14 research outputs found

    Blood biomarker changes following therapeutic hypothermia in ischemic stroke

    Get PDF
    Biomarkers; Hypothermia; IschemiaBiomarcadores; Hipotermia; IsquemiaBiomarcadors; Hipotèrmia; IsquèmiaIntroduction Therapeutic hypothermia is a promising candidate for stroke treatment although its efficacy has not yet been demonstrated in patients. Changes in blood molecules could act as surrogate markers to evaluate the efficacy and safety of therapeutic cooling. Methods Blood samples from 54 patients included in the EuroHYP-1 study (27 treated with hypothermia, and 27 controls) were obtained at baseline, 24 ± 2 h, and 72 ± 4 h. The levels of a panel of 27 biomarkers, including matrix metalloproteinases and cardiac and inflammatory markers, were measured. Results Metalloproteinase-3 (MMP-3), fatty-acid-binding protein (FABP), and interleukin-8 (IL-8) increased over time in relation to the hypothermia treatment. Statistically significant correlations between the minimum temperature achieved by each patient in the hypothermia group and the MMP-3 level measured at 72 h, FABP level measured at 24 h, and IL-8 levels measured at 24 and 72 h were found. No differential biomarker levels were observed in patients with poor or favorable outcomes according to modified Rankin Scale scores. Conclusion Although the exact roles of MMP3, FABP, and IL-8 in hypothermia-treated stroke patients are not known, further exploration is needed to confirm their roles in brain ischemia

    Peripheral inflammation preceeding ischemia impairs neuronal survival through mechanisms involving miR‐127 in aged animals

    Get PDF
    Envelliment; Inflamació; MicroARNEnvejecimiento; Inflamación; MicroARNAging; Inflammation; MicroRNAIschemic stroke, the third leading cause of death in the Western world, affects mainly the elderly and is strongly associated with comorbid conditions such as atherosclerosis or diabetes, which are pathologically characterized by increased inflammation and are known to influence the outcome of stroke. Stroke incidence peaks during influenza seasons, and patients suffering from infections such as pneumonia prior to stroke exhibit a worse stroke outcome. Earlier studies have shown that comorbidities aggravate the outcome of stroke, yet the mediators of this phenomenon remain obscure. Here, we show that acute peripheral inflammation aggravates stroke‐induced neuronal damage and motor deficits specifically in aged mice. This is associated with increased levels of plasma proinflammatory cytokines, rather than with an increase of inflammatory mediators in the affected brain parenchyma. Nascent transcriptomics data with mature microRNA sequencing were used to identify the neuron‐specific miRNome, in order to decipher dysregulated miRNAs in the brains of aged animals with stroke and co‐existing inflammation. We pinpoint a previously uninvestigated miRNA in the brain, miR‐127, that is highly neuronal, to be associated with increased cell death in the aged, LPS‐injected ischemic mice. Target prediction tools indicate that miR‐127 interacts with several basally expressed neuronal genes, and of these we verify miR‐127 binding to Psmd3. Finally, we report reduced expression of miR‐127 in human stroke brains. Our results underline the impact of peripheral inflammation on the outcome of stroke in aged subjects and pinpoint molecular targets for restoring endogenous neuronal capacity to combat ischemic stroke.This study was supported by Emil Aaltonen Foundation, Academy of Finland and Finnish Cultural Foundation

    Blood Biomarker Panels for the Early Prediction of Stroke‐Associated Complications

    Get PDF
    Biomarkers; Stroke; Stroke‐associated infectionBiomarcadors; Ictus; Infecció associada a un ictusBiomarcadores; Ictus; Infección asociada a un ictusBackground Acute decompensated heart failure (ADHF) and respiratory tract infections (RTIs) are potentially life‐threatening complications in patients experiencing stroke during hospitalization. We aimed to test whether blood biomarker panels might predict these complications early after admission. Methods and Results Nine hundred thirty‐eight patients experiencing ischemic stroke were prospectively recruited in the Stroke‐Chip study. Post‐stroke complications during hospitalization were retrospectively evaluated. Blood samples were drawn within 6 hours after stroke onset, and 14 biomarkers were analyzed by immunoassays. Biomarker values were normalized using log‐transformation and Z score. PanelomiX algorithm was used to select panels with the best accuracy for predicting ADHF and RTI. Logistic regression models were constructed with the clinical variables and the biomarker panels. The additional predictive value of the panels compared with the clinical model alone was evaluated by receiver operating characteristic curves. An internal validation through a 10‐fold cross‐validation with 3 repeats was performed. ADHF and RTI occurred in 19 (2%) and 86 (9.1%) cases, respectively. Three‐biomarker panels were developed as predictors: vascular adhesion protein‐1 >5.67, NT‐proBNP (N‐terminal pro‐B‐type natriuretic peptide) >4.98 and d‐dimer >5.38 (sensitivity, 89.5%; specificity, 71.7%) for ADHF; and interleukin‐6 >3.97, von Willebrand factor >3.67, and d‐dimer >4.58 (sensitivity, 82.6%; specificity, 59.8%) for RTI. Both panels independently predicted stroke complications (panel for ADHF: odds ratio [OR] [95% CI], 10.1 [3–52.2]; panel for RTI: OR, 3.73 [1.95–7.14]) after adjustment by clinical confounders. The addition of the panel to clinical predictors significantly improved areas under the curve of the receiver operating characteristic curves in both cases. Conclusions Blood biomarkers could be useful for the early prediction of ADHF and RTI. Future studies should assess the usefulness of these panels in front of patients experiencing stroke with respiratory symptoms such as dyspnea.This project received funding from Instituto de Salud Carlos III (ISCIII) [DTS14/00004, PI17/02130], co‐financed by the European Regional Development Fund (FEDER), and from Fundació La Marató de TV3 [201706] and the European Union's Horizon 2020 research and innovation program [754517]. Neurovascular Research Laboratory takes part into the Spanish stroke research network INVICTUS+ (RD16/0019/0021). The funders had no role in the study design and conduction

    The effects of mango leaf extract during adolescence and adulthood in a rat model of schizophrenia.

    Get PDF
    There is evidence that in schizophrenia, imbalances in inflammatory and oxidative processes occur during pregnancy and in the early postnatal period, generating interest in the potential therapeutic efficacy of anti-inflammatory and antioxidant compounds. Mangiferin is a polyphenolic compound abundant in the leaves of Mangifera indica L. that has robust antioxidant and anti-inflammatory properties, making it a potential candidate for preventive or co-adjuvant therapy in schizophrenia. Hence, this study set-out to evaluate the effect of mango leaf extract (MLE) in a model of schizophrenia based on maternal immune activation, in which Poly I:C (4 mg/kg) is administered intravenously to pregnant rats. Young adult (postnatal day 60-70) or adolescent (postnatal day 35-49) male offspring received MLE (50 mg/kg of mangiferin) daily, and the effects of MLE in adolescence were compared to those of risperidone, assessing behavior, brain magnetic resonance imaging (MRI), and oxidative/inflammatory and antioxidant mediators in the adult offspring. MLE treatment in adulthood reversed the deficit in prepulse inhibition (PPI) but it failed to attenuate the sensitivity to amphetamine and the deficit in novel object recognition (NOR) induced. By contrast, adolescent MLE treatment prevented the sensorimotor gating deficit in the PPI test, producing an effect similar to that of risperidone. This MLE treatment also produced a reduction in grooming behavior, but it had no effect on anxiety or novel object recognition memory. MRI studies revealed that adolescent MLE administration partially counteracted the cortical shrinkage, and cerebellum and ventricle enlargement. In addition, MLE administration in adolescence reduced iNOS mediated inflammatory activation and it promoted the expression of biomarkers of compensatory antioxidant activity in the prefrontal cortex and hippocampus, as witnessed through the reduction of Keap1 and the accumulation of NRF2 and HO1. Together, these findings suggest that MLE might be an alternative therapeutic or preventive add-on strategy to improve the clinical expression of schizophrenia in adulthood, while also modifying the time course of this disease at earlier stages in populations at high-risk.EB, JAG-P and ST-S work was supported by the “Fondo Europeo de Desarrollo Regional” (FEDER)-UE “A way to build Europe” from the “Ministerio de Economía y Competitividad” (RTI2018-099778-B-I00); from the “Plan Nacional sobre Drogas, Ministerio de Sanidad, Consumo y Bienestar Social” (2019I041); from the “Ministerio de Salud-Instituto de Salud Carlos III” (PI18/01691); from the “Programa Operativo de Andalucía FEDER, Iniciativa Territorial Integrada ITI 2014-2020 Consejería Salud y Familias, Junta de Andalucía” (PI-0080- 2017, PI-0009-2017), “Consejería de Salud y Familias, Junta de Andalucía” (PI-0134-2018 and PEMP-0008-2020); from the “Consejería de Transformación Económica, Industria, Conocimiento y Universidad, Junta de Andalucía” (P20_00958 and CTS-510); from the CEIMAR (CEIJ-003); from the “Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz-INiBICA” (LI19/06IN-CO22; IN-C09); from the “CIBERSAM”: CIBER-Consorcio Centro de Investigación Biomédica en Red- (CB07/09/0033), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación and from the European Union’s Horizon 2020 research and innovation programme under the Marie Sklodowska-Curie grant agreement No 955684. CM, LC and MTF-P were supported by the Spanish Ministry of Science and Technology (PID2020- 116229RB-I00) and European Regional Development Fund (ERDF). KM and JCL were supported by the “MICINN” (PID2019-109033RB-I00) and the “CIBERSAM”: CIBERConsorcio Centro de Investigación Biomédica en Red- (CB07/ 09/0026), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación. MLS-M was supported by the “Ministerio de Ciencia, Innovación, Instituto de Salud Carlos III” (PI17/ 01766, BA21/00030), co-financed by European Regional Development Fund (ERDF), “A way to make Europe”; from the “CIBERSAM”: CIBERConsorcio Centro de Investigación Biomédica en Red- (CB07/09/0031), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación; from the “Delegación del Gobierno para el Plan Nacional sobre Drogas” (2017/085); from the “Fundación Mapfre” and “Fundación Alicia Koplowitz.” MD work was supported by the “Ministerio de Ciencia e In review 18/ 28 Innovación” (MCIN) and “Instituto de Salud Carlos III” (ISCIII) (PT20/00044); from the “CIBERSAM” CIBERConsorcio Centro de Investigación Biomédica en Red-(CB07/ 09/0031). The CNIC is supported by the ISCIII, the MCIN and the Pro CNIC Foundation, and is a Severo Ochoa Center of Excellence (SEV-2015-0505).S

    The effects of mango leaf extract during adolescence and adulthood in a rat model of schizophrenia

    Get PDF
    There is evidence that in schizophrenia, imbalances in inflammatory and oxidative processes occur during pregnancy and in the early postnatal period, generating interest in the potential therapeutic efficacy of anti-inflammatory and antioxidant compounds. Mangiferin is a polyphenolic compound abundant in the leaves of Mangifera indica L. that has robust antioxidant and anti-inflammatory properties, making it a potential candidate for preventive or co-adjuvant therapy in schizophrenia. Hence, this study set-out to evaluate the effect of mango leaf extract (MLE) in a model of schizophrenia based on maternal immune activation, in which Poly I:C (4 mg/kg) is administered intravenously to pregnant rats. Young adult (postnatal day 60–70) or adolescent (postnatal day 35–49) male offspring received MLE (50 mg/kg of mangiferin) daily, and the effects of MLE in adolescence were compared to those of risperidone, assessing behavior, brain magnetic resonance imaging (MRI), and oxidative/inflammatory and antioxidant mediators in the adult offspring. MLE treatment in adulthood reversed the deficit in prepulse inhibition (PPI) but it failed to attenuate the sensitivity to amphetamine and the deficit in novel object recognition (NOR) induced. By contrast, adolescent MLE treatment prevented the sensorimotor gating deficit in the PPI test, producing an effect similar to that of risperidone. This MLE treatment also produced a reduction in grooming behavior, but it had no effect on anxiety or novel object recognition memory. MRI studies revealed that adolescent MLE administration partially counteracted the cortical shrinkage, and cerebellum and ventricle enlargement. In addition, MLE administration in adolescence reduced iNOS mediated inflammatory activation and it promoted the expression of biomarkers of compensatory antioxidant activity in the prefrontal cortex and hippocampus, as witnessed through the reduction of Keap1 and the accumulation of NRF2 and HO1. Together, these findings suggest that MLE might be an alternative therapeutic or preventive add-on strategy to improve the clinical expression of schizophrenia in adulthood, while also modifying the time course of this disease at earlier stages in populations at high-risk

    Identificación y uso de biomarcadores pronósticos en el ictus isquémico

    Get PDF
    El ictus es una enfermedad con una elevada morbimortalidad, suponiendo la segunda causa de muerte a nivel mundial y una de las principales causas de discapacidad en el adulto. Aun así, la heterogeneidad entre los pacientes con ictus dificulta anticipar el pronóstico de la enfermedad. Saber qué pacientes van a empeorar o cuáles se recuperarán sin secuelas ayudaría a facilitar información al paciente y su familia, valorar los riesgos y los beneficios del tratamiento trombolítico o la inclusión en ensayos clínicos y a optimizar los recursos sanitarios en cuanto a atención médica especializada, cuidado intensivo de los pacientes, duración de la estancia intrahospitalaria o terapias de rehabilitación tempranas. Actualmente, la predicción de la evolución clínica de los pacientes con ictus es poco precisa y suele basarse en la edad y la gravedad inicial del paciente. Aunque se han desarrollado diversos modelos predictivos basados en variables clínicas, en la práctica ninguno de ellos es aplicado al presentar valores de discriminación entre pacientes que, en el mejor de los casos, alcanzan el 70-80 % de precisión. Además, estos modelos suelen incluir variables clínicas que no son accesibles de forma inmediata o que requieren técnicas de neuroimagen. El uso de biomarcadores en la práctica clínica en otras enfermedades ha generado una corriente de investigación en torno a moléculas que puedan predecir la evolución de los pacientes con ictus. En la actualidad, pocos biomarcadores han demostrado tener un valor predictivo suficiente como para que se evalúe su utilidad en amplios estudios multicéntricos; para ello, los biomarcadores deberían añadir valor a las variables clínicas, aumentando la sensibilidad y la especificidad en la discriminación de los pacientes para influir en la toma de decisiones. Los trabajos incluidos en esta Tesis se centran, por un lado, en sistematizar el conocimiento sobre biomarcadores pronósticos en el ictus isquémico y la aplicación de criterios estadísticos que demuestren el valor añadido necesario para un futuro uso de los marcadores. Con ese propósito hemos desarrollado una página web donde se compila la información sobre las moléculas que se han asociado con el pronóstico del ictus y hemos aplicado técnicas de estadística comparativa y de metaanálisis para evaluar un biomarcador típico como el péptido natriurético de tipo B (BNP). Por otro lado, hemos querido evaluar moléculas involucradas en la fisiopatología del ictus, como las quimiocinas, explorándolas paralelamente a nivel cerebral y circulatorio. Finalmente, mediante técnicas de identificación masiva, como la proteómica, las librerías de anticuerpos y la transcriptómica, hemos identificado una serie de moléculas que han demostrado su asociación con el pronóstico del ictus en diferentes momentos de la evolución de los pacientes. Además, estas moléculas mejoran, en mayor o menor grado, la predicción basada solamente en variables clínicas. Valorar éstos u otros biomarcadores en las diferentes complicaciones secundarias al ictus isquémico en las que podrían tomarse decisiones que cambiasen el curso de la enfermedad, como la predicción de las infecciones post-ictus o la transformación hemorrágica, parece una excelente oportunidad de futuro para la aplicación real de los biomarcadores en el ictus.Stroke is a severe disease, being the second cause of death worldwide and one of the main causes of disability. Heterogeneity among stroke patients makes difficult to anticipate the prognosis of the disease. To identify patients who are going to worsen or will recover would help in giving valuable information to the patient and relatives; to evaluate risks and benefits of reperfusion therapies or the inclusion in clinical trials and to optimize public health resources in terms of specialized medical and intensive care, length of in-hospital stay or starting of early rehabilitation programs. Nowadays, clinical outcome prediction of stroke patients lacks accuracy and is usually based mainly on age and initial neurological severity. Although several predictive models have been developed including clinical variables, none of them is being applied in clinical practice. In the best-case scenario, these models differentiate patients with 70-80 % accuracy. Furthermore they tend to include clinical variables non-easily available or based on neuroimaging techniques. The use of biomarkers in the clinical practice in other diseases generated a trend in research around molecules that can predict stroke patients' outcome. Currently, few biomarkers have demonstrated enough predictive value to be evaluated in large multicenter studies; for that purpose, biomarkers should add value to clinical variables, increasing sensitivity and specificity in the discrimination of patients in order to influence decision-making processes. The papers comprised in this Thesis focus, on the one hand, on systematizing the knowledge about prognostic biomarkers in ischemic stroke and the application of statistical criteria that demonstrate the needed added value for a future use of the biomarkers. For that purpose we have developed a website to provide a data compilation of molecules that have been associated with stroke prognosis and we have applied comparative statistics and metaanalysis to evaluate a typical biomarker, such as natriuretic peptide type B (BNP). On the other hand, we aimed to evaluate molecules that are involved in the pathophysiology of stroke, such as chemokines, by analyzing them in parallel at both brain and blood levels. Finally, through massive discovery techniques, such as proteomics, antibody libraries and transcriptomics, we identified several molecules that demonstrated their association with stroke prognosis at different moments during the progression of the disease. Moreover, these molecules improved prediction based solely on clinical variables. To evaluate utility of these or other biomarkers in some of the secondary complications of ischemic stroke, where decisions can be made to change the evolution of the disease, such as the prediction of post-stroke infections or hemorrhagic transformation, seems an excellent opportunity for a real use of stroke biomarkers in the near future

    B-type natriuretic peptides and mortality after stroke A systematic review and meta-analysis

    No full text
    Saritas, Ayhan/0000-0002-4302-1093; Montaner, Joan/0000-0003-4845-2279; Saritas, Ayhan/0000-0002-4302-1093; GARCIA-BERROCOSO, TERESA/0000-0001-8072-8533; Whiteley, William/0000-0002-4816-8991; Khedri Jensen, Jesper/0000-0001-7426-4437WOS: 000330771700007PubMed: 24186915Objective: To measure the association of B-type natriuretic peptide (BNP) and N-terminal fragment of BNP (NT-proBNP) with all-cause mortality after stroke, and to evaluate the additional predictive value of BNP/NT-proBNP over clinical information. Methods: Suitable studies for meta-analysis were found by searching MEDLINE and EMBASE databases until October 26, 2012. Weighted mean differences measured effect size; meta-regression and publication bias were assessed. Individual participant data were used to estimate effects by logistic regression and to evaluate BNP/NT-proBNP additional predictive value by area under the receiver operating characteristic curves, and integrated discrimination improvement and categorical net reclassification improvement indexes. Results: Literature-based meta-analysis included 3,498 stroke patients from 16 studies and revealed that BNP/NT-proBNP levels were 255.78 pg/mL (95% confidence interval [CI] 105.10-406.47, p = 0.001) higher in patients who died; publication bias entailed the loss of this association. Individual participant data analysis comprised 2,258 stroke patients. After normalization of the data, patients in the highest quartile had double the risk of death after adjustment for clinical variables (NIH Stroke Scale score, age, sex) (odds ratio 2.30, 95% CI 1.32-4.01 for BNP; and odds ratio 2.63, 95% CI 1.75-3.94 for NT-proBNP). Only NT-proBNP showed a slight added value to clinical prognostic variables, increasing discrimination by 0.028 points (integrated discrimination improvement index; p < 0.001) and reclassifying 8.1% of patients into correct risk mortality categories (net reclassification improvement index; p = 0.003). Neither etiology nor time from onset to death affected the association of BNP/NT-proBNP with mortality. Conclusion: BNPs are associated with poststroke mortality independent of NIH Stroke Scale score, age, and sex. However, their translation to clinical practice seems difficult because BNP/NT-proBNP add only minor predictive value to clinical information.Instituto de Salud Carlos IIIInstituto de Salud Carlos III [FI09/00017]; FIS [11/0176]; Chief Scientist's Office [CAF/06/30]; UK Medical Research Council Clinician Scientist FellowshipMedical Research Council UK (MRC) [G0902303]; Medical Research CouncilMedical Research Council UK (MRC) [G0902303]; Chief Scientist Office [CAF/06/30]T. Garcia-Berrocoso is supported by a predoctoral fellowship (FI09/00017) from the Instituto de Salud Carlos III. Neurovascular Research Laboratory takes part in the Spanish stroke research network INVICTUS (RD12/0014/0005) and is supported on stroke biomarkers research by FIS 11/0176. D. Giralt, A. Bustamante, T. Etgen, J. Jensen, J. Sharma, K. Shibazaki, A. Saritas, and X. Chen report no disclosures. W. Whiteley was supported by the Chief Scientist's Office (CAF/06/30) and is now funded by a UK Medical Research Council Clinician Scientist Fellowship (G0902303). J. Montaner reports no disclosures. Go to Neurology.org for full disclosures

    A Mouse Brain-based Multi-omics Integrative Approach Reveals Potential Blood Biomarkers for Ischemic Stroke

    Full text link
    Stroke remains a leading cause of death and disability worldwide. Despite continuous advances, the identification of key molecular signatures in the hyper-acute phase of ischemic stroke is still a primary interest for translational research on stroke diagnosis, prognosis, and treatment. Data integration from high-throughput -omics techniques has become crucial to unraveling key interactions among different molecular elements in complex biological contexts, such as ischemic stroke. Thus, we used advanced data integration methods for a multi-level joint analysis of transcriptomics and proteomics data sets obtained from mouse brains at 2 h after cerebral ischemia. By modeling net-like correlation structures, we identified an integrated network of genes and proteins that are differentially expressed at a very early stage after stroke. We validated 10 of these deregulated elements in acute stroke, and changes in their expression pattern over time after cerebral ischemia were described. Of these, CLDN20, GADD45G, RGS2, BAG5, and CTNND2 were next evaluated as blood biomarkers of cerebral ischemia in mice and human blood samples, which were obtained from stroke patients and patients presenting stroke-mimicking conditions. Our findings indicate that CTNND2 levels in blood might potentially be useful for distinguishing ischemic strokes from stroke-mimicking conditions in the hyper-acute phase of the disease. Furthermore, circulating GADD45G content within the first 6 h after stroke could also play a key role in predicting poor outcomes in stroke patients. For the first time, we have used an integrative biostatistical approach to elucidate key molecules in the initial stages of stroke pathophysiology and highlight new notable molecules that might be further considered as blood biomarkers of ischemic stroke

    Antibodies Preventing the Interaction of Tissue-Type Plasminogen Activator With N-Methyl- d -Aspartate Receptors Reduce Stroke Damages and Extend the Therapeutic Window of Thrombolysis

    No full text
    International audienceBackground and Purpose— Tissue-type plasminogen activator (tPA) is the only drug approved for the acute treatment of ischemic stroke but with two faces in the disease: beneficial fibrinolysis in the vasculature and damaging effects on the neurovascular unit and brain parenchyma. To improve this profile, we developed a novel strategy, relying on antibodies targeting the proneurotoxic effects of tPA. Methods— After production and characterization of antibodies (αATD-NR1) that specifically prevent the interaction of tPA with the ATD-NR1 of N-methyl- d -aspartate receptors, we have evaluated their efficacy in a model of murine thromboembolic stroke with or without recombinant tPA-induced reperfusion, coupled to MRI, near-infrared fluorescence imaging, and behavior assessments. Results— In vitro, αATD-NR1 prevented the proexcitotoxic effect of tPA without altering N-methyl- d -aspartate-induced neurotransmission. In vivo, after a single administration alone or with late recombinant tPA-induced thrombolysis, antibodies dramatically reduced brain injuries and blood–brain barrier leakage, thus improving long-term neurological outcome. Conclusions— Our strategy limits ischemic damages and extends the therapeutic window of tPA-driven thrombolysis. Thus, the prospect of this immunotherapy is an extension of the range of treatable patients
    corecore