Review
The p53 pathway in vasculature revisited: A therapeutic target for pathological vascular remodeling?

https://doi.org/10.1016/j.phrs.2021.105683Get rights and content

Abstract

Pathological vascular remodeling contributes to the development of restenosis following intraluminal interventions, transplant vasculopathy, and pulmonary arterial hypertension. Activation of the tumor suppressor p53 may counteract vascular remodeling by inhibiting aberrant proliferation of vascular smooth muscle cells and repressing vascular inflammation. In particular, the development of different lines of small-molecule p53 activators ignites the hope of treating remodeling-associated vascular diseases by targeting p53 pharmacologically. In this review, we discuss the relationships between p53 and pathological vascular remodeling, and summarize current experimental data suggesting that drugging the p53 pathway may represent a novel strategy to prevent the development of vascular remodeling.

Introduction

The tumor suppressor protein p53 is encoded by the TP53 gene, which was discovered over 40 years ago in transformed malignant cells [1]. Mutations in p53 are related to about 50% of human cancers [2]. Despite the well documented roles of p53 in tumorigenesis, this protein is ubiquitously expressed and is thought to have major impacts on the physiology and/or pathophysiology of virtually all organs in the body, because it is a key transcription factor implicated in modulating the expression of ~60 genes involved in various very fundamental aspects of cell biology, including cell cycle arrest, apoptosis, cell senescence, and DNA repair [1], [3], [4]. Unsurprisingly, numerous studies have illustrated a tight link between p53 and the cardiovascular system [5]. In the heart, aberrant activation of the p53 pathway promotes the pathogenesis of various cardiac diseases, such as infarction-induced cardiac remodeling, hypertrophic and dilated cardiomyopathy, and chemotherapy-induced cardiotoxicity, all of which may result in heart failure [5]. In blood vessels, p53 is activated in atherosclerotic plaques, and is involved in regulating growth arrest, cell senescence and apoptosis of vascular smooth muscle cells (VSMCs) [6]. However, deletion of p53 gene in VSMCs appears to have no significant effect on atherogenesis [7]. In contrast, evidence suggests that the endogenous p53 protein expressed in macrophages has a protective role against the development of atherosclerosis [8], [9]. On the other hand, overexpression of p53 in the plaque may induce detrimental effects by promoting plaque destabilization [10]. Several studies have revealed that the inhibitory effect of p53 on VSMC proliferation can repress the development of pathological vascular remodeling caused by vascular injuries [11], [12]. This property of p53 in vascular remodeling is of a special translational interest currently, due to the emergence of novel small molecule activators of the p53 pathway [2]. Preclinical experiments in various animal models have suggested that drugging the p53 pathway may represent a promising strategy to prevent the development of pathological vascular remodeling with different etiologies. Here we summarize our current knowledge of the relationship between p53 and vascular remodeling, with a highlight on the emerging evidence demonstrating potential therapeutic efficacies of small molecule p53 activators.

Section snippets

Overview of the major mechanisms responsible for p53 activation

The p53 pathway is activated under various stress conditions, exemplified by genotoxic stress (DNA damages), oxidative stress, hypoxia, and nutrient depletion, which among others are mostly relevant to vascular cells [1], [13] (see Fig. 1). Of note, p53 is a short lived protein in cells. The ubiquitin ligase Mdm2 induces ubiquitination and degradation of the p53 protein in unstressed cells. Such stress stimuli can dissociate p53 from Mdm2, likely via Mdm2 dephosphorylation, leading to p53

Major cellular outcomes downstream of p53 activation

Activation of the p53 pathway promotes cell cycle arrest, cell senescence, and apoptosis [1] (Fig. 1). Induction of cell cycle arrest by p53 is attributable to transcriptional upregulation of several key cell cycle regulators including p21CIP1/WAF1, GADD45, and 14–3–3 proteins [23]. p21CIP1/WAF1 contributes to G1/S as well as G2/M blockade by inhibiting the functions of different cyclin/CDK complexes. GADD45 promotes G2/M arrest mainly by repressing the activity of cyclin B. On the other hand,

Development of small molecule activators of the p53 pathway

Because of the exceptional importance of p53 in repressing tumorigenesis, great efforts have been invested in identifying small molecule p53 activators, which are expected to be efficacious for treating cancers (reviewed in [2], [30], [31], [32]). Different targeting strategies have been explored to achieve this goal (see Fig. 1). Firstly, disruption of the interaction between p53 and its negative regulator Mdm2 causes stabilization and accumulation of wild type p53. Nutlins are a series of

Roles of p53 in pathological vascular remodeling and effects of p53 activating agents

Pathological remodeling of the vessel wall contributes to the development of several proliferative vascular diseases, including arterial restenosis, transplant vasculopathy (TV), and pulmonary arterial hypertension (PAH). Emerging evidence supports that activation of the p53 pathway has beneficial effects on vascular remodeling, primarily by repressing VSMC proliferation and vascular inflammation (see Fig. 2). In line with this notion, pre-clinical studies have revealed therapeutic effects of

p53 and vascular inflammation

In addition to VSMC activation, vascular inflammation is another equally important factor in advancing the remodeling process, regardless of the underlying etiologies [54], [55], [56], [57], [70], [73], [74], [75], [111], [112], [113]. The inflammatory reaction mediated by perivascular (adventitial) macrophages appears to be a key contributor, especially in the early phase of vascular inflammation [114]. Macrophage infiltration in the adventitia occurs early after angioplastic injury [115], and

Would p53 activation induce cell apoptosis in normal vascular cells?

It is a concern whether activation of the p53 pathway triggers apoptosis in normal vascular cells. Different studies have suggested that arterial VSMCs are fairly insensitive to p53-induced apoptosis. Overexpression of wild type p53 alone in arterial VSMCs does not induce apoptosis, whereas the pro-apoptotic activity of p53 is only observed in the presence of an additional apoptosis stimulus such as doxorubicin [59] or UV irradiation [60]. Instead, p53 primarily induces cell cycle arrest in

Perspectives on the translational value of small molecule p53 activators

One of the most concerning issues about the translational value of small molecule p53 activators is whether these agents would induce unwanted cell cycle arrest in normally proliferating cells (for example hematopoietic stem cells) [31]. Indeed, phase I clinical trials with RG7112 (a nutlin analogue) have shown that neutropenia and thrombocytopenia are among the observed adverse events [146], [147], although none of the cases of death were considered to be related to drug side effects. However,

Conflict of interest declaration

None.

Acknowledgements

This work was supported partially by the following grants: National Natural Science Foundation of China (82070265 and 81770469 for F.J.), Key Research and Development Program of Shandong Province (2019GSF108052 for X.C.), and National Science and Technology Program of China (2012ZX09303016-003 for H.G.).

References (150)

  • M.J. Bywater et al.

    Inhibition of RNA polymerase I as a therapeutic strategy to promote cancer-specific activation of p53

    Cancer Cell

    (2012)
  • S. Wan et al.

    Overexpression of p53 increases lumen size and blocks neointima formation in porcine interposition vein grafts

    Mol. Ther. J. Am. Soc. Gene Ther.

    (2004)
  • C. Ponticelli

    Progression of renal damage in chronic rejection

    Kidney Int. Suppl.

    (2000)
  • L.P. Wang et al.

    Inhibitory effect of wild-type P53 gene transfer on graft coronary artery disease

    Transpl. Immunol.

    (2018)
  • M. Humbert et al.

    Endothelial cell dysfunction and cross talk between endothelium and smooth muscle cells in pulmonary arterial hypertension

    Vasc. Pharmacol.

    (2008)
  • H. Liu et al.

    Up-regulation of cullin7 promotes proliferation and migration of pulmonary artery smooth muscle cells in hypoxia-induced pulmonary hypertension

    Eur. J. Pharm.

    (2019)
  • L.A. Dyer et al.

    The role of BMPs in endothelial cell function and dysfunction

    Trends Endocrinol. Metab. TEM

    (2014)
  • I. Diebold et al.

    BMPR2 preserves mitochondrial function and DNA during reoxygenation to promote endothelial cell survival and reverse pulmonary hypertension

    Cell Metab.

    (2015)
  • A.J. Levine

    The many faces of p53: something for everyone

    J. Mol. Cell Biol.

    (2019)
  • M. Ladds et al.

    Small molecule activators of the p53 response

    J. Mol. Cell Biol.

    (2019)
  • T. Riley et al.

    Transcriptional control of human p53-regulated genes

    Nat. Rev. Mol. Cell Biol.

    (2008)
  • H. Men et al.

    The regulatory roles of p53 in cardiovascular health and disease

    Cell. Mol. Life Sci. CMLS

    (2021)
  • J. Mercer et al.

    The role of p53 in atherosclerosis

    Cell Cycle

    (2006)
  • R.Y. Cao et al.

    Effects of p53-knockout in vascular smooth muscle cells on atherosclerosis in mice

    PLoS One

    (2017)
  • B.J. van Vlijmen et al.

    Macrophage p53 deficiency leads to enhanced atherosclerosis in APOE*3-Leiden transgenic mice

    Circ. Res.

    (2001)
  • A.J. Merched et al.

    Macrophage-specific p53 expression plays a crucial role in atherosclerosis development and plaque remodeling

    Arterioscler. Thromb. Vasc. Biol.

    (2003)
  • J.H. von der Thüsen et al.

    Induction of atherosclerotic plaque rupture in apolipoprotein E-/- mice after adenovirus-mediated transfer of p53

    Circulation

    (2002)
  • S.J. George et al.

    Wild-type p53 gene transfer inhibits neointima formation in human saphenous vein by modulation of smooth muscle cell migration and induction of apoptosis

    Gene Ther.

    (2001)
  • D.W. Meek

    Regulation of the p53 response and its relationship to cancer

    Biochem. J.

    (2015)
  • E. Appella et al.

    Post-translational modifications and activation of p53 by genotoxic stresses

    Eur. J. Biochem.

    (2001)
  • L.M. Jenkins et al.

    p53 N-terminal phosphorylation: a defining layer of complex regulation

    Carcinogenesis

    (2012)
  • C.L. Brooks et al.

    The impact of acetylation and deacetylation on the p53 pathway

    Protein Cell

    (2011)
  • N. Vlatkovic et al.

    Nucleolar control of p53: a cellular Achilles’ heel and a target for cancer therapy

    Cell. Mol. Life Sci. CMLS

    (2014)
  • K. Yang et al.

    Nucleolar Stress: hallmarks, sensing mechanism and diseases

    Cell Stress

    (2018)
  • C.P. Rubbi et al.

    Disruption of the nucleolus mediates stabilization of p53 in response to DNA damage and other stresses

    EMBO J.

    (2003)
  • M.T. Boyd et al.

    The nucleolus directly regulates p53 export and degradation

    J. Cell Biol.

    (2011)
  • X. Bi et al.

    Inhibition of nucleolar stress response by Sirt1: a potential mechanism of acetylation-independent regulation of p53 accumulation

    Aging Cell

    (2019)
  • L.E. Giono et al.

    The p53 tumor suppressor participates in multiple cell cycle checkpoints

    J. Cell. Physiol.

    (2006)
  • C.A. Tonnessen-Murray et al.

    The regulation of cellular functions by the p53 protein: cellular senescence

    Cold Spring Harb. Perspect. Med.

    (2017)
  • O.V. Leontieva et al.

    Hypoxia suppresses conversion from proliferative arrest to cellular senescence

    Proc. Natl. Acad. Sci. USA

    (2012)
  • L. Cao et al.

    A novel ATM/TP53/p21-mediated checkpoint only activated by chronic γ-irradiation

    PLoS One

    (2014)
  • D.A. Liebermann et al.

    p53 induced growth arrest versus apoptosis and its modulation by survival cytokines

    Cell Cycle

    (2007)
  • A.B. Williams et al.

    p53 in the DNA-damage-repair process

    Cold Spring Harb. Perspect. Med.

    (2016)
  • K.H. Khoo et al.

    Drugging the p53 pathway: understanding the route to clinical efficacy

    Nat. Rev. Drug Discov.

    (2014)
  • L.T. Vassilev et al.

    In vivo activation of the p53 pathway by small-molecule antagonists of MDM2

    Science

    (2004)
  • N. Issaeva et al.

    Small molecule RITA binds to p53, blocks p53-HDM-2 interaction and activates p53 function in tumors

    Nat. Med.

    (2004)
  • J. Ristau et al.

    RITA requires eIF2α-dependent modulation of mRNA translation for its anti-cancer activity

    Cell Death Dis.

    (2019)
  • S. Shangary et al.

    Temporal activation of p53 by a specific MDM2 inhibitor is selectively toxic to tumors and leads to complete tumor growth inhibition

    Proc. Natl. Acad. Sci. USA

    (2008)
  • K.L. Bill et al.

    SAR405838: a novel and potent inhibitor of the MDM2:p53 axis for the treatment of dedifferentiated Liposarcoma

    Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res.

    (2016)
  • D. Sun et al.

    Discovery of AMG 232, a potent, selective, and orally bioavailable MDM2-p53 inhibitor in clinical development

    J. Med. Chem.

    (2014)
  • Cited by (10)

    • Integrated network pharmacology and experimental verification to explore the mechanism of Sangqi Qingxuan formula against hypertensive vascular remodeling

      2022, Journal of Traditional Chinese Medical Sciences
      Citation Excerpt :

      Several recent studies have focused on the relationship between p53 and atherosclerosis and vascular remodeling. They found that the overexpression of p53 induced cell cycle arrest in VSMCs, while it had no significant effect on apoptosis.41 Dysregulated p53 plays different roles in macrophages, VSMCs, and endothelial cells in different stages of atherosclerosis, participating in biological processes, such as anti-angiogenesis, apoptosis, autophagy, cell cycle arrest, and altered cell metabolism, with bidirectional regulation.42

    • CX-5461 is a potent immunosuppressant which inhibits T cell-mediated alloimmunity via p53-DUSP5

      2022, Pharmacological Research
      Citation Excerpt :

      Moreover, the experiments with pharmacological p53 inhibitor have proved that the effects of CX-5461 on DUSP5 expression and T cell activation are mainly p53-dependent. In fact, p53-mediated anti-inflammatory actions have been repeatedly reported in different disease models (reviewed in [64]), while evidence suggests that p53-dependent regulation of T cell differentiation to the Treg phenotype may, at least partly, contribute to this effect [47,48]. Deletion of p53 gene in vivo reduces the abundance of Tregs, whereas p53 activation produces opposite effects [47,48].

    View all citing articles on Scopus
    View full text