Network


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

Hotspot


Dive into the research topics where Nathalie Thorin-Trescases is active.

Publication


Featured researches published by Nathalie Thorin-Trescases.


Mechanisms of Ageing and Development | 2007

Cellular senescence in endothelial cells from atherosclerotic patients is accelerated by oxidative stress associated with cardiovascular risk factors.

Guillaume Voghel; Nathalie Thorin-Trescases; Nada Farhat; Albert Nguyen; Louis Villeneuve; Aida M. Mamarbachi; Annik Fortier; Louis P. Perrault; Michel Carrier; Eric Thorin

Risk factors for cardiovascular diseases (CVD) increase oxidative stress, and they are proposed to hasten endothelial cell (EC) damage and dysfunction. Our objective was to elucidate the impact of chronic exposure to risk factors for CVD on senescence of EC isolated and cultured from internal mammary arterial segments of patients with severe coronary artery disease. Senescence induced by serial passages resulted in progressive telomere shortening, and short initial telomeres predicted early appearance of senescence in culture. Neither time course of senescence nor telomere length was age-dependent, suggesting that biological age, rather than chronological age, determined the dynamics. Senescence appeared earlier in patients with longer history of risk factor for CVD, and multivariate analysis suggested that hypertension hastened the onset of senescence. Risk factors for CVD override the effects of chronological aging likely by generating stress-dependent damage: senescent EC exhibited oxidative stress (increase in lipid peroxydation and caveolin-1 gene expression) and cell damage markers (loss of eNOS expression and increase in Cox2 mRNA, lower TRF1 protein level). Thus, cell senescence was triggered both by telomere-dependent and -independent pathways. In conclusion, chronic exposure to risk factors for CVD accelerated the development of endothelial senescence that could contribute to the pathogenesis of CVD.


Cardiovascular Research | 2009

Vascular endothelial ageing, heartbeat after heartbeat

Eric Thorin; Nathalie Thorin-Trescases

The vascular endothelium starts to age at the first heartbeat. There is no longer a need to demonstrate that an increased resting heart rate--above 70 b.p.m.--is associated with the onset of cardiovascular events and reduces lifespan in humans. Each cardiac cycle imposes a mechanical constraint on the arteries, and we would like to propose that this mechanical stress damages the vascular endothelium, its dysfunction being the prerequisite for atherogenesis. Consequently, reducing heart rate could protect the endothelium and slow the onset of atherosclerosis. The potential mechanisms by which reducing heart rate could be beneficial to the endothelium are likely a combination of a reduction in mechanical stress and tissue fatigue and a prolongation of the period of steady laminar flow, and thus sustained shear stress, between each systole. With age, irreparable damage accumulates in endothelial cells and leads to senescence, which is characterized by a pro-atherogenic phenotype. In the body, the highest mechanical stress occurs in the coronary vessels, where blood only flows during diastole and even reverses during systole; thus, coronary arteries are the prime site of atherosclerosis. All classical risk factors for cardiovascular diseases add up, to accelerate atherogenesis, but hypertension, which further raises mechanical stress, is likely the most damaging. By inducing flow through the arteries, the heart rate determines shear stress and its stability: mechanical stress and the associated damage induced by each systole are efficiently counteracted by the repair capacities of a healthy endothelium. The maintenance of a physiological, low heart rate may be key to prolonging the endothelial healthy lifespan and thus, vascular health.


American Journal of Physiology-heart and Circulatory Physiology | 2013

Endothelium-dependent control of cerebrovascular functions through age: exercise for healthy cerebrovascular aging

Virginie Bolduc; Nathalie Thorin-Trescases; Eric Thorin

Cognitive performances are tightly associated with the maximal aerobic exercise capacity, both of which decline with age. The benefits on mental health of regular exercise, which slows the age-dependent decline in maximal aerobic exercise capacity, have been established for centuries. In addition, the maintenance of an optimal cerebrovascular endothelial function through regular exercise, part of a healthy lifestyle, emerges as one of the key and primary elements of successful brain aging. Physical exercise requires the activation of specific brain areas that trigger a local increase in cerebral blood flow to match neuronal metabolic needs. In this review, we propose three ways by which exercise could maintain the cerebrovascular endothelial function, a premise to a healthy cerebrovascular function and an optimal regulation of cerebral blood flow. First, exercise increases blood flow locally and increases shear stress temporarily, a known stimulus for endothelial cell maintenance of Akt-dependent expression of endothelial nitric oxide synthase, nitric oxide generation, and the expression of antioxidant defenses. Second, the rise in circulating catecholamines during exercise not only facilitates adequate blood and nutrient delivery by stimulating heart function and mobilizing energy supplies but also enhances endothelial repair mechanisms and angiogenesis. Third, in the long term, regular exercise sustains a low resting heart rate that reduces the mechanical stress imposed to the endothelium of cerebral arteries by the cardiac cycle. Any chronic variation from a healthy environment will perturb metabolism and thus hasten endothelial damage, favoring hypoperfusion and neuronal stress.


Mechanisms of Ageing and Development | 2008

Chronic treatment with N-acetyl-cystein delays cellular senescence in endothelial cells isolated from a subgroup of atherosclerotic patients

Guillaume Voghel; Nathalie Thorin-Trescases; Nada Farhat; Aida M. Mamarbachi; Louis Villeneuve; Annik Fortier; Louis P. Perrault; Michel Carrier; Eric Thorin

Endothelial senescence may contribute to the pathogenesis of age-related vascular disorders. Furthermore, chronic exposure to risk factors for cardiovascular disease (CVD) accelerates the effects of chronological aging by generating stress-dependent damages, including oxidative stress, therefore promoting stress-induced premature senescence. Our objective was to determine whether a chronic treatment with an antioxidant (N-acetyl-cystein, NAC) could delay senescence of endothelial cells (EC) isolated and cultured from arterial segments of patients with severe coronary artery disease. If EC were considered as one population (n=26), chronic NAC treatment slightly shortened telomere attrition rate associated with senescence but did not significantly delay the onset of endothelial senescence. However, in a subgroup of NAC-treated EC (n=15) cellular senescence was significantly delayed, NAC decreased lipid peroxidation (HNE), activated the catalytic subunit of telomerase (hTERT) and inhibited telomere attrition. In contrast, in another subgroup of EC (n=11) characterized by initial short telomeres, no effect of NAC on HNE and high levels of DNA damages, the antioxidant was not beneficial on senescence, suggesting an irreversible stress-dependent damage. In conclusion, chronic exposure to NAC can delay senescence of diseased EC via hTERT activation and transient telomere stabilization, unless oxidative stress-associated cell damage has become irreversible.


Journal of the American Heart Association | 2013

Angiopoietin-Like 2 Promotes Atherogenesis in Mice

Nada Farhat; Nathalie Thorin-Trescases; Maya Mamarbachi; Louis Villeneuve; Carol Yu; Cécile Martel; Natacha Duquette; Mathieu Gayda; Anil Nigam; Martin Juneau; Bruce G. Allen; Eric Thorin

Background Angiopoietin like‐2 (angptl2), a proinflammatory protein, is overexpressed in endothelial cells (ECs) from patients with coronary artery disease (CAD). Whether angptl2 contributes to atherogenesis is unknown. We tested the hypothesis that angptl2 promotes inflammation and leukocyte adhesion onto ECs, thereby accelerating atherogenesis in preatherosclerotic dyslipidemic mice. Methods and Results In ECs freshly isolated from the aorta, basal expression of TNF‐α and IL‐6 mRNA was higher in 3‐month‐old severely dyslipidemic mice (LDLr−/−; hApoB100+/+ [ATX]) than in control healthy wild‐type (WT) mice (P<0.05) and was increased in both groups by exogenous angptl2 (100 nmol/L). Angptl2 stimulated the adhesion of leukocytes ex vivo on the native aortic endothelium of ATX, but not WT mice, in association with higher expression of ICAM‐1 and P‐selectin in ECs (P<0.05). Antibodies against these endothelial adhesion molecules prevented leukocyte adhesion. Intravenous administration of angptl2 for 1 month in preatherosclerotic 3‐month‐old ATX mice increased (P<0.05) total cholesterol and LDL‐cholesterol levels, strongly induced (P<0.05) the expression of endothelial proinflammatory cytokines and adhesion molecules while accelerating atherosclerotic lesion formation by 10‐fold (P<0.05). Plasma and aortic tissue levels of angptl2 increased (P<0.05) with age and were higher in 6‐ and 12‐month‐old ATX mice than in age‐matched WT mice. Angptl2 accumulated to high levels in the atherosclerotic lesions (P<0.05). Finally, angptl2 was greatly expressed (P<0.05) in ECs cultured from CAD patients, and circulating angptl2 levels were 6‐fold higher in CAD patients compared with age‐matched healthy volunteers. Conclusions Angptl2 contributes to the pathogenesis of atherosclerosis.


Mechanisms of Ageing and Development | 2010

Endogenous oxidative stress prevents telomerase-dependent immortalization of human endothelial cells

Guillaume Voghel; Nathalie Thorin-Trescases; Aida M. Mamarbachi; Louis Villeneuve; Frédérick A. Mallette; Gerardo Ferbeyre; Nada Farhat; Louis P. Perrault; Michel Carrier; Eric Thorin

INTRODUCTIONnWith aging, oxidative stress accelerates vascular endothelial cell (EC) telomere shortening-induced senescence, and may promote atherosclerosis in humans. Our aim was to investigate whether an antioxidant treatment combined with telomerase (hTERT) over-expression would prevent senescence of EC isolated from patients with severe atherosclerosis.nnnMETHODSnCells were isolated from internal mammary arteries (n=11 donors), cultured until senescence with or without N-acetylcystein (NAC) and infected, or not, with a lentivirus over-expressing hTERT.nnnRESULTSnCompared to control EC, hTERT-NAC cells had increased telomerase activity, longer telomeres and underwent more cell divisions. According to the donor, hTERT-NAC either delayed (n=5) or prevented (n=4) EC senescence, the latter leading to cell immortalization. Lack of cell immortalization by hTERT-NAC was accompanied by an absence of beneficial effect of NAC alone in paired EC. Accordingly, lack of EC immortalization by hTERT-NAC was associated with high endogenous susceptibility to oxidation. In EC where hTERT-NAC did not immortalize EC, p53, p21 and p16 expression increased with senescence, while oxidative-dependent DNA damage associated with senescence was not prevented.nnnCONCLUSIONnOur data suggest that irreversible oxidative stress-dependent damages associated with cardiovascular risk factors are responsible for senescence of EC from atherosclerotic patients.


Expert Reviews in Molecular Medicine | 2014

Angiopoietin-like-2: a multifaceted protein with physiological and pathophysiological properties

Nathalie Thorin-Trescases; Eric Thorin

Angptl2 is a multifaceted protein, displaying both physiological and pathological functions, in which scientific and clinical interest is growing exponentially within the past few years. Its physiological functions are not well understood, but angptl2 was first acknowledged for its pro-angiogenic and antiapoptotic capacities. In addition, angptl2 can be considered a growth factor, since it increases survival and expansion of hematopoietic stem cells and may promote vasculogenesis. Finally, angptl2 has an important, but largely unrecognised, physiological role: in the cytosol, angptl2 binds to type 1A angiotensin II receptors and induces their recycling, with recovery of the receptor signal functions. Despite these important physiological properties, angptl2 is better acknowledged for its deleterious pro-inflammatory properties and its contribution in multiple chronic diseases such as cancer, diabetes, atherosclerosis, metabolic disorders and many other chronic diseases. This review aims at presenting an updated description of both the beneficial and deleterious biological properties of angptl2, in addition to its molecular signalling pathways and transcriptional regulation. The multiplicity of diseases in which angptl2 contributes makes it a new highly relevant clinical therapeutic target.


Dose-response | 2012

Time-Dependent Beneficial Effect of Chronic Polyphenol Treatment with Catechin on Endothelial Dysfunction in Aging Mice

Marie-Ève Gendron; Nathalie Thorin-Trescases; Aida M. Mamarbachi; Louis Villeneuve; Jean-François Théorět; Yahye Mehri; Eric Thorin

A controlled redox environment is essential for vascular cell maturation and function. During aging, an imbalance occurs, leading to endothelial dysfunction. We hypothesized that, according to the concept of hormesis, exposure to physiologic oxidative stress during the maturation phase of the endothelium will activate protective pathways involved in stress resistance. C57Bl/6 mice were treated with the polyphenol catechin for the last 3 (post-maturation) or 9 months prior study at 12 months of age. Endothelial dysfunction, assessed by acetylcholine-induced dilations of isolated renal arteries, was present at 12 months (P<0.05). Only the 3-month treatment with catechin fully prevented the decline in efficacy and sensitivity to acetylcholine (P<0.05). Splenocytes adhesion to the native endothelium, expression of CD18 and shedding of CD62L and PSGL-1 augmented in 12 months old mice (P<0.05): only 3-month catechin fully normalized adhesion and prevented the expression of adhesion molecules on splenocytes (P<0.05). Aging was associated with vascular gene alterations, which were prevented by 3-month catechin treatment (P<0.05). In contrast, 9-month catechin further increased COX-2, p22phox and reduced MnSOD (P<0.05). In conclusion, we demonstrate a pivotal role of cellular redox equilibrium: exposure to physiologic oxidative stress during the maturation phase of the endothelium is essential for its function.


Pflügers Archiv: European Journal of Physiology | 2011

Up-regulation of thromboxane A2 impairs cerebrovascular eNOS function in aging atherosclerotic mice

Annick Drouin; Nada Farhat; Virginie Bolduc; Nathalie Thorin-Trescases; Marc-Antoine Gillis; Louis Villeneuve; Albert Nguyen; Eric Thorin

We previously reported that in healthy mouse cerebral arteries, endothelial nitric oxide synthase (eNOS) produces H2O2, leading to endothelium-dependent dilation. In contrast, thromboxane A2 (TXA2), a potent pro-oxidant and pro-inflammatory endogenous vasoconstrictor, is associated with eNOS dysfunction. Our objectives were to elucidate whether (1) the cerebrovascular eNOS–H2O2 pathway was sensitive to oxidative stress associated with aging and dyslipidemia and (2) TXA2 contributed to cerebral eNOS dysfunction. Atherosclerotic (ATXu2009=u2009LDLR−/−; hApoB+/+) and wild-type (WT) control mice were used at 3 and 12xa0months old (m/o). Three-m/o ATX mice were treated with the cardio-protective polyphenol catechin for 9xa0months. Dilations to ACh and the simultaneous eNOS-derived H2O2 production were recorded in isolated pressurized cerebral arteries. The age-associated decrease in cerebral eNOS–H2O2 pathway observed in WT was premature in ATX mice, decreasing at 3xa0m/o and abolished at 12xa0m/o. Thromboxane synthase inhibition by furegrelate increased dilations at 12xa0months in WT and at 3 and 12xa0months in ATX mice, suggesting an anti-dilatory role of TXA2 with age hastened by dyslipidemia. In addition, the non-selective NADP(H) oxidase inhibitor apocynin improved the eNOS–H2O2 pathway only in 12-m/o ATX mice. Catechin normalized the function of this pathway, which became sensitive to L-NNA and insensitive to furegrelate or apocynin; catechin also prevented the rise in TXA2 synthase expression. In conclusion, the age-dependent cerebral endothelial dysfunction is precocious in dyslipidemia and involves TXA2 production that limits eNOS activity. Preventive catechin treatment reduced the impact of endogenous TXA2 on the control of cerebral tone and maintained eNOS function.


Diabetologia | 2016

ANGPTL2 is associated with an increased risk of cardiovascular events and death in diabetic patients

Barnabas Gellen; Nathalie Thorin-Trescases; Philippe Sosner; Elise Gand; Pierre-Jean Saulnier; Stéphanie Ragot; Mathilde Fraty; Stéphanie Laugier; Gregory Ducrocq; David Montaigne; Pierre Llaty; V. Rigalleau; P. Zaoui; Jean-Michel Halimi; Ronan Roussel; Eric Thorin; Samy Hadjadj

Aims/hypothesisA high serum angiopoietin-like 2 (ANGPTL2) concentration is an independent risk factor for developing diabetes and is associated with insulin resistance and atherosclerosis. In this work, we have examined the impact of serum ANGPTL2 on improving cardiovascular (CV) risk stratification in patients with type 2 diabetes.MethodsA prospective, monocentric cohort of consecutive type 2 diabetes patients (the SURDIAGENE cohort; total of 1353 type 2 diabetes patients; 58% men, meanu2009±u2009SD age 64u2009±u200911xa0years) was followed for a median of 6.0xa0years for death as primary endpoint and major adverse CV events (MACE; i.e. CV death, myocardial infarction or stroke) as a secondary endpoint. Patients with end-stage renal disease, defined as a requirement for dialysis or a history of kidney transplantation, were excluded. Patients were grouped into quartiles according to ANGPTL2 concentrations at inclusion: <11.2 (Q1), 11.2–14.7 (Q2), 14.8–19.5 (Q3) or >19.5 (Q4) ng/ml.ResultsDuring follow up, 367 patients (representing 4.5% of the total person-years) died and 290 patients (representing 3.7% of the total person-years) presented with MACE. Both the survival and MACE-free survival rates were significantly different between ANGPTL2 quartiles (logrank 82.12, pu2009<u20090.0001 for death; and logrank 65.14, pu2009<u20090.0001 for MACE). Patients with ANGPTL2 concentrations higher than 19.5xa0ng/ml (Q4) had a significantly higher risk of death and MACE than those with ANGPTL2 levels of 19.5xa0ng/ml or less (Q1–3) (HR for death 2.44 [95% CI 1.98, 3.00], pu2009<u20090.0001; HR for MACE 2.43 [95% CI 1.92, 3.06], pu2009<u20090.0001) after adjustment for sex, age and established CV risk factors. Using ANGPTL2 concentrations, prediction of the risk of mortality, as assessed by integrated discrimination improvement (IDI), was significantly improved (IDI 0.006u2009±u20090.002, pu2009=u20090.0002).Conclusions/interpretationIn patients with type 2 diabetes, serum ANGPTL2 concentrations were independently associated with death and MACE. Therefore, ANGPTL2 is a promising candidate biomarker for improving risk stratification in type 2 diabetes patients, and may prove to be a valuable therapeutic target.

Collaboration


Dive into the Nathalie Thorin-Trescases's collaboration.

Top Co-Authors

Avatar

Eric Thorin

Montreal Heart Institute

View shared research outputs
Top Co-Authors

Avatar

Nada Farhat

Montreal Heart Institute

View shared research outputs
Top Co-Authors

Avatar

Carol Yu

Montreal Heart Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cécile Martel

Montreal Heart Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michel Carrier

Montreal Heart Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge