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

https://doi.org/10.1016/j.mad.2007.09.006Get rights and content

Abstract

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.

Introduction

Vascular aging is associated with a decrease in endothelial dilatory and antithrombotic functions (Cooper et al., 1994, Lakatta, 2002, Donato et al., 2007). This typical endothelial dysfunction, however, is also present in younger patients with cardiovascular diseases (CVD) (Cohen, 1995). At the cellular level, aging of healthy vascular endothelial cells (EC) leads to senescence, a state of permanent growth arrest in which cells remain alive and metabolically active for months but refractory to mitogenic stimuli (Ben-Porath and Weinberg, 2005, Chen and Goligorsky, 2006). Senescence can be triggered by cell divisions, leading to cumulative telomere attrition down to a threshold length at which cells enter the so-called replicative senescence (Allsopp et al., 1995). In healthy humans, telomere shortening is age-dependent (Slagboom et al., 1994, Chang and Harley, 1995, Brouilette et al., 2007) and likely the consequence of life-long reparative cell divisions. Telomere length has been proposed to contribute and to be a predictor of mortality in many aged-related diseases (Cawthon et al., 2003). Cellular senescence can occur prematurely, independent of replicative age, following exposure to multiple types of stress (stress-induced senescence), such as oxidative stress (Toussaint et al., 2000), DNA damage and mitogenic stress (Ben-Porath and Weinberg, 2005).

Senescence is characterized by specific changes in cell morphology and gene expression, which reduce EC function (Benetos et al., 2001, Wagner et al., 2001) and thus are proposed to be pro-atherogenic (Cohen, 1995, Serrano and Andres, 2004, Edo and Andres, 2005). Although mitotically inactive, senescent cells are not physiologically inert: they secrete degradative enzymes, inflammatory cytokines and growth factors (Chen and Goligorsky, 2006) and this could promote or contribute to the pathogenesis of human atherosclerosis (Minamino and Komuro, 2007). While the concept of senescence of EC is established, its origin is debated. Life-long cell repairs likely promotes instability and one consequence is an imbalance in the regulation of the redox environment towards pro-oxidation, a process accelerated by risk factors for CVD (Csiszar et al., 2002). The ensuing oxidative stress induces cellular damage (Lorenz et al., 2001, von Zglinicki et al., 2005) and further accelerates reparative cell division, promoting telomere instability (Kurz et al., 2004). Excessive telomere shortening in circulating white blood cells has been reported from patients with hypertension (Jeanclos et al., 2000), coronary atherosclerosis (Samani et al., 2001, Fitzpatrick et al., 2007, Brouilette et al., 2007), premature myocardial infarction (Brouilette et al., 2003), heart disease (Starr et al., 2007) and diabetes (Jeanclos et al., 1998). There is therefore a strong rationale for the hypothesis that risk factors for CVD accelerate the normal aging process as recently proposed by Chen and Goligorsky (2006). It is not known, however, if the association of short telomere length with the risk of coronary heart disease applies to other cell types, especially vascular tissue. The telomere-independent caveolin/p53 pathway has been reported to be activated by oxidative stress and lead to senescence (Galbiati et al., 2001, Volonte et al., 2002). In replicative senescence, however, telomere attrition following activation of ATM/p53-p21 pathway has been observed (Pandita, 2002, Herbig et al., 2004, Herbig and Sedivy, 2006).

The objective of the present study was therefore to elucidate the impact of chronic exposure to risk factors for CVD on the senescence of EC isolated and cultured from patients with severe coronary artery disease (CAD) and to find the pathway of choice leading to senescence. We demonstrate that the duration of exposure to risk factors for CVD, especially hypertension, positively correlates with the propensity of EC senescence, independent of the age of the donor. Thus, stress associated cell damages accelerated replicative senescence.

Section snippets

Clinical profile of the donors

Segments of human distal (close to the bifurcation) internal mammary artery (IMA, Table 1) discarded during primary CABG were harvested with low electrocautery energy and excised with cold scissors. The protocol has been approved by our institutional ethical committee.

Isolation and culture of EC

Endothelial cells were isolated by an explant technique (Shi et al., 2000, Thorin et al., 1997). Briefly, six to eight 1-mm2 segments of mammary artery were seeded in 35-mm2 culture dishes to obtain five to eight segments anchored

Senescence in endothelial cells from patients with coronary artery disease

Cell cultures were characterized by immunostaining for the von Willebrand factor and CD31. Senescence was induced in vitro by serial passage, and cells were positive for the specific endothelial markers whether or not they were senescent (Fig. 1A–D). Senescent EC exhibited phenotypic changes: cytoplasmic size was increased up to 20 times, associated with the inclusion of vacuoles and positive staining for senescence-associated β-galactosidase at pH 6 (Fig. 1E and F).

SA-beta-Gal staining was

Discussion

The literature suggests that risk factors for CVD accelerate the normal aging process of the endothelium (Cooper et al., 1994, Cohen, 1995, Lakatta, 2002, Edo and Andres, 2005, Chen and Goligorsky, 2006). Our study shows that senescence of EC from atherosclerotic patients is characterized by low EC growth potential, specific changes in cell phenotype, gene and protein expression and telomere shortening independent of the age of the donor. The duration of exposure to risk factors for CVD,

Acknowledgements

This work has been supported in part by the Foundation of the Montreal Heart Institute, the Heart and Stroke Foundation of Quebec, and the Canadian Institute for Health Research (MOP 14496). Guillaume Voghel is a fellow of the Fonds de la Recherche en Santé du Québec. We thank Guy Charron (Montreal Heart Institute) for fruitful discussions and technical advises, and the biological tissue bank (RETEB) of the Fonds de la Recherche en Santé du Québec for technical support.

References (53)

  • M. Narita et al.

    Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence

    Cell

    (2003)
  • N.J. Samani et al.

    Telomere shortening in atherosclerosis

    Lancet

    (2001)
  • D.N. Shelton et al.

    Microarray analysis of replicative senescence

    Curr. Biol.

    (1999)
  • J.M. Starr et al.

    Association between telomere length and heart disease in a narrow age cohort of older people

    Exp. Gerontol.

    (2007)
  • O. Toussaint et al.

    Cellular and molecular mechanisms of stress-induced premature senescence (SIPS) of human diploid fibroblasts and melanocytes

    Exp. Gerontol.

    (2000)
  • M. Wagner et al.

    Replicative senescence of human endothelial cells in vitro involves G1 arrest, polyploidization and senescence-associated apoptosis

    Exp. Gerontol.

    (2001)
  • N. Yokohori et al.

    Increased levels of cell death and proliferation in alveolar wall cells in patients with pulmonary emphysema

    Chest

    (2004)
  • L. Zhang et al.

    Loss of chromosome 13 in cultured human vascular endothelial cells

    Exp. Cell Res.

    (2000)
  • R.C. Allsopp et al.

    Telomere shortening in associated with cell division in vitro and in vivo

    Exp. Cell Res.

    (1995)
  • A. Benetos et al.

    Short telomeres are associated with increased carotid atherosclerotis in hypertensive patients

    Hypertension

    (2004)
  • A. Benetos et al.

    Telomere length as an indicator of biological aging: the gender effect and relation with pulse pressure and pulse wave velocity

    Hypertension

    (2001)
  • S. Brouilette et al.

    White cell telomere length and risk of premature myocardial infarction

    Arterioscler. Thromb. Vasc. Biol.

    (2003)
  • E. Chang et al.

    Telomere length and replicative aging in human vascular tissues

    Proc. Natl. Acad. Sci. U.S.A.

    (1995)
  • J. Chen et al.

    Premature senescence of endothelial cells: Methusaleh's dilemma

    Am. J. Physiol.

    (2006)
  • L.T. Cooper et al.

    The vasculopathy of ageing

    J. Gerontol.

    (1994)
  • A. Csiszar et al.

    Ageing-induced phenotypic changes and oxidative stress impair coronary arteriolar function

    Circ. Res.

    (2002)
  • Cited by (125)

    • Pharmacological inhibition of guanosine triphosphate cyclohydrolase1 elevates tyrosine phosphorylation of caveolin1 and cellular senescence

      2019, European Journal of Pharmacology
      Citation Excerpt :

      Caveolin1 is implicated in numerous cellular processes, including cell cycle regulation, cellular-senescence, apoptotic signalling and tissue reparative processes (Kurzchalia et al., 1992; Gvaramia et al., 2013; Cho et al., 2004; Ohno-Iwashita et al., 2010; Wheaton, 2011). Several evidences have demonstrated that caveolin1 has a critical role in premature senescence (Yu et al., 2017; Zou et al., 2011; Voghel et al., 2007) as it mediates apoptosis through its expression and post-translational-modification (PTM). OS is known to promote pY14 via p38-MAPK activation (Huot et al., 1996; Bellomo and Mirabelli, 1992), which in turn mediates apoptosis (Bulavin et al., 1999; Huang et al., 1999).

    View all citing articles on Scopus
    1

    These authors equally contributed to this work.

    View full text