Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(5)2022 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-35269975

RESUMO

Prenylcysteine Oxidase 1 (PCYOX1) is an enzyme involved in the degradation of prenylated proteins. It is expressed in different tissues including vascular and blood cells. We recently showed that the secretome from Pcyox1-silenced cells reduced platelet adhesion both to fibrinogen and endothelial cells, suggesting a potential contribution of PCYOX1 into thrombus formation. Here, we show that in vivo thrombus formation after FeCl3 injury of the carotid artery was delayed in Pcyox1-/- mice, which were also protected from collagen/epinephrine induced thromboembolism. The Pcyox1-/- mice displayed normal blood cells count, vascular procoagulant activity and plasma fibrinogen levels. Deletion of Pcyox1 reduced the platelet/leukocyte aggregates in whole blood, as well as the platelet aggregation, the alpha granules release, and the αIIbß3 integrin activation in platelet-rich plasma, in response to adenosine diphosphate (ADP) or thrombin receptor agonist peptide (TRAP). Washed platelets from the Pcyox1-/- and WT animals showed similar phosphorylation pathway activation, adhesion ability and aggregation. The presence of Pcyox1-/- plasma impaired agonist-induced WT platelet aggregation. Our findings show that the absence of PCYOX1 results in platelet hypo-reactivity and impaired arterial thrombosis, and indicates that PCYOX1 could be a novel target for antithrombotic drugs.


Assuntos
Liases de Carbono-Enxofre/metabolismo , Células Endoteliais , Trombose , Animais , Plaquetas/metabolismo , Cisteína/análogos & derivados , Células Endoteliais/metabolismo , Fibrinogênio/metabolismo , Camundongos , Oxirredutases/metabolismo , Ativação Plaquetária , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Trombose/metabolismo
2.
Int J Mol Sci ; 22(15)2021 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-34360827

RESUMO

The identification of new biomarkers allowing an early and more accurate characterization of patients with ST-segment elevation myocardial infarction (STEMI) is still needed, and exosomes represent an attractive diagnostic tool in this context. However, the characterization of their protein cargo in relation to cardiovascular clinical manifestation is still lacking. To this end, 35 STEMI patients (17 experiencing resuscitated out-of-hospital cardiac arrest (OHCA-STEMI) and 18 uncomplicated) and 32 patients with chronic coronary syndrome (CCS) were enrolled. Plasma exosomes were characterized by the nanoparticle tracking analysis and Western blotting. Exosomes from STEMI patients displayed a higher concentration and size and a greater expression of platelet (GPIIb) and vascular endothelial (VE-cadherin) markers, but a similar amount of cardiac troponin compared to CCS. In addition, a difference in exosome expression of acute-phase proteins (ceruloplasmin, transthyretin and fibronectin) between STEMI and CCS patients was found. GPIIb and brain-associated marker PLP1 accurately discriminated between OHCA and uncomplicated STEMI. In conclusion, the exosome profile of STEMI patients has peculiar features that differentiate it from that of CCS patients, reflecting the pathophysiological mechanisms involved in STEMI. Additionally, the exosome expression of brain- and platelet-specific markers might allow the identification of patients experiencing ischemic brain injury in STEMI.


Assuntos
Exossomos/metabolismo , Parada Cardíaca Extra-Hospitalar/sangue , Infarto do Miocárdio com Supradesnível do Segmento ST/sangue , Idoso , Biomarcadores/sangue , Ceruloplasmina/análise , Exossomos/química , Fibronectinas/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Pré-Albumina/análise , Infarto do Miocárdio com Supradesnível do Segmento ST/complicações , Troponina/sangue
3.
J Cell Physiol ; 235(12): 9667-9675, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32430940

RESUMO

Obesity, a rising public health burden, is a multifactorial disease with an increased risk for patients to develop several pathological conditions including type 2 diabetes mellitus, hypertension, and cardiovascular disease. Increasing evidence suggests a relationship between the human brain-derived neurotrophic factor (BDNF) Val66Met single-nucleotide polymorphism (SNP) and obesity, although the underlying mechanisms of this connection are still not completely understood. In the present study, we found that homozygous knock-in BDNFMet/Met mice were overweight and hyperphagic compared to wildtype BDNFVal/Val mice. Increased food intake was associated with reduction of total BDNF and BDNF1, BDNF4 and BDNF6 transcripts in the hypothalamus of BDNFMet/Met mice. In contrast, in the white adipose tissue total BDNF and Glut4 expression levels were augmented, while sirtuin 1 and leptin receptor (Ob-R) expression levels were reduced in BDNFMet/Met mice. Moreover, plasmatic leptin levels were decreased in BDNFMet/Met mice. However, BDNFVal/Val and BDNFMet/Met mice showed a similar response to the insulin tolerance test and glucose tolerance test. Altogether, these results suggest that BDNF Val66Met SNP strongly contributes to adipose tissue pathophysiology, resulting in reduced circulating leptin levels and hypothalamic expression of BDNF, which, in turn, promote increased food intake and overweight in BDNFMet/Met mice.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Diabetes Mellitus Tipo 2/genética , Ingestão de Alimentos/genética , Transportador de Glucose Tipo 4/genética , Obesidade/genética , Animais , Diabetes Mellitus Tipo 2/patologia , Regulação da Expressão Gênica/genética , Teste de Tolerância a Glucose , Humanos , Hipotálamo/metabolismo , Insulina/metabolismo , Camundongos , Obesidade/patologia , Sobrepeso/genética , Sobrepeso/patologia , Polimorfismo de Nucleotídeo Único/genética , Sirtuína 1/genética
4.
Int J Mol Sci ; 21(21)2020 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-33105629

RESUMO

Psychological stress induces different alterations in the organism in order to maintain homeostasis, including changes in hematopoiesis and hemostasis. In particular, stress-induced hyper activation of the autonomic nervous system and hypothalamic-pituitary-adrenal axis can trigger cellular and molecular alterations in platelets, coagulation factors, endothelial function, redox balance, and sterile inflammatory response. For this reason, mental stress is reported to enhance the risk of cardiovascular disease (CVD). However, contrasting results are often found in the literature considering differences in the response to acute or chronic stress and the health condition of the population analyzed. Since thrombosis is the most common underlying pathology of CVDs, the comprehension of the mechanisms at the basis of the association between stress and this pathology is highly valuable. The aim of this work is to give a comprehensive review of the studies focused on the role of acute and chronic stress in both healthy individuals and CVD patients, focusing on the cellular and molecular mechanisms underlying the relationship between stress and thrombosis.


Assuntos
Doenças Cardiovasculares/psicologia , Estresse Psicológico , Trombose/psicologia , Coagulação Sanguínea/fisiologia , Plaquetas/patologia , Plaquetas/fisiologia , Doenças Cardiovasculares/complicações , Doenças Cardiovasculares/fisiopatologia , Endotélio Vascular/fisiologia , Endotélio Vascular/fisiopatologia , Humanos , Sistema Hipotálamo-Hipofisário/fisiologia , Sistema Hipotálamo-Hipofisário/fisiopatologia , Inflamação/etiologia , Inflamação/psicologia , Estresse Psicológico/complicações , Estresse Psicológico/fisiopatologia , Trombose/etiologia
5.
Int J Mol Sci ; 21(20)2020 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-33066277

RESUMO

Depression is a major cause of morbidity and low quality of life among patients with cardiovascular disease (CVD), and it is now considered as an independent risk factor for major adverse cardiovascular events. Increasing evidence indicates not only that depression worsens the prognosis of cardiac events, but also that a cross-vulnerability between the two conditions occurs. Among the several mechanisms proposed to explain this interplay, platelet activation is the more attractive, seeing platelets as potential mirror of the brain function. In this review, we dissected the mechanisms linking depression and CVD highlighting the critical role of platelet behavior during depression as trigger of cardiovascular complication. In particular, we will discuss the relationship between depression and molecules involved in the CVD (e.g., catecholamines, adipokines, lipids, reactive oxygen species, and chemokines), emphasizing their impact on platelet activation and related mechanisms.


Assuntos
Doenças Cardiovasculares/sangue , Depressão/sangue , Ativação Plaquetária , Adipocinas/sangue , Animais , Doenças Cardiovasculares/complicações , Catecolaminas/sangue , Citocinas/sangue , Depressão/complicações , Humanos , Lipoproteínas LDL/sangue
6.
Int J Mol Sci ; 20(11)2019 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-31212641

RESUMO

Extracellular vesicles (EVs) are well-established mediators of cell-to-cell communication. EVs can be released by every cell type and they can be classified into three major groups according to their biogenesis, dimension, density, and predominant protein markers: exosomes, microvesicles, and apoptotic bodies. During their formation, EVs associate with specific cargo from their parental cell that can include RNAs, free fatty acids, surface receptors, and proteins. The biological function of EVs is to maintain cellular and tissue homeostasis by transferring critical biological cargos to distal or neighboring recipient cells. On the other hand, their role in intercellular communication may also contribute to the pathogenesis of several diseases, including thrombosis. More recently, their physiological and biochemical properties have suggested their use as a therapeutic tool in tissue regeneration as well as a novel option for drug delivery. In this review, we will summarize the impact of EVs released from blood and vascular cells in arterial and venous thrombosis, describing the mechanisms by which EVs affect thrombosis and their potential clinical applications.


Assuntos
Vesículas Extracelulares/metabolismo , Trombose/metabolismo , Animais , Biomarcadores/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Exossomos/metabolismo , Humanos
7.
Eur Heart J ; 38(18): 1426-1435, 2017 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-26705390

RESUMO

AIMS: Epidemiological studies strongly suggest a link between stress, depression, and cardiovascular diseases (CVDs); the mechanistic correlation, however, is poorly understood. A single-nucleotide polymorphism in the BDNF gene (BDNFVal66Met), associated with depression and anxiety, has been proposed as a genetic risk factor for CVD. Using a knock-in mouse carrying the BDNFVal66Met human polymorphism, which phenocopies psychiatric-related symptoms found in humans, we investigated the impact of this SNP on thrombosis. METHODS AND RESULTS: BDNFMet/Met mice displayed a depressive-like phenotype concomitantly with hypercoagulable state and platelet hyperreactivity. Proteomic analysis of aorta secretome from BDNFMet/Met and wild-type (WT) mice showed differential expression of proteins involved in the coagulation and inflammatory cascades. The BDNF Met allele predisposed to carotid artery thrombosis FeCl3-induced and to death after collagen/epinephrine injection. Interestingly, transfection with BDNFMet construct induced a prothrombotic/proinflammatory phenotype in WT cells. SIRT1 activation, using resveratrol and/or CAY10591, prevented thrombus formation and restored the physiological levels of coagulation and of platelet markers in BDNFMet/Met mice and/or cells transfected with the Met allele. Conversely, inhibition of SIRT1 by sirtinol and/or by specific siRNA induced the prothrombotic/proinflammatory phenotype in WT mice and cells. Finally, we found that BDNF Met homozygosity is associated with increased risk of acute myocardial infarction (AMI) in humans. CONCLUSION: Activation of platelets, alteration in coagulation pathways, and changes in vessel wall protein expression in BDNFMet/Met mice recapitulate well the features occurring in the anxiety/depression condition. Furthermore, our data suggest that the BDNFVal66Met polymorphism contribute to the individual propensity for arterial thrombosis related to AMI.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Transtorno Depressivo/genética , Polimorfismo de Nucleotídeo Único/genética , Trombose/genética , Animais , Transtornos de Ansiedade/genética , Aorta/fisiologia , Coagulação Sanguínea/genética , Artérias Carótidas/fisiologia , Trombose das Artérias Carótidas/genética , Modelos Animais de Doenças , Feminino , Heterozigoto , Homozigoto , Humanos , Masculino , Camundongos Transgênicos , Pessoa de Meia-Idade , Isquemia Miocárdica/genética , Proteínas do Tecido Nervoso/metabolismo , Ativação Plaquetária/genética , Inibidores da Agregação Plaquetária/farmacologia , Receptores de Superfície Celular/metabolismo , Resveratrol , Transdução de Sinais/fisiologia , Sirtuína 1/antagonistas & inibidores , Estilbenos/farmacologia
8.
Int J Mol Sci ; 19(8)2018 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-30081509

RESUMO

Reduction in brain-derived neurotrophic factor (BDNF) expression in the brain as well as mutations in BDNF gene and/or of its receptor are associated to obesity in both human and animal models. However, the association between circulating levels of BDNF and obesity is still not defined. To answer this question, we performed a meta-analysis carrying out a systematic search in electronic databases. Ten studies (307 obese patients and 236 controls) were included in the analysis. Our data show that obese patients have levels of BDNF similar to those of controls (SMD: 0.01, 95% CI: -0.28, 0.30, p = 0.94). The lack of difference was further confirmed both in studies in which BDNF levels were assessed in serum (MD: -0.93 ng/mL, 95% CI: -3.34, 1.48, p = 0.45) and in plasma (MD: 0.15 ng/mL, 95% CI: -0.09, 0.39, p = 0.23). Data evaluation has shown that some bias might affect BDNF measurements (e.g., subject recruitment, procedures of sampling, handling, and storage), leading to a difficult interpretation of the results. Standardization of the procedures is still needed to reach strong, affordable, and reliable conclusions.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/sangue , Animais , Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Bases de Dados Factuais , Feminino , Humanos , Masculino , Obesidade/sangue , Obesidade/metabolismo
9.
Int J Mol Sci ; 19(10)2018 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-30347685

RESUMO

Brain-Derived Neurotrophic Factor (BDNF) Val66Met polymorphism has been associated with increased susceptibility to develop mood disorders and recently it has been also linked with cardiovascular disease (CVD). Interestingly, stressful conditions unveil the anxious/depressive-like behavioral phenotype in heterozygous BDNFVal66Met (BDNFVal/Met) mice, suggesting an important relationship in terms of gene-environment interaction (GxE). However, the interplay between stress and BDNFVal/Met in relation to CVD is completely unknown. Here, we showed that BDNFVal/Met mice display a greater propensity to arterial thrombosis than wild type BDNFVal/Val mice after 7 days of restraint stress (RS). RS markedly increased the number of leukocytes and platelets, and induced hyper-responsive platelets as showed by increased circulating platelet/leukocyte aggregates and enhanced expression of P-selectin and GPIIbIIIa in heterozygous mutant mice. In addition, stressed BDNFVal/Met mice had a greater number of large and reticulated platelets but comparable number and maturation profile of bone marrow megakaryocytes compared to BDNFVal/Val mice. Interestingly, RS led to a significant reduction of BDNF expression accompanied by an increased activity of tissue factor in the aorta of both BDNFVal/Val and BDNFVal/Met mice. In conclusion, we provide evidence that sub-chronic stress unveils prothrombotic phenotype in heterozygous BDNF Val66Met mice affecting both the number and functionality of blood circulating cells, and the expression of key thrombotic molecules in aorta. Human studies will be crucial to understand whether this GxE interaction need to be taken into account in risk stratification of coronary artery disease (CAD) patients.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Trombose Coronária/genética , Interação Gene-Ambiente , Estresse Psicológico/complicações , Animais , Aorta/metabolismo , Aorta/patologia , Células da Medula Óssea/citologia , Trombose Coronária/etiologia , Trombose Coronária/patologia , Masculino , Camundongos , Mutação de Sentido Incorreto , Agregação Plaquetária
10.
Platelets ; 28(1): 60-65, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27494459

RESUMO

Cigarette smoke (CS) activates platelets, promotes vascular dysfunction, and enhances Tissue Factor (TF) expression in blood monocytes favoring pro-thrombotic states. Brain-derived neurotrophic factor (BDNF), a member of the family of neurotrophins involved in survival, growth, and maturation of neurons, is released by activated platelets (APLTs) and plays a role in the cardiovascular system. The effect of CS on circulating levels of BDNF is controversial and the function of circulating BDNF in atherothrombosis is not fully understood. Here, we have shown that human platelets, treated with an aqueous extract of CS (CSE), released BDNF in a dose-dependent manner. In addition, incubation of human monocytes with BDNF or with the supernatant of platelets activated with CSE increased TF activity by a Tropomyosin receptor kinase B (TrkB)-dependent mechanism. Finally, comparing serum and plasma samples of 12 male never smokers (NS) and 29 male active smokers (AS) we observed a significant increase in microparticle-associated TF activity (MP-TF) as well as BDNF in AS, while in serum, BDNF behaved oppositely. Taken together these findings suggest that platelet-derived BDNF is involved in the regulation of TF activity and that CS plays a role in this pathway by favoring a pro-atherothrombotic state.


Assuntos
Fatores de Coagulação Sanguínea , Plaquetas/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Fumar/efeitos adversos , Adulto , Idoso , Plaquetas/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Estudos de Casos e Controles , Micropartículas Derivadas de Células/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Monócitos/efeitos dos fármacos , Tromboplastina/metabolismo
11.
Int J Mol Sci ; 18(9)2017 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-28914800

RESUMO

Brain-derived neurothrophic factor (BDNF) is a neurotrophin expressed in different tissues and cells, including neurons, endothelial cells, leukocytes, megakaryocytes and platelets. Modifications of BDNF in plasma and/or in serum are associated with neurodegenerative and psychiatric disorders, cardiovascular diseases, metabolic syndrome and with mortality risk. Indeed, changes in blood levels of BDNF may reflect those of its tissue of origin and/or promote pathological dysfunctions. The measurement of BDNF amount in plasma or in serum has been characterized with particular attention in the impact of different anti-coagulants, clotting duration, temperature (≤21 °C) and delay in blood sample centrifugation as well as in stability of storage. However, the influences of normothermic conditions (37 °C) and of clotting duration on BDNF levels in human serum have not been investigated yet. Here, we showed that time and temperature during serum preparation could be taken into consideration to assess the association and/or impact of BDNF levels in the occurrence of pathological conditions.


Assuntos
Coagulação Sanguínea , Fator Neurotrófico Derivado do Encéfalo/sangue , Temperatura , Adulto , Plaquetas/metabolismo , Colágeno/metabolismo , Feminino , Humanos , Masculino , Tempo de Coagulação do Sangue Total
12.
FASEB J ; 29(9): 4001-10, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26065856

RESUMO

Cigarette smoke (CS) increases the incidence of atherothrombosis, the release of prostaglandin (PG) E2, and the amount of tissue factor (TF). The link between PGE2 and TF, and the impact of this interaction on CS-induced thrombosis, is unknown. Plasma from active smokers showed higher concentration of PGE2, TF total antigen, and microparticle-associated TF (MP-TF) activity compared with never smokers. Similar results were obtained in mice and in mouse cardiac endothelial cells (MCECs) after treatment with aqueous CS extracts (CSEs) plus IL-1ß [CSE (6.4 puffs/L)/IL-1ß (2 µg/L)]. A significant correlation between PGE2 and TF total antigen or MP-TF activity were observed in both human and mouse plasma or tissue. Inhibition of PGE synthase reduced TF in vivo and in vitro and prevented the arterial thrombosis induced by CSE/IL-1ß. Only PG E receptor 1 (EP1) receptor antagonists (SC51089:IC50 ∼ 1 µM, AH6809:IC50 ∼ 7.5 µM) restored the normal TF and sirtuin 1 (SIRT1) levels in MCECs before PGE2 (EC50 ∼ 2.5 mM) or CSE/IL-1ß exposure. Similarly, SIRT1 activators (CAY10591: IC50 ∼ 10 µM, resveratrol: IC50 ∼ 5 µM) or prostacyclin analogs (IC50 ∼ 5 µM) prevented SIRT1 inhibition and reduced TF induced by CSE/IL-1ß or by PGE2. In conclusion, PGE2 increases both TF expression and activity through the regulation of the EP1/SIRT1 pathway. These findings suggest that EP1 may represent a possible target to prevent prothrombotic states.


Assuntos
Dinoprostona/sangue , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Fumar/sangue , Tromboplastina/biossíntese , Trombose/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Células Endoteliais/patologia , Feminino , Humanos , Hidrazinas/farmacologia , Interleucina-1beta/sangue , Masculino , Camundongos , Pessoa de Meia-Idade , Miocárdio/metabolismo , Miocárdio/patologia , Oxazepinas/farmacologia , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/metabolismo , Fumar/efeitos adversos , Fumar/patologia , Trombose/etiologia , Xantonas/farmacologia
13.
Pharmacol Res ; 107: 415-425, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27063941

RESUMO

Recent trials suggest that Aspirin (ASA) reduces the incidence of venous thromboembolism in human. However, the molecular mechanisms underlying this effect are still unclear. In this study we assessed the effects of ASA in venous thrombosis mouse model induced by inferior vena cava (IVC) ligation and we investigated the mechanisms responsible for this effect. ASA (3mg/kg daily for 2 days) treatment decreased the thrombus size, the amounts of tissue factor activity in plasma microvesicles (TF-MP) and the levels of 2,3-dinor Thromboxane B2 (TXB-M) in urine compared to control mice. Interestingly, the thrombus size positively correlated with both TF-MP activity and TXB-M. In addition, positive correlation was observed between TF-MP activity and TXB-M. A reduced number of neutrophils and monocytes, and of TF-positive cells accompanied to a lower amount of fibrin and neutrophil extracellular traps (NETs) were also found in thrombi of ASA-treated mice. Similar results were obtained when mice were treated 24h before IVC ligation with SQ29548 (1mg/kg), a selective thromboxane receptor antagonist. In addition, transfusion of platelets in SQ29548 treated-mice excluded the likelihood of a redundant role of platelet-TP receptor in this context. Finally, incubation of macrophages and neutrophils with SQ29548 prevented TF activity and/or NETs formation induced by supernatant of activated platelets or by IBOP, a selective thromboxane analogue. In conclusion, ASA, suppressing TXA2, prevents macrophages and neutrophils activation and markedly reduces thrombus size with a mechanism most likely dependent of the inhibition of TF activity and NETs formation. These results provide a new link between platelet-produced thromboxane and the occurrence of venous thrombosis.


Assuntos
Aspirina , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária , Tromboxanos/metabolismo , Trombose Venosa/tratamento farmacológico , Animais , Aspirina/farmacologia , Aspirina/uso terapêutico , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Micropartículas Derivadas de Células , Modelos Animais de Doenças , Masculino , Camundongos , Neutrófilos/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Tromboplastina/metabolismo , Veia Cava Inferior/cirurgia , Trombose Venosa/metabolismo
14.
Redox Biol ; 70: 103060, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38310682

RESUMO

There is a complex interrelationship between the nervous system and the cardiovascular system. Comorbidities of cardiovascular diseases (CVD) with mental disorders, and vice versa, are prevalent. Adults with mental disorders such as anxiety and depression have a higher risk of developing CVD, and people with CVD have an increased risk of being diagnosed with mental disorders. Oxidative stress is one of the many pathways associated with the pathophysiology of brain and cardiovascular disease. Nicotinamide adenine dinucleotide phosphate oxidase (NOX) is one of the major generators of reactive oxygen species (ROS) in mammalian cells, as it is the enzyme that specifically produces superoxide. This review summarizes recent findings on the consequences of NOX activation in thrombosis and depression. It also discusses the therapeutic effects and pharmacological strategies of NOX inhibitors in CVD and brain disorders. A better comprehension of these processes could facilitate the development of new therapeutic approaches for the prevention and treatment of the comorbidity of thrombosis and depression.


Assuntos
Doenças Cardiovasculares , Trombose , Animais , Humanos , NADPH Oxidases/metabolismo , NADP/metabolismo , Depressão/tratamento farmacológico , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Trombose/tratamento farmacológico , Comorbidade , Mamíferos/metabolismo
15.
Circulation ; 126(11): 1373-84, 2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22865892

RESUMO

BACKGROUND: Selective inhibitors of cyclooxygenase (COX)-2 increase the risk of myocardial infarction and thrombotic events, but the responsible mechanisms are not fully understood. METHODS AND RESULTS: We found that ferric chloride-induced arterial thrombus formation was significantly greater in COX-2 knockout compared with wild-type mice. Cross-transfusion experiments excluded the likelihood that COX-2 knockout platelets, despite enhanced aggregation responses to collagen and thrombin, are responsible for increased arterial thrombus formation in COX-2 knockout mice. Importantly, we observed that COX-2 deletion decreased prostacyclin synthase and production and peroxisome proliferator-activated receptor- and sirtuin-1 (SIRT1) expression, with consequent increased upregulation of tissue factor (TF), the primary initiator of blood coagulation. Treatment of wild-type mice with a prostacyclin receptor antagonist or a peroxisome proliferator-activated receptor-δ antagonist, which predisposes to arterial thrombosis, decreased SIRT1 expression and increased TF activity. Conversely, exogenous prostacyclin or peroxisome proliferator-activated receptor-δ agonist completely reversed the thrombotic phenotype in COX-2 knockout mice, restoring normal SIRT1 levels and reducing TF activity. Furthermore, inhibition of SIRT1 increased TF expression and activity and promoted generation of occlusive thrombi in wild-type mice, whereas SIRT1 activation was sufficient to decrease abnormal TF activity and prothrombotic status in COX-2 knockout mice. CONCLUSIONS: Modulation of SIRT1 and hence TF by prostacyclin/peroxisome proliferator-activated receptor-δ pathways not only represents a new mechanism in controlling arterial thrombus formation but also might be a useful target for therapeutic intervention in the atherothrombotic complications associated with COX-2 inhibitors.


Assuntos
Trombose das Artérias Carótidas/epidemiologia , Trombose das Artérias Carótidas/metabolismo , Ciclo-Oxigenase 2/metabolismo , Epoprostenol/metabolismo , Sirtuína 1/metabolismo , Tromboplastina/antagonistas & inibidores , Animais , Plaquetas/fisiologia , Trombose das Artérias Carótidas/induzido quimicamente , Cloretos/efeitos adversos , Ciclo-Oxigenase 2/deficiência , Ciclo-Oxigenase 2/genética , Compostos Férricos/efeitos adversos , Incidência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , PPAR delta/agonistas , PPAR delta/antagonistas & inibidores , Receptores de Epoprostenol/agonistas , Receptores de Epoprostenol/antagonistas & inibidores , Fatores de Risco , Transdução de Sinais , Tromboplastina/metabolismo
16.
J Hum Lact ; 39(2): 278-287, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36945737

RESUMO

BACKGROUND: It is well known that the best nutritional option for infants is human milk, and that when breastfeeding is not possible, human milk banks are a possible alternative. However, in the case of infants with fat transport disorder like chylothorax, defatting of human milk is mandatory. RESEARCH AIM: The aim of the study was to reduce milk fat content without reducing other nutrients, increasing oxidative stress, or introducing harmful microorganisms. METHODS: In this prospective, cross-sectional, observational study, we examined the influence of defatting and pasteurization of 50 donor samples on fat, macro- and micronutrients, as well as on oxidative stress markers. RESULTS: Low-temperature centrifugation proved to be very efficient in defatting, reducing the concentration of triglycerides by 85% and cholesterol by 50%. The macronutrients (proteins, albumin, and Immunoglobulin A) did not undergo significant changes due to defatting and pasteurization procedures, while iron decreased by 36%. However, as the majority of iron is retained, this result does not remarkably change the milk composition. Furthermore, oxidative stress markers and antioxidant levels were unchanged, and the milk result was microbiologically safe. CONCLUSIONS: Cold milk centrifugation proved to be an effective technique that allows the reduction of human milk lipids. The determination of triglycerides and cholesterol can be used as an indicator of skimming. This procedure is not accompanied by substantial modifications of other components present in the milk.


Assuntos
Bancos de Leite Humano , Leite Humano , Lactente , Feminino , Humanos , Pasteurização/métodos , Estudos Transversais , Estudos Prospectivos , Aleitamento Materno , Nutrientes/análise , Triglicerídeos , Estresse Oxidativo
17.
Int J Cardiol ; 390: 131229, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37527756

RESUMO

BACKGROUND: Takotsubo cardiomyopathy (TTS) has long been considered a benign condition, despite recurrent events and long-term adverse outcomes are often reported. Endothelial damage, blood hyperviscosity, and platelet activation described in acute phase persist in long-term follow-up; however, TTS pathophysiology is still not fully understood. Here, we explored the hemostatic system at a median of 3.1 years after TTS to uncover additional long-lasting changes in these patients. METHODS: We assessed hemostatic parameters in women with TTS (n = 23) or coronary artery disease (CAD; n = 31) and in control women (n = 26) age-matched, by thromboelastographic analysis, prothrombin time (PT) and partial thromboplastin time (aPTT) coagulation assays and microparticle exposing Tissue Factor (MP-TF). Functional fibrinogen and fibrin polymerization were analyzed by Clauss method and spectrophotometry, respectively. Platelet reactivity was evaluated by light transmission aggregometry, whereas plasminogen activator inhibitor-1 (PAI-1) and brain-derived neurotrophic factor (BDNF) were measured by ELISA kit. RESULTS: Compared with control subjects, TTS patients exhibit an accelerated clot formation, higher percentage of fibrin polymerization and higher PAI-1 levels. Compared with CAD, TTS patients showed sustained residual platelet activation but decreased functional fibrinogen, fibrin polymerization and MP-TF levels, prolonged aPTT and a marked BDNF increase. CONCLUSIONS: The long-term activation of hemostatic system observed in TTS patients compared to control subjects suggests a persistent humoral abnormality that may be related to the propensity for TTS recurrence. The higher residual platelet activity observed in TTS than in CAD patients invites investigation on TTS-tailored antiplatelet therapy potentially needed to prevent TTS adverse outcomes.


Assuntos
Hemostáticos , Cardiomiopatia de Takotsubo , Humanos , Feminino , Inibidor 1 de Ativador de Plasminogênio , Fator Neurotrófico Derivado do Encéfalo , Fibrinogênio , Fibrina , Cardiomiopatia de Takotsubo/diagnóstico
18.
Biomolecules ; 13(10)2023 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-37892152

RESUMO

Circulating small extracellular vesicles (sEVs) contribute to inflammation, coagulation and vascular injury, and have great potential as diagnostic markers of disease. The ability of sEVs to reflect myocardial damage assessed by Cardiac Magnetic Resonance (CMR) in ST-segment elevation myocardial infarction (STEMI) is unknown. To fill this gap, plasma sEVs were isolated from 42 STEMI patients treated by primary percutaneous coronary intervention (pPCI) and evaluated by CMR between days 3 and 6. Nanoparticle tracking analysis showed that sEVs were greater in patients with anterior STEMI (p = 0.0001), with the culprit lesion located in LAD (p = 0.045), and in those who underwent late revascularization (p = 0.038). A smaller sEV size was observed in patients with a low myocardial salvage index (MSI, p = 0.014). Patients with microvascular obstruction (MVO) had smaller sEVs (p < 0.002) and lower expression of the platelet marker CD41-CD61 (p = 0.039). sEV size and CD41-CD61 expression were independent predictors of MVO/MSI (OR [95% CI]: 0.93 [0.87-0.98] and 0.04 [0-0.61], respectively). In conclusion, we provide evidence that the CD41-CD61 expression in sEVs reflects the CMR-assessed ischemic damage after STEMI. This finding paves the way for the development of a new strategy for the timely identification of high-risk patients and their treatment optimization.


Assuntos
Intervenção Coronária Percutânea , Infarto do Miocárdio com Supradesnível do Segmento ST , Humanos , Miocárdio/patologia , Imageamento por Ressonância Magnética , Inflamação/patologia
19.
FASEB J ; 25(10): 3731-40, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21737615

RESUMO

Tobacco smoke (TS) interacts with interleukin-1ß (IL-1ß) to modulate generation of reactive oxygen species (ROS) and expression of cyclooxygenase-2. We explored molecular mechanisms by which TS/IL-1ß alters expression and activity of microsomal-prostaglandin E synthase-1 (mPGES-1) and of prostacyclin synthase (PGIS) in mouse cardiac endothelial cells. TS (EC(50) ∼5 puffs/L) interacting with IL-1ß (2 µg/L) up-regulates PGE(2) production and mPGES-1 expression, reaching a plateau at 4-6 h, but down-regulates prostacyclin (PGI(2)) release by increasing IL-1ß-mediated PGIS tyrosine nitration. Inhibition of NADPH-oxidase, achieved pharmacologically and/or by silencing its catalytic subunit p47phox, or exogenous PGI(2) (carbaprostacyclin; IC(50) ∼5 µM) prevents production of both ROS and PGE(2), and negatively modulates mPGES-1 expression induced by TS/IL-1ß. Moreover, inhibiting PGI(2), either using PGIS siRNA and/or CAY10441 (EC(50) ∼20 nM), a PGI(2) receptor antagonist, increases NADPH-oxidase activation, mPGES-1 synthesis, and PGE(2) production. Finally, lower PGI(2) levels associated with higher PGIS tyrosine nitration, p47phox translocation to the membrane (an index of activation of NADPH-oxidase), and mPGES-1 expression and activity were detected in cardiovascular tissues of ApoE(-/-) mice exposed to cigarette smoke compared to control mice. In conclusion, cigarette smoke in association with cytokines alters the balance between PGI(2)/PGE(2), reducing PGI(2) production and increasing synthesis and activity of mPGES-1 via NADPH-oxidase activation, predisposing to development of pathological conditions.


Assuntos
Epoprostenol/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Oxirredutases Intramoleculares/metabolismo , NADPH Oxidases/metabolismo , Nicotiana/efeitos adversos , Fumaça/efeitos adversos , Animais , Compostos de Benzil/farmacologia , Linhagem Celular , Células Endoteliais/efeitos dos fármacos , Epoprostenol/análogos & derivados , Epoprostenol/genética , Epoprostenol/farmacologia , Imidazóis/farmacologia , Interleucina-1beta/metabolismo , Oxirredutases Intramoleculares/genética , Masculino , Camundongos , NADPH Oxidases/genética , Prostaglandina-E Sintases , Espécies Reativas de Oxigênio/metabolismo
20.
Front Biosci (Landmark Ed) ; 27(5): 161, 2022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35638428

RESUMO

BACKGROUND: Platelet-derived extracellular vesicles (PEVs) are small vesicles released by activated platelets that are gaining growing interest in the field of vascular biology. The mode of platelet activation is a critical determinant of PEVs release, phenotype and function. However, only very limited information is available concerning the impact of the platelet purification procedure on PEVs release. METHODS: Washed or isolated platelets were separated by differential centrifugations. For washed platelets, the platelet pellet was washed by resuspension in PIPES buffer and finally resuspended in HEPES buffer. Isolated platelets were obtained by directly resuspending the platelet pellet in HEPES, skipping the washing steps in PIPES buffer. PEVs release was induced in washed or isolated platelets by stimulation with different agonist and analysed by Nanoparticle Tracking Analysis. RESULTS: Isolated platelets showed a higher release of PEVs upon adenosine diphosphate (ADP) stimulation compared to washed platelets, whereas PEVs released upon stimulation with strong agonists (thrombin, collagen, A23187, U46619) were similar in the two groups. This different responsiveness to ADP was also observed as a higher α-granules release and protein kinase C activation in isolated platelets compared to washed ones. Residual plasma contamination appeared to be essential for the ability of platelets to release PEVs in response to ADP. CONCLUSIONS: In conclusion, our study strongly suggests that procedure adopted for platelets preparation is a critical determinant of PEVs release upon ADP stimulation.


Assuntos
Plaquetas , Vesículas Extracelulares , Difosfato de Adenosina/metabolismo , Difosfato de Adenosina/farmacologia , Plaquetas/metabolismo , Vesículas Extracelulares/metabolismo , HEPES/metabolismo , Ativação Plaquetária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA