scholarly journals Contact System Activation in Plasma from Dengue Patients Might Harness Endothelial Virus Replication through the Signaling of Bradykinin Receptors

2021 ◽  
Vol 14 (1) ◽  
pp. 56
Author(s):  
Sharton V. A. Coelho ◽  
Naiara M. Rust ◽  
Lucas Vellasco ◽  
Michelle P. Papa ◽  
Aline S. G. Pereira ◽  
...  

Since exacerbated inflammation and microvascular leakage are hallmarks of dengue virus (DENV) infection, here we interrogated whether systemic activation of the contact/kallikrein-kinin system (KKS) might hamper endothelial function. In vitro assays showed that dextran sulfate, a potent contact activator, failed to generate appreciable levels of activated plasma kallikrein (PKa) in the large majority of samples from a dengue cohort (n = 70), irrespective of severity of clinical symptoms. Impaired formation of PKa in dengue-plasmas correlated with the presence of cleaved Factor XII and high molecular weight kininogen (HK), suggesting that the prothrombogenic contact system is frequently triggered during the course of infection. Using two pathogenic arboviruses, DENV or Zika virus (ZIKV), we then asked whether exogenous BK could influence the outcome of infection of human brain microvascular endothelial cells (HBMECs). Unlike the unresponsive phenotype of Zika-infected HBMECs, we found that BK, acting via B2R, vigorously stimulated DENV-2 replication by reverting nitric oxide-driven apoptosis of endothelial cells. Using the mouse model of cerebral dengue infection, we next demonstrated that B2R targeting by icatibant decreased viral load in brain tissues. In summary, our study suggests that contact/KKS activation followed by BK-induced enhancement of DENV replication in the endothelium may underlie microvascular pathology in dengue.

Blood ◽  
2018 ◽  
Vol 131 (17) ◽  
pp. 1903-1909 ◽  
Author(s):  
Coen Maas ◽  
Thomas Renné

Abstract Combinations of proinflammatory and procoagulant reactions are the unifying principle for a variety of disorders affecting the cardiovascular system. The factor XII–driven contact system starts coagulation and inflammatory mechanisms via the intrinsic pathway of coagulation and the bradykinin-producing kallikrein-kinin system, respectively. The biochemistry of the contact system in vitro is well understood; however, its in vivo functions are just beginning to emerge. Challenging the concept of the coagulation balance, targeting factor XII or its activator polyphosphate, provides protection from thromboembolic diseases without interfering with hemostasis. This suggests that the polyphosphate/factor XII axis contributes to thrombus formation while being dispensable for hemostatic processes. In contrast to deficiency in factor XII providing safe thromboprotection, excessive FXII activity is associated with the life-threatening inflammatory disorder hereditary angioedema. The current review summarizes recent findings of the polyphosphate/factor XII–driven contact system at the intersection of procoagulant and proinflammatory disease states. Elucidating the contact system offers the exciting opportunity to develop strategies for safe interference with both thrombotic and inflammatory disorders.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. SCI-1-SCI-1
Author(s):  
Thomas Renné

Abstract Combinations of proinflammatory and procoagulant reactions are the unifying principle for a variety of disorders affecting the cardiovascular system. Factor XII (FXII, Hageman factor) is a plasma protease that initiates the contact system. This system starts a cascade of procoagulant and proinflammatory reactions via the intrinsic pathway of coagulation, and the bradykinin producing kallikrein-kinin system, respectively. The biochemistry of the contact system in vitro is well understood, however its in vivo functions are just beginning to emerge. This presentation will summarize roles of the FXII-driven contact system in vivo. Genetically altered mice and large animal models have shown that FXII is essential for thrombus formation while being dispensable for hemostatic processes that terminate blood loss. Challenging the dogma of a coagulation balance, targeting FXII protected from cerebral ischemia without interfering with hemostasis. In contrast, excess FXII activity is associated with a life threatening inflammatory disorder, hereditary angioedema. Platelet polyphosphate (an inorganic polymer), neutrophil extracellular traps (NETs) and mast cell heparin activate FXII with implications on the initiation of thrombosis and edema. A key aspect of the talk will be the analysis of common principles, interactions and cross-talk between coagulation and inflammation, and the use of the novel FXII blocking antibody 3F7 in cardiopulmonary bypass system. Elucidating the FXII-driven contact system offers the exciting opportunity to develop strategies for safe interference with both thrombotic and inflammatory diseases. Disclosures No relevant conflicts of interest to declare.


Author(s):  
R L Bick ◽  
R L Stedman ◽  
P L Kronick ◽  
E Hillman ◽  
J Fareed

Brown pigments from cured tobacco leaf, cigarette smoke condensates and saline extracts of smoke have been referred to as tobacco glycoprotein (TGP) and have been reported to produce allergic skin reactions, induce the formation of IgE antibodies, and to activate clotting factor XII and subsequently activate the fibrinolytic and complement system. These sequential pathological events have been claimed to represent the pathophysiological link between cigarette smoking and cardiopulmonary disease which is observed statisl- tically. To more carefully investigate these claims TGP was extracted by the previously published method of Becker et. al. (Proc. Natl. Acad. Sci. 73:1712-1716, 1976) and the resultant material subjected to in vitro assays related to the claimed biological activities of TGP. It was found that TGP cannot be isolated following the above procedure; the final product obtained by the isolation procedure contains a contaminant introduced by PAGE separation. The contaminant (CMT) appears to be a linear polyacrylate. When this isolated material was subjected to clotting system assays it was found to complex with factor XII, depleting about 15% of factor XII from plasma and,thus, giving the superficial appearance of activating factor XII however, no such activation occurred as noted by non-shortening of the non-activated PTT system, and coagulation factor assays. CMT was found to have no activity on the complement, fibrinolytic, or kinin system. Acrylates are known allergens and thrombus-inducing agents, and these studies suggest that CMT, instead of TGP, has been isolated from cigarette derivitives by the above reported method and CMT rather than TGP has many of the biological activities previously, and perhaps mistakenly, ascribed to TGP.


Endocrinology ◽  
2010 ◽  
Vol 151 (12) ◽  
pp. 5927-5934 ◽  
Author(s):  
Thayalini Ramaesh ◽  
James J. Logie ◽  
Antonia K. Roseweir ◽  
Robert P. Millar ◽  
Brian R. Walker ◽  
...  

Recent studies suggest that kisspeptin (a neuropeptide central to the regulation of gonadotrophin secretion) has diverse roles in human physiology, including a putative role in implantation and placental function. Kisspeptin and its receptor are present in human blood vessels, where they mediate vasoconstriction, and kisspeptin is known to inhibit tumor metastasis and trophoblast invasion, both processes involving angiogenesis. We hypothesized that kisspeptin contributes to the regulation of angiogenesis in the reproductive system. The presence of the kisspeptin receptor was confirmed in human placental blood vessels and human umbilical vein endothelial cells (HUVEC) using immunochemistry. The ability of kisspeptin-10 (KP-10) (a shorter biologically active processed peptide) to inhibit angiogenesis was tested in explanted human placental arteries and HUVEC using complementary ex vivo and in vitro assays. KP-10 inhibited new vessel sprouting from placental arteries embedded in Matrigel and tube-like structure formation by HUVEC, in a concentration-dependent manner. KP-10 had no effect on HUVEC viability or apoptosis but induced concentration-dependent inhibition of proliferation and migration. In conclusion, KP-10 has antiangiogenic effects and, given its high expression in the placenta, may contribute to the regulation of angiogenesis in this tissue.


2014 ◽  
Vol 112 (11) ◽  
pp. 868-875 ◽  
Author(s):  
Jenny Björkqvist ◽  
Katrin Nickel ◽  
Evi Stavrou ◽  
Thomas Renné

SummaryCombinations of proinflammatory and procoagulant reactions are the unifying principle for a variety of disorders affecting the cardiovascular system. Factor XII (FXII, Hageman factor) is a plasma protease that initiates the contact system. The biochemistry of the contact system in vitro is well understood; however, its in vivo functions are just beginning to emerge. The current review concentrates on activators and functions of the FXII-driven contact system in vivo. Elucidating its physiologic activities offers the exciting opportunity to develop strategies for the safe interference with both thrombotic and inflammatory diseases.


2015 ◽  
Vol 112 (13) ◽  
pp. 4068-4073 ◽  
Author(s):  
Daria Zamolodchikov ◽  
Zu-Lin Chen ◽  
Brooke A. Conti ◽  
Thomas Renné ◽  
Sidney Strickland

Alzheimer’s disease (AD) is characterized by accumulation of the β-amyloid peptide (Aβ), which likely contributes to disease via multiple mechanisms. Increasing evidence implicates inflammation in AD, the origins of which are not completely understood. We investigated whether circulating Aβ could initiate inflammation in AD via the plasma contact activation system. This proteolytic cascade is triggered by the activation of the plasma protein factor XII (FXII) and leads to kallikrein-mediated cleavage of high molecular-weight kininogen (HK) and release of proinflammatory bradykinin. Aβ has been shown to promote FXII-dependent cleavage of HK in vitro. In addition, increased cleavage of HK has been found in the cerebrospinal fluid of patients with AD. Here, we show increased activation of FXII, kallikrein activity, and HK cleavage in AD patient plasma. Increased contact system activation is also observed in AD mouse model plasma and in plasma from wild-type mice i.v. injected with Aβ42. Our results demonstrate that Aβ42-mediated contact system activation can occur in the AD circulation and suggest new pathogenic mechanisms, diagnostic tests, and therapies for AD.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1066-1066 ◽  
Author(s):  
Daniel J Sexton ◽  
Jon A Kenniston ◽  
Ryan R Faucette ◽  
Andrew E Nixon ◽  
Chris TenHoor ◽  
...  

Abstract Dysregulated plasma kallikrein proteolytic activity leads to edematous attacks in hereditary angioedema (HAE) and has been associated with inflammation and thrombosis. Plasma kallikrein (pKal) is a serine protease that circulates as prekallikrein, a zymogen, which, together with factor XII (FXII) and high molecular weight kininogen (HMWK), constitutes the contact system. Activation of the contact system following assembly of FXII, HMWK, and prekallikrein on a negatively charged surface promotes inflammation via the generation of bradykinin and triggers intrinsic pathway coagulation via formation of activated coagulation factor XIa. Normal hemostasis appears not to be mediated by the contact system as individuals deficient in contact system proteins are not at risk for bleeding. However, the contact system has been shown to be pathologically activated by agents that include misfolded proteins, platelet polyphosphate, and implanted devices. Therefore, pharmacologic modulation of the contact system may attenuate thrombosis and inflammation without disrupting normal hemostasis. C1 inhibitor (C1-INH) is a serpin and a key endogenous, protein-based, inhibitor of pKal activity. HAE is caused by autosomal dominant mutations in the C1-INH gene resulting in functional protein levels that are approximately 30% or less than normal (16-33 mg/dL or 1.6-3.3 μM). Prekallikrein is present in plasma at a concentration of approximately 500 nM and it has been estimated that only 30-110 nM is converted to active pKal during an HAE attack. This study investigates the requirement for super-stoichiometric amounts of endogenous C1-INH to adequately regulate pKal activity. In vitro enzyme inhibition kinetics experiments with purified proteins show that the need for high concentrations of C1-INH is likely due to its relatively slow association rate constant (1.7 x 104 M-1s-1). In contrast, DX-2930, a human monoclonal antibody inhibitor of pKal being developed for prophylactic treatment of HAE, potently inhibited pKal (Ki = 125 pM) with a faster association rate constant (3.4 x 106 M-1s-1). Contact activation was observed in human plasma activated by the addition of ellagic acid and monitored using a pKal-selective synthetic peptide substrate. Consistent with the data obtained using purified proteins, the apparent IC50 observed upon adding exogenous C1-INH to normal human plasma was approximately 100-fold higher than that of DX-2930. Using a Western blot assay to monitor endogenous HMWK cleavage in activated plasma we similarly observed that stoichiometric additions of DX-2930 were sufficient to prevent HMWK proteolysis by active pKal; whereas significantly higher concentrations of C1-INH (e.g. 1 µM) were required to block HMWK proteolysis. Active pKal can bind endothelial cells via interactions between the non-catalytic domain of pKal with HMWK, which binds receptors (urokinase receptor, cytokeratin 1, and the globular C1q receptor) present on endothelial cells. Cell bound pKal is likely to be a physiologically relevant form of the enzyme and may provide an explanation for attack localization in HAE. In this study, active pKal was assembled in vitro on cultured human umbilical vein endothelial cells (HUVEC) and binding of a range of concentrations of either biotinylated C1-INH or biotinylated DX-2930 was observed using streptavidin-horseradish peroxidase as detection. The data obtained demonstrates that C1-INH bound cell-associated pKal with > 200-fold less potency than DX-2930. Regarding protease inhibition specificity, while DX-2930 did not inhibit any of 20 tested serine proteases at a concentration of 1 µM, C1-INH is known to inhibit multiple serine proteases. This study demonstrates that effective regulation of pKal activity requires high concentrations of C1-INH (≥ 1 µM), which are necessary to drive the kinetics of this second order, irreversible interaction. These high inhibitory concentrations of C1-INH match the normal range and provide a potential kinetic mechanism for why HAE attacks can occur at C1-INH levels that exceed expected levels of activated pKal. Furthermore, the broad specificity of C1-INH towards other proteases that could be activated during disease could sufficiently deplete C1-INH levels and thereby lead to dysregulated pKal activity. Disclosures: Sexton: Dyax Corp: Employment. Kenniston:Dyax Corp: Employment. Faucette:Dyax Corp: Employment. Nixon:Dyax Corp: Employment. TenHoor:Dyax Corp: Employment. Chyung:Dyax Corp: Employment. Adelman:Dyax Corp: Employment.


2012 ◽  
Vol 57 (1) ◽  
pp. 15-25 ◽  
Author(s):  
Chelsea M. Byrd ◽  
Dongcheng Dai ◽  
Douglas W. Grosenbach ◽  
Aklile Berhanu ◽  
Kevin F. Jones ◽  
...  

ABSTRACTDengue viruses (DENV) infect 50 to 100 million people worldwide per year, of which 500,000 develop severe life-threatening disease. This mosquito-borne illness is endemic in most tropical and subtropical countries and has spread significantly over the last decade. While there are several promising vaccine candidates in clinical trials, there are currently no approved vaccines or therapeutics available for treatment of dengue infection. Here, we describe a novel small-molecule compound, ST-148, that is a potent inhibitor of all four serotypes of DENVin vitro. ST-148 significantly reduced viremia and viral load in vital organs and tended to lower cytokine levels in the plasma in a nonlethal model of DENV infection in AG129 mice. Compound resistance mapped to the DENV capsid (C) gene, and a direct interaction of ST-148 with C protein is suggested by alterations of the intrinsic fluorescence of the protein in the presence of compound. Thus, ST-148 appears to interact with the DENV C protein and inhibits a distinct step(s) of the viral replication cycle.


Blood ◽  
2012 ◽  
Vol 120 (10) ◽  
pp. 2133-2143 ◽  
Author(s):  
Roxane Darbousset ◽  
Grace M. Thomas ◽  
Soraya Mezouar ◽  
Corinne Frère ◽  
Rénaté Bonier ◽  
...  

AbstractFor a long time, blood coagulation and innate immunity have been viewed as interrelated responses. Recently, the presence of leukocytes at the sites of vessel injury has been described. Here we analyzed interaction of neutrophils, monocytes, and platelets in thrombus formation after a laser-induced injury in vivo. Neutrophils immediately adhered to injured vessels, preceding platelets, by binding to the activated endothelium via leukocyte function antigen-1–ICAM-1 interactions. Monocytes rolled on a thrombus 3 to 5 minutes postinjury. The kinetics of thrombus formation and fibrin generation were drastically reduced in low tissue factor (TF) mice whereas the absence of factor XII had no effect. In vitro, TF was detected in neutrophils. In vivo, the inhibition of neutrophil binding to the vessel wall reduced the presence of TF and diminished the generation of fibrin and platelet accumulation. Injection of wild-type neutrophils into low TF mice partially restored the activation of the blood coagulation cascade and accumulation of platelets. Our results show that the interaction of neutrophils with endothelial cells is a critical step preceding platelet accumulation for initiating arterial thrombosis in injured vessels. Targeting neutrophils interacting with endothelial cells may constitute an efficient strategy to reduce thrombosis.


2020 ◽  
Vol 161 (50) ◽  
pp. 2099-2103
Author(s):  
Sándor Sipka ◽  
Attila Tóth ◽  
Sándor Sipka jr.

Összefoglaló. Bevezetés: Egy új, számítógép által segített betegminta-asszociációs analízis eredménye szerint a COVID–19 tüneteinek kialakításában kiemelt tényezőként jelenik meg a bradikinin. Eszerint a bradikinin lebontása lelassul az angiotenzinkonvertáló enzim aktivitásának csökkenése miatt, ami jelentősen megemelkedő bradikininszinthez vezet a tüdőben. Nem merült fel azonban a véralvadási faktorok lehetséges szerepe a „bradikininviharban”, annak ellenére, hogy az idősebb cardiovascularis betegekben aktiválódó XII-es faktor és a C1-észteráz-inhibitor (C1INH) alacsony szintje nagy mennyiségű bradikinin képződéséhez vezethet. Módszer: Átfogó irodalmi áttekintés. Eredmények: 1) A vírus által fertőzött, sérült endotheliumsejtek felülete az a hely, amellyel érintkezve elindulhat a XII-es véralvadási faktor aktivációja – ez serkenti a prekallikrein/kallikrein/kinin rendszert, és bradikininképződést okoz. Ez a folyamat megtörténik a súlyos vese- és tüdőkárosodást okozó hantavírus-fertőzésekben. 2) Idős betegekben az atherosclerosis miatt többszörösen sérült, merev, „stiff” erek endotheliumfelszínein jóval magasabb lehet a XII-es faktor kontakt úton történő aktivációja, mint a fiatal egyének ereiben. Ez a tény egyik oka lehet az idős, cardiovascularis betegek körében tapasztalt magasabb halálozásnak. Következtetés: Az aktivált XII-es véralvadási faktor célzott gátlása újabb gyógyítási lehetőség lehet a SARS-CoV-2-fertőzött idős betegekben. Jelenleg már hatásosnak bizonyult a bradikininképzést gátló C1INH-nak, továbbá a bradikininreceptor-gátlóknak az adása is. Orv Hetil. 2020; 161(50): 2099–2103. Summary. Introduction: Bradykinin was implicated in a new complex model of pathomechanism leading to the symptoms of COVID-19 created by a computer-assisted association analysis. According to this model, the decrease in angiotensin-converting enzyme expression leads to impaired bradykinin elimination and subsequent enrichment in the lungs. However, there is no mentioning of the importance of blood coagulation factor XII in increased bradykinin production, in spite of its age-dependent activation and the lower level of C1-esterase inhibitor (C1INH). Activated factor XII may be an important contributor to the “bradykinin storm” in elder cardiovascular patients. Method: Literature review. Results: 1) Activation of the coagulation factor XII on the surface of SARS-CoV-2 infected endothelial cells may trigger the prekallikrein/kallikrein/kinin system producing bradykinin. Such process is taking place in hantavirus infections causing severe lung and kidney damages. 2) The endothelial system is dysregulated in elderly patients, resulting in potentially higher factor XII activities on the surface of damaged endothelial cells in the stiffened arteries. This can contribute to the higher mortality rates in the elderly. Conclusion: The targeted inhibition of activated blood coagulation factor XII may represent a new therapeutic target for COVID-19, especially for elder patients. Recently, beneficial results have already been observed by the clinical applications of recombinant C1INH and bradykinin receptor antagonists. Orv Hetil. 2020; 161(50): 2099–2103.


Sign in / Sign up

Export Citation Format

Share Document