Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Sci Rep ; 11(1): 15924, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34354123

ABSTRACT

Hereditary Angioedema (HAE) is a rare genetic disease generally caused by deficiency or mutations in the C1-inhibitor gene, SERPING1, a member of the Serpin family. HAE results in acute attacks of edema, vasodilation, GI pain and hypotension. C1INH is a key inhibitor of enzymes controlling complement activation, fibrinolysis and the contact system. In HAE patients, contact system activation leads to uncontrolled production of bradykinin, the vasodilator responsible for the characteristic symptoms of HAE. In this study, we present the first physiological in vivo model to mimic acute HAE attacks. We evaluate hypotension, one of the many hallmark symptoms of acute HAE attacks using Serping1 deficient mice (serping1-/-) and implanted telemetry. Attacks were induced by IV injection of a silica nanoparticle (SiNP) suspension. Blood pressure was measured in real time, in conscious and untethered mice using implanted telemetry. SiNP injection induced a rapid, reversible decrease in blood pressure, in the presence of angiotensin converting enzyme (ACE) inhibition. We also demonstrate that an HAE therapeutic, ecallantide, can prevent HAE attacks in this model. The in vivo murine model described here can facilitate the understanding of acute HAE attacks, support drug development and ultimately contribute to improved patient care.


Subject(s)
Angioedemas, Hereditary/physiopathology , Complement C1 Inhibitor Protein/genetics , Disease Models, Animal , Animals , Bradykinin/genetics , Complement Activation/genetics , Complement Activation/immunology , Complement C1 Inhibitor Protein/metabolism , Edema/drug therapy , Female , Fibrinolysis/genetics , Hypotension/physiopathology , Male , Mice , Mice, Inbred C57BL , Peptides , Serpins/genetics
2.
OMICS ; 25(7): 408-416, 2021 07.
Article in English | MEDLINE | ID: mdl-34191617

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus is anticipated to transition to an endemic state as vaccines are providing relief in some, but not all, countries. Drug discovery for COVID-19 can offer another tool in the fight against the pandemic. Additionally, COVID-19 impacts multiple organs that call for a systems medicine approach to planetary health and therapeutics innovation. In this context, innovation for drugs that prevent and treat COVID-19 is timely and much needed. As the virus variants emerge under different ecological conditions and contexts in the long haul, a broad array of vaccine and drug options will be necessary. This expert review article argues for a need to expand the COVID-19 interventions, including and beyond vaccines, to stimulate discovery and development of novel medicines against SARS-CoV-2 infection. The Renin-Angiotensin-Aldosterone System (RAAS) is known to play a major role in SARS-CoV-2 infection. Neprilysin (NEP) and angiotensin-converting enzyme (ACE) have emerged as the pharmaceutical targets of interest in the search for therapeutic interventions against COVID-19. While the NEP/ACE inhibitors offer promise for repurposing against COVID-19, they may display a multitude of effects in different organ systems, some beneficial, and others adverse, in modulating the inflammation responses in the course of COVID-19. This expert review offers an analysis and discussion to deepen our present understanding of the pathophysiological function of neprilysin in multiple organs, and the possible effects of NEP inhibitor-induced inflammatory responses in COVID-19-infected patients.


Subject(s)
Neprilysin/chemistry , Bradykinin/genetics , Bradykinin/metabolism , Renin-Angiotensin System/genetics , Renin-Angiotensin System/physiology , SARS-CoV-2
3.
J Biol Chem ; 296: 100329, 2021.
Article in English | MEDLINE | ID: mdl-33497625

ABSTRACT

Recent functional and proteomic studies in eukaryotes (www.openprot.org) predict the translation of alternative open reading frames (AltORFs) in mature G-protein-coupled receptor (GPCR) mRNAs, including that of bradykinin B2 receptor (B2R). Our main objective was to determine the implication of a newly discovered AltORF resulting protein, termed AltB2R, in the known signaling properties of B2R using complementary methodological approaches. When ectopically expressed in HeLa cells, AltB2R presented predominant punctate cytoplasmic/perinuclear distribution and apparent cointeraction with B2R at plasma and endosomal/vesicular membranes. The presence of AltB2R increases intracellular [Ca2+] and ERK1/2-MAPK activation (via phosphorylation) following B2R stimulation. Moreover, HEK293A cells expressing mutant B2R lacking concomitant expression of AltB2R displayed significantly decreased maximal responses in agonist-stimulated Gαq-Gαi2/3-protein coupling, IP3 generation, and ERK1/2-MAPK activation as compared with wild-type controls. Conversely, there was no difference in cell-surface density as well as ligand-binding properties of B2R and in efficiencies of cognate agonists at promoting B2R internalization and ß-arrestin 2 recruitment. Importantly, both AltB2R and B2R proteins were overexpressed in prostate and breast cancers, compared with their normal counterparts suggesting new associative roles of AltB2R in these diseases. Our study shows that BDKRB2 is a dual-coding gene and identifies AltB2R as a novel positive modulator of some B2R signaling pathways. More broadly, it also supports a new, unexpected alternative proteome for GPCRs, which opens new frontiers in fields of GPCR biology, diseases, and drug discovery.


Subject(s)
Alternative Splicing/genetics , Bradykinin/genetics , Protein Isoforms/genetics , Receptor, Bradykinin B2/genetics , Bradykinin/metabolism , Endocytosis/genetics , Endosomes/genetics , HEK293 Cells , HeLa Cells , Humans , MAP Kinase Signaling System/genetics , Open Reading Frames/genetics , Proteomics , Signal Transduction/genetics
4.
Cells ; 9(9)2020 09 21.
Article in English | MEDLINE | ID: mdl-32967374

ABSTRACT

During the last three decades, timely myocardial reperfusion using either thrombolytic therapy or primary percutaneous intervention (pPCI) has allowed amazing improvements in outcomes with a more than halving in 1-year ST-elevation myocardial infarction (STEMI) mortality. However, mortality and left ventricle (LV) remodeling remain substantial in these patients. As such, novel therapeutic interventions are required to reduce myocardial infarction size, preserve LV systolic function, and improve survival in reperfused-STEMI patients. Myocardial ischemia-reperfusion injury (MIRI) prevention represents the main goal to reach in order to reduce STEMI mortality. There is currently no effective therapy for MIRI prevention in STEMI patients. A significant reason for the weak and inconsistent results obtained in this field may be the presence of multiple, partially redundant, mechanisms of cell death during ischemia-reperfusion, whose relative importance may depend on the conditions. Therefore, it is always more recognized that it is important to consider a "multi-targeted cardioprotective therapy", defined as an additive or synergistic cardioprotective agents or interventions directed to distinct targets with different timing of application (before, during, or after pPCI). Given that some neprilysin (NEP) substrates (natriuretic peptides, angiotensin II, bradykinin, apelins, substance P, and adrenomedullin) exert a cardioprotective effect against ischemia-reperfusion injury, it is conceivable that antagonism of proteolytic activity by this enzyme may be considered in a multi-targeted strategy for MIRI prevention. In this review, by starting from main pathophysiological mechanisms promoting MIRI, we discuss cardioprotective effects of NEP substrates and the potential benefit of NEP pharmacological inhibition in MIRI prevention.


Subject(s)
Aminobutyrates/therapeutic use , Angiotensin II/genetics , Cardiotonic Agents/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Neprilysin/antagonists & inhibitors , ST Elevation Myocardial Infarction/drug therapy , Tetrazoles/therapeutic use , Adrenomedullin/genetics , Adrenomedullin/metabolism , Angiotensin II/metabolism , Animals , Apelin/genetics , Apelin/metabolism , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Biphenyl Compounds , Bradykinin/genetics , Bradykinin/metabolism , Drug Combinations , Gene Expression Regulation , Humans , Mice , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/physiopathology , Neprilysin/genetics , Neprilysin/metabolism , ST Elevation Myocardial Infarction/enzymology , ST Elevation Myocardial Infarction/genetics , ST Elevation Myocardial Infarction/physiopathology , Substance P/genetics , Substance P/metabolism , Survival Analysis , Valsartan , Ventricular Remodeling/drug effects
5.
Nat Commun ; 11(1): 2132, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32358539

ABSTRACT

Brown adipose tissue (BAT) is known to secrete regulatory factors in response to thermogenic stimuli. Components of the BAT secretome may exert local effects that contribute to BAT recruitment and activation. Here, we found that a thermogenic stimulus leads to enhanced secretion of kininogen (Kng) by BAT, owing to induction of kininogen 2 (Kng2) gene expression. Noradrenergic, cAMP-mediated signals induce KNG2 expression and release in brown adipocytes. Conversely, the expression of kinin receptors, that are activated by the Kng products bradykinin and [Des-Arg9]-bradykinin, are repressed by thermogenic activation of BAT in vivo and of brown adipocytes in vitro. Loss-of-function models for Kng (the circulating-Kng-deficient BN/Ka rat) and bradykinin (pharmacological inhibition of kinin receptors, kinin receptor-null mice) signaling were coincident in showing abnormal overactivation of BAT. Studies in vitro indicated that Kng and bradykinin exert repressive effects on brown adipocyte thermogenic activity by interfering the PKA/p38 MAPK pathway of control of Ucp1 gene transcription, whereas impaired kinin receptor expression enhances it. Our findings identify the kallikrein-kinin system as a relevant component of BAT thermogenic regulation that provides auto-regulatory inhibitory signaling to BAT.


Subject(s)
Adipose Tissue, Brown/metabolism , Kallikreins/metabolism , Kinins/metabolism , Animals , Bradykinin/genetics , Bradykinin/metabolism , Endocrine System/metabolism , Fluorescent Antibody Technique , Kallikreins/genetics , Kininogens/genetics , Kininogens/metabolism , Kinins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology
6.
Int J Mol Sci ; 21(2)2020 Jan 08.
Article in English | MEDLINE | ID: mdl-31936257

ABSTRACT

Bradykinin (BK), a hormone inducing pain and inflammation, is known to inhibit potassium M-currents (IM) and to increase the excitability of the superior cervical ganglion (SCG) neurons by activating the Ca2+-calmodulin pathway. M-current is also reduced by muscarinic agonists through the depletion of membrane phosphatidylinositol 4,5-biphosphate (PIP2). Similarly, the activation of muscarinic receptors inhibits the current through two-pore domain potassium channels (K2P) of the "Tandem of pore-domains in a Weakly Inward rectifying K+ channel (TWIK)-related channels" (TREK) subfamily by reducing PIP2 in mouse SCG neurons (mSCG). The aim of this work was to test and characterize the modulation of TREK channels by bradykinin. We used the perforated-patch technique to investigate riluzole (RIL) activated currents in voltage- and current-clamp experiments. RIL is a drug used in the palliative treatment of amyotrophic lateral sclerosis and, in addition to blocking voltage-dependent sodium channels, it also selectively activates the K2P channels of the TREK subfamily. A cell-attached patch-clamp was also used to investigate TREK-2 single channel currents. We report here that BK reduces spike frequency adaptation (SFA), inhibits the riluzole-activated current (IRIL), which flows mainly through TREK-2 channels, by about 45%, and reduces the open probability of identified single TREK-2 channels in cultured mSCG cells. The effect of BK on IRIL was precluded by the bradykinin receptor (B2R) antagonist HOE-140 (d-Arg-[Hyp3, Thi5, d-Tic7, Oic8]BK) but also by diC8PIP2 which prevents PIP2 depletion when phospholipase C (PLC) is activated. On the contrary, antagonizing inositol triphosphate receptors (IP3R) using 2-aminoethoxydiphenylborane (2-APB) or inhibiting protein kinase C (PKC) with bisindolylmaleimide did not affect the inhibition of IRIL by BK. In conclusion, bradykinin inhibits TREK-2 channels through the activation of B2Rs resulting in PIP2 depletion, much like we have demonstrated for muscarinic agonists. This mechanism implies that TREK channels must be relevant for the capture of information about pain and visceral inflammation.


Subject(s)
Bradykinin/metabolism , Neurons/drug effects , Phosphatidylinositol 4,5-Diphosphate/metabolism , Potassium Channels, Tandem Pore Domain/genetics , Sympathetic Nervous System/drug effects , Action Potentials/drug effects , Animals , Bradykinin/administration & dosage , Bradykinin/analogs & derivatives , Bradykinin/genetics , Bradykinin/pharmacology , Cells, Cultured , Humans , Mice , Muscarinic Agonists/pharmacology , Neurons/pathology , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/genetics , Potassium/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Receptors, Muscarinic/genetics , Riluzole/pharmacology , Sodium Channel Blockers/pharmacology , Superior Cervical Ganglion/drug effects , Sympathetic Nervous System/metabolism , Type C Phospholipases
7.
Clin Exp Allergy ; 49(11): 1395-1402, 2019 11.
Article in English | MEDLINE | ID: mdl-31574187

ABSTRACT

Since the Osler's identification of the inherited nature of hereditary angioedema, a huge array of information was collected on pathogenetic mechanisms of the disease. Over the last years, information grew fast, and mutations in different genes, in addition to C1-inhibitor, were found to be causative. All types are inherited as autosomal-dominant traits with incomplete penetrance and little or no genotype-phenotype correlation. As a result, the clinical expression is characterized by a large heterogeneity. The acknowledgement of mechanisms leading to heterogeneity of the clinical phenotype is likely to provide important information not only for a better understanding of the pathogenesis but also for therapy. Regardless of which gene is mutated, similar pathways seem to play a pivotal role, triggering the up-regulation of contact activation system/kallikrein kinin system and giving rise to an unbalanced increase of bradykinin. However, notwithstanding the increase of bradykinin in bloodstream, the phenomenon is localized and no general vascular leakage and oedema is recognized. Thus, it is conceivable that there exist one or more localized factors that stimulate the production of bradykinin, which does not become a systemically event. Uncovering of these factors may shed lights on the missing part of the pathogenesis of hereditary angioedema. The present review, collecting information on pathogenesis from biochemical and genetics investigations, tries to provide a comprehensive view of the pathogenesis of hereditary angioedema. This can allow for a better understanding of the disease and lead to focused investigations that can further improve our knowledge.


Subject(s)
Angioedemas, Hereditary , Bradykinin , Capillary Permeability/genetics , Angioedemas, Hereditary/genetics , Angioedemas, Hereditary/metabolism , Angioedemas, Hereditary/pathology , Bradykinin/genetics , Bradykinin/metabolism , Humans
8.
Nucleic Acid Ther ; 29(2): 82-91, 2019 04.
Article in English | MEDLINE | ID: mdl-30817230

ABSTRACT

Kallikrein is the key contact system mediator responsible for the conversion of high-molecular-weight kininogen into the inflammatory vasodilator peptide bradykinin, a process regulated by C1-esterase inhibitor (C1-INH). In hereditary angioedema (HAE), genetic mutations result in deficient or dysfunctional C1-INH and dysregulation of the contact system leading to recurrent, sometimes fatal, angioedema attacks. IONIS-PKKRx is a second-generation 2'-O-(2-methoxyethyl)-modified chimeric antisense oligonucleotide, designed to bind and selectively reduce prekallikrein (PKK) mRNA in the liver. IONIS-PKKRx demonstrated dose-dependent reduction of human prekallikrein hepatic mRNA and plasma protein in transgenic mice and dose- and time-dependent reductions of plasma PKK in Cynomolgus monkeys. Similar dose-dependent reductions of plasma PKK levels were observed in healthy human volunteers accompanied by decreases in bradykinin generation capacity with an acceptable safety and tolerability profile. These results highlight a novel and specific approach to target PKK for the treatment of HAE and other diseases involving contact system activation and overproduction of bradykinin.


Subject(s)
Angioedemas, Hereditary/therapy , Bradykinin/genetics , Complement C1s/genetics , Prekallikrein/genetics , Angioedemas, Hereditary/blood , Angioedemas, Hereditary/genetics , Animals , Animals, Genetically Modified/blood , Bradykinin/blood , Complement C1 Inhibitor Protein/pharmacology , Complement C1s/antagonists & inhibitors , Dose-Response Relationship, Drug , Healthy Volunteers , Humans , Liver/drug effects , Liver/metabolism , Macaca fascicularis/blood , Mice , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Prekallikrein/antagonists & inhibitors
9.
Clin Exp Allergy ; 49(2): 142-154, 2019 02.
Article in English | MEDLINE | ID: mdl-30537422

ABSTRACT

Angioedema is a rare adverse effect of the commonly used angiotensin-converting enzyme inhibitors (ACEi) and is reported to occur with a prevalence of 0.1%-0.7%. Although most ACEi-induced angioedema (ACEi-A) cases are mild, severe cases requiring intensive care and even resulting in death have been reported in the literature. The mechanisms underlying ACEi-A are not yet fully understood, but bradykinin and/or substance P accumulation resulting from inhibition of ACE is believed to play a crucial role. ACEi-A occurs at variable frequencies across different racial groups, suggesting a genetic association with the development of ACEi-A. To date, one genome-wide association study and several candidate gene studies have been published on the association of genetic variation with ACEi-A. Genetic associations reported have been attributed to several distinct mechanisms: (a) genes coding for alternative enzymes responsible for the degradation of bradykinin and/or substance P in the diminution of ACE activity (b) ACE gene function, (c) bradykinin receptor genes, (d) genes implicated in immune and inflammation regulation and (e) genes in the fibrinolytic and coagulation pathway. Despite several plausible genetic associations, there are currently no genetic variants with sufficient effect to be clinically useful. The low incidence of ACEi-A suggests that a combination of genomic approaches with the capability to detect potentially important variants might be required to shed light on the mechanism of this adverse reaction. Additionally, many non-genetic risk factors associated with ACEi-A suggest the potential contribution of epigenetic dysregulation.


Subject(s)
Angioedema , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Polymorphism, Genetic , Angioedema/blood , Angioedema/chemically induced , Angioedema/epidemiology , Angioedema/genetics , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Bradykinin/blood , Bradykinin/genetics , Genome-Wide Association Study , Humans , Pharmacogenetics , Prevalence , Substance P/blood , Substance P/genetics
10.
Protein J ; 37(4): 324-332, 2018 08.
Article in English | MEDLINE | ID: mdl-30008150

ABSTRACT

In recent decades, various types of bioactive substances have been identified from amphibian skin and its secretions. Bradykinin-related peptides (BRPs) are among these compounds that make up the host defence system of amphibians. In the present study, we identified six novel BRPs, amolopkinin-GN1, amolopkinin-RK1, amolopkinin-TR1, amolopkinin-LF1, ranakinin-MS1, and ranakinin-MS2, from five East Asian amphibians, Amolops granulosus, Amolops ricketti, Amolops torrentis, Amolops lifanensis, and Hylarana maosonensis. This is the first report on BRPs in the skin of these species. Physiological assays reveal that these peptides have a contractive effect on the smooth muscle of rat ileum.


Subject(s)
Bradykinin/pharmacology , Ileum/drug effects , Muscle Contraction/drug effects , Peptide Fragments/pharmacology , Skin/chemistry , Amino Acid Sequence , Animals , Anura , Bradykinin/genetics , Bradykinin/isolation & purification , Cloning, Molecular , Peptide Fragments/genetics , Peptide Fragments/isolation & purification , Rats , Sequence Homology , Skin/metabolism
11.
Clin Exp Med ; 18(3): 355-361, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29623491

ABSTRACT

Hereditary angioedema (HAE) is a rare autosomic-dominant disorder characterized by a deficiency of C1 esterase inhibitor which causes episodic swellings of subcutaneous tissues, bowel walls and upper airways that are disabling and potentially life-threatening. We evaluated n = 17 patients with confirmed HAE diagnosis during attack and remission state and n = 19 healthy subjects. The samples were tested for a panel of IL (Interleukin)-17-type cytokines (IL-1ß, IL-6, IL-10, granulocyte-macrophage colony stimulating factor (GM-CSF), IL-17, IL-21, IL-22, IL-23) and transforming growth factor-beta (TGF-ß) subtypes. Data indicate that there are variations of cytokine levels in HAE subjects comparing the condition during the crisis respect to the value in the remission phase, in particular type 17 signature cytokines are increased, whereas IL-23 is unmodified and TGF-ß3 is significantly reduced. When comparing healthy and HAE subjects in the remission state, we found a significant difference for IL-17, GM-CSF, IL-21, TGF-ß1 and TGF-ß2 cytokines. These results confirm and extend our previous findings indicating that in HAE there is operating an inflammatory activation process, which involves also T helper 17 (Th17) cytokines and TGF-ß isoforms, associated with localized angioedema attacks and characterized by elevated bradykinin levels.


Subject(s)
Angioedemas, Hereditary/diagnosis , Angioedemas, Hereditary/immunology , Gene Expression Regulation/immunology , Interleukin-17/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/immunology , Adolescent , Adult , Aged , Angioedemas, Hereditary/genetics , Angioedemas, Hereditary/pathology , Bradykinin/genetics , Bradykinin/immunology , Bronchi/immunology , Bronchi/pathology , Case-Control Studies , Child , Complement C1 Inhibitor Protein/genetics , Complement C1 Inhibitor Protein/immunology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Interleukin-17/genetics , Interleukin-23/genetics , Interleukin-23/immunology , Interleukins/genetics , Interleukins/immunology , Intestines/immunology , Intestines/pathology , Male , Middle Aged , Subcutaneous Tissue/immunology , Subcutaneous Tissue/pathology , Th17 Cells/pathology , Transforming Growth Factor beta/genetics , Interleukin-22
12.
J Am Chem Soc ; 140(7): 2401-2404, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29412650

ABSTRACT

Mutational analysis is widely used to study the relationship between sequence and structure of proteins and peptides. It is often assumed that substituting a proline with another amino acid "locks" the peptide bond in the trans conformation, allowing only a subset of the initial molecular geometries to be observed. To test this assumption, we assess the result of substituting two prolines in the bradykinin sequence with alanine using field-asymmetric ion mobility spectrometry combined with cryogenic ion spectroscopy in the gas phase. While the structure of the mutant coincides with a part of the conformational space of the original peptide, the higher flexibility of the alanine backbone compared to proline allows it to access additional structures. We conclude that proline-to-nonproline substitutions are helpful to assign structures, but they should be used in conjunction with spectroscopic techniques that allow detailed comparison of the structures of the mutant and the native peptide.


Subject(s)
Bradykinin/genetics , Alanine/chemistry , Bradykinin/chemistry , Mutation , Proline/chemistry , Protein Conformation
13.
Biochim Biophys Acta Mol Cell Res ; 1864(10): 1855-1866, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28757212

ABSTRACT

In recent years a wide range of studies have shown that G protein-coupled receptors modulate a variety of cell functions through the formation of dimers. For instance, there is growing evidence for the dimerization of bradykinin or dopamine receptors, both as homodimers and heterodimers. A discovery of direct interactions of angiotensin II receptors with bradykinin 2 receptor (B2R) or dopamine D2 (D2R) receptor has led to a hypothesis on a potential dimerization between two latter receptors. In this study, we have demonstrated a constitutive colocalization of receptors on the membranes of HEK293 cells transiently transfected with plasmid vectors encoding B2R and D2R, fused with fluorescent proteins. The receptor colocalization was significantly enhanced by specific agonists of B2R or D2R after 5min following the addition, whereas simultaneous stimulation with these agonists did not influence the B2R/D2R colocalization level. In addition, B2R-D2R heterodimerization was confirmed with FLIM-FRET technique. The most characteristic signaling pathways for B2R and D2R, dependent on intracellular Ca2+ and cAMP concentration, respectively, were analyzed in cells presenting similar endogenous expression of B2R and D2R. Significant changes in receptors' signaling were observed after simultaneous stimulation with agonists, suggesting transformations in proteins' conformation after dimerization. The evidence of B2R-D2R dimerization may open new perspectives in the modulation of diverse cellular functions which depend on their activation.


Subject(s)
Bradykinin/chemistry , Dimerization , Receptor, Bradykinin B2/chemistry , Receptors, Dopamine D2/chemistry , Bradykinin/genetics , Bradykinin/metabolism , HEK293 Cells , Humans , Protein Conformation , Receptor, Bradykinin B2/agonists , Receptor, Bradykinin B2/genetics , Receptors, Dopamine D2/genetics , Signal Transduction/genetics
14.
Biochim Biophys Acta Mol Cell Res ; 1864(11 Pt B): 2118-2127, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28743596

ABSTRACT

The contact system is a potent procoagulant and proinflammatory plasma protease cascade that is initiated by binding ("contact")-induced, auto-activation of factor XII zymogen. Formed active serine protease FXIIa then cleaves plasma prekallikrein to kallikrein that in turn liberates the mediator bradykinin from its precursor high molecular weight kininogen. Bradykinin induces inflammation with implications for host defense and innate immunity. FXIIa also triggers the intrinsic pathway of coagulation that has been shown to critically contribute to thrombosis. Vice versa, FXII deficiency impairs thrombosis in animal models without inducing abnormal excessive bleeding. Recent work has established the FXIIa-driven contact system as promising target for anticoagulant and anti-inflammatory drugs. This review focuses on the biochemistry of the contact system, its regulation by endogenous and exogenous inhibitors, and roles in disease states. This article is part of a Special Issue entitled: Proteolysis as a Regulatory Event in Pathophysiology edited by Stefan Rose-John.


Subject(s)
Blood Coagulation/genetics , Factor XII Deficiency/genetics , Factor XIIa/genetics , Inflammation/genetics , Bradykinin/genetics , Factor XII Deficiency/blood , Factor XII Deficiency/pathology , Humans , Immunity, Innate/genetics , Inflammation/blood , Inflammation/pathology , Kallikreins/genetics , Thrombosis/blood , Thrombosis/genetics , Thrombosis/pathology
15.
Physiol Res ; 66(3): 425-439, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28730837

ABSTRACT

Transient receptor potential A1 (TRPA1) is an excitatory ion channel that functions as a cellular sensor, detecting a wide range of proalgesic agents such as environmental irritants and endogenous products of inflammation and oxidative stress. Topical application of TRPA1 agonists produces an acute nociceptive response through peripheral release of neuropeptides, purines and other transmitters from activated sensory nerve endings. This, in turn, further regulates TRPA1 activity downstream of G-protein and phospholipase C-coupled signaling cascades. Despite the important physiological relevance of such regulation leading to nociceptor sensitization and consequent pain hypersensitivity, the specific domains through which TRPA1 undergoes post-translational modifications that affect its activation properties are yet to be determined at a molecular level. This review aims at providing an account of our current knowledge on molecular basis of regulation by neuronal inflammatory signaling pathways that converge on the TRPA1 channel protein and through modification of its specific residues influence the extent to which this channel may contribute to pain.


Subject(s)
Nociceptors/physiology , Pain/metabolism , TRPA1 Cation Channel/physiology , Animals , Bradykinin/genetics , Bradykinin/metabolism , Humans , Pain/genetics , Signal Transduction/physiology
16.
Semin Thromb Hemost ; 43(8): 814-826, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28346966

ABSTRACT

Contact activation is the surface-induced conversion of factor XII (FXII) zymogen to the serine protease FXIIa. Blood-circulating FXII binds to negatively charged surfaces and this contact to surfaces triggers a conformational change in the zymogen inducing autoactivation. Several surfaces that have the capacity for initiating FXII contact activation have been identified, including misfolded protein aggregates, collagen, nucleic acids, and platelet and microbial polyphosphate. Activated FXII initiates the proinflammatory kallikrein-kinin system and the intrinsic coagulation pathway, leading to formation of bradykinin and thrombin, respectively. FXII contact activation is well characterized in vitro and provides the mechanistic basis for the diagnostic clotting assay, activated partial thromboplastin time. However, only in the past decade has the critical role of FXII contact activation in pathological thrombosis been appreciated. While defective FXII contact activation provides thromboprotection, excess activation underlies the swelling disorder hereditary angioedema type III. This review provides an overview of the molecular basis of FXII contact activation and FXII contact activation-associated disease states.


Subject(s)
Blood Coagulation , Bradykinin/metabolism , Factor XIIa/metabolism , Hereditary Angioedema Type III/metabolism , Thrombin/metabolism , Animals , Bradykinin/genetics , Enzyme Activation , Factor XIIa/genetics , Hereditary Angioedema Type III/genetics , Humans , Thrombin/genetics
17.
Peptides ; 85: 46-55, 2016 11.
Article in English | MEDLINE | ID: mdl-27628189

ABSTRACT

Bradykinin exerts its vascular actions via two types of receptors, the non-constitutively expressed bradykinin receptor type 1 (BR1) and the constitutive type 2 receptor (BR2). Bradykinin-induced vasorelaxation is age-dependent, a phenomenon related to the varying amounts of BR1 and BR2 in the vasculature. Isoleucine-proline-proline (Ile-Pro-Pro), a bioactive tripeptide, lowers elevated blood pressure and improves impaired endothelium-dependent vasorelaxation in hypertensive rats. It inhibits angiotensin converting enzyme 1 (ACE1). Other mechanisms of action have also been postulated. The aims of the study were to clarify the underlying mechanisms of the age-dependency of bradykinin-induced vasodilatation such as the roles of the two bradykinin receptors, the mas-receptor and synergism with Ile-Pro-Pro. The vascular response studies were conducted using mesenteric artery and aorta rings from normotensive 6 wk. (young) and 22 wk. (old) Wistar rats. Cumulative dosing of acetylcholine, bradykinin and angiotensin(1-7) (Ang(1-7))were tested in phenylephrine-induced vasoconstriction with or without 10min pre-incubation with antagonists against BR1-, BR2- or mas-receptors, Ang(1-7) or ACE1-inhibitors captopril and Ile-Pro-Pro. The bradykinin-induced vasorelaxation in vitro was age-dependent and it was improved by pre-incubation with Ile-Pro-Pro, especially in old rats with endothelial dysfunction. The mas-receptor antagonist, D-Pro7-Ang(1-7) abolished bradykinin-induced relaxation totally. Interestingly, BR1 and BR2 antagonists only slightly reduced bradykinin-induced vasorelaxation, as an evidence for the involvement of other mechanisms in addition to receptor activation. In conclusion, bradykinin-induced vasorelaxation was age-dependent and Ile-Pro-Pro improved it. Mas receptor antagonist abolished relaxation while bradykinin receptor antagonist only slightly reduced it, suggesting that bradykinin-induced vasorelaxation is regulated also by other mechanisms than the classical BR1/BR2 pathway.


Subject(s)
Hypertension/drug therapy , Oligopeptides/administration & dosage , Proto-Oncogene Proteins/metabolism , Receptor, Bradykinin B1/metabolism , Receptor, Bradykinin B2/metabolism , Receptors, G-Protein-Coupled/metabolism , Angiotensin I/metabolism , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/metabolism , Animals , Blood Pressure/drug effects , Bradykinin/genetics , Bradykinin/metabolism , Captopril/administration & dosage , Humans , Hypertension/genetics , Hypertension/pathology , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Mesenteric Arteries/pathology , Oligopeptides/metabolism , Peptide Fragments/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/antagonists & inhibitors , Rats , Rats, Wistar , Receptor, Bradykinin B1/drug effects , Receptor, Bradykinin B2/drug effects , Receptors, G-Protein-Coupled/antagonists & inhibitors , Vasodilation/drug effects
18.
J Pain ; 17(11): 1183-1197, 2016 11.
Article in English | MEDLINE | ID: mdl-27544818

ABSTRACT

Chronic vulvar pain is alarmingly common in women of reproductive age and is often accompanied by psychological distress, sexual dysfunction, and a significant reduction in quality of life. Localized provoked vulvodynia (LPV) is associated with intense vulvar pain concentrated in the vulvar vestibule (area surrounding vaginal opening). To date, the origins of vulvodynia are poorly understood, and treatment for LPV manages pain symptoms, but does not resolve the root causes of disease. Until recently, no definitive disease mechanisms had been identified; our work indicates LPV has inflammatory origins, although additional studies are needed to understand LPV pain. Bradykinin signaling is one of the most potent inducers of inflammatory pain and is a candidate contributor to LPV. We report that bradykinin receptors are expressed at elevated levels in LPV patient versus healthy control vestibular fibroblasts, and patient vestibular fibroblasts produce elevated levels of proinflammatory mediators with bradykinin stimulation. Inhibiting expression of one or both bradykinin receptors significantly reduces proinflammatory mediator production. Finally, we determined that bradykinin activates nuclear factor (NF)κB signaling (a major inflammatory pathway), whereas inhibition of NFκB successfully ablates this response. These data suggest that therapeutic agents targeting bradykinin sensing and/or NFκB may represent new, more specific options for LPV therapy. PERSPECTIVE: There is an unmet need for the development of more effective vulvodynia therapies. As we explore the mechanisms by which human vulvar fibroblasts respond to proinflammatory/propain stimuli, we move closer to understanding the origins of chronic vulvar pain and identifying new therapeutic targets, knowledge that could significantly improve patient care.


Subject(s)
Bradykinin/metabolism , Pelvic Pain/metabolism , Signal Transduction/physiology , Adult , Bradykinin/analogs & derivatives , Bradykinin/genetics , Bradykinin/pharmacology , Bradykinin Receptor Antagonists/pharmacology , Case-Control Studies , Cells, Cultured , Chronic Pain , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Interleukin-6/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Pelvic Pain/drug therapy , Pelvic Pain/pathology , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Bradykinin/genetics , Receptors, Bradykinin/metabolism
19.
Biochem Pharmacol ; 97(2): 203-14, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26235942

ABSTRACT

Phosphorylated kininogen and some of its fragments containing serine phosphorylated bradykinin ([pS(6)]-Bk) were identified in human serum and plasma by a phosphoproteomic approach. We report the kininogenase ability of human tissue and plasma kallikreins and tryptase to generate [pS(6)]-Bk or Lys-[pS(6)]-Bk having as substrate the synthetic human kininogen fluorescent fragment Abz-MISLMKRPPGF[pS(386)]PFRSSRI-NH2. The pharmacological assays of [pS(6)]-Bk showed it as a full B2 bradykinin receptor agonist in smooth muscle, it produces a portal liver hypertensive response in rat and mouse paw edema that lasts longer than Bk. The rat hypotensive response to infusions of Bk is greater than that of [pS(6)]Bk, both if injected through femoral vein or aorta. [pS(6)]-Bk was more resistant than Bk to kininase digestion performed with angiotensin converting enzyme, neprilysin, thimet oligopeptidase, aminopeptidase P and carboxypeptidase M. (1)H-NMR experiments indicated that [pS(6)]-Bk has lower flexibility, with the pS(6)-P(7) bond restricted to the trans conformation, and can explain [pS(6)]-Bk resistance to hydrolysis. In conclusion, [pS(6)]-Bk presenting lower activity than Bk, with longer lasting effects and being slowly released by kininogenases from synthetic Abz-MISLMKRPPGF[pS(386)]PFRSSRI-NH2, suggests that phosphorylation of the kininogens can be an efficient kallikrein-kinin system regulator.


Subject(s)
Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Peptide Hydrolases/pharmacology , Amino Acid Sequence , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Bradykinin/genetics , Guinea Pigs , Humans , Hydrolysis/drug effects , Mice , Molecular Sequence Data , Organ Culture Techniques , Peptide Hydrolases/genetics , Rabbits , Rats , Rats, Sprague-Dawley , Rats, Wistar
20.
J Clin Invest ; 125(8): 3132-46, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26193639

ABSTRACT

Hereditary angioedema type III (HAEIII) is a rare inherited swelling disorder that is associated with point mutations in the gene encoding the plasma protease factor XII (FXII). Here, we demonstrate that HAEIII-associated mutant FXII, derived either from HAEIII patients or recombinantly produced, is defective in mucin-type Thr309-linked glycosylation. Loss of glycosylation led to increased contact-mediated autoactivation of zymogen FXII, resulting in excessive activation of the bradykinin-forming kallikrein-kinin pathway. In contrast, both FXII-driven coagulation and the ability of C1-esterase inhibitor to bind and inhibit activated FXII were not affected by the mutation. Intravital laser-scanning microscopy revealed that, compared with control animals, both F12-/- mice reconstituted with recombinant mutant forms of FXII and humanized HAEIII mouse models with inducible liver-specific expression of Thr309Lys-mutated FXII exhibited increased contact-driven microvascular leakage. An FXII-neutralizing antibody abolished bradykinin generation in HAEIII patient plasma and blunted edema in HAEIII mice. Together, the results of this study characterize the mechanism of HAEIII and establish FXII inhibition as a potential therapeutic strategy to interfere with excessive vascular leakage in HAEIII and potentially alleviate edema due to other causes.


Subject(s)
Blood Coagulation , Factor XII/metabolism , Hereditary Angioedema Type III/metabolism , Mutation, Missense , Adult , Amino Acid Substitution , Animals , Antibodies, Neutralizing/pharmacology , Bradykinin/genetics , Bradykinin/metabolism , Disease Models, Animal , Factor XII/genetics , Female , Glycosylation/drug effects , Hereditary Angioedema Type III/drug therapy , Hereditary Angioedema Type III/genetics , Hereditary Angioedema Type III/pathology , Humans , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...