Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
J Immunol ; 207(12): 3038-3049, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34799423

RESUMEN

Neutrophil migration requires ß2 integrins and chemoattractant receptor signaling for motility and directionality. G protein subunit Gα13 can facilitate cell migration by mediating RhoA activation induced by G protein-coupled receptors. However, the possible role of Gα13-integrin interaction in migration is unclear. In this study, we show that Gα13 -/- neutrophils are deficient in transendothelial migration and migration on ß2 integrin ligand ICAM-1. However, unlike G protein-coupled receptors and integrin inside-out signaling pathways, Gα13 is important in migration velocity and neutrophil spreading but not in directionality nor cell adhesion. Importantly, neutrophil recruitment in vivo was also inhibited in Gα13 -/- mice, suggesting the importance of Gα13 in transendothelial migration of neutrophils in vitro and in vivo. Furthermore, a synthetic peptide (MB2mP6) derived from the Gα13 binding site of ß2 inhibited Gα13-ß2 interaction and Gα13-mediated transient RhoA inhibition in neutrophils, suggesting that this peptide inhibited integrin outside-in signaling. MB2mP6 inhibited migration of control neutrophils through endothelial cell monolayers or ICAM-1-coated filters, but was without further effect on Gα13 -/- neutrophils. It also inhibited integrin-dependent neutrophil migration velocity without affecting directionality. In vivo, MB2mP6 markedly inhibited neutrophil infiltration into the cardiac tissues induced by ischemia/reperfusion injury. Thus, Gα13-dependent outside-in signaling enables integrin-dependent neutrophil motility without affecting directionality and may be a new therapeutic target for inhibiting neutrophil trafficking but not adhesion.


Asunto(s)
Neutrófilos , Migración Transendotelial y Transepitelial , Animales , Antígenos CD18/metabolismo , Adhesión Celular/fisiología , Integrinas/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Neutrófilos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
2.
J Biol Chem ; 290(19): 12058-67, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25802336

RESUMEN

Pathological cardiac hypertrophy (an increase in cardiac mass resulting from stress-induced cardiac myocyte growth) is a major factor underlying heart failure. Src homology 2 domain-containing phosphatase (Shp2) is critical for cardiac function because mutations resulting in loss of Shp2 catalytic activity are associated with congenital cardiac defects and hypertrophy. We identified a novel mechanism of Shp2 inhibition that may promote cardiac hypertrophy. We demonstrate that Shp2 is a component of the protein kinase A anchoring protein (AKAP)-Lbc complex. AKAP-Lbc facilitates PKA phosphorylation of Shp2, which inhibits Shp2 phosphatase activity. We identified two key amino acids in Shp2 that are phosphorylated by PKA. Thr-73 contributes a helix cap to helix αB within the N-terminal SH2 domain of Shp2, whereas Ser-189 occupies an equivalent position within the C-terminal SH2 domain. Utilizing double mutant PKA phosphodeficient (T73A/S189A) and phosphomimetic (T73D/S189D) constructs, in vitro binding assays, and phosphatase activity assays, we demonstrate that phosphorylation of these residues disrupts Shp2 interaction with tyrosine-phosphorylated ligands and inhibits its protein-tyrosine phosphatase activity. Overall, our data indicate that AKAP-Lbc integrates PKA and Shp2 signaling in the heart and that AKAP-Lbc-associated Shp2 activity is reduced in hypertrophic hearts in response to chronic ß-adrenergic stimulation and PKA activation. Therefore, although induction of cardiac hypertrophy is a multifaceted process, inhibition of Shp2 activity through AKAP-Lbc-anchored PKA is a previously unrecognized mechanism that may promote this compensatory response.


Asunto(s)
Cardiomegalia/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Animales , Células Cultivadas , Células HEK293 , Humanos , Ligandos , Mutación , Fosforilación , Unión Proteica , Ratas , Receptores Adrenérgicos beta/metabolismo , Serina/química , Transducción de Señal , Treonina/química , Tirosina/química , Dominios Homologos src
3.
J Biol Chem ; 288(46): 33226-40, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24108126

RESUMEN

Ligand binding to extracellular domains of G protein-coupled receptors can result in novel and nuanced allosteric effects on receptor signaling. We previously showed that the protein-protein interaction of carboxypeptidase M (CPM) and kinin B1 receptor (B1R) enhances B1R signaling in two ways; 1) kinin binding to CPM causes a conformational activation of the B1R, and 2) CPM-generated des-Arg-kinin agonist is efficiently delivered to the B1R. Here, we show CPM is also a positive allosteric modulator of B1R signaling to its agonist, des-Arg(10)-kallidin (DAKD). In HEK cells stably transfected with B1R, co-expression of CPM enhanced DAKD-stimulated increases in intracellular Ca(2+) or phosphoinositide turnover by a leftward shift of the dose-response curve without changing the maximum. CPM increased B1R affinity for DAKD by ∼5-fold but had no effect on basal B1R-dependent phosphoinositide turnover. Soluble, recombinant CPM bound to HEK cells expressing B1Rs without stimulating receptor signaling. CPM positive allosteric action was independent of enzyme activity but depended on interaction of its C-terminal domain with the B1R extracellular loop 2. Disruption of the CPM/B1R interaction or knockdown of CPM in cytokine-treated primary human endothelial cells inhibited the allosteric enhancement of CPM on B1R DAKD binding or ERK1/2 activation. CPM also enhanced the DAKD-induced B1R conformational change as detected by increased intramolecular fluorescence or bioluminescence resonance energy transfer. Thus, CPM binding to extracellular loop 2 of the B1R results in positive allosteric modulation of B1R signaling, and disruption of this interaction could provide a novel therapeutic approach to reduce pathological B1R signaling.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Metaloendopeptidasas/metabolismo , Receptor de Bradiquinina B1/metabolismo , Regulación Alostérica/fisiología , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Células HEK293 , Humanos , Metaloendopeptidasas/genética , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptor de Bradiquinina B1/genética
4.
J Biol Chem ; 288(6): 4174-93, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23255592

RESUMEN

High levels of NO generated in the vasculature under inflammatory conditions are usually attributed to inducible nitric-oxide synthase (iNOS), but the role of the constitutively expressed endothelial NOS (eNOS) is unclear. In normal human lung microvascular endothelial cells (HLMVEC), bradykinin (BK) activates kinin B2 receptor (B2R) signaling that results in Ca(2+)-dependent activation of eNOS and transient NO. In inflamed HLMVEC (pretreated with interleukin-1ß and interferon-γ), we found enhanced binding of eNOS to calcium-calmodulin at basal Ca(2+) levels, thereby increasing its basal activity that was dependent on extracellular l-Arg. Furthermore, B2R stimulation generated prolonged high output eNOS-derived NO that is independent of increased intracellular Ca(2+) and is mediated by a novel Gα(i)-, MEK1/2-, and JNK1/2-dependent pathway. This high output NO stimulated with BK was blocked with a B2R antagonist, eNOS siRNA, or eNOS inhibitor but not iNOS inhibitor. Moreover, B2R-mediated NO production and JNK phosphorylation were inhibited with MEK1/2 and JNK inhibitors or MEK1/2 and JNK1/2 siRNA but not with ERK1/2 inhibitor. BK induced Ca(2+)-dependent eNOS phosphorylation at Ser(1177), Thr(495), and Ser(114) in cytokine-treated HLMVEC, but these modifications were not dependent on JNK1/2 activation and were not responsible for prolonged NO output. Cytokine treatment did not alter the expression of B2R, Gα(q/11), Gα(i1,2), JNK, or eNOS. B2R activation in control endothelial cells enhanced migration, but in cytokine-treated HLMVEC it reduced migration. Both responses were NO-dependent. Understanding how JNK regulates prolonged eNOS-derived NO may provide new therapeutic targets for the treatment of disorders involving vascular inflammation.


Asunto(s)
Células Endoteliales/enzimología , Endotelio Vascular/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/biosíntesis , Vasculitis/enzimología , Arginina/metabolismo , Bradiquinina/metabolismo , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Células Endoteliales/patología , Endotelio Vascular/metabolismo , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/biosíntesis , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Inflamación/patología , Inflamación/terapia , Interleucina-1beta/metabolismo , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Receptor de Bradiquinina B2/metabolismo , Vasculitis/tratamiento farmacológico , Vasculitis/patología
5.
Blood ; 120(9): 1942-52, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22806890

RESUMEN

Polymorphonuclear neutrophil (PMN) extravasation requires selectin-mediated tethering, intercellular adhesion molecule-1 (ICAM-1)-dependent firm adhesion, and platelet/endothelial cell adhesion molecule 1 (PECAM-1)-mediated transendothelial migration. An important unanswered question is whether ICAM-1-activated signaling contributes to PMN transmigration mediated by PECAM-1. We tested this concept and the roles of endothelial nitric oxide synthase (eNOS) and Src activated by PMN ligation of ICAM-1 in mediating PECAM-1-dependent PMN transmigration. We observed that lung PMN infiltration in vivo induced in carrageenan-injected WT mice was significantly reduced in ICAM-1(-/-) and eNOS(-/-) mice. Crosslinking WT mouse ICAM-1 expressed in human endothelial cells (ECs), but not the phospho-defective Tyr(518)Phe ICAM-1 mutant, induced SHP-2-dependent Src Tyr530 dephosphorylation that resulted in Src activation. ICAM-1 activation also stimulated phosphorylation of Akt (p-Ser473) and eNOS (p-Ser1177), thereby increasing NO production. PMN migration across EC monolayers was abolished in cells expressing the Tyr(518)Phe ICAM-1 mutant or by pretreatment with either the Src inhibitor PP2 or eNOS inhibitor L-NAME. Importantly, phospho-ICAM-1 induction of Src signaling induced PECAM-1 Tyr686 phosphorylation and increased EC surface anti-PECAM-1 mAb-binding activity. These results collectively show that ICAM-1-activated Src and eNOS signaling sequentially induce PECAM-1-mediated PMN transendothelial migration. Both Src and eNOS inhibition may be important therapeutic targets to prevent or limit vascular inflammation.


Asunto(s)
Células Endoteliales/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Neutrófilos/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Familia-src Quinasas/metabolismo , Animales , Western Blotting , Carragenina/farmacología , Adhesión Celular , Células Cultivadas , Activación Enzimática , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inmunoprecipitación , Molécula 1 de Adhesión Intercelular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/citología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Fosforilación , Unión Proteica , Transducción de Señal , Migración Transendotelial y Transepitelial/efectos de los fármacos
6.
Biol Chem ; 394(3): 335-45, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23183746

RESUMEN

The G protein-coupled receptors (GPCRs) are the largest class of membrane proteins that play key roles in transducing extracellular signals to intracellular proteins to generate cellular responses. The kinin GPCRs, named B1 (B1R) and B2 (B2R), are responsible for mediating the biological responses to kinin peptides released from the precursor kininogens. Bradykinin (BK) or kallidin (KD) are agonists for B2Rs, whereas their carboxypeptidase (CP)-generated metabolites, des-Arg(9)-BK or des-Arg(10)-KD, are specific agonists for B1Rs. Here, we review the evidence for a critical role of membrane-bound CPM in facilitating B1R signaling by its ability to directly activate the receptor via conformational crosstalk as well as generate its specific agonist. In endothelial cells, the CPM/B1R interaction facilitates B1R-dependent high-output nitric oxide under inflammatory conditions.


Asunto(s)
Células Endoteliales/metabolismo , Metaloendopeptidasas/metabolismo , Óxido Nítrico/metabolismo , Receptor de Bradiquinina B1/metabolismo , Transducción de Señal , Células Endoteliales/enzimología , Proteínas Ligadas a GPI/metabolismo , Humanos , Unión Proteica
7.
Nat Commun ; 14(1): 4966, 2023 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-37587112

RESUMEN

The integrins and G protein-coupled receptors are both fundamental in cell biology. The cross talk between these two, however, is unclear. Here we show that ß3 integrins negatively regulate G protein-coupled signaling by directly inhibiting the Gα13-p115RhoGEF interaction. Furthermore, whereas ß3 deficiency or integrin antagonists inhibit integrin-dependent platelet aggregation and exocytosis (granule secretion), they enhance G protein-coupled RhoA activation and integrin-independent secretion. In contrast, a ß3-derived Gα13-binding peptide or Gα13 knockout inhibits G protein-coupled RhoA activation and both integrin-independent and dependent platelet secretion without affecting primary platelet aggregation. In a mouse model of myocardial ischemia/reperfusion injury in vivo, the ß3-derived Gα13-binding peptide inhibits platelet secretion of granule constituents, which exacerbates inflammation and ischemia/reperfusion injury. These data establish crucial integrin-G protein crosstalk, providing a rationale for therapeutic approaches that inhibit exocytosis in platelets and possibly other cells without adverse effects associated with loss of cell adhesion.


Asunto(s)
Proteínas de Unión al GTP , Transducción de Señal , Animales , Ratones , Exocitosis , Factores de Intercambio de Guanina Nucleótido Rho , Integrina beta3
8.
J Biol Chem ; 286(21): 18547-61, 2011 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-21454694

RESUMEN

G protein-coupled receptor (GPCR) signaling is affected by formation of GPCR homo- or heterodimers, but GPCR regulation by other cell surface proteins is not well understood. We reported that the kinin B1 receptor (B1R) heterodimerizes with membrane carboxypeptidase M (CPM), facilitating receptor signaling via CPM-mediated conversion of bradykinin or kallidin to des-Arg kinin B1R agonists. Here, we found that a catalytically inactive CPM mutant that still binds substrate (CPM-E264Q) also facilitates efficient B1R signaling by B2 receptor agonists bradykinin or kallidin. This response required co-expression of B1R and CPM-E264Q in the same cell, was disrupted by antibody that dissociates CPM from B1R, and was not found with a CPM-E264Q-B1R fusion protein. An additional mutation that reduced the affinity of CPM for C-terminal Arg and increased the affinity for C-terminal Lys inhibited the B1R response to bradykinin (with C-terminal Arg) but generated a response to Lys(9)-bradykinin. CPM-E264Q-mediated activation of B1Rs by bradykinin resulted in increased intramolecular fluorescence resonance energy transfer (FRET) in a B1R FRET construct, similar to that generated directly by a B1R agonist. In cytokine-treated human lung microvascular endothelial cells, disruption of B1R-CPM heterodimers inhibited B1R-dependent NO production stimulated by bradykinin and blocked the increased endothelial permeability caused by treatment with bradykinin and pyrogallol (a superoxide generator). Thus, CPM and B1Rs on cell membranes form a critical complex that potentiates B1R signaling. Kinin peptide binding to CPM causes a conformational change in the B1R leading to intracellular signaling and reveals a new mode of GPCR activation by a cell surface peptidase.


Asunto(s)
Permeabilidad Capilar/fisiología , Células Endoteliales/metabolismo , Metaloendopeptidasas/metabolismo , Receptor de Bradiquinina B1/metabolismo , Transducción de Señal/fisiología , Sustitución de Aminoácidos , Bradiquinina/genética , Bradiquinina/metabolismo , Células Endoteliales/citología , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Células HEK293 , Humanos , Calidina/genética , Calidina/metabolismo , Pulmón/citología , Pulmón/metabolismo , Metaloendopeptidasas/genética , Mutación Missense , Multimerización de Proteína/fisiología , Receptor de Bradiquinina B1/agonistas , Receptor de Bradiquinina B1/genética
9.
J Cell Mol Med ; 15(2): 258-69, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20015194

RESUMEN

Inducible nitric oxide synthase (iNOS) is a major source of nitric oxide during inflammation whose activity is thought to be controlled primarily at the expression level. The B1 kinin receptor (B1R) post-translationally activates iNOS beyond its basal activity via extracellular signal regulated kinase (ERK)-mediated phosphorylation of Ser(745) . Here we identified the signalling pathway causing iNOS activation in cytokine-treated endothelial cells or HEK293 cells transfected with iNOS and B1R. To allow kinetic measurements of nitric oxide release, we used a sensitive porphyrinic microsensor (response time = 10 msec.; 1 nM detection limit). B1Rs signalled through Gαi coupling as ERK and iNOS activation were inhibited by pertussis toxin. Furthermore, transfection of constitutively active mutant Gαi Q204L but not Gαq Q209L resulted in high basal iNOS-derived nitric oxide. G-ßγ subunits were also necessary as transfection with the ß-adrenergic receptor kinase C-terminus inhibited the response. B1R-dependent iNOS activation was also inhibited by Src family kinase inhibitor PP2 and trans-fection with dominant negative Src. Other ERK-MAP kinase members were involved as the response was inhibited by dominant negative H-Ras, Raf kinase inhibitor, ERK activation inhibitor and MEK inhibitor PD98059. In contrast, PI3 kinase inhibitor LY94002, calcium chelator 1,2-bis-(o-Aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid, tetraacetoxymethyl ester (BAPTA-AM), protein kinase C inhibitor calphostin C and protein kinase C activator PMA had no effect. Angiotensin converting enzyme inhibitor enalaprilat also directly activated B1Rs to generate high output nitric oxide via the same pathway. These studies reveal a new mechanism for generating receptor-regulated high output nitric oxide in inflamed endothelium that may play an important role in the development of vascular inflammation.


Asunto(s)
Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/biosíntesis , Receptor de Bradiquinina B1/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Línea Celular , Citocinas/farmacología , Enalaprilato/farmacología , Células Endoteliales/metabolismo , Activación Enzimática , Flavonoides/farmacología , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Pirimidinas/farmacología , Receptor de Bradiquinina B1/agonistas , Receptor de Bradiquinina B1/genética , Transfección , Quinasas de Receptores Adrenérgicos beta/metabolismo , Familia-src Quinasas/antagonistas & inhibidores
10.
FASEB J ; 24(7): 2475-83, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20228252

RESUMEN

A major source of "high-output" NO in inflammation is inducible nitric oxide synthase (iNOS). iNOS is primarily transcriptionally regulated and is thought to function as an uncontrolled generator of high NO. We found that iNOS in cytokine-stimulated human lung microvascular endothelial cells (HLMVECs) is highly regulated post-translationally via activation of the B1 kinin G protein-coupled receptor (B1R). We report here that B1R-mediated iNOS activation was significantly inhibited by knockdown of beta-arrestin 2 with siRNA in cytokine-treated HLMVECs or HEK293 cells transfected with iNOS and B1R. In contrast, beta-arrestin 1 siRNA had no effect. The prolonged phase of B1R-dependent ERK activation was also inhibited by beta-arrestin 2 knockdown. Furthermore, robust ERK activation by the epidermal growth factor receptor (a beta-arrestin 2 independent pathway) had no effect on iNOS-derived NO production. beta-arrestin 2 and iNOS coimmunoprecipitated, and there was significant fluorescence resonance energy transfer between CFP-iNOS and beta-arrestin 2-YFP (but not beta-arrestin 1-YFP) that increased 3-fold after B1R stimulation. These data show that beta-arrestin 2 mediates B1R-dependent high-output NO by scaffolding iNOS and ERK to allow post-translational activation of iNOS. This could play a critical role in mediating endothelial function in inflammation.


Asunto(s)
Arrestinas/fisiología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Receptor de Bradiquinina B1/metabolismo , Línea Celular , Endotelio Vascular/citología , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Inflamación , Pulmón/irrigación sanguínea , beta-Arrestina 1 , Arrestina beta 2 , beta-Arrestinas
11.
Nat Commun ; 12(1): 3185, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-34045461

RESUMEN

Systemic inflammation as manifested in sepsis is an excessive, life-threatening inflammatory response to severe bacterial or viral infection or extensive injury. It is also a thrombo-inflammatory condition associated with vascular leakage/hemorrhage and thrombosis that is not effectively treated by current anti-inflammatory or anti-thrombotic drugs. Here, we show that MB2mP6 peptide nanoparticles, targeting the Gα13-mediated integrin "outside-in" signaling in leukocytes and platelets, inhibited both inflammation and thrombosis without causing hemorrhage/vascular leakage. MB2mP6 improved mouse survival when infused immediately or hours after onset of severe sepsis. Furthermore, platelet Gα13 knockout inhibited septic thrombosis whereas leukocyte Gα13 knockout diminished septic inflammation, each moderately improving survival. Dual platelet/leukocyte Gα13 knockout inhibited septic thrombosis and inflammation, further improving survival similar to MB2mP6. These results demonstrate that inflammation and thrombosis independently contribute to poor outcomes and exacerbate each other in systemic inflammation, and reveal a concept of dual anti-inflammatory/anti-thrombotic therapy without exacerbating vascular leakage.


Asunto(s)
Antígenos CD18/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Fragmentos de Péptidos/farmacología , Sepsis/tratamiento farmacológico , Trombosis/tratamiento farmacológico , Animales , Antiinflamatorios , Plaquetas/efectos de los fármacos , Plaquetas/inmunología , Plaquetas/metabolismo , Antígenos CD18/metabolismo , Cloruros/administración & dosificación , Cloruros/toxicidad , Modelos Animales de Enfermedad , Compuestos Férricos/administración & dosificación , Compuestos Férricos/toxicidad , Fibrinolíticos , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Humanos , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Leucocitos/metabolismo , Macrófagos , Ratones , Ratones Noqueados , Nanopartículas/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Cultivo Primario de Células , Unión Proteica/efectos de los fármacos , Sepsis/sangre , Sepsis/complicaciones , Sepsis/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Células THP-1 , Trombosis/sangre , Trombosis/inducido químicamente
12.
Biochem Biophys Res Commun ; 399(4): 629-36, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20691153

RESUMEN

Type 1 diabetes (T1D) is caused by a T cell-mediated autoimmune response that leads to the loss of insulin-producing beta cells. The optimal preclinical testing of promising therapies would be aided by a humanized immune-mediated T1D model. We develop this model in NOD-scid IL2rgamma(null) mice. The selective destruction of pancreatic islet beta cells was mediated by human T lymphocytes after an initial trigger was supplied by the injection of irradiated spleen mononuclear cells (SMC) from diabetic nonobese diabetic (NOD) mice. This resulted in severe insulitis, a marked loss of total beta-cell mass, and other related phenotypes of T1D. The migration of human T cells to pancreatic islets was controlled by the beta cell-produced highly conserved chemokine stromal cell-derived factor 1 (SDF-1) and its receptor C-X-C chemokine receptor (CXCR) 4, as demonstrated by in vivo blocking experiments using antibody to CXCR4. The specificity of humanized T cell-mediated immune responses against islet beta cells was generated by the local inflammatory microenvironment in pancreatic islets including human CD4(+) T cell infiltration and clonal expansion, and the mouse islet beta-cell-derived CD1d-mediated human iNKT activation. The selective destruction of mouse islet beta cells by a human T cell-mediated immune response in this humanized T1D model can mimic those observed in T1D patients. This model can provide a valuable tool for translational research into T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Células Secretoras de Insulina/inmunología , Ratones , Linfocitos T/inmunología , Animales , Movimiento Celular , Quimiocina CXCL12/metabolismo , Diabetes Mellitus Tipo 1/patología , Humanos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/efectos de la radiación , Leucocitos Mononucleares/trasplante , Ratones Endogámicos NOD , Ratones SCID , Bazo/citología , Bazo/inmunología , Bazo/efectos de la radiación , Linfocitos T/trasplante , Investigación Biomédica Traslacional
13.
Sci Transl Med ; 12(552)2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32669423

RESUMEN

Inefficient delivery is a major obstacle to the development of peptide-based drugs targeting the intracellular compartment. We recently showed that selectively inhibiting integrin outside-in signaling using a peptide (mP6) derived from the Gα13-binding ExE motif within the integrin ß3 cytoplasmic domain had antithrombotic effects. Here, we engineered lipid-stabilized, high-loading peptide nanoparticles (HLPN), in which a redesigned ExE peptide (M3mP6) constituted up to 70% of the total nanoparticle molarity, allowing efficient in vivo delivery. We observed that M3mP6 HLPN inhibited occlusive thrombosis more potently than a clopidogrel/aspirin combination without adverse effects on hemostasis in rodents. Furthermore, M3mP6 HLPN synergized with P2Y12 receptor inhibitors or the clopidogrel/aspirin combination in preventing thrombosis, without exacerbating hemorrhage. M3mP6 HLPN also inhibited intravascular coagulation more potently than the P2Y12 inhibitor cangrelor. Postischemia injection of M3mP6 HLPN protected the heart from myocardial ischemia-reperfusion injury in a mouse model. This study demonstrates an efficient in vivo peptide delivery strategy for a therapeutic that not only efficaciously prevented thrombosis with minimal bleeding risk but also protected from myocardial ischemia-reperfusion injury in mice.


Asunto(s)
Daño por Reperfusión Miocárdica , Nanopartículas , Preparaciones Farmacéuticas , Trombosis , Animales , Isquemia , Ratones , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/prevención & control , Péptidos , Trombosis/prevención & control
14.
J Mol Biol ; 366(2): 504-16, 2007 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-17157876

RESUMEN

Human carboxypeptidase N (CPN), a member of the CPN/E subfamily of "regulatory" metallo-carboxypeptidases, is an extracellular glycoprotein synthesized in the liver and secreted into the blood, where it controls the activity of vasoactive peptide hormones, growth factors and cytokines by specifically removing C-terminal basic residues. Normally, CPN circulates in blood plasma as a hetero-tetramer consisting of two 83 kDa (CPN2) domains each flanked by a 48 to 55 kDa catalytic (CPN1) domain. We have prepared and crystallized the recombinant C-terminally truncated catalytic domain of human CPN1, and have determined and refined its 2.1 A crystal structure. The structural analysis reveals that CPN1 has a pear-like shape, consisting of a 319 residue N-terminal catalytic domain and an abutting, cylindrically shaped 79 residue C-terminal beta-sandwich transthyretin (TT) domain, more resembling CPD-2 than CPM. Like these other CPN/E members, two surface loops surrounding the active-site groove restrict access to the catalytic center, offering an explanation for why some larger protein carboxypeptidase inhibitors do not inhibit CPN. Modeling of the Pro-Phe-Arg C-terminal end of the natural substrate bradykinin into the active site shows that the S1' pocket of CPN1 might better accommodate P1'-Lys than Arg residues, in agreement with CPN's preference for cleaving off C-terminal Lys residues. Three Thr residues at the distal TT edge of CPN1 are O-linked to N-acetyl glucosamine sugars; equivalent sites in the membrane-anchored CPM are occupied by basic residues probably involved in membrane interaction. In tetrameric CPN, each CPN1 subunit might interact with the central leucine-rich repeat tandem of the cognate CPN2 subunit via a unique hydrophobic surface patch wrapping around the catalytic domain-TT interface, exposing the two active centers.


Asunto(s)
Dominio Catalítico , Lisina Carboxipeptidasa/química , Estructura Terciaria de Proteína , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sitios de Unión , Bradiquinina/química , Cristalografía por Rayos X , Humanos , Lisina Carboxipeptidasa/genética , Lisina Carboxipeptidasa/aislamiento & purificación , Modelos Moleculares , Datos de Secuencia Molecular , Prealbúmina/química , Análisis de Secuencia de Proteína , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
15.
Circ Res ; 99(8): 870-7, 2006 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-16973909

RESUMEN

Caveolin-1, the caveolae scaffolding protein, binds to and negatively regulates eNOS activity. As caveolin-1 also regulates caveolae-mediated endocytosis after activation of the 60-kDa albumin-binding glycoprotein gp60 in endothelial cells, we addressed the possibility that endothelial NO synthase (eNOS)-dependent NO production was functionally coupled to caveolae internalization. We observed that gp60-induced activation of endocytosis increased NO production within 2 minutes and up to 20 minutes. NOS inhibitor N(G)-nitro-L-arginine (L-NNA) prevented the NO production. To determine the role of caveolae internalization in the mechanism of NO production, we expressed dominant-negative dynamin-2 mutant (K44A) or treated cells with methyl-beta-cyclodextrin. Both interventions inhibited caveolae-mediated endocytosis and NO generation induced by gp60. We determined the role of signaling via Src kinase in the observed coupling of endocytosis to eNOS activation. Src activation induced the phosphorylation of caveolin-1, Akt and eNOS, and promoted dissociation of eNOS from caveolin-1. Inhibitors of Src kinase and Akt also prevented NO production. In isolated perfused mouse lungs, gp60 activation induced NO-dependent vasodilation, whereas the response was attenuated in eNOS(-/-) or caveolin-1(-/-) lungs. Together, these results demonstrate a critical role of caveolae-mediated endocytosis in regulating eNOS activation in endothelial cells and thereby the NO-dependent vasomotor tone.


Asunto(s)
Caveolas/fisiología , Endocitosis/fisiología , Células Endoteliales/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Calcio/fisiología , Caveolina 1/deficiencia , Caveolina 1/metabolismo , Células Cultivadas , Células Endoteliales/metabolismo , Activación Enzimática/fisiología , Subunidades beta de la Proteína de Unión al GTP/fisiología , Subunidades gamma de la Proteína de Unión al GTP/fisiología , Pulmón/irrigación sanguínea , Ratones , Ratones Noqueados , Óxido Nítrico/biosíntesis , Proteínas Proto-Oncogénicas c-akt/fisiología , Ratas , Sialoglicoproteínas/fisiología , Factores de Tiempo , Distribución Tisular , Vasodilatación , Familia-src Quinasas/fisiología
16.
Int Immunopharmacol ; 7(14): 1888-99, 2007 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-18039526

RESUMEN

Human carboxypeptidase N (CPN) was discovered in the early 1960s as a plasma enzyme that inactivates bradykinin and was identified 8 years later as the major "anaphylatoxin inactivator" of blood. CPN plays an important role in protecting the body from excessive buildup of potentially deleterious peptides that normally act as local autocrine or paracrine hormones. This review summarizes the structure, enzymatic properties and function of this important human enzyme, including insights gained by the recent elucidation of the crystal structure of the CPN catalytic subunit and structural modeling of the non-catalytic regulatory 83 kDa subunit. We also discuss its physiological role in cleaving substrates such as kinins, anaphylatoxins, creatine kinase, plasminogen receptors, hemoglobin and stromal cell-derived factor-1alpha (SDF-1alpha).


Asunto(s)
Lisina Carboxipeptidasa/sangre , Lisina Carboxipeptidasa/química , Estructura Terciaria de Proteína , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Lisina Carboxipeptidasa/genética , Modelos Moleculares , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
17.
Biochem J ; 388(Pt 1): 81-91, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15617514

RESUMEN

Human CPN (carboxypeptidase N) is a tetrameric plasma enzyme containing two glycosylated 83 kDa non-catalytic/regulatory subunits that carry and protect two active catalytic subunits. Because CPN can regulate the level of plasminogen binding to cell surface proteins, we investigated how plasmin cleaves CPN and the consequences. The products of hydrolysis were analysed by activity assays, Western blotting, gel filtration and sequencing. When incubated with intact CPN tetramer, plasmin rapidly cleaved the 83 kDa subunit at the Arg457-Ser458 bond near the C-terminus to produce fragments of 72 and 13 kDa, thereby releasing an active 142 kDa heterodimer, and also cleaved the active subunit, decreasing its size from 55 kDa to 48 kDa. Further evidence for the heterodimeric form of CPN was obtained by re-complexing the non-catalytic 72 kDa fragment with recombinant catalytic subunit or by immunoprecipitation of the catalytic subunit after plasmin treatment of CPN using an antibody specific for the 83 kDa subunit. Upon longer incubation, plasmin cleaved the catalytic subunit at Arg218-Arg219 to generate fragments of 27 kDa and 21 kDa, held together by non-covalent bonds, that were more active than the native enzyme. These data show that plasmin can alter CPN structure and activity, and that the C-terminal 13 kDa fragment of the CPN 83 kDa subunit is a docking peptide that is necessary to maintain the stable active tetrameric form of human CPN in plasma.


Asunto(s)
Fibrinolisina/química , Lisina Carboxipeptidasa/química , Lisina Carboxipeptidasa/metabolismo , Estructura Cuaternaria de Proteína , Secuencia de Aminoácidos , Fibrinolisina/fisiología , Humanos , Hidrólisis , Datos de Secuencia Molecular , Subunidades de Proteína
18.
J Mol Biol ; 338(2): 257-69, 2004 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-15066430

RESUMEN

Carboxypeptidase M (CPM), an extracellular glycosylphosphatidyl-inositol(GPI)-anchored membrane glycoprotein belonging to the CPN/E subfamily of "regulatory" metallo-carboxypeptidases, specifically removes C-terminal basic residues from peptides and proteins. Due to its wide distribution in human tissues, CPM is believed to play important roles in the control of peptide hormone and growth factor activity at the cell surface, and in the membrane-localized degradation of extracellular proteins. We have crystallized human GPI-free CPM, and have determined and refined its 3.0A crystal structure. The structure analysis reveals that CPM consists of a 295 residue N-terminal catalytic domain similar to that of duck CPD-2 (but only distantly related to CPA/B), an adjacent 86 residue beta-sandwich C-terminal domain characteristic of the CPN/E family but more conically shaped than the equivalent domain in CPD-2, and a unique, partially disordered 25 residue C-terminal extension to which the GPI membrane-anchor is post-translationally attached. Through this GPI anchor, and presumably via some positively charged side-chains of the C-terminal domain, the CPM molecule may interact with the membrane in such a way that its active centre will face alongside, i.e. well suited to interact with other membrane-bound protein substrates or small peptides. Modelling of the C-terminal part of the natural substrate Arg(6)-Met-enkephalin into the active site shows that the S1' pocket of CPM is particularly well designed to accommodate P1'-Arg residues, in agreement with the preference of CPM for cleaving C-terminal Arg.


Asunto(s)
Metaloendopeptidasas/química , Hormonas Peptídicas/metabolismo , Estructura Terciaria de Proteína , Secuencia de Aminoácidos , Animales , Dominio Catalítico , Membrana Celular/metabolismo , Cristalografía por Rayos X , Proteínas Ligadas a GPI , Glicosilfosfatidilinositoles , Humanos , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Alineación de Secuencia
19.
Cell Signal ; 27(1): 90-103, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25289859

RESUMEN

Signaling through the G protein-coupled kinin receptors B1 (kB1R) and B2 (kB2R) plays a critical role in inflammatory responses mediated by activation of the kallikrein-kinin system. The kB2R is constitutively expressed and rapidly desensitized in response to agonist whereas kB1R expression is upregulated by inflammatory stimuli and it is resistant to internalization and desensitization. Here we show that the kB1R heterodimerizes with kB2Rs in co-transfected HEK293 cells and natively expressing endothelial cells, resulting in significant internalization and desensitization of the kB1R response in cells pre-treated with kB2R agonist. However, pre-treatment of cells with kB1R agonist did not affect subsequent kB2R responses. Agonists of other G protein-coupled receptors (thrombin, lysophosphatidic acid) had no effect on a subsequent kB1R response. The loss of kB1R response after pretreatment with kB2R agonist was partially reversed with kB2R mutant Y129S, which blocks kB2R signaling without affecting endocytosis, or T342A, which signals like wild type but is not endocytosed. Co-endocytosis of the kB1R with kB2R was dependent on ß-arrestin and clathrin-coated pits but not caveolae. The sorting pathway of kB1R and kB2R after endocytosis differed as recycling of kB1R to the cell surface was much slower than that of kB2R. In cytokine-treated human lung microvascular endothelial cells, pre-treatment with kB2R agonist inhibited kB1R-mediated increase in transendothelial electrical resistance (TER) caused by kB1R stimulation (to generate nitric oxide) and blocked the profound drop in TER caused by kB1R activation in the presence of pyrogallol (a superoxide generator). Thus, kB1R function can be downregulated by kB2R co-endocytosis and signaling, suggesting new approaches to control kB1R signaling in pathological conditions.


Asunto(s)
Regulación hacia Abajo , Multimerización de Proteína , Receptor de Bradiquinina B1/metabolismo , Receptor de Bradiquinina B2/metabolismo , Transducción de Señal , Animales , Células CHO , Calcio/metabolismo , Bovinos , Permeabilidad de la Membrana Celular , Vesículas Cubiertas por Clatrina/metabolismo , Cricetinae , Cricetulus , Endocitosis , Células Endoteliales/metabolismo , Células HEK293 , Humanos , Cinética , Unión Proteica , Receptor de Bradiquinina B2/agonistas
20.
Cell Signal ; 27(5): 908-22, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25683917

RESUMEN

Hypertrophy increases the risk of heart failure and arrhythmia. Prevention or reversal of the maladaptive hypertrophic phenotype has thus been proposed to treat heart failure. Chronic ß-adrenergic receptor (ß-AR) stimulation induces cardiomyocyte hypertrophy by elevating 3',5'-cyclic adenosine monophosphate (cAMP) levels and activating downstream effectors such protein kinase A (PKA). Conversely, hydrolysis of cAMP by phosphodiesterases (PDEs) spatiotemporally restricts cAMP signaling. Here, we demonstrate that PDE4, but not PDE3, is critical in regulating cardiomyocyte hypertrophy, and may represent a potential target for preventing maladaptive hypertrophy. We identify a sequence within the upstream conserved region 1 of PDE4D, termed UCR1C, as a novel activator of PDE4 long isoforms. UCR1C activates PDE4 in complex with A-kinase anchoring protein (AKAP)-Lbc resulting in decreased PKA signaling facilitated by AKAP-Lbc. Expression of UCR1C in cardiomyocytes inhibits hypertrophy in response to chronic ß-AR stimulation. This effect is partially due to inhibition of nuclear PKA activity, which decreases phosphorylation of the transcription factor cAMP response element-binding protein (CREB). In conclusion, PDE4 activation by UCR1C attenuates cardiomyocyte hypertrophy by specifically inhibiting nuclear PKA activity.


Asunto(s)
Cardiomegalia/tratamiento farmacológico , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Activación Enzimática/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Péptidos/química , Péptidos/farmacología , Proteínas de Anclaje a la Quinasa A/metabolismo , Secuencia de Aminoácidos , Animales , Cardiomegalia/metabolismo , Cardiomegalia/patología , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/química , Células HEK293 , Humanos , Datos de Secuencia Molecular , Miocitos Cardíacos/metabolismo , Fosforilación/efectos de los fármacos , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda