunology , Glomerular Basement Membrane/metabolism , Receptors, Interleukin-17/immunology , Receptors, Interleukin-17/metabolism , Signal Transduction/immunology , Acute Kidney Injury/microbiology , Adoptive Transfer , Animals , Bradykinin/pharmacology , Candida albicans , Epithelial Cells/metabolism , Female , Genetic Predisposition to Disease , Glomerular Basement Membrane/cytology , Kallikrein-Kinin System/drug effects , Kallikrein-Kinin System/physiology , Kidney Tubules/metabolism , Male , Mice , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Interleukin-17/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
16.
Asia Pac J Ophthalmol (Phila) ; 7(1): 28-35, 2018.
Article in English | MEDLINE | ID: mdl-29473719

ABSTRACT

Diabetic retinopathy and diabetic macular edema comprise a major source of visual disability throughout the developed world. The etiology and pathogenesis of macular edema is intricate and multifactorial, in which the hyperglycemic state in diabetes induces a microangiopathy. Through several inflammatory and vasogenic mediators, including vascular endothelial growth factor (VEGF) upregulation and inflammatory cytokines and chemokines, pathologic changes are induced in the vascular endothelium triggering breakdown of the blood retinal barrier, causing extravasation of fluid into the extracellular space and manifesting clinically as macular edema, resulting in visual loss. The advent of medications targeting the VEGF pathway has led to great clinical improvements compared with the previous standard of care of laser therapy alone, as shown in studies such as RISE, RIDE, VIVID, VISTA, and DRCR. However, analyses have shown that many patients have inadequate response or are nonresponders to anti-VEGF therapy, demonstrating the need for additional therapies to more comprehensively treat this disease. Although corticosteroid treatments and implants have demonstrated some efficacy in adjunctive and supplemental treatment, the need to more adequately treat macular edema remains. Our knowledge of diabetic macular edema continues to grow, leading to new currently available and emerging pharmacotherapies to further enhance our treatment and restore vision in those affected by diabetic macular edema. This review will discuss the pathogenesis of diabetic macular edema and the pharmacologic therapies available for its treatment, including anti-VEGF, steroids, and newer therapies still in development, such as angiopoietin antagonists, Tie2 agonists, kallikrein inhibitors, interleukin inhibitors, and others.


Subject(s)
Diabetic Retinopathy/drug therapy , Macular Edema/drug therapy , Adrenal Cortex Hormones/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Angiopoietins/antagonists & inhibitors , Anti-Inflammatory Agents/therapeutic use , Diabetic Retinopathy/physiopathology , Humans , Kallikrein-Kinin System/drug effects , Macular Edema/physiopathology , Protein Kinase Inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors
17.
Chemosphere ; 190: 191-200, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28987408

ABSTRACT

Per- and polyfluoroalkyl substances (PFASs) are ubiquitous and high persistent in human blood, thus potentially inducing a myriad of deleterious consequences. Plasma kallikrein-kinin system (KKS), which physiologically regulates vascular permeability, is vulnerable to exogenous stimulators, like PFASs with long-chain alkyl backbone substituted by electronegative fluorine. The study on the interactions of PFASs with the KKS and the subsequent effects on vascular permeability would be helpful to illustrate how the chemicals penetrate the biological vascular barriers to reach different tissues. In present study, three representative PFASs, including perfluorooctane sulfonic acid (PFOS), perfluorooctanoic acid (PFOA) and perfluorohexadecanoic acid (PFHxDA), were investigated for their effects on the activation of the KKS, paracellular permeability in human retina endothelial cells (HRECs) and integrity of the adherens junctions. In contrast to either PFOS or PFOA, PFHxDA efficiently triggered KKS activation in a concentration-dependent manner based on protease activity assays. The plasma activated by PFHxDA significantly increased paracellular permeability of HRECs through the degradation of adherens junctions. As evidenced by the antagonistic effect of aprotinin, PFHxDA-involved effects on vascular permeability were mediated by KKS activation. The results herein firstly revealed the mechanistic pathway for PFHxDA induced effects on vascular endothelial cells. Regarding the possible structure-related activities of the chemicals, this finding would be of great help in the risk assessment of PFASs.


Subject(s)
Capillary Permeability/drug effects , Endothelial Cells/metabolism , Fluorocarbons/pharmacology , Kallikrein-Kinin System/drug effects , Palmitic Acid/pharmacology , Adherens Junctions/metabolism , Alkanesulfonic Acids/pharmacology , Caprylates/pharmacology , Cells, Cultured , Endothelial Cells/physiology , Humans , Kallikrein-Kinin System/physiology , Plasma/drug effects , Retina/cytology
18.
Afr J Tradit Complement Altern Med ; 14(2): 383-406, 2017.
Article in English | MEDLINE | ID: mdl-28573254

ABSTRACT

BACKGROUND: The commercially available synthetic angiotensin-I-converting enzyme (ACE) inhibitors are known to exert negative side effects which have driven many research groups globally to discover the novel ACE inhibitors. METHOD: Literature search was performed within the PubMed, ScienceDirect.com and Google Scholar. RESULTS: The presence of proline at the C-terminal tripeptide of ACE inhibitor can competitively inhibit the ACE activity. The effects of other amino acids are less studied leading to difficulties in predicting potent peptide sequences. The broad specificity of the enzyme may be due to the dual active sites observed on the somatic ACE. The inhibitors may not necessarily competitively inhibit the enzyme which explains why some reported inhibitors do not have the common ACE inhibitor characteristics. Finally, the in vivo assay has to be carried out before the peptides as the antihypertensive agents can be claimed. The peptides must be absorbed into circulation without being degraded, which will affect their bioavailability and potency. Thus, peptides with strong in vitro IC50 values do not necessarily have the same effect in vivo and vice versa. CONCLUSION: The relationship between peptide amino acid sequence and inhibitory activity, in vivo studies of the active peptides and bioavailability must be studied before the peptides as antihypertensive agents can be claimed.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Antihypertensive Agents/pharmacology , Kallikrein-Kinin System/drug effects , Peptides/pharmacology , Peptidyl-Dipeptidase A/metabolism , Renin-Angiotensin System/drug effects , Amino Acids , Angiotensin-Converting Enzyme Inhibitors/chemistry , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Antihypertensive Agents/chemistry , Antihypertensive Agents/therapeutic use , Catalytic Domain , Humans , Hypertension/drug therapy , Hypertension/metabolism , Peptides/chemistry , Peptides/therapeutic use , Structure-Activity Relationship
19.
Sci Rep ; 7(1): 4308, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28655904

ABSTRACT

Although Danhong injection (DHI) is one of the most prescribed cardiovascular medicines in China, its therapeutic indications and mechanisms remain partially defined. We now identify molecular targets of DHI in resistance vasculatures and demonstrate its role in vascular function and blood pressure (BP) regulation. BP was determined in DHI, Losartan, and placebo- treated Spontaneously Hypertensive Rats (SHR) by both noninvasive and invasive measurements. Vasorelaxation was examined both in conduit and resistance vasculature by ex vivo aortic rings. Microarray analysis was performed and gene expression changes were verified by RT-qPCR and ELISA. Diastolic, systolic and mean BPs were significantly lower in DHI-treated SHR than controls by both tail-cuff and invasive BP measurements. In ex vivo rings, aortic and mesenteric vessels from SHR treated with DHI exhibited significantly greater acetylcholine-mediated relaxation. Among the 282 genes that are differentially expressed in microarray analysis, DHI treatment up-regulated the expression of kallikrein and plasma kallikrein B genes. DHI also significantly increased serum kallikrein content in SHR. Treatment with DHI significantly increased the ratio of aortic lumen to outer diameter. Therefore, the reduction of vascular remodeling and the up-regulation of Kallikrein-kinin system contribute, at least in part, to the antihypertensive effect of DHI in SHR.


Subject(s)
Drugs, Chinese Herbal/administration & dosage , Hypertension/etiology , Hypertension/physiopathology , Kallikrein-Kinin System/drug effects , Vascular Remodeling/drug effects , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Biomarkers , Blood Pressure/drug effects , Disease Models, Animal , Endothelial Cells/metabolism , Hypertension/drug therapy , Kallikreins/blood , Male , Rats , Rats, Inbred SHR , Vasodilation/drug effects
20.
Curr Opin Nephrol Hypertens ; 26(5): 351-357, 2017 09.
Article in English | MEDLINE | ID: mdl-28538015

ABSTRACT

PURPOSE OF REVIEW: Diabetic kidney disease (DKD) is one of the most common complications in diabetes mellitus and accounts for a large proportion of clinical nephrology practice. Studies have shown that the kallikrein-kinin system (KKS) may be involved in several pathogenic mechanisms that contribute to DKD, including oxidative stress, inflammatory cytokines, and profibrotic autacoids. This review focuses on recent research advance on the potential role of the KKS in the development of DKD and its clinical relevance. RECENT FINDINGS: A number of recent studies support the idea that there is a protective role of the KKS in diabetes. For example, agents that activate the KKS have shown strong renal protective effects that might highlight its potential to change the clinical practice. In addition, diabetic mice lacking both bradykinin B2 and B1 receptors have worse kidney lesions as compared with wild-type diabetic mice. SUMMARY: Current basic research has demonstrated that pharmacological activation of the KKS improves renal outcomes in diabetes. These findings suggest that this system may be a therapeutic target in preventing and treating DKD.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/metabolism , Kallikrein-Kinin System , Animals , Diabetes Mellitus, Experimental/genetics , Diabetic Nephropathies/drug therapy , Humans , Kallikrein-Kinin System/drug effects , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...