Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jacinto López-Sagaseta is active.

Publication


Featured researches published by Jacinto López-Sagaseta.


Journal of Thrombosis and Haemostasis | 2007

Binding of factor VIIa to the endothelial cell protein C receptor reduces its coagulant activity

Jacinto López-Sagaseta; Ramón Montes; Cristina Puy; N. Díez; Kenji Fukudome; José Hermida

Summary.  Background: Endothelial cell protein C receptor (EPCR) binds protein C through its γ‐carboxyglutamic acid (Gla) domain and enhances its thrombin–thrombomodulin complex‐dependent activation. So far, only protein C/activated protein C has been shown to interact with EPCR. Factor VII (FVII), the coagulation trigger upon tissue factor (TF) interaction, is a serine protease whose Gla domain is highly homologous to the Gla domain of protein C. Objectives:  To characterize the binding of FVII/FVIIa to EPCR and its functional consequences. Methods and results:  We demonstrated by surface plasmon resonance (SPR) that FVII/FVIIa binds to EPCR through its Gla domain. At therapeutic concentrations, FVIIa reduced the activation of protein C by 40%. Soluble EPCR (sEPCR) was also able to prolong dose‐dependently the clotting time induced by the FVIIa–TF complex. SPR and amidolytic experiments showed that FVIIa is able to interact simultaneously with TF and EPCR, thus ruling out the possibility that the effect of EPCR on clotting time was due to the inhibition of the binding between FVIIa and TF. sEPCR inhibited dose‐dependently the activation of FX by the FVIIa–TF complex. Notably, blocking the binding site of EPCR on the endothelial surface increased the generation of FXa 2‐fold. Conclusions: EPCR binds to FVII/FVIIa and inhibits the procoagulant activity of the FVIIa–TF complex.


Journal of Immunology | 2010

A Peptide Inhibitor of FOXP3 Impairs Regulatory T Cell Activity and Improves Vaccine Efficacy in Mice

Noelia Casares; Francesc Rudilla; Laura Arribillaga; Diana Llopiz; José Ignacio Riezu-Boj; Teresa Lozano; Jacinto López-Sagaseta; Laura Guembe; Pablo Sarobe; Jesús Prieto; Francisco Borrás-Cuesta; Juan José Lasarte

Immunosuppressive activity of regulatory T cells (Treg) may contribute to the progression of cancer or infectious diseases by preventing the induction of specific immune responses. Using a phage-displayed random peptide library, we identified a 15-mer synthetic peptide, P60, able to bind to forkhead/winged helix transcription factor 3 (FOXP3), a factor required for development and function of Treg. P60 enters the cells, inhibits FOXP3 nuclear translocation, and reduces its ability to suppress the transcription factors NF-κB and NFAT. In vitro, P60 inhibited murine and human-derived Treg and improved effector T cell stimulation. P60 administration to newborn mice induced a lymphoproliferative autoimmune syndrome resembling the reported pathology in scurfy mice lacking functional Foxp3. However, P60 did not cause toxic effects in adult mice and, when given to BALB/c mice immunized with the cytotoxic T cell epitope AH1 from CT26 tumor cells, it induced protection against tumor implantation. Similarly, P60 improved the antiviral efficacy of a recombinant adenovirus expressing NS3 protein from hepatitis C virus. Functional inhibition of Treg by the FOXP3-inhibitory peptide P60 constitutes a strategy to enhance antitumor and antiviral immunotherapies.


Journal of Immunology | 2008

In vitro and in vivo down-regulation of regulatory T cell activity with a peptide inhibitor of TGF-beta1

Lucía Gil-Guerrero; Javier Dotor; Inge L. Huibregtse; Noelia Casares; Ana Belén López-Vázquez; Francesc Rudilla; José Ignacio Riezu-Boj; Jacinto López-Sagaseta; José Hermida; Sander J. H. van Deventer; Jaione Bezunartea; Diana Llopiz; Pablo Sarobe; Jesús Prieto; Francisco Borrás-Cuesta; Juan José Lasarte

Down-regulation of CD4+CD25+ regulatory T (Treg) cell function might be beneficial to enhance the immunogenicity of viral and tumor vaccines or to induce breakdown of immunotolerance. Although the mechanism of suppression used by Treg cells remains controversial, it has been postulated that TGF-β1 mediates their immunosuppressive activity. In this study, we show that P17, a short synthetic peptide that inhibits TGF-β1 and TGF-β2 developed in our laboratory, is able to inhibit Treg activity in vitro and in vivo. In vitro studies demonstrate that P17 inhibits murine and human Treg-induced unresponsiveness of effector T cells to anti-CD3 stimulation, in an MLR or to a specific Ag. Moreover, administration of P17 to mice immunized with peptide vaccines containing tumor or viral Ags enhanced anti-vaccine immune responses and improved protective immunogenicity against tumor growth or viral infection or replication. When CD4+ T cells purified from OT-II transgenic mice were transferred into C57BL/6 mice bearing s.c. EG.7-OVA tumors, administration of P17 improved their proliferation, reduced the number of CD4+Foxp3+ T cells, and inhibited tumor growth. Also, P17 prevented development of immunotolerance induced by oral administration of OVA by genetically modified Lactococcus lactis in DO11.10 transgenic mice sensitized by s.c. injection of OVA. These findings demonstrate that peptide inhibitors of TGF-β may be a valuable tool to enhance vaccination efficacy and to break tolerance against pathogens or tumor Ags.


Hypertension | 2009

Losartan Metabolite EXP3179 Blocks NADPH Oxidase-Mediated Superoxide Production by Inhibiting Protein Kinase C: Potential Clinical Implications in Hypertension

Ana Fortuño; Julen Bidegain; Pablo A. Robador; José Hermida; Jacinto López-Sagaseta; Oscar Beloqui; Javier Díez; Guillermo Zalba

Oxidative stress plays a critical role in the pathogenesis of hypertension. The NADPH oxidase constitutes a major source of superoxide anion in phagocytic cells, and its activation is associated with matrix metalloproteinase (MMP)-9 secretion by these cells. We investigated the effects of the angiotensin II type 1 receptor antagonist losartan and its metabolites (EXP3174 and EXP3179) on NADPH oxidase activity and MMP-9 secretion in human phagocytic cells. EXP3179, but not losartan and EXP3174, dose-dependently inhibited (P<0.05) phorbol myristate acetate and insulin-stimulated NADPH oxidase activity. EXP3179 also inhibited phorbol myristate acetate–induced NADPH oxidase in endothelial cells. In addition, EXP3179 inhibited (P<0.05) both phorbol myristate acetate–stimulated p47phox translocation from cytosol to membranes and protein kinase C activity. Affinity experiments and enzymatic assays confirmed that EXP3179 inhibited several protein kinase C isoforms. EXP3179 also inhibited (P<0.05) phorbol myristate acetate–stimulated MMP-9 secretion. In a study performed in 153 hypertensive patients, phagocytic NADPH oxidase activity was lower (P<0.05) in losartan-treated compared with untreated patients and in patients treated with other angiotensin II type 1 receptor antagonists or with angiotensin-converting enzyme inhibitors. Plasma levels of MMP-9 were lower (P<0.05) in losartan-treated hypertensives compared with the other group of patients. Thus, EXP3179 acts as a blocker of the NADPH oxidase in phagocytic cells by a potential mechanism that targets the protein kinase C signaling pathway. This effect can be involved in reduced MMP-9 secretion by these cells. It is proposed that the EXP3179 metabolite may confer to losartan the specific capacity to reduce oxidative stress mediated by phagocytic cells in hypertensive patients.


Thrombosis and Haemostasis | 2010

Blocking endothelial protein C receptor (EPCR) accelerates thrombus development in vivo

Miguel N. Centelles; Cristina Puy; Jacinto López-Sagaseta; Kenji Fukudome; Ramón Montes; José Hermida

The endothelial protein C receptor (EPCR) plays an anticoagulant role by improving protein C activation. Although low levels of activated protein C (APC) constitute a thrombosis risk factor, the relationship between modulating EPCR function and thrombosis has not been addressed so far. Monoclonal antibodies (mAb) against murine EPCR were raised, and their ability to block protein C/APC binding was tested. The ferric chloride carotid artery injury model in mice was chosen to test the effect of anti-EPCR mAb on thrombus formation. The time to total occlusion of the vessel was analysed in three groups, given an isotype control mAb (IC), a blocking (RCR-16) or a non-blocking (RCR-20) anti-EPCR mAb. RCR-16 prevented the interaction between protein C/APC and EPCR as demonstrated by surface plasmon resonance and flow cytometry, and inhibited the activation of protein C on the endothelium. IC and RCR-20 were unable to induce such effects. In vivo , RCR-16 shortened the time to total vessel occlusion with respect to IC [13.4 +/- 1.0 (mean +/- SD) and 17.8 +/- 3.2 minutes, respectively, p<0.001]. Occlusive thrombi lasting for more than one hour were observed in all RCR-16-treated animals, but only in 43% of IC-treated ones. Results with RCR-20 were indistinguishable from those observed with IC. For the first time, a direct relationship between blocking EPCR and thrombosis is demonstrated. Blocking anti-EPCR autoantibodies can predispose to thrombosis episodes and may constitute a new therapeutic target.


Blood | 2012

sPLA2-V inhibits EPCR anticoagulant and antiapoptotic properties by accommodating lysophosphatidylcholine or PAF in the hydrophobic groove

Jacinto López-Sagaseta; Cristina Puy; Ibai Tamayo; Mikel Allende; Jorge Cerveró; Susana E. Velasco; Charles T. Esmon; Ramón Montes; José Hermida

The endothelial protein C receptor (EPCR) plays an important role in cardiovascular disease by binding protein C/activated protein C (APC). EPCR structure contains a hydrophobic groove filled with an unknown phospholipid needed to perform its function. It has not been established whether lipid exchange takes place in EPCR as a regulatory mechanism of its activity. Our objective was to identify this phospholipid and to explore the possibility of lipid exchange as a regulatory mechanism of EPCR activity driven by the endothelially expressed secretory group V phospholipase A(2) (sPLA(2)-V). We identified phosphatidylcholine (PCh) as the major phospholipid bound to human soluble EPCR (sEPCR). PCh in EPCR could be exchanged for lysophosphatidylcholine (lysoPCh) and platelet activating factor (PAF). Remarkably, lysoPCh and PAF impaired the protein C binding ability of sEPCR. Inhibition of sPLA(2)-V, responsible for lysoPCh and PAF generation, improved APC binding to endothelial cells. EPCR-dependent protein C activation and APC antiapoptotic effect were thus significantly enhanced. In contrast, endothelial cell supplementation with sPLA(2)-V inhibited both APC generation and its antiapoptotic effects. We conclude that APC generation and function can be modulated by changes in phospholipid occupancy of its endothelial cell receptor.


Hepatology | 2011

Improved dendritic cell‐based immunization against hepatitis C virus using peptide inhibitors of interleukin 10

Nancy Díaz-Valdés; Lorea Manterola; Virginia Belsúe; José Ignacio Riezu-Boj; Esther Larrea; Itziar Echeverria; Diana Llopiz; Jacinto López-Sagaseta; Hervé Lerat; Jean-Michel Pawlotsky; Jesús Prieto; Juan J. Lasarte; Francisco Borrás-Cuesta; Pablo Sarobe

The high levels of interleukin 10 (IL‐10) present in chronic hepatitis C virus (HCV) infection have been suggested as responsible for the poor antiviral cellular immune responses found in these patients. To overcome the immunosuppressive effect of IL‐10 on antigen‐presenting cells such as dendritic cells (DCs), we developed peptide inhibitors of IL‐10 to restore DC functions and concomitantly induce efficient antiviral immune responses. Two IL‐10‐binding peptides (p9 and p13) were selected using a phage‐displayed library and their capacity to inhibit IL‐10 was assessed in a bioassay and in STAT‐3 (signal transducer and activator of transcription 3) phosphorylation experiments in vitro. In cultures of human leukocytes where HCV core protein induces the production of IL‐10, p13 restored the ability of plasmacytoid DC to produce interferon alpha (IFN‐α) after Toll‐like receptor 9 (TLR9) stimulation. Similarly, when myeloid DCs were stimulated with CD40L in the presence of HCV core, p9 enhanced IL‐12 production by inhibiting HCV core‐induced as well as CD40L‐induced IL‐10. Moreover, in vitro, p13 potentiated the effect of maturation stimuli on human and murine DC, increasing their IL‐12 production and stimulatory activity, which resulted in enhanced proliferation and IFN‐γ production by responding T‐cells. Finally, immunization with p13‐treated murine DC induced stronger anti‐HCV T‐cell responses not only in wildtype mice but also in HCV transgenic mice and in mice transiently expressing HCV core in the liver. Conclusion: These results suggest that IL‐10 inhibiting peptides may have important applications to enhance anti‐HCV immune responses by restoring the immunostimulatory capabilities of DC. (HEPATOLOGY 2011.)


British Journal of Haematology | 2010

The endothelial cells downregulate the generation of factor VIIa through EPCR binding

Cristina Puy; Jacinto López-Sagaseta; José Hermida; Ramón Montes

Traces of activated factor VII (FVIIa) are required to maintain haemostasis. Activated factor X (FXa) is the main activator of FVII in the absence of tissue factor. However, little is known about how this mechanism is regulated. We and others reported the interaction between FVII and the endothelial cell protein C receptor (EPCR). We have analysed the role of EPCR in the FXa‐dependent FVIIa generation. Activation was performed on the surface of human aortic endothelial cells in the presence or absence of a blocking anti‐EPCR monoclonal antibody (mAb). Western‐blot analyses revealed that FVII activation was increased twofold upon EPCR blocking. Kinetic analyses revealed that blocking doubled the catalytic efficiency for activation. Protein C was unable to mimic the effect of the anti‐EPCR mAb on activation. Surface plasmon resonance experiments revealed that binding of EPCR and phospholipids to FVII were mutually exclusive. The 50% inhibitory concentration value for phospholipids to reduce the binding of FVIIa to EPCR was 57·67 ± 0·11 μmol/l. Immunofluorescence experiments showed that EPCR and phosphatidylserine are located at different regions of the cell surface. We propose that EPCR downregulates FVII activation by moving it from phosphatidylserine‐rich regions. In summary, this study described a new anticoagulant role for EPCR.


Haematologica | 2008

The functional properties of a truncated form of endothelial cell protein C receptor generated by alternative splicing

Eva Molina; José Hermida; Jacinto López-Sagaseta; Cristina Puy; Ramón Montes

Background A soluble form of endothelial cell protein C receptor (sEPCR) is generated by shedding of the cellular form. sEPCR binds to protein C and factor VIIa and inhibits both the activation of protein C and the activity of activated protein C and factor VIIa. High sEPCR levels may increase the risk of thrombosis.We wanted to explore the possibility of detecting soluble endothelial cell protein C receptor forms generated by alternative splicing. Design and Methods Reverse transcriptase polymerase chain reaction was used to look for new forms of endothelial cell protein C receptor. A yeast expression system was used to generate sufficient amounts of the distinct sEPCR forms. Surface plasmon resonance experiments, chromogenic assays, clotting assays and immunoassays were subsequently performed to characterize a new form of sEPCR that was found. Results We demonstrated, by reverse transcriptase polymerase chain reaction and sequencing, the existence of a new, soluble form of endothelial cell protein C receptor generated by alternative splicing, in which the transmembrane region is replaced by a 56-residue tail (tEPCR). Its cDNA was present in human umbilical vein endothelial cells and in most tissues as well as in lung cancer cells. tEPCR was not located in the membrane of transfected cells.We demonstrated that tEPCR binds to protein C and factor VIIa. tEPCR blocked the generation of activated protein C and inhibited the activity of both activated protein C and factor VIIa. tEPCR was detected, by immunoassays, in the supernatant of lung cancer cells and human umbilical vein endothelial cells. Conclusions A truncated form of alternatively spliced endothelial cell protein C receptor was detected in the endothelium and cancer cells. tEPCR behaves as sEPCR generated by shedding of the cellular endothelial cell protein C receptor.


Protein Expression and Purification | 2009

Recombinant expression of biologically active murine soluble EPCR

Jacinto López-Sagaseta; Ramón Montes; José Hermida

Endothelial cell protein C receptor (EPCR) downregulates the coagulation system and prevents thrombosis by binding to protein C/activated protein C (APC) and factor VII/activated factor VII (VIIa). Recombinant APC and factor VIIa have been shown to be useful in a variety of clinical conditions. Murine models could prove extremely helpful in order to study in vivo actions of these drugs. It is therefore crucial to demonstrate the interaction between these and murine EPCR. We expressed the extracellular region of the murine EPCR in a yeast expression system and obtained a colony of Pichia pastoris that produce high amounts of recombinant extracellular murine EPCR, which we purified by liquid chromatography to homogeneity. The analysis of the interaction of EPCR with APC and factor VIIa was carried out using surface plasmon resonance technology. Murine EPCR binds to APC and factor VIIa with similar affinity than human EPCR. As for human EPCR, the binding is Ca2+ dependent while Mg2+ ions optimize it. In conclusion, we succeeded in establishing a system to produce enough recombinant soluble murine EPCR to perform biochemical studies. Murine EPCR binds to human APC and factor VIIa, which opens up new possibilities for characterizing the in vivo effect of APC and factor VII by using murine models.

Collaboration


Dive into the Jacinto López-Sagaseta's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge