Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
2.
Biol Chem ; 401(8): 901-902, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32436856
3.
FASEB J ; 33(2): 2599-2609, 2019 02.
Article in English | MEDLINE | ID: mdl-30281335

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 B2 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., Renné, T., Vieira, M. L., Müller-Esterl, W., Herwald, H., Lindbom, L. Neutrophils engage the kallikrein-kinin system to open up the endothelial barrier in acute inflammation.


Subject(s)
Cell Membrane Permeability , Endothelium, Vascular/physiology , Inflammation/pathology , Kallikrein-Kinin System/physiology , Neutrophils/metabolism , Pulmonary Edema/pathology , Animals , Bradykinin/metabolism , Endothelium, Vascular/cytology , Factor XII/metabolism , Female , Humans , Inflammation/metabolism , Kininogen, High-Molecular-Weight/metabolism , Male , Mesocricetus , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Pulmonary Edema/etiology , Pulmonary Edema/metabolism
4.
FASEB J ; 33(2): p. 2599-2609, 2019.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib15819

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.

5.
FASEB J, v. 33, n. 2, p. 2599-2609, jan. 2019
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-2666

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.

6.
PLoS One ; 9(12): e116150, 2014.
Article in English | MEDLINE | ID: mdl-25546391

ABSTRACT

Holoprosencephaly is a common developmental disorder in humans characterised by incomplete brain hemisphere separation and midface anomalies. The etiology of holoprosencephaly is heterogeneous with environmental and genetic causes, but for a majority of holoprosencephaly cases the genes associated with the pathogenesis could not be identified so far. Here we report the generation of knockout mice for the ubiquitin E3 ligase NOSIP. The loss of NOSIP in mice causes holoprosencephaly and facial anomalies including cleft lip/palate, cyclopia and facial midline clefting. By a mass spectrometry based protein interaction screen we identified NOSIP as a novel interaction partner of protein phosphatase PP2A. NOSIP mediates the monoubiquitination of the PP2A catalytic subunit and the loss of NOSIP results in an increase in PP2A activity in craniofacial tissue in NOSIP knockout mice. We conclude, that NOSIP is a critical modulator of brain and craniofacial development in mice and a candidate gene for holoprosencephaly in humans.


Subject(s)
Face/embryology , Protein Phosphatase 2/metabolism , Skull/embryology , Skull/enzymology , Ubiquitin-Protein Ligases/metabolism , Animals , Animals, Newborn , Catalytic Domain , Cleft Palate/embryology , Cleft Palate/enzymology , Face/abnormalities , Holoprosencephaly/embryology , Holoprosencephaly/enzymology , Holoprosencephaly/pathology , Methylation , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Skull/abnormalities , Ubiquitination
7.
EMBO J ; 31(15): 3309-22, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22751148

ABSTRACT

F-BAR proteins are multivalent adaptors that link plasma membrane and cytoskeleton and coordinate cellular processes such as membrane protrusion and migration. Yet, little is known about the function of F-BAR proteins in vivo. Here we report, that the F-BAR protein NOSTRIN is necessary for proper vascular development in zebrafish and postnatal retinal angiogenesis in mice. The loss of NOSTRIN impacts on the migration of endothelial tip cells and leads to a reduction of tip cell filopodia number and length. NOSTRIN forms a complex with the GTPase Rac1 and its exchange factor Sos1 and overexpression of NOSTRIN in cells induces Rac1 activation. Furthermore, NOSTRIN is required for fibroblast growth factor 2 dependent activation of Rac1 in primary endothelial cells and the angiogenic response to fibroblast growth factor 2 in the in vivo matrigel plug assay. We propose a novel regulatory circuit, in which NOSTRIN assembles a signalling complex containing FGFR1, Rac1 and Sos1 thereby facilitating the activation of Rac1 in endothelial cells during developmental angiogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Blood Vessels/embryology , DNA-Binding Proteins/physiology , Fibroblast Growth Factors/metabolism , Neovascularization, Physiologic/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Genetically Modified , Animals, Newborn , Blood Vessels/growth & development , Blood Vessels/physiology , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Embryo, Mammalian , Embryo, Nonmammalian , Fibroblast Growth Factors/physiology , Mice , Mice, Knockout , Models, Biological , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 1/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Zebrafish/embryology , Zebrafish/genetics
8.
PLoS One ; 7(5): e37485, 2012.
Article in English | MEDLINE | ID: mdl-22629405

ABSTRACT

Treatment of malignant glioma with chemotherapy is limited mostly because of delivery impediment related to the blood-brain tumor barrier (BTB). B1 receptors (B1R), inducible prototypical G-protein coupled receptors (GPCR) can regulate permeability of vessels including possibly that of brain tumors. Here, we determine the extent of BTB permeability induced by the natural and synthetic peptide B1R agonists, LysdesArg(9)BK (LDBK) and SarLys[dPhe(8)]desArg(9)BK (NG29), in syngeneic F98 glioma-implanted Fischer rats. Ten days after tumor inoculation, we detected the presence of B1R on tumor cells and associated vasculature. NG29 infusion increased brain distribution volume and uptake profiles of paramagnetic probes (Magnevist and Gadomer) at tumoral sites (T(1)-weighted imaging). These effects were blocked by B1R antagonist and non-selective cyclooxygenase inhibitors, but not by B2R antagonist and non-selective nitric oxide synthase inhibitors. Consistent with MRI data, systemic co-administration of NG29 improved brain tumor delivery of Carboplatin chemotherapy (ICP-Mass spectrometry). We also detected elevated B1R expression in clinical samples of high-grade glioma. Our results documented a novel GPCR-signaling mechanism for promoting transient BTB disruption, involving activation of B1R and ensuing production of COX metabolites. They also underlined the potential value of synthetic biostable B1R agonists as selective BTB modulators for local delivery of different sized-therapeutics at (peri)tumoral sites.


Subject(s)
Bradykinin/analogs & derivatives , Brain Neoplasms/metabolism , Glioma/metabolism , Kallidin/analogs & derivatives , Receptor, Bradykinin B1/agonists , Adult , Aged , Animals , Biological Transport/drug effects , Bradykinin/pharmacology , Brain Neoplasms/blood supply , Brain Neoplasms/drug therapy , Capillary Permeability/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Glioma/blood supply , Glioma/drug therapy , Humans , Kallidin/pharmacology , Male , Middle Aged , Permeability/drug effects , Rats , Rats, Inbred F344 , Receptor, Bradykinin B1/metabolism
9.
Immunity ; 34(2): 258-68, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21349432

ABSTRACT

Activated mast cells trigger edema in allergic and inflammatory disease. We report a paracrine mechanism by which mast cell-released heparin increases vascular permeability in vivo. Heparin activated the protease factor XII, which initiates bradykinin formation in plasma. Targeting factor XII or kinin B2 receptors abolished heparin-triggered leukocyte-endothelium adhesion and interfered with a mast cell-driven drop in blood pressure in rodents. Intravital laser scanning microscopy and tracer measurements showed heparin-driven fluid extravasation in mouse skin microvessels. Ablation of factor XII or kinin B2 receptors abolished heparin-induced skin edema and protected mice from allergen-activated mast cell-driven leakage. In contrast, heparin and activated mast cells induced excessive edema in mice deficient in the major inhibitor of factor XII, C1 esterase inhibitor. Allergen exposure triggered edema attacks in hereditary angioedema patients, lacking C1 esterase inhibitor. The data indicate that heparin-initiated bradykinin formation plays a fundamental role in mast cell-mediated diseases.


Subject(s)
Bradykinin/biosynthesis , Capillary Leak Syndrome/physiopathology , Capillary Permeability/physiology , Heparin/physiology , Mast Cells/metabolism , Passive Cutaneous Anaphylaxis/physiology , Animals , Bradykinin/genetics , Capillary Leak Syndrome/etiology , Cell Adhesion , Complement C1 Inhibitor Protein/physiology , Edema/etiology , Edema/physiopathology , Endothelial Cells/pathology , Enzyme Activation , Factor XII/physiology , Heparin/metabolism , Hypotension/etiology , Hypotension/physiopathology , Immunoglobulin E/immunology , Kallikrein-Kinin System/physiology , Leukocytes/physiology , Male , Mice , Paracrine Communication/physiology , Plasma , Rats , Signal Transduction/physiology , Skin/blood supply
10.
Microbiology (Reading) ; 156(Pt 12): 3660-3668, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20705662

ABSTRACT

Since the late 1980s, a worldwide increase of severe Streptococcus pyogenes infections has been associated with strains of the M1 serotype, strains which all secrete the streptococcal inhibitor of complement-mediated lysis (SIC). Previous work has shown that SIC blocks complement-mediated haemolysis, inhibits the activity of antibacterial peptides and has affinity for the human plasma proteins clusterin and histidine-rich glycoprotein; the latter is a member of the cystatin protein family. The present work demonstrates that SIC binds to cystatin C, high-molecular-mass kininogen (HK) and low-molecular-mass kininogen, which are additional members of this protein family. The binding sites in HK are located in the cystatin-like domain D3 and the endothelial cell-binding domain D5. Immobilization of HK to cellular structures plays a central role in activation of the human contact system. SIC was found to inhibit the binding of HK to endothelial cells, and to reduce contact activation as measured by prolonged blood clotting time and impaired release of bradykinin. These results suggest that SIC modifies host defence systems, which may contribute to the virulence of S. pyogenes strains of the M1 serotype.


Subject(s)
Bacterial Proteins/immunology , Streptococcal Infections/microbiology , Streptococcus pyogenes/immunology , Streptococcus pyogenes/pathogenicity , Bacterial Proteins/genetics , Complement System Proteins , Cystatin C/immunology , Host-Pathogen Interactions , Humans , Kininogen, High-Molecular-Weight/immunology , Protein Binding , Streptococcal Infections/immunology , Streptococcus pyogenes/genetics , Virulence
11.
J Cereb Blood Flow Metab ; 30(1): 130-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19773800

ABSTRACT

Inflammatory mechanisms are known to contribute to the pathophysiology of traumatic brain injury (TBI). Since bradykinin is one of the first mediators activated during inflammation, we investigated the role of bradykinin and its receptors in posttraumatic secondary brain damage. We subjected wild-type (WT), B(1)-, and B(2)-receptor-knockout mice to controlled cortical impact (CCI) and analyzed tissue bradykinin as well as kinin receptor mRNA and protein expression up to 48 h thereafter. Brain edema, contusion volume, and functional outcome were assessed 24 h and 7 days after CCI. Tissue bradykinin was maximally increased 2 h after trauma (P<0.01 versus sham). Kinin B(1) receptor mRNA was upregulated up to four-fold 24 h after CCI. Immunohistochemistry showed that B(1) and B(2) receptors were expressed in the brain and were significantly upregulated in the traumatic penumbra 1 to 24 h after CCI. B(2)R(-/-) mice had significantly less brain edema (-51% versus WT, 24 h; P<0.001), smaller contusion volumes ( approximately 50% versus WT 24 h and 7 d after CCI; P<0.05), and better functional outcome 7 days after TBI as compared with WT mice (P<0.05). The present results show that bradykinin and its B(2) receptors play a causal role for brain edema formation and cell death after TBI.


Subject(s)
Brain Injuries/pathology , Receptor, Bradykinin B1/physiology , Receptor, Bradykinin B2/physiology , Animals , Bradykinin/metabolism , Brain Edema/pathology , Contusions/pathology , Immunohistochemistry , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Bradykinin B1/biosynthesis , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B2/biosynthesis , Receptor, Bradykinin B2/genetics , Reverse Transcriptase Polymerase Chain Reaction
12.
Nat Cell Biol ; 11(7): 873-80, 2009 07.
Article in English | MEDLINE | ID: mdl-19503073

ABSTRACT

Epidermal growth factor-like domain 7 (EGFL7) is a secreted factor implicated in cellular responses such as cell migration and blood vessel formation; however the molecular mechanisms underlying the effects of EGFL7 are largely unknown. Here we have identified transmembrane receptors of the Notch family as EGFL7-binding molecules. Secreted EGFL7 binds to a region in Notch involved in ligand-mediated receptor activation, thus acting as an antagonist of Notch signalling. Expression of EGFL7 in neural stem cells (NSCs) in vitro decreased Notch-specific signalling and consequently, reduced proliferation and self-renewal of NSCs. Such altered Notch signalling caused a shift in the differentiation pattern of cultured NSCs towards an excess of neurons and oligodendrocytes. We identified neurons as a source of EGFL7 in the brain, suggesting that brain-derived EGFL7 acts as an endogenous antagonist of Notch signalling that regulates proliferation and differentiation of subventricular zone-derived adult NSCs.


Subject(s)
Endothelial Growth Factors/physiology , Neurons/cytology , Neurons/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Calcium-Binding Proteins , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cell Proliferation , Cells, Cultured , EGF Family of Proteins , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Humans , Mice , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Two-Hybrid System Techniques
13.
J Immunol ; 182(12): 7906-15, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494315

ABSTRACT

The kinin-forming pathway is activated on endothelial cells and neutrophils when high-molecular weight kininogen (HK) is cleaved by plasma kallikrein liberating bradykinin, a potent mediator of inflammation. Kinins are released during inflammatory conditions such as vasculitis, associated with neutrophil influx around blood vessels. Some patients with vasculitis have elevated plasma levels of neutrophil-derived proteinase 3 (PR3) and anti-PR3 Abs. This study investigated if neutrophil-derived PR3 could induce activation of the kinin pathway. PR3 incubated with HK, or a synthetic peptide derived from HK, induced breakdown and release of a novel tridecapeptide termed PR3-kinin, NH(2)-MKRPPGFSPFRSS-COOH, consisting of bradykinin with two additional amino acids on each terminus. The reaction was specific and inhibited by anti-PR3 and alpha(1)-antitrypsin. Recombinant wild-type PR3 incubated with HK induced HK breakdown, whereas mutated PR3, lacking enzymatic activity, did not. PR3-kinin bound to and activated human kinin B(1) receptors, but did not bind to B(2) receptors, expressed by transfected HEK293 cells in vitro. In human plasma PR3-kinin was further processed to the B(2) receptor agonist bradykinin. PR3-kinin exerted a hypotensive effect in vivo through both B(1) and B(2) receptors as demonstrated using wild-type and B(1) overexpressing rats as well as wild-type and B(2) receptor knockout mice. Neutrophil extracts from vasculitis patients and healthy controls contained comparable amounts of PR3 and induced HK proteolysis, an effect that was abolished when PR3 was immunoadsorbed. Neutrophil-derived PR3 can proteolyze HK and liberate PR3-kinin, thereby initiating kallikrein-independent activation of the kinin pathway.


Subject(s)
Kinins/metabolism , Myeloblastin/metabolism , Neutrophils/enzymology , Adolescent , Aged , Aged, 80 and over , Animals , Bradykinin/blood , Child , Humans , Kallikreins/metabolism , Kininogens/metabolism , Male , Mice , Middle Aged , Myeloblastin/genetics , Protein Binding , Rats
14.
Circ Res ; 105(1): 33-41, 2009 Jul 02.
Article in English | MEDLINE | ID: mdl-19478201

ABSTRACT

Nitric oxide (NO) is an essential vasodilator. In vascular diseases, oxidative stress attenuates NO signaling by both chemical scavenging of free NO and oxidation and downregulation of its major intracellular receptor, the alphabeta heterodimeric heme-containing soluble guanylate cyclase (sGC). Oxidation can also induce loss of the heme of sGC, as well as the responsiveness of sGC to NO. sGC activators such as BAY 58-2667 bind to oxidized/heme-free sGC and reactivate the enzyme to exert disease-specific vasodilation. Here, we show that oxidation-induced downregulation of sGC protein extends to isolated blood vessels. Mechanistically, degradation was triggered through sGC ubiquitination and proteasomal degradation. The heme-binding site ligand BAY 58-2667 prevented sGC ubiquitination and stabilized both alpha and beta subunits. Collectively, our data establish oxidation-ubiquitination of sGC as a modulator of NO/cGMP signaling and point to a new mechanism of action for sGC activating vasodilators by stabilizing their receptor, oxidized/heme-free sGC.


Subject(s)
Guanylate Cyclase/metabolism , Heme/metabolism , Nitric Oxide/pharmacology , Proteasome Endopeptidase Complex/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Vasodilator Agents/pharmacology , Blood Vessels , Cell Line , Cyclic GMP/metabolism , Humans , Oxidation-Reduction , Soluble Guanylyl Cyclase , Ubiquitination
15.
Differentiation ; 77(4): 377-85, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19281786

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 Reaction
16.
J Immunol ; 182(4): 2063-73, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19201859

ABSTRACT

Persistent mitochondrial hyperpolarization (MHP) and enhanced calcium fluxing underlie aberrant T cell activation and death pathway selection in systemic lupus erythematosus. Treatment with rapamycin, which effectively controls disease activity, normalizes CD3/CD28-induced calcium fluxing but fails to influence MHP, suggesting that altered calcium fluxing is downstream or independent of mitochondrial dysfunction. In this article, we show that activity of the mammalian target of rapamycin (mTOR), which is a sensor of the mitochondrial transmembrane potential, is increased in lupus T cells. Activation of mTOR was inducible by NO, a key trigger of MHP, which in turn enhanced the expression of HRES-1/Rab4, a small GTPase that regulates recycling of surface receptors through early endosomes. Expression of HRES-1/Rab4 was increased in CD4(+) lupus T cells, and in accordance with its dominant impact on the endocytic recycling of CD4, it was inversely correlated with diminished CD4 expression. HRES-1/Rab4 overexpression was also inversely correlated with diminished TCRzeta protein levels. Pull-down studies revealed a direct interaction of HRES-1/Rab4 with CD4 and TCRzeta. Importantly, the deficiency of the TCRzeta chain and of Lck and the compensatory up-regulation of FcepsilonRIgamma and Syk, which mediate enhanced calcium fluxing in lupus T cells, were reversed in patients treated with rapamcyin in vivo. Knockdown of HRES-1/Rab4 by small interfering RNA and inhibitors of lysosomal function augmented TCRzeta protein levels in vitro. The results suggest that activation of mTOR causes the loss of TCRzeta in lupus T cells through HRES-1/Rab4-dependent lysosomal degradation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lupus Erythematosus, Systemic/immunology , Protein Kinases/immunology , Receptors, Antigen, T-Cell/immunology , rab4 GTP-Binding Proteins/immunology , Adolescent , Adult , Blotting, Western , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Female , Flow Cytometry , Gene Expression/drug effects , Humans , Immunosuppressive Agents/therapeutic use , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Microscopy, Confocal , Middle Aged , Nitric Oxide/metabolism , Oligonucleotide Array Sequence Analysis , Protein Kinases/metabolism , RNA, Small Interfering , Receptors, Antigen, T-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases , Transfection , rab4 GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/biosynthesis
17.
Traffic ; 10(1): 26-34, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18980613

ABSTRACT

We recently observed that a novel, shortened variant of eNOS trafficking inducer (NOSTRIN) is expressed in cirrhotic liver. This shortened variant (NOSTRINbeta) lacks the first 78 amino acids of full-length NOSTRIN (NOSTRINalpha) and thus a substantial part of its F-BAR domain. In contrast to NOSTRINalpha, NOSTRINbeta mainly localizes to the cell nucleus. In this study, we show that nuclear import of NOSTRINbeta depends on two nuclear localization signals (aa 32-36: KKRK and aa 57-61: KAKKK). Each of the sequences is independently functional, but both are required to sustain nuclear localization of NOSTRINbeta. Export of NOSTRINbeta from the nucleus is facilitated by a CRM1-dependent mechanism relying on the nuclear export sequence LELEKERIQL (aa 135-145). Unlike NOSTRINbeta, the full-length variant NOSTRINalpha was conspicuously absent from the nucleus. This is most likely because of the fact that its N-terminal F-BAR domain, which is truncated in NOSTRINbeta, facilitates association with cellular membranes. NOSTRINbeta directly binds to the 5'-regulatory region of the NOSTRIN gene (bp -200 to -1), and overexpression of NOSTRINbeta strongly decreases transcription of a reporter gene under control of this DNA region. Taken together, our results suggest that nuclear NOSTRINbeta may negatively regulate transcription of the NOSTRIN gene.


Subject(s)
Alternative Splicing/genetics , Gene Expression Regulation/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Transcription, Genetic/genetics , Active Transport, Cell Nucleus , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Cell Line , Cell Nucleus/metabolism , DNA-Binding Proteins , Genes, Reporter/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
18.
J Cell Physiol ; 216(1): 234-44, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18264983

ABSTRACT

Bradykinin (BK) represents a pro-inflammatory mediator that partakes in many inflammatory diseases. The mechanism of action of BK is thought to be primarily mediated by specific cell surface membrane B2 receptors (B2Rs). Some evidence has suggested, however, the existence of an intracellular/nuclear B2R population. Whether these receptors are functional and contribute to BK signaling remains to be determined. In this study, by mean of Western blotting, 3D-confocal microscopy, receptor autoradiography and radioligand binding analysis, we showed that plasma membrane and highly purified nuclei from isolated rat hepatocytes contain specific B2R that bind BK. The results depicting B2R nuclear expression in isolated nuclear organelles were reproduced in situ on hepatic sections by immunogold labeling and transmission electron microscopy. Functional tests on single nuclei, by means of confocal microscopy and the calcium-sensitive probe fluo-4AM, showed that BK induces concentration-dependent transitory mobilization of nucleoplasmic calcium; these responses were blocked by B2R antagonist HOE 140, not by the B1R antagonist R954 and, were also found in wild-type C57/Bl6 mice, but not in B2R-KO mice. In isolated nuclei, BK elicited activation/phosphorylation of Akt, acetylation of histone H3 and ensuing pro-inflammatory iNOS gene induction as determined by Western blot and RT-PCR. ChIP assay confirmed binding of acetylated-histone H3 complexes, but not B2R, to promoter region of iNOS gene suggesting that B2R-mediated gene expression is bridged with accessory downstream effectors. This study discloses a previously undescribed mechanism in BK-induced transcriptional events, via intracrine B2R-mediated signaling, occurring in rat autologous hepatic cells.


Subject(s)
Cell Membrane/metabolism , Cell Nucleus/metabolism , Gene Expression Regulation , Genes, Immediate-Early , Receptor, Bradykinin B2/metabolism , Signal Transduction/physiology , Animals , Bradykinin/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Kininogen, High-Molecular-Weight/genetics , Kininogen, High-Molecular-Weight/metabolism , Kininogen, Low-Molecular-Weight/genetics , Kininogen, Low-Molecular-Weight/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptor, Bradykinin B2/genetics , Transcriptional Activation , Vasodilator Agents/metabolism
19.
Am J Physiol Lung Cell Mol Physiol ; 294(3): L505-15, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18192590

ABSTRACT

Ferritin is a protein principally known for its role in iron storage. We have previously shown that ferritin can bind high-molecular-weight kininogen (HK). Upon proteolytic cleavage by the protease kallikrein, HK releases the proinflammatory peptide bradykinin (BK) and other biologically active products, such as two-chain high-molecular-weight kininogen, HKa. At inflammatory sites, HK is oxidized, which renders it a poor substrate for kallikrein. However, oxidized HK remains a good substrate for elastase and tryptase, thereby providing an alternative cleavage mechanism for HK during inflammation. Here we report that ferritin can retard the cleavage of both native HK and oxidized HK by elastase and tryptase. Initial rates of cleavage were reduced 45-75% in the presence of ferritin. Ferritin is not a substrate for elastase or tryptase and does not interfere with the ability of either protease to digest a synthetic substrate, suggesting that ferritin may impede HK cleavage through direct interaction with HK. Immunoprecipitation and solid phase binding studies reveal that ferritin and HK bind directly with a Kd of 134 nM. To test whether ferritin regulates HK cleavage in vivo, we used THP-1 cells, a human monocyte/macrophage cell line that has been used to model pulmonary inflammatory cells. We observed that ferritin impedes the cleavage of HK by secretory proteases in stimulated macrophages. Furthermore, ferritin, HK, and elastase are all present in or on alveolar macrophages in a mouse model of pulmonary inflammation. Collectively, these results implicate ferritin in the modulation of HK cleavage at sites of inflammation.


Subject(s)
Ferritins/pharmacology , Kininogen, High-Molecular-Weight/metabolism , Pancreatic Elastase/antagonists & inhibitors , Pancreatic Elastase/metabolism , Tryptases/antagonists & inhibitors , Tryptases/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Ferritins/metabolism , Humans , Immunoprecipitation , Inflammation/physiopathology , Macrophages, Alveolar/metabolism , Mice , Oxidation-Reduction , Peroxidase/metabolism , Protein Binding , Succinimides/chemistry , Zinc/pharmacology
20.
PLoS Pathog ; 3(11): e185, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18052532

ABSTRACT

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/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...