ABSTRACT
Neutrophil recruitment and plasma exudation are key elements in the immune response to injury or infection. Activated neutrophils stimulate opening of the endothelial barrier; however, the underlying mechanisms have remained largely unknown. In this study, we identified a pivotal role of the proinflammatory kallikrein-kinin system and consequent formation of bradykinin in neutrophil-evoked vascular leak. In mouse and hamster models of acute inflammation, inhibitors of bradykinin generation, and signaling markedly reduced plasma exudation in response to chemoattractant activation of neutrophils. The neutrophil-driven leak was likewise suppressed in mice deficient in either the bradykinin B-2 receptor or factor XII (initiator of the kallikrein-kinin system). In human endothelial cell monolayers, material secreted from activated neutrophils induced cytoskeletal rearrangement, leading to paracellular gap formation in a bradykinin-dependent manner. As a mechanistic basis, we found that a neutrophil-derived heparin-binding protein (HBP/azurocidin) displaced the bradykinin precursor high-molecular-weight kininogen from endothelial cells, thereby enabling proteolytic processing of kininogen into bradykinin by neutrophil and plasma proteases. These data provide novel insight into the signaling pathway by which neutrophils open up the endothelial barrier and identify the kallikrein-kinin system as a target for therapeutic interventions in acute inflammatory reactions.Kenne, E., Rasmuson, J., Renne, T., Vieira, M. L., Muller-Esterl, W., Herwald, H., Lindbom, L. Neutrophils engage the kallikrein-kinin system to open up the endothelial barrier in acute inflammation.
ABSTRACT
The V(2) vasopressin receptor gene contains an alternative splice site in exon-3, which leads to the generation of two splice variants (V(2a) and V(2b)) first identified in the kidney. The open reading frame of the alternatively spliced V(2b) transcript encodes a truncated receptor, showing the same amino acid sequence as the canonical V(2a) receptor up to the sixth transmembrane segment, but displaying a distinct sequence to the corresponding seventh transmembrane segment and C-terminal domain relative to the V(2a) receptor. Here, we demonstrate the postnatal expression of V(2a) and V(2b) variants in the rat cerebellum. Most importantly, we showed by in situ hybridization and immunocytochemistry that both V(2) splice variants were preferentially expressed in Purkinje cells, from early to late postnatal development. In addition, both variants were transiently expressed in the neuroblastic external granule cells and Bergmann fibers. These results indicate that the cellular distributions of both splice variants are developmentally regulated, and suggest that the transient expression of the V(2) receptor is involved in the mechanisms of cerebellar cytodifferentiation by AVP. Finally, transfected CHO-K1 expressing similar amounts of both V(2) splice variants, as that found in the cerebellum, showed a significant reduction in the surface expression of V(2a) receptors, suggesting that the differential expression of the V(2) splice variants regulates the vasopressin signaling in the cerebellum.
Subject(s)
Cerebellum/metabolism , Gene Expression Regulation, Developmental , Receptors, Vasopressin/metabolism , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Female , Genetic Variation , Immunohistochemistry , In Situ Hybridization , Protein Isoforms/metabolism , Purkinje Cells/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Vasopressin/classification , Receptors, Vasopressin/genetics , Reverse Transcriptase Polymerase Chain ReactionABSTRACT
Although the concept that dendritic cells (DCs) recognize pathogens through the engagement of Toll-like receptors is widely accepted, we recently suggested that immature DCs might sense kinin-releasing strains of Trypanosoma cruzi through the triggering of G-protein-coupled bradykinin B2 receptors (B2R). Here we report that C57BL/6.B2R-/- mice infected intraperitoneally with T. cruzi display higher parasitemia and mortality rates as compared to B2R+/+ mice. qRT-PCR revealed a 5-fold increase in T. cruzi DNA (14 d post-infection [p.i.]) in B2R-/- heart, while spleen parasitism was negligible in both mice strains. Analysis of recall responses (14 d p.i.) showed high and comparable frequencies of IFN-gamma-producing CD4+ and CD8+ T cells in the spleen of B2R-/- and wild-type mice. However, production of IFN-gamma by effector T cells isolated from B2R-/- heart was significantly reduced as compared with wild-type mice. As the infection continued, wild-type mice presented IFN-gamma-producing (CD4+CD44+ and CD8+CD44+) T cells both in the spleen and heart while B2R-/- mice showed negligible frequencies of such activated T cells. Furthermore, the collapse of type-1 immune responses in B2R-/- mice was linked to upregulated secretion of IL-17 and TNF-alpha by antigen-responsive CD4+ T cells. In vitro analysis of tissue culture trypomastigote interaction with splenic CD11c+ DCs indicated that DC maturation (IL-12, CD40, and CD86) is controlled by the kinin/B2R pathway. Further, systemic injection of trypomastigotes induced IL-12 production by CD11c+ DCs isolated from B2R+/+ spleen, but not by DCs from B2R-/- mice. Notably, adoptive transfer of B2R+/+ CD11c+ DCs (intravenously) into B2R-/- mice rendered them resistant to acute challenge, rescued development of type-1 immunity, and repressed TH17 responses. Collectively, our results demonstrate that activation of B2R, a DC sensor of endogenous maturation signals, is critically required for development of acquired resistance to T. cruzi infection.
Subject(s)
Chagas Disease/immunology , Dendritic Cells/immunology , Kinins/metabolism , Receptor, Bradykinin B2/immunology , Th1 Cells/immunology , Adoptive Transfer , Animals , Chagas Disease/metabolism , Dendritic Cells/metabolism , Flow Cytometry , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Interleukin-17/biosynthesis , Kinins/immunology , Lymphocyte Activation/immunology , Mice , Mice, Mutant Strains , Polymerase Chain Reaction , Receptor, Bradykinin B2/metabolism , Th1 Cells/metabolism , Trypanosoma cruzi , Tumor Necrosis Factor-alpha/biosynthesisABSTRACT
We have previously reported that exogenous bradykinin activates immature dendritic cells (DCs) via the bradykinin B(2) receptor (B(2)R), thereby stimulating adaptive immunity. In this study, we show that these premises are met in a model of s.c. infection by Trypanosoma cruzi, a protozoan that liberates kinins from kininogens through its major protease, cruzipain. Intensity of B(2)R-dependent paw edema evoked by trypomastigotes correlated with levels of IL-12 produced by CD11c(+) dendritic cells isolated from draining lymph nodes. The IL-12 response induced by endogenously released kinins was vigorously increased in infected mice pretreated with inhibitors of angiotensin converting enzyme (ACE), a kinin-degrading metallopeptidase. Furthermore, these innate stimulatory effects were linked to B(2)R-dependent up-regulation of IFN-gamma production by Ag-specific T cells. Strikingly, the trypomastigotes failed to up-regulate type 1 immunity in TLR2(-/-) mice, irrespective of ACE inhibitor treatment. Analysis of the dynamics of inflammation revealed that TLR2 triggering by glycosylphosphatidylinositol-anchored mucins induces plasma extravasation, thereby favoring peripheral accumulation of kininogens in sites of infection. Further downstream, the parasites generate high levels of innate kinin signals in peripheral tissues through the activity of cruzipain. The demonstration that the deficient type 1 immune responses of TLR2(-/-) mice are rescued upon s.c. injection of exogenous kininogens, along with trypomastigotes, supports the notion that generation of kinin "danger" signals is intensified through cooperative activation of TLR2 and B(2)R. In summary, we have described a s.c. infection model where type 1 immunity is vigorously up-regulated by bradykinin, an innate signal whose levels in peripheral tissues are controlled by an intricate interplay of TLR2, B(2)R, and ACE.
Subject(s)
Chagas Disease/immunology , Kinins/metabolism , Receptor, Bradykinin B2/agonists , Toll-Like Receptor 2/agonists , Trypanosoma cruzi , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Bradykinin/pharmacology , CD11c Antigen/analysis , Cell Differentiation , Cysteine Endopeptidases/metabolism , Dendritic Cells/chemistry , Dendritic Cells/immunology , Disease Models, Animal , Immunity, Innate , Interleukin-12/metabolism , Kininogens/administration & dosage , Kininogens/metabolism , Mice , Mice, Mutant Strains , Peptidyl-Dipeptidase A/metabolism , Protozoan Proteins , Receptor, Bradykinin B2/genetics , Skin/immunology , Skin/parasitology , Toll-Like Receptor 2/geneticsABSTRACT
In an effort to define the varied expression of three vasoactive markers in the clinical models of normal placenta/ normal invasion (n = 11), preeclampsia/restricted trophoblast invasion (n = 15), and placenta accreta/exaggerated invasion (n = 6), we performed semiquantitative immunohistochemistry for kallikrein, bradykinin B2 receptor, and endothelial nitric oxide synthase (eNOS). In the floating villi, the syncytiotrophoblast expressed more kallikrein in placenta accreta (p < 0.05), than in normal and preeclamptic placentas, while the bradykinin B2 receptor and eNOS were similarly expressed in all groups; in the fetal endothelium, the bradykinin B2 receptor was enhanced in placenta accreta (p < 0.005), but kallikrein and eNOS were similarly expressed in the other two groups. In the extravillous trophoblast, both kallikrein and eNOS expression were higher in placenta accreta (p < 0.001), while the bradykinin B2 receptor signal was only enhanced in preeclampsia (p < 0.05). The presence and localization of kallikrein, the bradykinin B2 receptor, and eNOS in the fetomaternal interface in the three study conditions supports a local role for interrelated vasodilatory/antiaggregating systems. This first report of the variations observed in kallikrein and eNOS in a condition of exaggerated trophoblast invasion supports the participation of vasodilatation in trophoblast migration.
Subject(s)
Kallikreins/metabolism , Nitric Oxide Synthase Type III/metabolism , Placenta Accreta/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Receptor, Bradykinin B2/metabolism , Chorionic Villi/metabolism , Endothelial Cells/metabolism , Female , Fetus/metabolism , Gene Expression , Gestational Age , Humans , Kallikreins/urine , Maternal-Fetal Exchange , Models, Biological , Placenta/blood supply , Pregnancy , Pregnancy Complications, Hematologic/metabolism , Pregnancy Outcome , Trophoblasts/metabolismABSTRACT
Kinins, the vasoactive peptides proteolytically liberated from kininogens, were recently recognized as signals alerting the innate immune system. Here we demonstrate that Leishmania donovani and Leishmania chagasi, two etiological agents of visceral leishmaniasis (VL), activate the kinin system. Intravital microscopy in the hamster cheek pouch showed that topically applied promastigotes induced macromolecular leakage (FITC-dextran) through postcapillary venules. Peaking at 15 min, the parasite-induced leakage was drastically enhanced by captopril (Cap), an inhibitor of angiotensin-converting enzyme (ACE), a kinin-degrading metallopeptidase. The enhanced microvascular responses were cancelled by HOE-140, an antagonist of the B2 bradykinin receptor (B2R), or by pre-treatment of promastigotes with the irreversible cysteine proteinase inhibitor N-methylpiperazine-urea-Phe-homoPhe-vinylsulfone-benzene (N-Pip-hF-VSPh). In agreement with the above-mentioned data, the promastigotes vigorously induced edema in the paw of Cap-treated J129 mice, but not Cap-B2R-/- mice. Analysis of parasite-induced breakdown of high molecular weight kininogens (HK), combined with active site-affinity-labeling with biotin-N-Pip-hF-VSPh, identified 35-40 kDa proteins as kinin-releasing cysteine peptidases. We then checked if macrophage infectivity was influenced by interplay between these kinin-releasing parasite proteases, kininogens, and kinin-degrading peptidases (i.e. ACE). Our studies revealed that full-fledged B2R engagement resulted in vigorous increase of L. chagasi uptake by resident macrophages. Evidence that inflammatory macrophages treated with HOE-140 became highly susceptible to amastigote outgrowth, assessed 72 h after initial macrophage interaction, further suggests that the kinin/B2R activation pathway may critically modulate inflammation and innate immunity in visceral leishmaniasis.
Subject(s)
Capillary Permeability/physiology , Cysteine Endopeptidases/metabolism , Kinins/metabolism , Leishmania donovani/enzymology , Leishmania infantum/enzymology , Macrophages/metabolism , Macrophages/parasitology , Animals , Cricetinae , Gene Deletion , Male , Mice , Mice, Inbred BALB C , Peptidyl-Dipeptidase A/metabolism , Receptors, Bradykinin/genetics , Receptors, Bradykinin/metabolism , Time FactorsABSTRACT
BACKGROUND: Ontogeny and cellular distribution of vasopressin receptors in the kidney are key factors determining the role of vasopressin in renal physiology. Expression of vasopressin V(2) receptor (V(2)R) mRNA and the immunoreactive protein in rat kidney were investigated. METHODS: An antiserum directed to epitope TLD25 of the rat V(2)R sequence was characterized by Western blotting. Expression of V(2)R mRNA was assessed by reverse transcription-polymerase chain reaction (RT-PCR), and on protein level by immunohistochemistry. RESULTS: Specificity of the antiserum was documented by Western blots from cells expressing a fusion protein of V(2)R and GFP. Using lysates of rat kidney and of native cell lines expressing V(2)R but not V(1)R, our antiserum to peptide TLD25 revealed a major band of 55 kD corresponding to the monomeric form of V(2)R, and a band of 110 kD most likely representing the homodimeric form of the receptor. This highly specific antiserum allowed us to localize the V(2)R in thick ascending limbs, distal convoluted and connecting tubules, and in collecting ducts. During ontogeny, immunoreactivity was first observed at the luminal membrane on prenatal day 20, emerging at the basolateral side from postnatal day 5 on. RT-PCR demonstrated V(2)R transcripts from prenatal day 18 to gradually increasing thereafter. CONCLUSION: Expression of V(2)R is first detectable in the late embryonic stage of rat ontogeny starting from day E18 and gradually increasing with kidney maturation. In the adult kidney, V(2)R is differentially distributed in the various nephron segments.
Subject(s)
Nephrons/embryology , Nephrons/physiology , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , Age Factors , Animals , Antibody Specificity , Cell Membrane/metabolism , Female , Gene Expression Regulation, Developmental , Gestational Age , Immunohistochemistry , Kidney Tubules, Collecting/embryology , Kidney Tubules, Collecting/physiology , Kidney Tubules, Distal/embryology , Kidney Tubules, Distal/physiology , Loop of Henle/embryology , Loop of Henle/physiology , Male , Pregnancy , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Vasopressin/immunologyABSTRACT
Trypanosoma cruzi, the protozoan that causes Chagas' heart disease, invades endothelial cells in vitro by activating the B2 kinin receptor (B2R). Here, we demonstrate that mice infected with trypomastigotes develop potent edema after treatment with the angiotensin-converting enzyme (ACE) (or kininase II) inhibitor captopril. Experiments performed with specific kinin receptor (B2R/B1R) antagonists and knockout mice revealed that the early-phase (3-h) edema is mediated by the constitutive B2R, whereas the late-phase (24-h) response depends on stimulation of the up-regulated B1R. Given previous evidence that parasite invasion of cells expressing B2R is potentiated by captopril, we investigated the prerequisites for in vitro infection of Chinese hamster ovary cells overexpressing either B1R or B2R, human umbilical vein endothelial cells activated by lipopolysaccharide, and neonatal rat cardiomyocytes. Our results indicate that captopril potentiates parasite invasion regardless of the kinin (B2/B1) activation pathways, whereas DL-2-mercaptomethyl-3-guanidino-ethylthiopropanoic acid (MGTA), an inhibitor of kininase I (carboxypeptidase M/N), selectively decreases parasite infectivity for B1R-expressing cells. These data suggest that formation of the B1R agonist, i.e., [des-Arg] kinins, critically depends on the processing action of kininase I, here proposed as a potential pathogenesis cofactor. Collectively, our data suggest that fluctuations in the levels of kininases may modulate parasite infectivity and pathological outcome in Chagas' disease.
Subject(s)
Chagas Disease/etiology , Edema/parasitology , Heart/parasitology , Receptors, Bradykinin/metabolism , Trypanosoma cruzi/pathogenicity , Animals , Animals, Newborn , CHO Cells , Cells, Cultured , Chagas Disease/metabolism , Cricetinae , Edema/metabolism , Endothelium, Vascular/parasitology , Extremities/parasitology , Kinetics , Lipopolysaccharides/pharmacology , Lysine Carboxypeptidase/physiology , Mice , Models, Biological , Receptor, Bradykinin B1 , Receptor, Bradykinin B2 , Receptors, Bradykinin/physiology , Trypanosoma cruzi/growth & developmentABSTRACT
Trypanosoma cruzi activates the kinin pathway through the activity of its major cysteine proteinase, cruzipain. Because kininogen molecules may be displayed on cell surfaces by binding to glycosaminoglycans, we examined whether the ability of cruzipain to release kinins from high molecular weight kininogen (HK) is modulated by heparan sulfate (HS). Kinetic assays show that HS reduces the cysteine proteinase inhibitory activity (K(i app)) of HK about 10-fold. Conversely, the catalytic efficiency of cruzipain on kinin-related synthetic fluorogenic substrates is enhanced up to 6-fold in the presence of HS. Analysis of the HK breakdown products generated by cruzipain indicated that HS changes the pattern of HK cleavage products. Direct measurements of bradykinin demonstrated an up to 35-fold increase in cruzipain-mediated kinin liberation in the presence of HS. Similarly, kinin release by living trypomastigotes increased up to 10-fold in the presence of HS. These studies suggest that the efficiency of T. cruzi to initiate kinin release is potently enhanced by the mutual interactions between cruzipain, HK, and heparan sulfate proteoglycans.
Subject(s)
Cysteine Endopeptidases/metabolism , Heparitin Sulfate/pharmacology , Kinins/metabolism , Trypanosoma cruzi/enzymology , Amino Acid Motifs , Amino Acid Sequence , Animals , Enzyme Activation/drug effects , Kinetics , Kininogens/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protozoan Proteins , Substrate Specificity , Trypanosoma cruzi/drug effectsABSTRACT
-Bradykinin, a major vasodilator peptide, plays an important role in the local regulation of blood pressure, blood flow, and vascular permeability; however, the cellular distribution of the major bradykinin B(2) receptor in the cardiovascular system is not precisely known. Immunoblot analysis with an anti-peptide antibody to the bradykinin B(2) receptor or chemical cross-linkage with [(125)I]Tyr(0)-bradykinin revealed a band of 69+/-3 kDa at varying intensity in the homogenates of the endothelium and tunica media of the rat aorta and endocardium. Immunostaining showed that the B(2) receptor is abundant in the endothelial linings of the aorta, other elastic arteries, muscular arteries, capillaries, venules, and large veins, where it localizes preferentially to the luminal face of the endothelial cells. In marked contrast, small arterioles (ie, the principal blood-pressure regulating vessels) of the mesenterium, heart, urinary bladder, brain, salivary gland, and kidney had a different staining pattern in which B(2) receptor was prominent in the perivascular smooth muscle cells of the tunica media. A similar distribution pattern was found in mouse as well as in human tissues, indicating that the particular distribution pattern of the B(2) receptor in arterioles is not a species-specific phenomenon. During development, the distribution of B(2) receptor in the heart changes; for example, in the heart of newborn rats, the B(2) receptor was abundant in the myocardium, whereas in the adult heart, the receptor was present in the endocardium of atria, atrioventricular valves, and ventricles but not in the myocardium. Thus, B(2) receptors are localized differentially in different parts of the cardiovascular system: the arterioles have smooth muscle-localized B(2) receptors, and large elastic vessels have endothelium-localized receptors.