Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Cardiovasc Res ; 1: 1056-1071, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36523570

RESUMEN

Dissecting the pathways regulating the adaptive immune response in atherosclerosis is of particular therapeutic interest. Here we report that the lipid G-protein coupled receptor GPR55 is highly expressed by splenic plasma cells (PC), upregulated in mouse spleens during atherogenesis and human unstable or ruptured compared to stable plaques. Gpr55-deficient mice developed larger atherosclerotic plaques with increased necrotic core size compared to their corresponding controls. Lack of GPR55 hyperactivated B cells, disturbed PC maturation and resulted in immunoglobulin (Ig)G overproduction. B cell-specific Gpr55 depletion or adoptive transfer of Gpr55-deficient B cells was sufficient to promote plaque development and elevated IgG titers. In vitro, the endogenous GPR55 ligand lysophsophatidylinositol (LPI) enhanced PC proliferation, whereas GPR55 antagonism blocked PC maturation and increased their mitochondrial content. Collectively, these discoveries provide previously undefined evidence for GPR55 in B cells as a key modulator of the adaptive immune response in atherosclerosis.

2.
PLoS One ; 17(7): e0260173, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35802710

RESUMEN

For most cell culture experiments, it is indispensable that the cells are firmly anchored to culture plates, withstanding rinsing steps that can create shear forces and tolerating temperature changes without detaching. For semi-adherent cells such as the common HEK 293 or PC-12 cells, this could so far be obtained by time-consuming plate pre-coating with cationic polymer solutions. We report here, that i) pre-coating with the cheaper poly-ethylenimine (PEI) works as well as the commonly used poly-D-lysine (PDL), but more importantly and novel ii) that simple direct addition of either PEI (1.5 µg/ml) or PDL (2 µg/ml) to the cell culture medium results in strongly anchored HEK 293 cells, indistinguishable from ones seeded on pre-coated plates. Therefore, the replacement of plate pre-coating by direct addition of either PEI or PDL gives comparable excellent results, but is highly labour-, time-, and cost-efficient. Moreover, we could show that addition of PDL or PEI also works similarly well in animal-free culture using human platelet lysate instead of fetal bovine serum. Interestingly, additional experiments showed that strong cell attachment requires only cationic polymers but not fetal bovine serum or human platelet lysate added to the medium.


Asunto(s)
Lisina , Polímeros , Aziridinas , Técnicas de Cultivo de Célula/métodos , Células HEK293 , Humanos , Polietileneimina
3.
Int Immunopharmacol ; 105: 108523, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35086057

RESUMEN

PHA-022121 is a novel small molecule bradykinin B2 receptor antagonist, in clinical development for the treatment and prevention of hereditary angioedema attacks. The present study describes the in vitro pharmacological characteristics of PHA-022121 and its active metabolite, PHA-022484 (M2-D). In mammalian cell lines, PHA-022121 and PHA-022484 show high affinity for the recombinant human bradykinin B2 receptor with Ki values of 0.47 and 0.70 nM, respectively, and potent antagonism of the human bradykinin B2 receptor with Kb values of 0.15 and 0.26 nM, respectively (calcium mobilization assay). Antagonist potency at the recombinant cynomolgus monkey bradykinin B2 receptor is similarly high (Kb values of 1.42 and 1.12 nM for PHA-022121 and PHA-022484, respectively), however, potency at rat, mouse, rabbit and dog bradykinin B2 receptors is at least 100-fold lower than the potency at the human receptor for both compounds. In the human umbilical vein contractility assay, both PHA-022121 and PHA-022484 show a potent, surmountable and reversible B2 antagonist activity with pA2 values of 0.35 and 0.47 nM, respectively. The in vitro off-target profile of PHA-022121 and PHA-022484 demonstrates a high degree of selectivity over a wide range of molecular targets, including the bradykinin B1 receptor. It is concluded that PHA-022121 is a novel, low-molecular weight, competitive antagonist of the human bradykinin B2 receptor with high affinity, high antagonist potency, and high selectivity. It is about 20-fold more potent than icatibant at the human bradykinin B2 receptor as assessed using recombinant or endogenously expressed receptors.


Asunto(s)
Antagonistas de los Receptores de Bradiquinina , Bradiquinina , Animales , Unión Competitiva , Bradiquinina/metabolismo , Antagonistas de los Receptores de Bradiquinina/metabolismo , Antagonistas de los Receptores de Bradiquinina/farmacología , Perros , Macaca fascicularis/metabolismo , Mamíferos , Ratones , Conejos , Ratas , Receptor de Bradiquinina B1/metabolismo , Receptor de Bradiquinina B2/metabolismo , Venas Umbilicales/metabolismo
4.
EMBO Rep ; 21(4): e47852, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32080959

RESUMEN

Chemokines and galectins are simultaneously upregulated and mediate leukocyte recruitment during inflammation. Until now, these effector molecules have been considered to function independently. Here, we tested the hypothesis that they form molecular hybrids. By systematically screening chemokines for their ability to bind galectin-1 and galectin-3, we identified several interacting pairs, such as CXCL12 and galectin-3. Based on NMR and MD studies of the CXCL12/galectin-3 heterodimer, we identified contact sites between CXCL12 ß-strand 1 and Gal-3 F-face residues. Mutagenesis of galectin-3 residues involved in heterodimer formation resulted in reduced binding to CXCL12, enabling testing of functional activity comparatively. Galectin-3, but not its mutants, inhibited CXCL12-induced chemotaxis of leukocytes and their recruitment into the mouse peritoneum. Moreover, galectin-3 attenuated CXCL12-stimulated signaling via its receptor CXCR4 in a ternary complex with the chemokine and receptor, consistent with our structural model. This first report of heterodimerization between chemokines and galectins reveals a new type of interaction between inflammatory mediators that can underlie a novel immunoregulatory mechanism in inflammation. Thus, further exploration of the chemokine/galectin interactome is warranted.


Asunto(s)
Galectinas , Inflamación , Animales , Quimiotaxis , Galectinas/genética , Galectinas/metabolismo , Inflamación/genética , Leucocitos/metabolismo , Ratones , Transducción de Señal
6.
Arterioscler Thromb Vasc Biol ; 38(5): 1007-1019, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29567680

RESUMEN

OBJECTIVE: Cardiovascular diseases and depression are the leading causes of disability in Western countries. Clinical data on potential cardiovascular effects of serotonin reuptake inhibitors (SSRIs), the most commonly used antidepressant drugs, are controversial. In addition to blocking serotonin reuptake transporter in the brain, SSRIs deplete the major peripheral serotonin (5-hydroxytryptamine [5-HT]) storage by inhibiting serotonin reuptake transporter-mediated uptake in platelets. In this study, we aimed to investigate the effect of chronic SSRI intake on the development of atherosclerosis. APPROACH AND RESULTS: Treatment of apolipoprotein E-deficient mice with the SSRI fluoxetine for 2, 4, or 16 weeks increased atherosclerotic lesion formation, with most pronounced effect during early plaque development. Intravital microscopy of carotid arteries revealed enhanced myeloid cell adhesion on fluoxetine treatment. Mechanistically, we found that fluoxetine augmented vascular permeability and increased chemokine-induced integrin-binding activity of circulating leukocytes. In vitro stimulation of murine blood demonstrated that fluoxetine, but not 5-HT, could directly promote ß1 and ß2 integrin activation provided C-C motif chemokine ligand 5 was also present. Similar effects were observed with the SSRI escitalopram. Enhanced C-C motif chemokine ligand 5-induced integrin activation by fluoxetine was also confirmed in a human neutrophil-like cell line. In contrast to the proatherogenic properties of fluoxetine, pharmacological inhibition of the peripheral 5-HT synthesizing enzyme tryptophan hydroxylase 1 did not promote atherosclerosis, suggesting that the proatherogenic effect of fluoxetine occurs independent of peripheral 5-HT depletion. CONCLUSIONS: SSRI intake may promote atherosclerosis and therefore potentially increase the risk for acute cardiovascular events by a mechanism that is independent of 5-HT depletion.


Asunto(s)
Aorta/efectos de los fármacos , Enfermedades de la Aorta/inducido químicamente , Aterosclerosis/inducido químicamente , Arterias Carótidas/efectos de los fármacos , Enfermedades de las Arterias Carótidas/inducido químicamente , Fluoxetina/toxicidad , Placa Aterosclerótica , Inhibidores Selectivos de la Recaptación de Serotonina/toxicidad , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/sangre , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/patología , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Antígenos CD18/sangre , Permeabilidad Capilar/efectos de los fármacos , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/sangre , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/patología , Adhesión Celular/efectos de los fármacos , Quimiocina CCL5/sangre , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Esquema de Medicación , Fluoxetina/administración & dosificación , Células HEK293 , Células HL-60 , Humanos , Integrina beta1/sangre , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , Serotonina/sangre , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Transducción de Señal , Factores de Tiempo
7.
Eur Heart J ; 38(48): 3590-3599, 2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29045618

RESUMEN

Aims: The co-stimulatory receptor CD27 modulates responses of T cells, B cells, and NK cells. Various T cell subsets participate in atherogenesis. However, the role of CD27 in atherosclerosis remains unexplored. Methods and results: Here we investigated the effect of bone marrow-derived and systemic CD27 deficiency in Apolipoprotein E-deficient (Apoe-/-) mice in early and advanced stages of atherosclerosis. Lethally-irradiated Apoe-/- mice reconstituted with Cd27-/-Apoe-/- bone marrow and consuming an atherogenic diet displayed a markedly increased plaque size and lesional inflammation compared to mice receiving Cd27+/+Apoe-/- bone marrow. Accordingly, chow diet-fed Cd27-/-Apoe-/- mice showed exacerbated lesion development and increased inflammation at the age of 18 weeks. At a more advanced stage of atherosclerosis (28 weeks), lesion size and phenotype did not differ between the two groups. Systemic and bone marrow-derived CD27 deficiency reduced the abundance of regulatory T cells (Treg) in blood, lymphoid organs, and the aorta. Numbers of other immune cells were not affected while expression of inflammatory cytokine genes (e.g. IL-1ß and IL-6) was increased in the aorta when haematopoietic CD27 was lacking. In vitro, Tregs of CD27-deficient mice showed similar suppressive capacity compared with their wild-type controls and migrated equally towards CCL19 and CCL21. However, thymic Cd27-/- Tregs underwent increased apoptosis and expressed fewer markers of proliferation in vivo. Reconstitution of Cd27-/-Apoe-/- mice with Cd27+/+Apoe-/- Tregs reversed the increase in atherosclerosis. Conclusion: We demonstrate that CD27 co-stimulation increases the number of Tregs and limits lesion development and inflammation in experimental atherosclerosis, particularly during early stages of disease. Thus, our study suggests that promotion of CD27 function may mitigate atherosclerosis.


Asunto(s)
Aterosclerosis/inmunología , Hiperlipidemias/complicaciones , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Animales , Apoptosis , Aterosclerosis/etiología , Aterosclerosis/prevención & control , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Hiperlipidemias/inmunología , Hiperlipidemias/metabolismo , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/inmunología
8.
Sci Transl Med ; 9(384)2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28381538

RESUMEN

Chemokines orchestrate leukocyte trafficking and function in health and disease. Heterophilic interactions between chemokines in a given microenvironment may amplify, inhibit, or modulate their activity; however, a systematic evaluation of the chemokine interactome has not been performed. We used immunoligand blotting and surface plasmon resonance to obtain a comprehensive map of chemokine-chemokine interactions and to confirm their specificity. Structure-function analyses revealed that chemokine activity can be enhanced by CC-type heterodimers but inhibited by CXC-type heterodimers. Functional synergism was achieved through receptor heteromerization induced by CCL5-CCL17 or receptor retention at the cell surface via auxiliary proteoglycan binding of CCL5-CXCL4. In contrast, inhibitory activity relied on conformational changes (in CXCL12), affecting receptor signaling. Obligate CC-type heterodimers showed high efficacy and potency and drove acute lung injury and atherosclerosis, processes abrogated by specific CCL5-derived peptide inhibitors or knock-in of an interaction-deficient CXCL4 variant. Atheroprotective effects of CCL17 deficiency were phenocopied by a CCL5-derived peptide disrupting CCL5-CCL17 heterodimers, whereas a CCL5 α-helix peptide mimicked inhibitory effects on CXCL12-driven platelet aggregation. Thus, formation of specific chemokine heterodimers differentially dictates functional activity and can be exploited for therapeutic targeting.


Asunto(s)
Quimiocinas/metabolismo , Inflamación/metabolismo , Inflamación/patología , Mapeo de Interacción de Proteínas , Enfermedad Aguda , Animales , Plaquetas/metabolismo , Enfermedad Crónica , Modelos Animales de Enfermedad , Espectroscopía de Resonancia Magnética , Ratones Endogámicos C57BL , Péptidos/metabolismo , Unión Proteica , Multimerización de Proteína
9.
Thromb Haemost ; 117(1): 164-175, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-27786334

RESUMEN

The co-stimulatory molecule CD70 is expressed on activated immune cells and is known to modulate responses of T, B, and NK cells via its receptor CD27. Until now, there is only limited data describing the role of CD70 in atherosclerosis. We observed that ruptured human carotid atherosclerotic plaques displayed higher CD70 expression than stable carotid atherosclerotic plaques, and that CD70 expression in murine atheroma localized to macrophages. Lack of CD70 impaired the inflammatory capacity (e. g. reactive oxygen species and nitric oxide production) of bone marrow-derived macrophages, increased both M1-like and M2-like macrophage markers, and rendered macrophages metabolically inactive and prone to apoptosis. Moreover, CD70-deficient macrophages expressed diminished levels of scavenger receptors and ABC-transporters, impairing uptake of oxidised low-density lipoprotein (oxLDL) and cholesterol efflux, respectively. Hyperlipidaemic Apoe-/- mice reconstituted with CD70-deficient bone marrow displayed a profound increase in necrotic core size, plaque area, and number of lesional macrophages as compared to mice receiving control bone marrow. Accordingly, 18 week-old, chow diet-fed CD70-deficient Apoe-/- mice displayed larger atheroma characterised by lower cellularity and more advanced plaque phenotype than Apoe-/- mice. In conclusion, CD70 promotes macrophage function and viability and is crucial for effective phagocytosis and efflux of oxLDL. Deficiency in CD70 results in more advanced atheroma. Our data suggest that CD70 mitigates atherosclerosis at least in part by modulating macrophage function.


Asunto(s)
Aterosclerosis/metabolismo , Ligando CD27/metabolismo , Enfermedades de las Arterias Carótidas/metabolismo , Macrófagos/metabolismo , Placa Aterosclerótica , Anciano , Animales , Apoptosis , Aterosclerosis/inmunología , Aterosclerosis/patología , Aterosclerosis/prevención & control , Trasplante de Médula Ósea , Ligando CD27/deficiencia , Ligando CD27/genética , Enfermedades de las Arterias Carótidas/inmunología , Enfermedades de las Arterias Carótidas/patología , Células Cultivadas , Colesterol/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones Noqueados para ApoE , Necrosis , Óxido Nítrico/metabolismo , Fagocitosis , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo
10.
JACC Basic Transl Sci ; 1(3): 131-142., 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-27766315

RESUMEN

To enhance the antithrombotic properties of recombinant glycoprotein VI fragment crystallizable (GPVI-Fc), the authors incubated GPVI-Fc with anti-human Fc antibodies to cross-link the Fc tails of GPVI-Fc. Cross-linking potentiated the inhibition of human plaque- and collagen-induced platelet aggregation by GPVI-Fc under static and flow conditions without increasing bleeding time in vitro. Cross-linking with anti-human-Fc Fab2 was even superior to anti-human-Fc immunoglobulin G (IgG). Advanced optical imaging revealed a continuous sheath-like coverage of collagen fibers by cross-linked GPVI-Fc complexes. Cross-linking of GPVI into oligomeric complexes provides a new, highly effective, and probably safe antithrombotic treatment as it suppresses platelet GPVI-plaque interaction selectively at the site of acute atherothrombosis.

11.
PLoS One ; 10(8): e0134176, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26252890

RESUMEN

BACKGROUND AND AIMS: Plasmacytoid dendritic cells (pDCs) are a small subset of dendritic cells and the main producers of type I interferons. Besides their contribution to tolerance, they are known to be involved in autoimmune diseases and have recently been implicated in atherosclerosis. However, their precise involvement, particularly in advanced lesion development, remains elusive. Hence, we investigated the role of pDCs in atherogenesis vs atheroprogression by specifically depleting this cell population using the BDCA2-DTR mouse model bred to Apolipoprotein E (Apoe-/-) deficient mice. METHODS AND RESULTS: Our results revealed that continuous diphtheria toxin-induced pDC depletion in Apoe-/- BDCA2-DTR mice receiving a high-fat diet (HFD) for 4 weeks did not alter lesion size or composition. Instead, these mice displayed increased B cell numbers and altered levels of inflammatory cytokines. Analysis of depletion efficiency showed that complete pDC depletion could only be sustained for one week and reoccurring pDCs sorted after 4 weeks did not express DTR anymore. Consequently, we analyzed lesion development in a model of partial carotid ligation, inducing established lesions after 5 weeks of HFD feeding, and only depleted pDCs during the last week of 5 weeks HFD feeding. Despite short-term, but efficient pDC depletion, we observed no differences in atherosclerotic lesion development, but changes in inflammatory cytokine titers. To assure the functionality of the BDCA2-DTR model in acute settings, we additionally examined the effect of pDC depletion in an indirect acute lung injury (iALI) model. This time, efficient pDC depletion resulted in a significantly reduced macrophage and neutrophil accumulation in the lung 12 hours after LPS challenge, underlining a pro-inflammatory role of pDCs in the innate immune response in iALI. CONCLUSION: Taken together, the BDCA2-DTR mouse model only allows efficient pDC depletion for one week, which subsequently restricts its usability to more acute but not chronic inflammatory disease models.


Asunto(s)
Inflamación/patología , Enfermedad Aguda , Lesión Pulmonar Aguda/patología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/metabolismo , Aterosclerosis/sangre , Aterosclerosis/patología , Médula Ósea/patología , Recuento de Células , Enfermedad Crónica , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Dieta Alta en Grasa , Toxina Diftérica/inmunología , Modelos Animales de Enfermedad , Antígenos Comunes de Leucocito/metabolismo , Lípidos/sangre , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/efectos de los fármacos , Células Mieloides/patología , Bazo/patología , Transgenes
12.
J Am Coll Cardiol ; 65(22): 2404-15, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26046734

RESUMEN

BACKGROUND: Glycoprotein VI (GPVI) is the essential platelet collagen receptor in atherothrombosis, but its inhibition causes only a mild bleeding tendency. Thus, targeting this receptor has selective antithrombotic potential. OBJECTIVES: This study sought to compare compounds interfering with platelet GPVI-atherosclerotic plaque interaction to improve current antiatherothrombotic therapy. METHODS: Human atherosclerotic plaque-induced platelet aggregation was measured in anticoagulated blood under static and arterial flow conditions (550/s, 1,100/s, and 1,500/s). Inhibition by dimeric GPVI fragment crystallizable region of IgG (Fc) masking GPVI binding sites on collagen was compared with that of 3 anti-GPVI antibodies: BLO8-1, a human domain antibody; 5C4, a fragment antigen-binding (Fab fragment) of monoclonal rat immunoglobulin G; and m-Fab-F, a human recombinant sFab against GPVI dimers. RESULTS: GPVI-Fc reduced plaque-triggered platelet aggregation in static blood by 51%, BLO8-1 by 88%, and 5C4 by 93%. Under arterial flow conditions, BLO8-1 and 5C4 almost completely inhibited platelet aggregation while preserving platelet adhesion on plaque. Inhibition by GPVI-Fc, even at high concentrations, was less marked but increased with shear rate. Advanced optical imaging revealed rapid persistent GPVI-Fc binding to collagen under low and high shear flow, upstream and downstream of plaque fragments. At low shear particularly, platelets adhered in plaque flow niches to GPVI-Fc-free segments of collagen fibers and recruited other platelets onto aggregates via ADP and TxA2 release. CONCLUSIONS: Anti-GPVI antibodies inhibit atherosclerotic plaque-induced platelet aggregation under static and flow conditions more effectively than GPVI-Fc. However, potent platelet inhibition by GPVI-Fc at a higher shear rate (1,500/s) suggests localized antithrombotic efficacy at denuded or fissured stenotic high-risk lesions without systemic bleeding. The compound-specific differences have relevance for clinical trials targeting GPVI-collagen interaction combined with established antiplatelet therapies in patients with spontaneous plaque rupture or intervention-associated plaque injury.


Asunto(s)
Velocidad del Flujo Sanguíneo/fisiología , Arterias Carótidas/fisiopatología , Fragmentos Fab de Inmunoglobulinas/farmacología , Placa Aterosclerótica/tratamiento farmacológico , Activación Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/farmacología , Animales , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/patología , Estenosis Carotídea/tratamiento farmacológico , Estenosis Carotídea/etiología , Estenosis Carotídea/fisiopatología , Humanos , Factores Inmunológicos/farmacología , Placa Aterosclerótica/complicaciones , Placa Aterosclerótica/diagnóstico , Ratas
13.
BMC Microbiol ; 15: 60, 2015 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-25879450

RESUMEN

BACKGROUND: Candida albicans yeast produces 10 distinct secreted aspartic proteases (Saps), which are some of the most important virulence factors of this pathogenic fungus. One of the suggested roles of Saps is their deregulating effect on various proteolytic cascades that constitute the major homeostatic systems in human hosts, including blood coagulation, fibrinolysis, and kallikrein-kinin systems. This study compared the characteristics of the action of all 10 Saps on human kininogens, which results in generating proinflammatory bradykinin-related peptides (kinins). RESULTS: Recombinant forms of Saps, heterologously overexpressed in Pichia pastoris were applied. Except for Sap7 and Sap10, all Saps effectively cleaved the kininogens, with the highest hydrolytic activity toward the low-molecular-mass form (LK). Sap1-6 and 8 produced a biologically active kinin-Met-Lys-bradykinin-and Sap3 was exceptional in terms of the kinin-releasing yield (>60% LK at pH 5.0 after 24 hours). Des-Arg(1)-bradykinin was released from LK by Sap9 at a comparably high yield, but this peptide was assumed to be biologically inactive because it was unable to interact with cellular B2-type kinin receptors. However, the collaborative actions of Sap9 and Sap1, -2, -4-6, and -8 on LK rerouted kininogen cleavage toward the high-yield release of the biologically active Met-Lys-bradykinin. CONCLUSIONS: Our present results, together with the available data on the expression of individual SAP genes in candidal infection models, suggest a biological potential of Saps to produce kinins at the infection foci. The kinin release during candidiasis can involve predominant and complementary contributions of two different Sap3- and Sap9-dependent mechanisms.


Asunto(s)
Proteasas de Ácido Aspártico/química , Autacoides/química , Candida albicans/química , Proteínas Fúngicas/química , Quininógenos/química , Cininas/química , Secuencia de Aminoácidos , Proteasas de Ácido Aspártico/genética , Bradiquinina/análogos & derivados , Bradiquinina/química , Candida albicans/enzimología , Candida albicans/patogenicidad , Proteínas Fúngicas/genética , Expresión Génica , Humanos , Concentración de Iones de Hidrógeno , Isoenzimas/química , Isoenzimas/genética , Datos de Secuencia Molecular , Pichia/genética , Pichia/metabolismo , Proteolisis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Virulencia
14.
Cell Signal ; 26(2): 260-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24269653

RESUMEN

The WNT/ß-catenin signalling cascade is the best-investigated frizzled receptor (FZD) pathway, however, whether and how specific combinations of WNT/FZD and co-receptors LRP5 and LRP6 differentially affect this pathway are not well understood. This is mostly due to the fact that there are 19 WNTs, 10 FZDs and at least two co-receptors. In our attempt to identify the signalling capabilities of specific WNT/FZD/LRP combinations we made use of our previously reported TCF/LEF Gaussia luciferase reporter gene HEK293 cell line (Ring et al., 2011). Generation of WNT/FZD fusion constructs - but not their separate transfection - without or with additional isogenic overexpression of LRP5 and LRP6 in our reporter cells permitted the investigation of specific WNT/FZD/LRP combinations. The canonical WNT3a in fusion to almost all FZDs was able to induce ß-catenin-dependent signalling with strong dependency on LRP6 but not LRP5. Interestingly, noncanonical WNT ligands, WNT4 and WNT5a, were also able to act "canonically" but only in fusion with specific FZDs and with selective dependence on LRP5 or LRP6. These data and extension of this experimental setup to the poorly characterized other WNTs should facilitate deeper insight into the complex WNT/FZD signalling system and its function.


Asunto(s)
Receptores Frizzled/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Receptores Frizzled/genética , Genes Reporteros , Células HEK293 , Humanos , Ligandos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Wnt/genética , Proteína Wnt-5a , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo , Proteína Wnt4/genética , Proteína Wnt4/metabolismo
15.
Peptides ; 48: 114-23, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23954712

RESUMEN

In terms of infection incidence, the yeast Candida parapsilosis is the second after Candida albicans as causative agent of candidiases in humans. The major virulence factors of C. parapsilosis are secreted aspartic proteases (SAPPs) which help the pathogen to disseminate, acquire nutrients and dysregulate the mechanisms of innate immunity of the host. In the current work we characterized the action of two major extracellular proteases of C. parapsilosis, SAPP1 and SAPP2, on human kininogens, proteinaceous precursors of vasoactive and proinflammatory bradykinin-related peptides, collectively called the kinins. The kininogens, preferably the form with lower molecular mass, were effectively cleaved by SAPPs, with the release of two uncommon kinins, Met-Lys-bradykinin and Leu-Met-Lys-bradykinin. While optimal at acidic pH (4-5), the kinin release yield was only 2-3-fold lower at neutral pH. These peptides were able to interact with cellular kinin receptors of B2 subtype and to stimulate the human endothelial cells HMEC-1 to increased secretion of proinflammatory interleukins (ILs), IL-1ß and IL-6. The analysis of the stability of SAPP-generated kinins in plasma suggested that they are biologically equivalent to bradykinin, the best agonist of B2 receptor subtype and can be quickly converted to des-Arg(9)-bradykinin, the agonist of inflammation-inducible B1 receptors.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Bradiquinina/análogos & derivados , Candidiasis/enzimología , Proteínas Fúngicas/metabolismo , Quininógenos/metabolismo , Proteasas de Ácido Aspártico , Bradiquinina/química , Bradiquinina/metabolismo , Candida/enzimología , Candida/patogenicidad , Candidiasis/microbiología , Candidiasis/patología , Células Endoteliales , Humanos , Quininógenos/química , Oligopéptidos , Péptidos/química , Péptidos/metabolismo
16.
Acta Biochim Pol ; 60(3): 299-305, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23757449

RESUMEN

Kinins, a group of important pro-inflammatory peptides, are abundantly found in tissues and biological fluids of cancer patients. Bradykinin, the major representative of kinins, induces vascular permeability and, in consequence, promotes tumor expansion. Additionally, the kinin-induced inflammatory responses, especially those mediated by kinin metabolites without the C-terminal arginine residue, lead to enhanced tumor growth. The present study aimed at analyzing the ability of the human glioblastoma cell line U-373, derived from a malignant tumor, to produce kinin peptides. The proteins involved in kinin generation, i.e., the kininogens and the kallikreins, were shown to be expressed in these cells. Moreover, tumor necrosis factor α, a proinflammatory cytokine that mediates tumorigenesis, was found to enhance the expression of enzymes associated with kinin production. The strong binding of kininogen to the cell surface and the enzymatic degradation of this protein by cells suggest the activation of kinin-generating systems. Indeed, glioblastoma cells, pre-treated with tumor necrosis factor α, released kinin peptides from exogenous kininogen. The expression of kinin receptors in these cells was also shown to increase under the influence of this cytokine. Our results suggest that the human glioblastoma cell line U-373 constitutes a good cellular model that can be helpful in cancer research focused on kinin-induced inflammation. Furthermore, our findings can contribute to new approaches in cancer treatment with the use of kinin receptor antagonists and inhibitors of kinin production.


Asunto(s)
Bradiquinina/genética , Neoplasias Encefálicas/genética , Citocinas/genética , Glioblastoma/genética , Calicreínas/genética , Quininógenos/genética , Receptores de Bradiquinina/genética , Bradiquinina/inmunología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Citocinas/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Glioblastoma/inmunología , Glioblastoma/patología , Humanos , Inflamación , Calicreínas/inmunología , Quininógenos/inmunología , Modelos Biológicos , Receptores de Bradiquinina/inmunología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
17.
J Pharmacol Exp Ther ; 344(1): 85-95, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23086229

RESUMEN

The DRY motif with the highly conserved R3.50 is a hallmark of family A G protein-coupled receptors (GPCRs). The crystal structure of rhodopsin revealed a salt bridge between R135(3.50) and another conserved residue, E247(6.30), in helix 6. This ionic lock was shown to maintain rhodopsin in its inactive state. Thus far, little information is available on how interruption of this ionic bond affects signaling properties of nonrhodopsin GPCRs, because the focus has been on mutations of R3.50, although this residue is indispensable for G protein activation. To investigate the importance of an ionic lock for overall receptor activity in a nonrhodopsin GPCR, we mutated R128(3.50) and E238(6.30) in the bradykinin (BK) B(2) receptor (B(2)R) and stably expressed the constructs in HEK293 cells. As expected, mutation of R3.50 resulted in lack of G protein activation. In addition, this mutation led to considerable constitutive receptor internalization. Mutation of E6.30 (mutants E6.30A and E6.30R) also caused strong constitutive internalization. Most intriguingly, however, although the two E6.30 mutants displayed no increased basal phosphatidylinositol hydrolysis, they gave a response to three different B(2)R antagonists that was almost comparable to that obtained with BK. In contrast, swapping of R3.50 and E6.30, thus allowing the formation of an inverse ionic bond, resulted in rescue of the wild type phenotype. These findings demonstrate for the first time, to our knowledge, that interruption of the ionic lock in a family A GPCR can have distinctly different effects on receptor internalization and G protein stimulation, shedding new light on its role in the activation process.


Asunto(s)
Receptor de Bradiquinina B2/efectos de los fármacos , Aminoácidos/metabolismo , Biotinilación , Bradiquinina/metabolismo , Antagonistas del Receptor de Bradiquinina B2 , Proteínas de Unión al GTP/metabolismo , Expresión Génica , Células HEK293 , Humanos , Hidrólisis , Fosfatos de Inositol/metabolismo , Iones/metabolismo , Fosforilación , Mutación Puntual , Piridonas/farmacología , Quinolinas/farmacología , Receptor de Bradiquinina B2/agonistas , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Temperatura
18.
Biol Chem ; 393(8): 829-39, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22944684

RESUMEN

Bradykinin-related peptides, universal mediators of inflammation collectively referred to as the kinins, are often produced in excessive amounts during microbial infections. We have recently shown that the yeast Candida albicans, the major fungal pathogen to humans, can exploit two mechanisms to enhance kinin levels at the sites of candidial infection, one depending on adsorption and activation of the endogenous kinin-generating system of the host on the fungal cell wall and the other relying on cleavage of kinin precursors, the kininogens, by pathogen-secreted proteases. This work aimed at assigning this kininogenase activity to the major secreted aspartic protease of C. albicans (SAP2). The purified SAP2 was shown to cleave human kininogens, preferably the low molecular mass form (LK) and optimally in an acidic environment (pH 3.5-4.0), and to produce two kinins, Met-Lys-bradykinin and its derivative, [Hydroxyproline(3)]-Met-Lys-bradykinin, both of which are capable of interacting with cellular bradykinin receptors of the B2 subtype. Additionally, albeit with a lower yield, des-Arg(9)-Met-Lys-bradykinin, an effective agonist of B1-subtype receptors, was released. The pathophysiological potential of these kinins and des-Arg-kinin was also proven by presenting their ability to stimulate human promonocytic cells U937 to release proinflammatory interleukin 1ß (IL-1ß) and IL-6.


Asunto(s)
Ácido Aspártico Endopeptidasas/inmunología , Bradiquinina/análogos & derivados , Candida albicans/inmunología , Candidiasis/inmunología , Proteínas Fúngicas/inmunología , Quininógenos/inmunología , Secuencia de Aminoácidos , Ácido Aspártico Endopeptidasas/metabolismo , Bradiquinina/química , Bradiquinina/inmunología , Bradiquinina/metabolismo , Candida albicans/enzimología , Candidiasis/enzimología , Candidiasis/metabolismo , Línea Celular , Proteínas Fúngicas/metabolismo , Humanos , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Quininógenos/metabolismo , Datos de Secuencia Molecular , Receptores de Bradiquinina/inmunología
19.
J Biol Chem ; 286(50): 43282-93, 2011 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-22016392

RESUMEN

Upon activation the human bradykinin B(2) receptor (B(2)R) acts as guanine nucleotide exchange factor for the G proteins G(q/11) and G(i). Thereafter, it gets phosphorylated by G protein-coupled receptor kinases (GRKs) and recruits ß-arrestins, which block further G protein activation and promote B(2)R internalization via clathrin-coated pits. As for most G protein-coupled receptors of family A, an intracellular helix 8 after transmembrane domain 7 is also predicted for the B(2)R. We show here that disruption of helix 8 in the B(2)R by either C-terminal truncation or just by mutation of a central amino acid (Lys-315) to a helix-breaking proline resulted in strong reduction of surface expression. Interestingly, this malfunction could be overcome by the addition of the membrane-permeable B(2)R antagonist JSM10292, suggesting that helix 8 has a general role for conformational stabilization that can be accounted for by an appropriate antagonist. Intriguingly, an intact helix 8, but not the C terminus with its phosphorylation sites, was indispensable for receptor sequestration and for interaction of the B(2)R with GRK2/3 and ß-arrestin2 as shown by co-immunoprecipitation. Recruitment of ß-arrestin1, however, required the presence of the C terminus. Taken together, our results demonstrate that helix 8 of the B(2)R plays a crucial role not only in efficient trafficking to the plasma membrane or the activation of G proteins but also for the interaction of the B(2)R with GRK2/3 and ß-arrestins. Additional data obtained with chimera of B(2)R with other G protein-coupled receptors of family A suggest that helix 8 might have similar functions in other GPCRs as well.


Asunto(s)
Transporte de Proteínas/fisiología , Receptor de Bradiquinina B2/química , Receptor de Bradiquinina B2/metabolismo , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Línea Celular , Humanos , Immunoblotting , Inmunoprecipitación , Fosfatos de Inositol/metabolismo , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Transporte de Proteínas/genética , Receptor de Bradiquinina B2/genética , Homología de Secuencia de Aminoácido , Transducción de Señal/genética , Relación Estructura-Actividad
20.
Biol Chem ; 392(11): 1011-20, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21864196

RESUMEN

WNT/Frizzled receptor (FZD) signaling pathways are pivotal for physiological and pathophysiological processes. In humans, the complexity of WNT/FZD signaling is based on 19 WNTs, 10 FZDs and at least two (co)receptors (LRP5/6) mediating supposably four different signaling cascades. The detailed investigation of the specific function of the different initiating components is primarily hampered by the lack of most WNT proteins in a purified form. Therefore, we constructed and examined a chimeric protein of WNT3a and FZD4 as a suitable approach to overcome this obstacle for future studies of the specificity of other WNT/FZD combinations. Furthermore, we produced four different reporter HEK 293 cell lines to quantify the induced activation of the proposed signaling cascades, the ß-catenin-, the NFAT-, the AP-1- and the CRE-regulated pathways. The chimera WNT3aFZD4 efficiently induced ß-catenin-mediated luciferase activity. This activity was increased 40-fold compared with basal when LRP6 was stably cotransfected, proving that the chimera WNT3aFZD4 can also interact efficiently with LRP6. Our results demonstrate that the approach of using reporter gene cell lines in combination with WNT/FZD chimeras is efficient to study the ß-catenin-mediated pathway and should also allow clarifying the specificity of WNT/FZD combinations in the activation of the other pathways.


Asunto(s)
Receptores Frizzled/genética , Genes Reporteros , Proteínas Recombinantes de Fusión/genética , Proteínas Wnt/genética , Vía de Señalización Wnt , Secuencia de Bases , Receptores Frizzled/metabolismo , Células HEK293 , Humanos , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...