Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Elife ; 102021 03 08.
Article in English | MEDLINE | ID: mdl-33683204

ABSTRACT

Extensive fibrin deposition in the lungs and altered levels of circulating blood coagulation proteins in COVID-19 patients imply local derangement of pathways that limit fibrin formation and/or promote its clearance. We examined transcriptional profiles of bronchoalveolar lavage fluid (BALF) samples to identify molecular mechanisms underlying these coagulopathies. mRNA levels for regulators of the kallikrein-kinin (C1-inhibitor), coagulation (thrombomodulin, endothelial protein C receptor), and fibrinolytic (urokinase and urokinase receptor) pathways were significantly reduced in COVID-19 patients. While transcripts for several coagulation proteins were increased, those encoding tissue factor, the protein that initiates coagulation and whose expression is frequently increased in inflammatory disorders, were not increased in BALF from COVID-19 patients. Our analysis implicates enhanced propagation of coagulation and decreased fibrinolysis as drivers of the coagulopathy in the lungs of COVID-19 patients.


Subject(s)
Blood Coagulation/genetics , COVID-19/pathology , Fibrin/genetics , Lung/pathology , SARS-CoV-2 , Anticoagulants/metabolism , Bronchoalveolar Lavage Fluid , COVID-19/genetics , COVID-19/metabolism , Endothelial Protein C Receptor/genetics , Endothelial Protein C Receptor/metabolism , Fibrin/metabolism , Gene Expression , Humans , Kallikrein-Kinin System/genetics , Kallikreins/genetics , Kallikreins/metabolism , Kinins/genetics , Kinins/metabolism , Lung/metabolism , RNA, Messenger/metabolism , Sequence Analysis, RNA , Thrombomodulin/genetics , Thrombomodulin/metabolism , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
2.
Sci Rep ; 10(1): 19522, 2020 11 11.
Article in English | MEDLINE | ID: mdl-33177594

ABSTRACT

SARS-CoV-2, the pathogenic agent of COVID-19, employs angiotensin converting enzyme-2 (ACE2) as its cell entry receptor. Clinical data reveal that in severe COVID-19, SARS-CoV-2 infects the lung, leading to a frequently lethal triad of respiratory insufficiency, acute cardiovascular failure, and coagulopathy. Physiologically, ACE2 plays a role in the regulation of three systems that could potentially be involved in the pathogenesis of severe COVID-19: the kinin-kallikrein system, resulting in acute lung inflammatory edema; the renin-angiotensin system, promoting cardiovascular instability; and the coagulation system, leading to thromboembolism. Here we assembled a healthy human lung cell atlas meta-analysis with ~ 130,000 public single-cell transcriptomes and show that key elements of the bradykinin, angiotensin and coagulation systems are co-expressed with ACE2 in alveolar cells and associated with their differentiation dynamics, which could explain how changes in ACE2 promoted by SARS-CoV-2 cell entry result in the development of the three most severe clinical components of COVID-19.


Subject(s)
Betacoronavirus/genetics , Blood Coagulation , Gene Expression Profiling , Kallikrein-Kinin System/genetics , Peptidyl-Dipeptidase A/genetics , Pulmonary Alveoli/cytology , Renin-Angiotensin System/genetics , Angiotensin-Converting Enzyme 2 , Betacoronavirus/enzymology , Betacoronavirus/physiology , Humans , Pulmonary Alveoli/metabolism , SARS-CoV-2 , Serine Endopeptidases/genetics
3.
J Renin Angiotensin Aldosterone Syst ; 21(2): 1470320319900038, 2020.
Article in English | MEDLINE | ID: mdl-32458737

ABSTRACT

BACKGROUND: There is some evidence that components of the renin-angiotensin system and kallikrein-kinin system are not similarly regulated in both sexes. The aim of this work was to analyze the expression of angiotensin-converting enzyme, angiotensin-converting enzyme 2, angiotensin 1 receptor, angiotensin 2 receptor, beta-1 receptor, and beta-2 receptor during the evolution of myocardial infarction. METHODS: Thirty-six male and 36 female Wistar rats were used. Myocardial infarction was induced. Six groups of both sexes were formed, (n=6): (a) sham; (b) 48 h myocardial infarction; (c) one week myocardial infarction; (d) two weeks myocardial infarction; (e) three weeks myocardial infarction and (f) four weeks myocardial infarction. The expression was evaluated by real-time polymerase chain reaction on the penumbra of left ventricle. RESULTS: The mRNA expression of most biomarkers was lower in females than in males. During acute infarction, an increase of all protein expression was found in female and at two weeks while in the male only biomarker changes occurred at three weeks. In addition, in male biomarkers mRNA expression decreased during chronic infarction while in females it did not. CONCLUSIONS: The renin-angiotensin system and kallikrein-kinin system biomarkers expression occurs at earlier times in the female than in the male rat. In addition, during chronic myocardial infarction these biomarkers remained unchanged in females while in males they decreased.


Subject(s)
Kallikrein-Kinin System/genetics , Myocardial Infarction/physiopathology , Renin-Angiotensin System/genetics , Angiotensin-Converting Enzyme 2/biosynthesis , Angiotensin-Converting Enzyme 2/genetics , Animals , Biomarkers , Body Weight , Chronic Disease , Female , Gene Expression Regulation , Heart Ventricles/metabolism , Male , Peptidyl-Dipeptidase A/biosynthesis , Peptidyl-Dipeptidase A/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/biosynthesis , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 2/biosynthesis , Receptor, Angiotensin, Type 2/genetics , Sex Characteristics
4.
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
6.
Biomed Res Int ; 2017: 6761549, 2017.
Article in English | MEDLINE | ID: mdl-29201909

ABSTRACT

Essential hypertension (EH) is a risk factor for some severe diseases. This study aimed to screen out serum special proteins and seek interaction between them, which would provide new therapeutic targets and elucidate the comprehensive pathophysiological mechanism for EH. Patients with EH (Group A, n = 47) and healthy controls (HC) (Group B, n = 47) were recruited in this study. Serums from the two groups were analyzed with isobaric tags for relative and absolute quantitation coupled two-dimensional liquid chromatography followed by electrospray ionization-tandem mass spectrometry technique, while the candidate special proteins were verified with ELISA and western blot. A total of 404 proteins were identified, of which 30 proteins were upregulated (>1.2-fold, p < 0.05) and 81 proteins were downregulated (<0.833-fold, p < 0.05) compared with HC group. With GO, KEGG analysis, and literature retrieval, 4 proteins, cathepsin G, transforming growth factor beta-1, hyaluronidase-1, and kininogen-1, were found jointly involved in the renin-angiotensin-aldosterone system and kallikrein-kinin system. The profiles of these 4 candidate proteins were confirmed with ELISA and western blot. The concentration variation of these 4 proteins could better predict the occurrence and illustrate the pathophysiological mechanism of EH. And their discovery may help pave the way for exploring new therapies of EH.


Subject(s)
Blood Proteins/genetics , Essential Hypertension/blood , Proteome/genetics , Proteomics/methods , Adult , Chromatography, Liquid , Essential Hypertension/physiopathology , Female , Gene Expression Regulation/genetics , Humans , Kallikrein-Kinin System/genetics , Male , Middle Aged , Renin-Angiotensin System/genetics , Risk Factors , Tandem Mass Spectrometry
7.
Expert Opin Biol Ther ; 17(11): 1345-1361, 2017 11.
Article in English | MEDLINE | ID: mdl-28780894

ABSTRACT

INTRODUCTION: The control of hypertension and the resulting cardiovascular events is still insufficient. Thus, the search for novel means for blood pressure (BP) reduction remains worth further clinical and research attention. The advances in vector and construct design sketch the use of gene therapy in hypertension. Areas covered: We have searched for studies using gene therapy in hypertension reporting BP outcomes. We have identified 63 experimental studies demonstrating feasible targeting of the classical and new renin-angiotensin-aldosterone system, ß1-adrenergic receptor, NO-cGMP axis, endothelin, natriuretic peptides, kallikrein system, cytochrome P-450 hydroxylase, oncogenes, growth factors, interleukins, angiopoietin-1, adrenomedullin or Klotho in small rodents. Expert opinion: The usual BP reduction was by 10-30 mmHg for up to several months. Some studies reported target organ damage attenuation or even survival prolongation. However, the concept did not reach the clinical phase, in contrast to other cardiovascular conditions. Increased gene transfection efficacy necessary for a systemic treatment, personalized identification of the implied aetiology from the multifactorial background and evidence from larger mammals are required for gene therapy to compete with the broad spectrum of current therapeutic options in hypertension. Until then, in the field of hypertension, gene modulation will provide a valuable research tool.


Subject(s)
Hypertension/therapy , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Animals , Blood Pressure , Cytochrome P-450 CYP4A/genetics , Cytochrome P-450 CYP4A/metabolism , Endothelin-1/genetics , Endothelin-1/metabolism , Genetic Therapy , Hypertension/genetics , Kallikrein-Kinin System/genetics , Receptors, Adrenergic/genetics , Receptors, Adrenergic/metabolism , Renin-Angiotensin System/genetics
8.
Vasa ; 46(1): 37-45, 2017 01.
Article in English | MEDLINE | ID: mdl-27960614

ABSTRACT

BACKGROUND: Individual shear rate therapy (ISRT) has been designed as a novel non-invasive treatment option for peripheral artery disease (PAD) patients and has been shown to improve endothelial function and walking distance. The aim of this study was to elucidate the impact of ISRT on the level of nitric oxide in patient blood plasma and the expression of related molecular markers in peripheral blood mononuclear cells (PBMCs). Molecular diagnostic tests were performed for two ISRT trials. PATIENTS AND METHODS: In ISRT-1 26 healthy subjects underwent one session of treadmill training and one session of ISRT respectively in a cross-over design. In ISRT-2 14 PAD patients with a stable intermittent claudication underwent a 30 hours long-term treatment. Plasma nitrite release as well as the mRNA expression of NOS2 and key regulators of the kallikrein-kinin system were measured in PBMCs at different time points. RESULTS: Short-term ISRT revealed significantly decreased NOS2 expression in PBMCs of healthy volunteers and PAD patients. Long-term ISRT, in turn, demonstrated a significant plasma nitrite increase in PAD patients. CONCLUSIONS: We verified that long-term ISRT stimulates the vascular system and exerts a comparable effect to physical exercise in regards to NO release, which coincide with recent findings regarding an improvement of endothelial function. However, further studies are necessary to investigate the role for circulating leukocytes.
.


Subject(s)
Counterpulsation/methods , Intermittent Claudication/therapy , Nitrites/blood , Peripheral Arterial Disease/therapy , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Cross-Over Studies , Exercise Test , Female , Humans , Intermittent Claudication/blood , Intermittent Claudication/diagnosis , Intermittent Claudication/physiopathology , Kallikrein-Kinin System/genetics , Leukocytes, Mononuclear/enzymology , Male , Middle Aged , Nitric Oxide/blood , Nitric Oxide Synthase Type II/blood , Nitric Oxide Synthase Type II/genetics , Peripheral Arterial Disease/blood , Peripheral Arterial Disease/diagnosis , Peripheral Arterial Disease/physiopathology , RNA, Messenger/blood , RNA, Messenger/genetics , Stress, Mechanical , Time Factors , Treatment Outcome , Young Adult
9.
Nefrologia ; 37(1): 5-8, 2017.
Article in English, Spanish | MEDLINE | ID: mdl-27469036

ABSTRACT

Renal tubular calcium reabsorption is one of the principal factors that determine serum calcium concentration and calcium excretion. Calcium excretion is regulated by the distal convoluted tubule and connecting tubule, where the epithelial calcium channel TRPV5 can be found, which limits the rate of transcellular calcium transport. The dynamic presence of the TRPV5 channel on the surface of the tubular cell is mediated by an endosomal recycling process. Different intrarenal factors are involved in calcium channel fixation in the apical membrane, including the anti-ageing hormone klotho and tissue kallikrein (TK). Both proteins are synthesised in the distal tubule and secreted in the tubular fluid. TK stimulates active calcium reabsorption through the bradykinin receptor B2 that compromises TRPV5 activation through the protein kinase C pathway. TK-deficient mice show hypercalciuria of renal origin comparable to that seen in TRPV5 knockout mice. There is a polymorphism with loss of function of the human TK gene R53H (allele H) that causes a marked decrease in enzymatic activity. The presence of the allele H seems to be common at least in the Japanese population (24%). These individuals have a tendency to greater calcium and sodium excretion in urine that is more evident during furosemide infusion. Future studies should analyse if manipulating the renal kallikrein-kinin system can correct idiopathic hypercalciuria with drugs other than thiazide diuretics.


Subject(s)
Calcium/urine , Kallikrein-Kinin System/physiology , Alleles , Animals , Calcium Channels/deficiency , Cross-Over Studies , Genetic Predisposition to Disease , Glucuronidase/physiology , Humans , Kallikrein-Kinin System/genetics , Kidney Tubules, Distal/physiopathology , Klotho Proteins , Male , Mice , Mice, Knockout , Polymorphism, Single Nucleotide , Randomized Controlled Trials as Topic , TRPV Cation Channels/deficiency , Tissue Kallikreins/deficiency , Tissue Kallikreins/genetics
10.
Thromb Haemost ; 116(6): 1041-1049, 2016 Nov 30.
Article in English | MEDLINE | ID: mdl-27656708

ABSTRACT

L-arginine is the essential precursor of nitric oxide, and is involved in multiple key physiological processes, including vascular and immune function. The genetic regulation of blood L-arginine levels is largely unknown. We performed a genome-wide association study (GWAS) to identify genetic factors determining serum L-arginine levels, amongst 901 Europeans and 1,394 Indian Asians. We show that common genetic variations at the KLKB1 and F12 loci are strongly associated with serum L-arginine levels. The G allele of single nucleotide polymorphism (SNP) rs71640036 (T/G) in KLKB1 is associated with lower serum L-arginine concentrations (10 µmol/l per allele copy, p=1×10-24), while allele T of rs2545801 (T/C) near the F12 gene is associated with lower serum L-arginine levels (7 µmol/l per allele copy, p=7×10-12). Together these two loci explain 7 % of the total variance in serum L-arginine concentrations. The associations at both loci were replicated in independent cohorts with plasma L-arginine measurements (p<0.004). The two sentinel SNPs are in nearly complete LD with the nonsynonymous SNP rs3733402 at KLKB1 and the 5'-UTR SNP rs1801020 at F12, respectively. SNPs at both loci are associated with blood pressure. Our findings provide new insight into the genetic regulation of L-arginine and its potential relationship with cardiovascular risk.


Subject(s)
Arginine/blood , Genome-Wide Association Study , Kallikrein-Kinin System/genetics , Polymorphism, Single Nucleotide , Adult , Aged , Cardiovascular Diseases/genetics , Female , Humans , Kallikreins/genetics , Male , Middle Aged , Risk Factors
11.
Sci Rep ; 6: 22078, 2016 Feb 22.
Article in English | MEDLINE | ID: mdl-26898917

ABSTRACT

Melanoma is a very aggressive tumor that arises from melanocytes. Late stage and widely spread diseases do not respond to standard therapeutic approaches. The kallikrein-kinin system (KKS) participates in biological processes such as vasodilatation, pain and inflammatory response. However, the role of KKS in tumor formation and progression is not completely understood. The role of the host kinin B1 receptor in melanoma development was evaluated using a syngeneic melanoma model. Primary tumors and metastasis were respectively induced by injecting B16F10 melanoma cells, which are derived from C57BL/6 mice, subcutaneously or in the tail vein in wild type C57BL/6 and B1 receptor knockout mice (B1(-/-)). Tumors developed in B1(-/-) mice presented unfavorable prognostic factors such as increased incidence of ulceration, higher levels of IL-10, higher activation of proliferative pathways such as ERK1/2 and Akt, and increased mitotic index. Furthermore, in the metastasis model, B1(-/-) mice developed larger metastatic colonies in the lung and lower CD8(+)immune effector cells when compared with WT animals. Altogether, our results provide evidences that B1(-/-) animals developed primary tumors with multiple features associated with poor prognosis and unfavorable metastatic onset, indicating that the B1 receptor may contribute to improve the host response against melanoma progression.


Subject(s)
Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Melanoma, Experimental/genetics , Receptor, Bradykinin B1/genetics , Skin Neoplasms/genetics , Animals , Disease Progression , Female , Interleukin-10/genetics , Interleukin-10/metabolism , Kallikrein-Kinin System/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Melanoma, Experimental/metabolism , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mitotic Index , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Bradykinin B1/deficiency , Signal Transduction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
12.
Part Fibre Toxicol ; 12: 17, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-26113123

ABSTRACT

BACKGROUND: Particulate matter (PM) adverse effects on health include lung and heart damage. The renin-angiotensin-aldosterone (RAAS) and kallikrein-kinin (KKS) endocrine systems are involved in the pathophysiology of cardiovascular diseases and have been found to impact lung diseases. The aim of the present study was to evaluate whether PM exposure regulates elements of RAAS and KKS. METHODS: Sprague-Dawley rats were acutely (3 days) and subchronically (8 weeks) exposed to coarse (CP), fine (FP) or ultrafine (UFP) particulates using a particulate concentrator, and a control group exposed to filtered air (FA). We evaluated the mRNA of the RAAS components At1, At2r and Ace, and of the KKS components B1r, B2r and Klk-1 by RT-PCR in the lungs and heart. The ACE and AT1R protein were evaluated by Western blot, as were HO-1 and γGCSc as indicators of the antioxidant response and IL-6 levels as an inflammation marker. We performed a binding assay to determinate AT1R density in the lung, also the subcellular AT1R distribution in the lungs was evaluated. Finally, we performed a histological analysis of intramyocardial coronary arteries and the expression of markers of heart gene reprogramming (Acta1 and Col3a1). RESULTS: The PM fractions induced the expression of RAAS and KKS elements in the lungs and heart in a time-dependent manner. CP exposure induced Ace mRNA expression and regulated its protein in the lungs. Acute and subchronic exposure to FP and UFP induced the expression of At1r in the lungs and heart. All PM fractions increased the AT1R protein in a size-dependent manner in the lungs and heart after subchronic exposure. The AT1R lung protein showed a time-dependent change in subcellular distribution. In addition, the presence of AT1R in the heart was accompanied by a decrease in HO-1, which was concomitant with the induction of Acta1 and Col3a1 and the increment of IL-6. Moreover, exposure to all PM fractions increased coronary artery wall thickness. CONCLUSION: We demonstrate that exposure to PM induces the expression of RAAS and KKS elements, including AT1R, which was the main target in the lungs and the heart.


Subject(s)
Kallikrein-Kinin System/drug effects , Lung/drug effects , Myocardium/metabolism , Particulate Matter/toxicity , Renin-Angiotensin System/drug effects , Actins/genetics , Actins/metabolism , Animals , Antioxidants/metabolism , Collagen Type III/genetics , Collagen Type III/metabolism , Cytokines/metabolism , Gene Expression Regulation , Inhalation Exposure/adverse effects , Kallikrein-Kinin System/genetics , Lung/metabolism , Lung/pathology , Myocardium/pathology , Particle Size , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Renin-Angiotensin System/genetics , Time Factors
13.
Brain Behav Immun ; 33: 90-101, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23777652

ABSTRACT

Multiple sclerosis (MS) is a progressive T cell-mediated autoimmune demyelinating inflammatory disease of the central nervous system (CNS). Although it is recognized that cognitive deficits represent a manifestation of the disease, the underlying pathogenic mechanisms remain unknown. Here we provide evidence of spatial reference memory impairments during the pre-motor phase of experimental autoimmune encephalomyelitis (EAE) in mice. Specifically, these cognitive deficits were accompanied by down-regulation of choline acetyltransferase (ChAT) mRNA expression on day 5 and 11 post-immunization, and up-regulation of inflammatory cytokines in the hippocampus and prefrontal cortex. Moreover, a marked increase in B1R mRNA expression occurred selectively in the hippocampus, whereas protein level was up-regulated in both brain areas. Genetic deletion of kinin B1R attenuated cognitive deficits and cholinergic dysfunction, and blocked mRNA expression of both IL-17 and IFN-γ in the prefrontal cortex, lymph node and spleen of mice subjected to EAE. The discovery of kinin receptors, mainly B1R, as a target for controlling neuroinflammatory response, as well as the cognitive deficits induced by EAE may foster the therapeutic exploitation of the kallikrein-kinin system (KKS), in particular for the treatment of autoimmune disorders, such as MS, mainly during pre-symptomatic phase.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Kallikrein-Kinin System/immunology , Memory Disorders/immunology , Memory Disorders/physiopathology , Movement Disorders/immunology , Movement Disorders/physiopathology , Spatial Behavior , Animals , Choline O-Acetyltransferase/antagonists & inhibitors , Choline O-Acetyltransferase/biosynthesis , Down-Regulation/genetics , Down-Regulation/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Hippocampus/enzymology , Hippocampus/immunology , Hippocampus/pathology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/genetics , Interleukin-17/antagonists & inhibitors , Interleukin-17/genetics , Kallikrein-Kinin System/genetics , Memory Disorders/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Movement Disorders/genetics , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Random Allocation , Receptor, Bradykinin B1/deficiency , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B2/agonists , Receptor, Bradykinin B2/deficiency , Receptor, Bradykinin B2/genetics , Up-Regulation/genetics , Up-Regulation/immunology
14.
Hypertension ; 61(3): 602-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23381795

ABSTRACT

Endothelin-1 (ET-1) and adrenomedullin (ADM) are circulating vasoactive peptides involved in vascular homeostasis and endothelial function. Elevated levels of plasma ET-1 and ADM, and their biologically stable surrogates, C-terminal-pro-endothelin-1 (CT-pro-ET-1) and midregional proadrenomedullin (MR-pro-ADM), are predictors of cardiac death and heart failure. We studied the association of common genetic variation with MR-pro-ADM and CT-pro-ET-1 by genome-wide association analyses in 3444 participants of European ancestry. We performed follow-up genotyping of single nucleotide polymorphisms (SNPs) that showed suggestive or significant association in the discovery stage in additional 3230 participants. The minor variants in KLKB1 (rs4253238) and F12 (rs2731672), both part of the kallikrein-kinin system, were associated with higher MR-pro-ADM (P=4.46E-52 and P=5.90E-24, respectively) and higher CT-pro-ET-1 levels (P=1.23E-122 and P=1.26E-67, respectively). Epistasis analyses showed a significant interaction between the sentinel SNP of F12 and KLKB1 for both traits. In addition, a variant near the ADM gene (rs2957692) was associated with MR-pro-ADM (P=1.05E-12) and a variant in EDN-1 (rs5370) was associated with CT-pro-ET-1 (P=1.49E-27). The total phenotypic variation explained by the genetic variants was 7.2% for MR-pro-ADM and 14.6% for CT-pro-ET-1. KLKB1 encodes plasma kallikrein, a proteolytic enzyme known to cleave high-molecular-weight kininogen to bradykinin and prorenin to renin. We cloned the precursors of ADM and ET-1 and demonstrated that purified plasma kallikrein can cleave these recombinant proteins into multiple smaller peptides. The discovery of genetic variants in the kallikrein-kinin system and in the genes encoding pre-pro-ET-1 and pre-pro-ADM provides novel insights into the (co-)regulation of these vasoactive peptides in the vascular system.


Subject(s)
Adrenomedullin/blood , Endothelin-1/blood , Genome-Wide Association Study , Peptide Fragments/blood , Protein Precursors/blood , Adrenomedullin/genetics , Adult , Aged , Endothelin-1/genetics , Epistasis, Genetic , Female , Humans , Kallikrein-Kinin System/genetics , Kallikreins/genetics , Male , Middle Aged , Peptide Fragments/genetics , Polymorphism, Single Nucleotide , Protein Precursors/genetics , White People/genetics , White People/statistics & numerical data
15.
Am J Epidemiol ; 176 Suppl 7: S72-80, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-23035147

ABSTRACT

The current study comprehensively examined the association between common genetic variants of the kallikrein-kinin system (KKS) and blood pressure salt sensitivity. A 7-day low-sodium followed by a 7-day high-sodium dietary intervention was conducted among 1,906 Han Chinese participants recruited from 2003 to 2005. Blood pressure was measured by using a random-zero sphygmomanometer through the study. A total of 205 single nucleotide polymorphisms (SNPs) covering 11 genes of the KKS were selected for the analyses. Genetic variants of the bradykinin receptor B2 gene (BDKRB2) and the endothelin converting enzyme 1 gene (ECE1) showed significant associations with the salt-sensitivity phenotypes even after adjustment for multiple testing. Compared with the major G allele, the BDKRB2 rs11847625 minor C allele was significantly associated with increased systolic blood pressure responses to low-sodium intervention (P = 0.0001). Furthermore, a haplotype containing allele C was associated with an increased systolic blood pressure response to high-sodium intervention (P = 0.0009). Seven highly correlated ECE1 SNPs were shown to increase the diastolic blood pressure response to low-sodium intervention (P values ranged from 0.0003 to 0.002), with 2 haplotypes containing these 7 SNPs also associated with this same phenotype (P values ranged from 0.0004 to 0.002). In summary, genetic variants of the genes involved in the regulation of KKS may contribute to the salt sensitivity of blood pressure.


Subject(s)
Blood Pressure/genetics , Kallikrein-Kinin System/genetics , Sodium, Dietary/pharmacology , Adult , Alleles , Aspartic Acid Endopeptidases/genetics , Blood Pressure/drug effects , China , Diet, Sodium-Restricted , Endothelin-Converting Enzymes , Female , Genotype , Haplotypes , Humans , Kallikrein-Kinin System/drug effects , Male , Metalloendopeptidases/genetics , Polymorphism, Single Nucleotide/genetics , Receptor, Bradykinin B2/genetics
16.
PLoS One ; 7(7): e40573, 2012.
Article in English | MEDLINE | ID: mdl-22829877

ABSTRACT

The Kallikrein-Kinin System (KKS) has been implicated in several aspects of metabolism, including the regulation of glucose homeostasis and adiposity. Kinins and des-Arg-kinins are the major effectors of this system and promote their effects by binding to two different receptors, the kinin B2 and B1 receptors, respectively. To understand the influence of the KKS on the pathophysiology of obesity and type 2 diabetes (T2DM), we generated an animal model deficient for both kinin receptor genes and leptin (obB1B2KO). Six-month-old obB1B2KO mice showed increased blood glucose levels. Isolated islets of the transgenic animals were more responsive to glucose stimulation releasing greater amounts of insulin, mainly in 3-month-old mice, which was corroborated by elevated serum C-peptide concentrations. Furthermore, they presented hepatomegaly, pronounced steatosis, and increased levels of circulating transaminases. This mouse also demonstrated exacerbated gluconeogenesis during the pyruvate challenge test. The hepatic abnormalities were accompanied by changes in the gene expression of factors linked to glucose and lipid metabolisms in the liver. Thus, we conclude that kinin receptors are important for modulation of insulin secretion and for the preservation of normal glucose levels and hepatic functions in obese mice, suggesting a protective role of the KKS regarding complications associated with obesity and T2DM.


Subject(s)
Glucose/metabolism , Homeostasis/physiology , Liver/metabolism , Receptor, Bradykinin B1/deficiency , Receptor, Bradykinin B1/metabolism , Receptor, Bradykinin B2/deficiency , Receptor, Bradykinin B2/metabolism , Animals , Blood Glucose/metabolism , Body Composition/genetics , Body Composition/physiology , Homeostasis/genetics , Hyperglycemia/blood , Hyperglycemia/genetics , Hyperglycemia/metabolism , Insulin Resistance/genetics , Insulin Resistance/physiology , Kallikrein-Kinin System/genetics , Kallikrein-Kinin System/physiology , Mice , Mice, Knockout , Mice, Obese , Phosphorylation , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B2/genetics
17.
Eur Heart J ; 31(15): 1854-64, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20538738

ABSTRACT

AIMS: The efficacy of angiotensin-converting enzyme (ACE)-inhibitors in stable coronary artery disease (CAD) may be increased by targeting the therapy to those patients most likely to benefit. However, these patients cannot be identified by clinical characteristics. We developed a genetic profile to predict the treatment benefit of ACE-inhibitors exist and to optimize therapy with ACE-inhibitors. METHODS AND RESULTS: In 8907 stable CAD patients participating in the randomized placebo-controlled EUROPA-trial, we analysed 12 candidate genes within the pharmacodynamic pathway of ACE-inhibitors, using 52 haplotype-tagging-single nucleotide polymorphisms (SNPs). The primary outcome was the reduction in cardiovascular mortality, non-fatal myocardial infarction, and resuscitated cardiac arrest during 4.2 years of follow-up. Multivariate Cox regression was performed with multiple testing corrections using permutation analysis. Three polymorphisms, located in the angiotensin-II type I receptor and bradykinin type I receptor genes, were significantly associated with the treatment benefit of perindopril after multivariate adjustment for confounders and correction for multiple testing. A pharmacogenetic score, combining these three SNPs, demonstrated a stepwise reduction of risk in the placebo group and a stepwise decrease in treatment benefit of perindopril with an increasing scores (interaction P < 0.0001). A pronounced treatment benefit was observed in a subgroup of 73.5% of the patients [hazard ratio (HR) 0.67; 95% confidence interval (CI) 0.56-0.79], whereas no benefit was apparent in the remaining 26.5% (HR 1.26; 95% CI 0.97-1.67) with a trend towards a harmful effect. In 1051 patients with cerebrovascular disease from the PROGRESS-trial, treated with perindopril or placebo, an interaction effect of similar direction and magnitude, although not statistically significant, was observed. CONCLUSION: The current study is the first to identify genetic determinants of treatment benefit of ACE-inhibitor therapy. We developed a genetic profile which predicts the treatment benefit of ACE-inhibitors and which could be used to optimize therapy.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Coronary Artery Disease/genetics , Perindopril/therapeutic use , Polymorphism, Single Nucleotide/genetics , Receptor, Angiotensin, Type 1/genetics , Receptor, Bradykinin B1/genetics , Coronary Artery Disease/drug therapy , Female , Gene Frequency , Genotype , Heart Arrest/prevention & control , Humans , Kallikrein-Kinin System/genetics , Male , Middle Aged , Myocardial Infarction/prevention & control , Pharmacogenetics , Renin-Angiotensin System/genetics
18.
J Clin Invest ; 119(4): 768-71, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19348047

ABSTRACT

The kidney kallikrein-kinin system plays important roles in inflammation, coagulation, angiogenesis, and regulation of vessel tone and permeability. In this issue of the JCI, Liu et al. provide data that suggest a protective role for kallikrein in animal models of anti-glomerular basement membrane(GBM) antibody-induced nephritis, an experimental model of Goodpasture disease (see the related article beginning on page 911). Furthermore, human systemic lupus erythematosus and lupus nephritis were shown to be associated with kallikrein 1 (KLK1) and the KLK3 promoter. The authors suggest that kallikrein genes are involved in the development of SLE and lupus nephritis and may exert a renoprotective role. It is possible, however, that the kallikrein-kinin system may play dual roles: protecting the kidney against ischemia and interstitial fibrosis while also mediating vasodilation, inflammation, and activation of the innate immune response.


Subject(s)
Kallikreins/physiology , Lupus Nephritis/etiology , Animals , Disease Models, Animal , Humans , Hypertension/etiology , Hypertension/physiopathology , Kallikrein-Kinin System/genetics , Kallikrein-Kinin System/physiology , Kallikreins/genetics , Kidney/physiopathology , Lupus Erythematosus, Systemic/etiology , Lupus Nephritis/genetics , Lupus Nephritis/physiopathology , Lupus Nephritis/prevention & control , Mice , Models, Biological
20.
Int Immunopharmacol ; 9(6): 653-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19010452

ABSTRACT

The Kallikrein-kinin system works through activation of two receptors. One constitutive, named B2 receptor (B2R) and another inducible, denominated B1 receptor (B1R). In renal fibrosis, B2R receptor activation appears to be protective, however B1R participation is unveiled. The aim of this study was to analyze how the deletion of the B1R would modify tissue responses after unilateral ureteral obstruction (UUO). For that, B1R knockout (B1KO) and wild-type mice (B1B2WT) were subjected to UUO and sacrificed at days 1, 5 and 14. Renal dysfunction was assayed by urine proteinuria/creatinine ratio and percentage of tubulointerstitial fibrosis. Kidneys were harvested at day 5 to analyze anti and pro-inflammatory molecules expression by real-time PCR. We demonstrated that at all time points, B1KO mice presented lower proteinuria/creatinine ratio from bladder urine. B1KO protection was reinforced by its lower tubular interstitial fibrosis percentage at day 14 (B1B2WT: 12.16+/-1.53% vs. B1KO: 6.73+/-1.07%, p<0.02). UUO was able to induce B1R expression and its highest transcription was achieved at day 5. At this day, B1KO had significant lower expression of pro-inflammatory molecules such as TGF-beta, MCP-1, OPN and IL-6 and higher anti-inflammatory components, as IL-10 and HO-1. Herein, we observed that B1R deletion may be an important component in renal fibrosis prevention.


Subject(s)
Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney/pathology , Receptor, Bradykinin B1/genetics , Animals , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Creatinine/blood , Creatinine/urine , Fibrosis , Gene Deletion , Heme Oxygenase-1/immunology , Heme Oxygenase-1/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Kallikrein-Kinin System/genetics , Kidney Diseases/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteinuria/urine , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Ureteral Obstruction/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...