Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Int J Cardiol ; 391: 131291, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37619880

RESUMO

BACKGROUND: Myocardial ischemia induces intracellular accumulation of non-glycosylated apolipoprotein J that results in a reduction of circulating glycosylated ApoJ (ApoJ-Glyc). The latter has been suggested to be a marker of transient myocardial ischemia. OBJECTIVE: This proof-of-concept clinical study aimed to assess whether changes in circulating ApoJ-Glyc could detect myocardial ischemia in patients attending the emergency department (ED) with chest pain suggestive of acute coronary syndrome (ACS). METHODS: In suspected ACS patients, EDICA (Early Detection of Myocardial Ischemia in Suspected Acute Coronary Syndromes by ApoJ-Glyc a Novel Pathologically based Ischemia Biomarker), a multicentre, international, cohort study assessed changes in 2 glycosylated variants of ApoJ-Glyc, (ApoJ-GlycA2 and ApoJ-GlycA6), in serum samples obtained at ED admission (0 h), and 1 h and 3 h thereafter, blinded to the clinical diagnosis (i.e. STEMI, NSTEMI, unstable angina, non-ischemic). RESULTS: 404 patients were recruited; 291 were given a clinical diagnosis of "non-ischemic" chest pain and 113 were considered to have had an ischemic event. ApoJ-GlycA6 was lower on admission in ischemic compared with "non-ischemic" patients (66 [46-90] vs. 73 [56-95] µg/ml; P = 0.04). 74% of unstable angina patients (all with undetectable hs-Tn), had ischemic changes in ApoJ-Glyc at 0 h and 89% at 1 h. Initially low ApoJ-Glyc levels in 62 patients requiring coronary revascularization increased significantly after successful percutaneous intervention. CONCLUSIONS: Circulating ApoJ-Glyc concentrations decrease early in ED patients with myocardial ischemia compared with "non-ischemic" patients, even in the absence of troponin elevations. ApoJ-Glyc may be a useful marker of myocardial ischemia in the ED setting.

2.
J Appl Lab Med ; 8(5): 917-930, 2023 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-37473435

RESUMO

BACKGROUND: Previous studies have pointed out a potential role of ApoJ-Glyc as a biomarker of cardiac ischemia. The aim of this study was to validate the analytical performance of 2 novel ELISAs against 2 different glycosylated ApoJ variants, ApoJ-GlycA2 and ApoJ-GlycA6. METHODS: The analytical measuring range, limit of blank (LoB), lower limit of quantification (LoQ), precision, accuracy, recovery, cross-reactivity, and stability were evaluated in serum samples. RESULTS: The analytical measuring range was 500-16 000 ng/mL for ApoJ-GlycA2 and 125-4000 ng/mL for ApoJ-GlycA6, with a LoB of 455 ng/mL and 121 ng/mL for ApoJ-GlycA2 and ApoJ-GlycA6, respectively. The LoQ was 500 ng/mL for ApoJ-GlycA2 and 125 ng/mL for ApoJ-GlycA6. The assay performance fulfills the acceptance criteria established in the European Medicines Agency Guideline on bioanalytical method validation. Specifically, the calibration range variability is <15% for ApoJ-GlycA2 and ApoJ-GlycA6; the accuracy is <15% for ApoJ-GlycA2 and ApoJ-GlycA6; the between- and within-run precision is <15% for ApoJ-GlycA6 and ≤20% for ApoJ-GlycA2; and the total allowable error is <30% for ApoJ-GlycA2 and ApoJ-GlycA6. Cross-reactivity studies revealed the absence of cross-reactivity with endogenous components of the matrix (using ApoJ-depleted serum), with nonglycosylated ApoJ and with transferrin (as a high abundant N-glycosylated serum protein). Both ApoJ-GlycA2 and ApoJ-GlycA6 measurements were stable after storage of serum samples at -80°C for 24 months. CONCLUSIONS: The newly developed ELISAs to quantify ApoJ-GlycA2 and ApoJ-GlycA6 serum levels showed an acceptable analytical performance according to European Medicines Agency guidelines on bioanalytical method validation in terms of precision, accuracy, recovery, cross-reactivity, and stability.


Assuntos
Doença da Artéria Coronariana , Isquemia Miocárdica , Humanos , Clusterina , Isquemia Miocárdica/diagnóstico , Biomarcadores , Ensaio de Imunoadsorção Enzimática
3.
Eur Heart J ; 43(2): 153-163, 2022 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-34580705

RESUMO

AIM: Using proteomics, we previously found that serum levels of glycosylated (Glyc) forms of apolipoprotein J (ApoJ), a cytoprotective and anti-oxidant protein, decrease in the early phase of acute myocardial infarction (AMI). We aimed to investigate: (i) ApoJ-Glyc intracellular distribution and secretion during ischaemia; (ii) the early changes in circulating ApoJ-Glyc during AMI; and (iii) associations between ApoJ-Glyc and residual ischaemic risk post-AMI. METHODS AND RESULTS: Glycosylated apolipoprotein J was investigated in: (i) cells from different organ/tissue origin; (ii) a pig model of AMI; (iii) de novo AMI patients (n = 38) at admission within the first 6 h of chest pain onset and without troponin T elevation at presentation (early AMI); (iv) ST-elevation myocardial infarction patients (n = 212) who were followed up for 6 months; and (v) a control group without any overt cardiovascular disease (n = 144). Inducing simulated ischaemia in isolated cardiac cells resulted in an increased intracellular accumulation of non-glycosylated ApoJ forms. A significant decrease in ApoJ-Glyc circulating levels was seen 15 min after ischaemia onset in pigs. Glycosylated apolipoprotein J levels showed a 45% decrease in early AMI patients compared with non-ischaemic patients (P < 0.0001), discriminating the presence of the ischaemic event (area under the curve: 0.934; P < 0.0001). ST-elevation myocardial infarction patients with lower ApoJ-Glyc levels at admission showed a higher rate of recurrent ischaemic events and mortality after 6-month follow-up (P = 0.008). CONCLUSIONS: These results indicate that ischaemia induces an intracellular accumulation of non-glycosylated ApoJ and a reduction in ApoJ-Glyc secretion. Glycosylated apolipoprotein J circulating levels are reduced very early after ischaemia onset. Its continuous decrease indicates a worsening in the evolution of the cardiac event, likely identifying patients with sustained ischaemia after AMI.


Assuntos
Clusterina , Doença da Artéria Coronariana , Infarto do Miocárdio , Animais , Clusterina/sangue , Clusterina/química , Doença da Artéria Coronariana/sangue , Glicosilação , Humanos , Isquemia , Infarto do Miocárdio/sangue , Suínos , Troponina T
4.
Transl Res ; 235: 1-14, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33887528

RESUMO

The molecular understanding of the pathophysiological changes elicited by diabetes in platelets may help in further elucidating the involvement of this pseudo-cell in the increased risk of developing cardiovascular disease and thrombosis in diabetic subjects. We aimed to investigate the differential characteristics of platelets from diabetic patients and nondiabetic controls to unveil the molecular mechanisms behind the increased platelet reactivity in diabetes. We compared platelets from diabetic and control subjects by 2 dimensional-electrophoresis followed by mass spectrometry. Changes in selected differential proteins were validated by immunoprecipitation assays and western blot. Platelet aggregation was measured by light transmittance aggregometry induced by collagen and ADP, and dynamic coagulation analysis of whole blood was measured by thromboelastometry. We observed significant differences in proteins related to platelet aggregation, cell migration, and cell homeostasis. Subjects with diabetes showed higher platelet aggregation and thrombogenicity and higher contents of the stress-related protein complex HSPA8/Hsp90/CSK2α than nondiabetic subjects. Changes in the chaperones HSPA8 and Hsp90, and in CSK2α protein contents correlated with changes in platelet aggregation and blood coagulation activity. In conclusion, the complex HSPA8/Hsp90/CSK2α is involved in diabetes-related platelet hyperreactivity. The role of the HSPA8/Hsp90/CSK2α complex may become a molecular target for the development of future preventive and therapeutic strategies for platelet dysfunction associated with diabetes and its complications.


Assuntos
Plaquetas/fisiologia , Proteína Tirosina Quinase CSK/fisiologia , Diabetes Mellitus/sangue , Proteínas de Choque Térmico HSC70/fisiologia , Proteínas de Choque Térmico HSP90/fisiologia , Agregação Plaquetária , Adulto , Idoso , Feminino , Proteínas de Choque Térmico HSC70/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Masculino , Pessoa de Meia-Idade , Glicoproteínas da Membrana de Plaquetas/análise
6.
Nutrients ; 10(3)2018 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-29495516

RESUMO

BACKGROUND: Patients with type 2 diabetes mellitus (T2DM) have a higher incidence of cardiovascular (CV) events. The ingestion of high-glycemic index (GI) diets, specially sweetened beverage consumption, has been associated with the development of T2DM and CV disease. OBJECTIVE: We investigated the effects of the intake of a sweetened beverage, obtained from natural carbohydrates containing pinitol (PEB) compared to a sucrose-enriched beverage (SEB) in the context of impaired glucose tolerance (IGT) and diabetes. METHODS: The study was divided in three different phases: (1) a discovery phase where the plasma proteomic profile was investigated by 2-DE (two-dimensional electrophoresis) followed by mass spectrometry (matrix-assisted laser desorption/ionization time-of-flight-MALDI-TOF/TOF) in healthy and IGT volunteers; (2) a verification phase where the potential mechanisms behind the observed protein changes were investigated in the discovery cohort and in an additional group of T2DM volunteers; and (3) the results were validated in a proof-of-concept interventional study in an animal model of diabetic rats with complementary methodologies. RESULTS: Six weeks of pinitol-enriched beverage (PEB) intake induced a significant increase in two proteins involved in the insulin secretion pathway, insulin-like growth factor acid labile subunit (IGF1BP-ALS; 1.3-fold increase; P = 0.200) and complement C4A (1.83-fold increase; P = 0.007) in IGT subjects but not in healthy volunteers. Changes in C4A were also found in the serum samples of Zucker diabetic fatty (ZDF) rats after four weeks of PEB intake compared to basal levels (P = 0.042). In addition, an increased expression of the glucose transporter-2 (GLUT2) gene was observed in the jejunum (P = 0.003) of inositol-supplemented rats when compared to sucrose supplementation. This change was correlated with the observed change in C4A (P = 0.002). CONCLUSIONS: Our results suggest that the substitution of a common sugar source, such as sucrose, by a naturally-based, pinitol-enriched beverage induces changes in the insulin secretion pathway that could help to reduce blood glucose levels by protecting ß-cells and by stimulating the insulin secretion pathway. This mechanism of action could have a relevant role in the prevention of insulin resistance and diabetes progression.


Assuntos
Glicemia/metabolismo , Fabaceae/química , Inositol/análogos & derivados , Adoçantes Calóricos/administração & dosagem , Extratos Vegetais/administração & dosagem , Adolescente , Adulto , Idoso , Animais , Bebidas/análise , Índice de Massa Corporal , Complemento C4a/metabolismo , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2/sangue , Modelos Animais de Doenças , Método Duplo-Cego , Intolerância à Glucose/sangue , Hemoglobinas Glicadas/metabolismo , Voluntários Saudáveis , Humanos , Inositol/administração & dosagem , Insulina/sangue , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Pessoa de Meia-Idade , Sobrepeso/sangue , Proteômica , Ratos , Ratos Zucker , Adulto Jovem
8.
Thromb Haemost ; 117(10): 1970-1980, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28837209

RESUMO

The role of erythrocytes in thrombus formation has been often neglected, but some studies have highlighted their active role in thrombotic events. Free-haemoglobin (Hb) has shown to induce oxidative-stress damage. Herein we have investigated the coordinated changes in heme-related proteins in patients with acute-coronary-syndromes (ACS), their association to ongoing thrombosis and their impact on patients' prognosis. The serum proteome of STEMI-patients (N=27) within the first 6h after event-onset and 3d after were compared to controls (N=60). Changes in heme-metabolism were characterized in a second STEMI-group by a dual proteomic approach analyzing in-vivo aspirated coronary thrombi at PCI (N=24) and the associated peripheral-blood changes (N=10). A third STEMI-group (N=132) was studied to analyze the impact of the observed changes in prognosis at 6-months-follow-up. The haptoglobin/hemopexin(Hpg/Hpx)-scavenging-system revealed a time-dependent response after STEMI with an early increase in Hpg circulating levels in the acute phase (P=0.01) and a late increase in Hpx levels 3d after (P=0.045). Beta-Hb content in coronary thrombi was directly correlated with systemic beta-Hb and Hpg (R=0.804,P=0.0029; R=0.859,P=0.0007) levels. The presence of a fully-occlusive thrombus was associated to higher circulating levels of beta-Hb (P=0.03) and unbound-Hpg (P=0.03). ELISA validation demonstrated a decreased survival rate at 6-months follow-up in STEMI-patients with lower Hpg plasma levels at admission (P=0.027). Our results show active Hb-release form erythrocytes in ACS. This release is followed by a systemic early increase in Hpg levels and a late increase in Hpx levels that can co-ordinately help to prevent systemic pro-oxidative effects. The Hb-scavenging ability of haptoglobin is related to patients' prognosis.


Assuntos
Síndrome Coronariana Aguda/sangue , Trombose Coronária/sangue , Eritrócitos/metabolismo , Hemeproteínas/metabolismo , Infarto do Miocárdio com Supradesnível do Segmento ST/sangue , Síndrome Coronariana Aguda/diagnóstico , Síndrome Coronariana Aguda/terapia , Biomarcadores/sangue , Estudos de Casos e Controles , Trombose Coronária/diagnóstico , Trombose Coronária/terapia , Feminino , Haptoglobinas/metabolismo , Hemoglobinas/metabolismo , Hemopexina/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Estresse Oxidativo , Intervenção Coronária Percutânea , Prognóstico , Proteômica/métodos , Infarto do Miocárdio com Supradesnível do Segmento ST/diagnóstico , Infarto do Miocárdio com Supradesnível do Segmento ST/terapia , Trombectomia , Fatores de Tempo
9.
Curr Opin Lipidol ; 28(5): 427-433, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28682808

RESUMO

PURPOSE OF REVIEW: Familial hypercholesterolemia, represents one of the most extreme clinical entities associated with premature coronary artery disease (CAD). However, clinical manifestation of CAD varies across cohorts and individual patients suggesting the existence of additional non-LDL factors potentially contributing to their cardiovascular burden. RECENT FINDINGS: Changes in HDL-associated proteins appear as one of the potential additional factors contributing to the cardiovascular risk in familial hypercholesterolemia. Specifically, the content of Apo M-SP1 in HDL3 has been directly associated with cholesterol efflux capacity. In addition, a coordinated decrease in the content of Apo L1 and LCAT in HDL3 has been related to the presence of corneal arcus and to bad prognosis in familial hypercholesterolemia patients after an acute ischemic event. In fact, HDL3 particles of familial hypercholesterolemia patients have diminished antioxidant and anti-inflammatory function. SUMMARY: The identification of the specific changes in HDL-associated proteins that contribute to the increased cardiovascular risk of familial hypercholesterolemia patients could be useful for the development of novel therapeutic targets. These novel strategies, in combination with current lipid-lowering therapies, may help to reduce the residual risk found in these patients.


Assuntos
Doenças Cardiovasculares/complicações , Hiperlipoproteinemia Tipo II/complicações , Hiperlipoproteinemia Tipo II/metabolismo , Lipoproteínas/metabolismo , Animais , Humanos , Hiperlipoproteinemia Tipo II/terapia , Fatores de Risco
10.
J Am Coll Cardiol ; 70(2): 165-178, 2017 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-28683964

RESUMO

BACKGROUND: Beneficial effects of high-density lipoproteins (HDL) seem altered in patients with symptomatic cardiovascular disease. We recently demonstrated in a swine model of ischemia-reperfusion (IR) that hypercholesterolemia abolishes HDL-related cardioprotection. OBJECTIVES: This study sought to investigate, using the same animal model, whether the reported impairment of HDL cardioprotective function was associated with alterations in HDL remodeling and functionality. METHODS: Pigs were fed a normocholesterolemic (NC) or hypercholesterolemic (HL) diet for 10 days, reaching non-HDL cholesterol concentrations of 38.2 ± 3.5 mg/dl and 218.6 ± 27.6 mg/dl, respectively (p < 0.0001). HDLs were isolated, and lipidomics and differential proteomics tests were performed to determine HDL molecular changes. HDL functionality and particle size were determined. RESULTS: Using principal component analysis, we identified 255 molecular lipid species differentially clustered in NC-HDL and HL-HDL. Ninety lipid metabolites were differentially expressed, and 50 showed at least 1.5-fold variation (false discovery rate adjustment q value <0.05). HL-HDLs presented a core enriched in cholesteryl esters and a surface depleted of phosphatidylcholine species containing polyunsaturated and long-chain fatty acids, indicating the presence of mature HDL particles with low surface fluidity. Hypercholesterolemia induced an important change in HDL-transported proteins (576 spots in HL-HDL vs. 621 spots in NC-HDL). HL-HDLs showed a reduced content of lipocalin retinol binding protein 4 and apolipoprotein M and in the retinoic acid-transporter cellular retinoic acid binding protein 1 (p < 0.05 vs. NC-HDL). No changes were observed in apolipoprotein A-I content and profile. Functionally, HL-HDL showed lower antioxidant activity (-35%) and a reduced capacity to efflux cholesterol (-60%) compared to NC-HDL (p < 0.05). Hypercholesterolemia induced larger HDL particles. CONCLUSIONS: We demonstrate that hypercholesterolemia induces HDL lipidomic changes, losing phosphatidylcholine-lipid species and gaining cholesteryl esters, and proteomic changes, with losses in cardioprotective proteins. These remodeling changes shifted HDL particles toward a dysfunctional state.


Assuntos
Doença da Artéria Coronariana/sangue , Hipercolesterolemia/sangue , Lipoproteínas HDL/química , Proteômica/métodos , Animais , Modelos Animais de Doenças , Lipoproteínas HDL/sangue , Tamanho da Partícula , Suínos
11.
Thromb Haemost ; 117(9): 1816-1827, 2017 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-28640320

RESUMO

The clinical impact of in-stent thrombosis is high because it is associated with high mortality and 20 % of the patients suffer a recurrent event within the two following years. The aim of this study was to characterise the morphologic and proteomic profile of in-stent thrombi (IST) in comparison to thrombi developed on native coronary arteries (CT) to identify a differential molecular signature. The study included 45 patients with ST-elevation-myocardial-infarction (STEMI) treated by primary-percutaneous-intervention and thrombus aspiration: 21 had IST and 24 had CT. Thrombi were characterised by morphologic immunohistochemical analysis and differential proteomic profiling (2-DE+MALDI-TOF/TOF). Bioinformatic analysis revealed differences in proteins related to oxidative-stress and cell death/survival. IST showed a higher content of structural proteins (gelsolin, actin-cytoplasmic-1, tropomyosin, and myosin) together with an imbalance in redox-homeostasis related proteins (increased superoxide-dismutase and decreased peroxiredoxin-2 thrombus content), and a coordinated increase of chaperones (HSP60 and HSC70) and cellular quality control-related proteins (26S-protease-regulatory-subunit-7). These changes were reflected into a significant decrease in HSC70 systemic levels and a significant increase in advanced-oxidation-protein-products (AOPP) indicative of increased oxidative stress-mediated protein damage in IST. Our results reveal an imbalance in redox-related proteins indicative of an exacerbated oxidative-stress that leads to an accumulation of AOPP serum levels in IST. Moreover, the coordinated increase in chaperones and regulatory proteins reflects the activation of intracellular protection mechanisms to maintain protein integrity in IST. The failure to counterbalance the stress situation could trigger cellular apoptosis leading to the destabilization of the thrombus and to a worse prognosis of IST-STEMI-patients.


Assuntos
Trombose Coronária/metabolismo , Imunidade Inata , Estresse Oxidativo , Intervenção Coronária Percutânea/instrumentação , Infarto do Miocárdio com Supradesnível do Segmento ST/terapia , Stents , Biomarcadores/metabolismo , Biologia Computacional , Trombose Coronária/diagnóstico por imagem , Trombose Coronária/etiologia , Trombose Coronária/terapia , Feminino , Fibrinogênio/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Chaperonas Moleculares/metabolismo , Intervenção Coronária Percutânea/efeitos adversos , Proteômica/métodos , Infarto do Miocárdio com Supradesnível do Segmento ST/diagnóstico por imagem , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Trombectomia , Resultado do Tratamento
12.
Nutrients ; 9(6)2017 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-28608804

RESUMO

Cardiovascular disease (CVD) remains one of the major causes of death and disability worldwide. In addition to drug treatment, nutritional interventions or supplementations are becoming a health strategy for CVD prevention. Phytosterols (PhyS) are natural components that have been shown to reduce cholesterol levels; while poly-unsaturated fatty acids (PUFA), mainly omega-3 (ω3) fatty acids, have shown to reduce triglyceride levels. Here we aimed to investigate whether the proteins in the main lipoproteins (low density lipoproteins (LDL) and high density lipoproteins (HDL)) as well as proteins in the lipid free plasma fraction (LPDP) were regulated by the intake of PhyS-milk or ω3-milk, in overweight healthy volunteers by a proteomic based systems biology approach. The study was a longitudinal crossover trial, including thirty-two healthy volunteers with body mass index (BMI) 25-35 kg/m² (Clinical Trial: ISRCTN78753338). Basal samples before any intervention and after 4 weeks of intake of PhyS or ω3-milk were analyzed. Proteomic profiling by two dimensional electrophoresis (2-DE) followed by mass spectrometry-(MALDI/TOF), ELISA, Western blot, conventional biochemical analysis, and in-silico bioinformatics were performed. The intake of PhyS-milk did not induce changes in the lipid associated plasma protein fraction, whereas ω3-milk significantly increased apolipoprotein (Apo)- E LDL content (p = 0.043) and induced a coordinated increase in several HDL-associated proteins, Apo A-I, lecitin cholesterol acyltransferase (LCAT), paraoxonase-1 (PON-1), Apo D, and Apo L1 (p < 0.05 for all). Interestingly, PhyS-milk intake induced a reduction in inflammatory molecules not seen after ω3-milk intake. Serum amyloid P component (SAP) was reduced in the LPDP protein fraction (p = 0.001) of subjects taking PhyS-milk and C-C motif chemokine 2 (CCL2)expression detected by reverse transcription polymerase chain reaction (RT-PCR) analysis in white blood cells was significantly reduced (p = 0.013). No changes were observed in the lipid-free plasma proteome with ω3-milk. Our study provides novel results and highlights that the PhyS-milk induces attenuation of the pro-inflammatory pathways, whereas ω3-milk induces improvement in lipid metabolic pathways.


Assuntos
Ácidos Graxos Ômega-3/farmacologia , Inflamação/tratamento farmacológico , Fitosteróis/farmacologia , Animais , Estudos Cross-Over , Gorduras na Dieta , Método Duplo-Cego , Ácidos Graxos Ômega-3/administração & dosagem , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/metabolismo , Masculino , Pessoa de Meia-Idade , Leite , Fitosteróis/administração & dosagem , Transdução de Sinais/fisiologia
13.
Transl Res ; 185: 34-46.e9, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28506697

RESUMO

Social changes and medical advances have increased longevity, but the conditions governing healthy vs unhealthy cardiovascular (CV) aging are not fully known. Factors beyond classical CV risk factors may have an important unrecognized value. We sought to identify proteins differentially expressed in healthy octogenarians (HOs) without a history of cardiovascular disease (CVD) and preserved functional and cognitive state compared with octogenarians with a history of CVD and cognitive decline (UHOs) using a systems biology approach, and investigated how these proteins relate to CV mortality at 5-year follow-up. Plasmas obtained from older octogenarians (87 ± 0 years) were analyzed by 2-DE + MS and bioinformatic pathway analysis in HOs (N = 38) and UHOs with cognitive (MEC<25) and functional (Barthel<90) decline and a previous ischemic event (acute myocardial infarction and/or stroke; N = 27). Results were validated by ELISA in HOs and UHOs and in an additional group of older octogenarians without cognitive impairment but with a previous CVD manifestation (HO-CVD; N = 35). UHOs showed a coordinated change in several inflammation-related proteins (AMBP, RBP4, and ITIH4; P < 0.05), together with a significant increase in the major inducer of the acute-phase reaction, interleukin-6 (P = 0.03). UHOs also showed a coordinated increase in hemostatic proteins that was associated with an impairment of fibrinolysis and an increased 5-year CV mortality (P = 0.003). The combination of inflammation (ITIH4 and interleukin-6) and hemostatic markers (D-dimer, A2AP, and coagulation factor XIII) was able to discriminate the presence of an unhealthy phenotype in the elderly (AUC = 0.750; P = 0.001). Unhealthy older octogenarians show increased levels of several plasma proteins of inflammation and coagulation. In older octogenarians, the increase in hemostatic markers indicated an increase in 5-year CV mortality at follow-up.


Assuntos
Hemostasia/fisiologia , Inflamação/metabolismo , Idoso de 80 Anos ou mais , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Análise de Sobrevida
14.
Cardiovasc Res ; 113(9): 1064-1073, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28498891

RESUMO

Adipose tissue (AT) is a highly heterogeneous organ. Beside the heterogeneity associated to different tissue types (white, brown, and 'brite') and its location-related heterogeneity (subcutaneous, visceral, epicardial, and perivascular, etc.), AT composition, structure, and functionality are highly dependent on individual-associated factors. As such, the pro-inflammatory state associated to the presence of obesity and other cardiovascular risk factors (CVRFs) directly affects AT metabolism. Furthermore, the adipose-derived stem cells (ASCs) that reside in the stromal vascular fraction of AT, besides being responsible for most of the plasticity attributed to AT, is an additional source of heterogeneity. Thus, ASCs directly contribute to AT homeostasis, cell renewal, and spontaneous repair. These ASCs share many properties with the bone-marrow mesenchymal stem cells (i.e. potential to differentiate towards multiple tissue lineages, and angiogenic, antiapoptotic, and immunomodulatory properties). Moreover, ASCs show clear advantages in terms of accessibility and quantity of available sample, their easy in vitro expansion, and the possibility of having an autologous source. All these properties point out towards a potential use of ASCs in regenerative medicine. However, the presence of obesity and other CVRFs induces a pro-inflammatory state that directly impacts ASCs proliferation and differentiation capacities affecting their regenerative abilities. The focus of this review is to summarize how inflammation affects the different AT depots and the mechanisms by which these changes further enhance the obesity-associated metabolic disturbances. Furthermore, we highlight the impact of obesity-induced inflammation on ASCs properties and how those effects impair their plasticity.


Assuntos
Tecido Adiposo/patologia , Linhagem da Célula , Plasticidade Celular , Inflamação/patologia , Células-Tronco Mesenquimais/patologia , Obesidade/patologia , Adipocinas/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo/fisiopatologia , Adiposidade , Animais , Metabolismo Energético , Humanos , Inflamação/metabolismo , Inflamação/fisiopatologia , Mediadores da Inflamação/metabolismo , Células-Tronco Mesenquimais/metabolismo , Obesidade/metabolismo , Obesidade/fisiopatologia , Fenótipo , Transdução de Sinais
15.
Stem Cell Res Ther ; 8(1): 52, 2017 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-28279225

RESUMO

BACKGROUND: Myocardial microvascular loss after myocardial infarction (MI) remains a therapeutic challenge. Autologous stem cell therapy was considered as an alternative; however, it has shown modest benefits due to the impairing effects of cardiovascular risk factors on stem cells. Allogenic adipose-derived stem cells (ASCs) may overcome such limitations, and because of their low immunogenicity and paracrine potential may be good candidates for cell therapy. In the present study we investigated the effects of allogenic ASCs and their released products on cardiac rarefaction post MI. METHODS: Pig subcutaneous adipose tissue ASCs were isolated, expanded and GFP-labeled. ASC angiogenic function was assessed by the in-vivo chick chorioallantoic membrane (CAM) model. Pigs underwent MI induction and 7 days after were randomized to receive: allogenic ASCs (intracoronary infusion); conditioned media (CM; intravenous infusion); ASCs + CM; or PBS/placebo (control). Cardiac damage and function were monitored by 3-T cardiac magnetic resonance imaging upon infusion (baseline CMR) and 1 and 3 weeks thereafter. We assessed in the myocardium: microvessel density; angiogenic markers (CD105, CD31, TF, VEGFR2, VEGFR1, vWF, eNOS, CD62); collagen deposition; and reparative fibrosis (TGFß/TßRII/collagen). Differential proteomics of ASCs and CM was performed to characterize the ASC protein signature. RESULTS: CAM indicated a significant ASC proangiogenic capacity. In pigs after MI, only PBS/placebo animals displayed an impaired cardiac function 3 weeks after infusion (p < 0.05 vs baseline). Administration of ASCs + CM significantly enhanced neovessel formation and favored cardiac repair post MI (p < 0.05 vs the other groups). Molecular markers of angiogenesis were significantly upregulated both at transcriptional and protein levels (p < 0.05). The in-silico bioinformatics analysis of the ASC and CM proteome (interactome) indicated activation of a coordinated protein network involved in the formation of microvessels and the resolution of rarefaction. CONCLUSION: Coadministration of allogenic ASCs and their CM synergistically contribute to the neovascularization of the infarcted myocardium through a coordinated upregulation of the proangiogenic protein interactome.


Assuntos
Infarto do Miocárdio/terapia , Isquemia Miocárdica/terapia , Transplante de Células-Tronco , Transplante Autólogo , Tecido Adiposo/citologia , Animais , Terapia Baseada em Transplante de Células e Tecidos , Humanos , Microvasos/crescimento & desenvolvimento , Microvasos/fisiopatologia , Infarto do Miocárdio/fisiopatologia , Isquemia Miocárdica/fisiopatologia , Miocárdio/patologia , Fatores de Risco , Suínos , Biologia de Sistemas
16.
Eur Heart J ; 38(1): 51-52, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27106954
18.
Int J Cardiol ; 222: 1105-1109, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27499220

RESUMO

Elderly patients represent an important proportion of the acute coronary syndrome (ACS) population. Furthermore, this group of ACS patients is continuously growing because of the progressive ageing of the population. The ageing process implies marked changes in patient physiology that directly impact in their risk. However, there is a differential distribution in the risk of elderly patients, revealing the existence of a discrepancy between the chronological and the "biological age". This discrepancy has highlighted the need of performing individual risk assessment in order to identify those patients at higher risk. In addition, the lack of representation of elderly patients in clinical trials leads to the underutilization of evidence-based therapies in this group of patients. All these factors influence not only the high prevalence of ACS presentation in the elderly but also their worse prognosis after suffering an ischaemic event. Herein we will explore the pathophysiological mechanisms behind the age-related changes at the vascular and the cardiac level that explain the high risk of elderly subjects of suffering ACS and their worse prognosis.


Assuntos
Síndrome Coronariana Aguda/epidemiologia , Envelhecimento , Medição de Risco , Idoso , Saúde Global , Humanos , Prevalência , Fatores de Risco
20.
Int J Cardiol ; 215: 406-16, 2016 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-27128573

RESUMO

AIMS: Molecular chaperones constitute protectors of intracellular protein integrity and seem to confer short-term defence against various cell insults. Myocardial damage is associated to a loss of protective chaperones. Ischemic post-conditioning (IPost-Co) is a procedure that seems to protect against reperfusion injury. However, little is known on alpha-crystallin-B-chain (cryab/HspB5) evolution in IPost-Co. Here we have investigated cryab in myocardial ischemia and IPost-Co. METHODS AND RESULTS: Pigs underwent closed-chest 1.5h mid-left anterior descending (LAD) balloon occlusion and were either sacrificed without reperfusion (I;N=10), subjected to 2.5h of reperfusion and sacrificed (I/R; N=5); or subjected to IPost-Co before reperfusion and sacrificed 2.5h afterwards (IPost-Co; N=5). A sham-operated group was included (N=6). Proteomic analysis (2-D-electrophoresis/MALDI-TOF/TOF) revealed cryab as a single spot (20kDa; pI7.6). Myocardial cryab-20-protein and cryab-gene expression levels were decreased after ischemia and I/R(P<0.05). After IPost-Co, cryab-20-protein and cryab-gene expression levels were similar to those found in the heart of sham-operated animals (P<0.05). There was a direct correlation between LVEF-improvement after IPost-Co and myocardial cryab-20-protein levels. In a mice proof-of-principle study, cryab-20-peptide was synthesized and administered 1h before LAD-ligation and ECG-proven MI. A 59% reduction in infarct size was achieved in cryab-20-treated animals (P<0.05). CONCLUSIONS: Ischemia and reperfusion induce a decrease in myocardial cryab-20-protein levels together with a clinical impairment of cardiac function. IPost-Co induces a clinical improvement of cardiac function and a preservation of cryab-20 levels. Intervention studies on a mice-MI model showed that cryab-20-peptide administration reduces infarct size. All together our results show a significant cardioprotective effect of cryab.


Assuntos
Pós-Condicionamento Isquêmico/métodos , Isquemia Miocárdica/terapia , alfa-Cristalinas/farmacologia , Animais , Cardiotônicos/farmacologia , Hipóxia Celular/fisiologia , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C3H , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteômica/métodos , Distribuição Aleatória , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...