Next Article in Journal
Genome-Wide Identification and Expression Analysis of the 4-Coumarate: CoA Ligase Gene Family in Solanum tuberosum
Next Article in Special Issue
Zebrafish Models to Study Ectopic Calcification and Calcium-Associated Pathologies
Previous Article in Journal
Synapse Dysfunctions in Multiple Sclerosis
Previous Article in Special Issue
Vascular Smooth Muscle Cell Neutral Sphingomyelinase 2 in the Release of Exosomes and Vascular Calcification
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

p53 Regulates Mitochondrial Dynamics in Vascular Smooth Muscle Cell Calcification

1
The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, UK
2
Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
3
MRC Protein Phosphorylation & Ubiquitylation Unit, Sir James Black Centre, University of Dundee, Dundee DD1 5EH, UK
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(2), 1643; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24021643
Submission received: 6 October 2022 / Revised: 6 January 2023 / Accepted: 9 January 2023 / Published: 13 January 2023

Abstract

:
Arterial calcification is an important characteristic of cardiovascular disease. It has key parallels with skeletal mineralization; however, the underlying cellular mechanisms responsible are not fully understood. Mitochondrial dynamics regulate both bone and vascular function. In this study, we therefore examined mitochondrial function in vascular smooth muscle cell (VSMC) calcification. Phosphate (Pi)-induced VSMC calcification was associated with elongated mitochondria (1.6-fold increase, p < 0.001), increased mitochondrial reactive oxygen species (ROS) production (1.83-fold increase, p < 0.001) and reduced mitophagy (9.6-fold decrease, p < 0.01). An increase in protein expression of optic atrophy protein 1 (OPA1; 2.1-fold increase, p < 0.05) and a converse decrease in expression of dynamin-related protein 1 (DRP1; 1.5-fold decrease, p < 0.05), two crucial proteins required for the mitochondrial fusion and fission process, respectively, were noted. Furthermore, the phosphorylation of DRP1 Ser637 was increased in the cytoplasm of calcified VSMCs (5.50-fold increase), suppressing mitochondrial translocation of DRP1. Additionally, calcified VSMCs showed enhanced expression of p53 (2.5-fold increase, p < 0.05) and β-galactosidase activity (1.8-fold increase, p < 0.001), the cellular senescence markers. siRNA-mediated p53 knockdown reduced calcium deposition (8.1-fold decrease, p < 0.01), mitochondrial length (3.0-fold decrease, p < 0.001) and β-galactosidase activity (2.6-fold decrease, p < 0.001), with concomitant mitophagy induction (3.1-fold increase, p < 0.05). Reduced OPA1 (4.1-fold decrease, p < 0.05) and increased DRP1 protein expression (2.6-fold increase, p < 0.05) with decreased phosphorylation of DRP1 Ser637 (3.20-fold decrease, p < 0.001) was also observed upon p53 knockdown in calcifying VSMCs. In summary, we demonstrate that VSMC calcification promotes notable mitochondrial elongation and cellular senescence via DRP1 phosphorylation. Furthermore, our work indicates that p53-induced mitochondrial fusion underpins cellular senescence by reducing mitochondrial function.

1. Introduction

Arterial calcification involves the deposition of hydroxyapatite crystals within blood vessels including the aorta, carotid and femoral arteries [1]. The process of arterial calcification shares key similarities with physiological bone formation [2,3,4]. Indeed, numerous reports have demonstrated that in a calcified environment, vascular smooth muscle cells (VSMCs), the principal cell type underpinning arterial calcification, can transdifferentiate to osteoblast-like cells [5,6]. Furthermore, inorganic phosphate (Pi) accelerates this transition, as evidenced by the upregulation of osteogenic markers, such as osteocalcin (OCN), sodium-dependent phosphate co-transporter (PiT-1) and tissue-nonspecific alkaline phosphatase (TNAP) [7]. Arterial calcification additionally proceeds via the reciprocal loss of recognised calcification suppressors, including fetuin, inorganic pyrophosphate (PPi) and matrix Gla protein (MGP) [8]. Furthermore, there is the growing evidence that elevated Pi levels can also induce VSMC senescence, with human aortic VSMCs showing upregulated expression of p53 and its downstream target p21, in combination with elevated senescence-associated β-galactosidase (β-gal) activity [9]. Indeed, senescent VSMCs appear to play an important role in mediating the process of arterial calcification [10,11,12].
Mitochondria are crucial bioenergetics hubs within cells, generating toxic reactive oxygen species (ROS) following exposure to oxidative stress [13]. Mitochondrial physiology and function are likely to be affected by this pathological process. Impaired mitochondria function, oxidative stress, modulated mitochondrial metabolism and mitochondrial DNA damage have been noted in disorders that display arterial calcification including type 2 diabetes, end stage renal disease and atherosclerosis [14]. Furthermore, cells undergoing senescence have been reported to show altered mitochondrial function [15,16]. Recent studies by our laboratory have shown that calcifying VSMCs show reduced oxygen consumption rate, ATP-linked respiration and maximal respiration, suggestive of dysregulated mitochondrial activity [17]. However, molecular mechanisms linking mitochondrial dysfunction, cellular senescence and arterial calcification have yet to be determined. In this study, we tested the hypothesis that the senescence regulator p53 plays a functional role in arterial calcification through the regulation of mitochondrial dynamics.

2. Results

2.1. High Phosphate Treatment Alters Mitochondrial Dynamics in VSMCs

Treatment with 3 mM Pi produced an increase in VSMC calcium deposition by day 14 compared with cells cultured in control medium (p < 0.001; Figure 1A). In alignment with recent reports from our laboratory, 3 mM Pi-induced osteogenic transdifferentiation in VSMCs [18,19], as demonstrated by the upregulated protein expression of OCN (2.1-fold increase, p < 0.01; Figure 1B,C) and TNAP (1.5-fold increase, p < 0.05; Figure 1B,D).
Our previous work has shown that VSMCs cultured in calcification medium have impaired respiratory status, marked with low ATP levels and reduced oxygen consumption rate [17]. These data suggest altered mitochondrial function in the arterial calcification process. We now extend these findings and report an increase in mitochondrial reactive oxygen species (ROS) production (1.83-fold increase, p < 0.001; Figure 2A), with a concomitant decrease in mitochondrial membrane potential (as determined by MitoTracker Red CMXRos, 1.6-fold increase, p < 0.01; Figure 2B) in calcified versus control VSMCs.
We next sought to visualize mitochondrial morphology and assess mitophagy (a selective type of autophagy that necessitates the removal of dysfunctional mitochondria) [20,21] in calcified VSMCs isolated from mito-QC transgenic mice. Mito-QC mice utilize a binary fluorescence system whereby a globally expressed tandem mCherry–GFP tag targets mitochondria [22], resulting in these organelles fluorescing red and green within mito-QC mice. Following capture by lysosomes, the low pH reduces the fluorescence of GFP, resulting in mitochondria showing as mCherry foci. Quantification of confocal microscopy images confirmed that calcified mito-QC VSMCs contained hyperfused mitochondria of increased length (1.6-fold increase, p < 0.001; Figure 2C,D), with an associated reduction in the mitophagy index (9.6-fold decrease, p < 0.01; Figure 2E) measured as mCherry-only puncta compared to control cells. Hyperfused mitochondria in calcified VSMCs were also observed using translocase of mitochondrial outer membrane protein (TOM20) staining (Figure S1 (Supplementary Materials)). Together, our data show for the first time that mitochondria within calcified VSMCs are elongated, damaged and do not undergo the process of mitophagic degradation.

2.2. Regulators of Mitochondrial Fusion and Fission Drive Mitochondrial Elongation in Calcified VSMCs

The capacity of mitochondria to alter their morphology is linked to the intricate processes of fission or fusion. We therefore next sought to elucidate the protein expression levels of optic atrophy protein 1 (OPA1) and dynamin-related protein 1 (DRP1), two crucial proteins needed for mitochondrial fusion and fission, respectively [23,24]. Immunoblotting studies revealed an enhanced expression of OPA1 (2.1-fold increase, p < 0.05; Figure 3A,B) and a reduced expression of DRP1 (1.5-fold decrease, p < 0.05; Figure 3A,C) in whole cell lysates produced from VSMCs cultured in calcification versus control medium.
Next, we undertook mitochondrial fractionation (enrichment confirmed by TOM20 expression, Figure 3D), to determine DRP1 expression levels specifically associated with mitochondria. Reduced expression of DRP1 was also observed in the enriched mitochondrial fraction produced from calcified versus control VSMCs (Figure 3E). However, DRP1 function is also modulated by a number of post-translational modifications, such as phosphorylation. In particular, the phosphorylation of DRP1 at Ser637 has been shown to impede its mitochondrial translocation, thus promoting mitochondrial elongation [15]. Consistent with these reports, the ratio of cytoplasmic DRP1 Ser637 phosphorylation to mitochondrial DRP1 was notably elevated in calcified VSMCs (Figure 3E,F).
Together these data strongly suggest that mitochondrial elongation in calcified VSMCs is driven by (i) increased expression of the fusion protein OPA1, (ii) loss of the fission protein DRP1 required for mitophagic degradation and (iii) enhanced mitochondrial fusion via inhibitory phosphorylation of DRP1 at Ser637.

2.3. Calcified VSMCs Show Increased Expression of Senescence Markers

Hyperfused dysfunctional mitochondria are a hallmark of senescence and are frequently observed in natural aging and diseases associated with aging [25,26,27]. We therefore next investigated if calcified VSMCs also show an upregulated expression of established senescence markers. Indeed, VSMCs cultured in calcification medium showed increased mRNA and protein expression levels of p53 (2.5-fold increase, p < 0.05; Figure 4A–C) and p21 (1.9-fold increase, p < 0.05; Figure 4A,B,D), key transcription factors involved in cell cycle regulation, DNA damage and stress [28]. More calcified cells showed the localisation of p53 in the nuclei compared to control VSMCs (5.5-fold increase, p < 0.05; Figure 4E,F). Furthermore, cellular senescence-associated β-galactosidase activity, a typical feature of cellular senescence, was also noted in calcified VSMCs compared to control cells (1.8-fold increase, p < 0.001; Figure 4G,H).

2.4. p53 Suppression Blunts VSMC Calcification and Senescence

Increasing numbers of studies have demonstrated critical roles for the tumour suppressor p53 in regulating not only cellular senescence but also mitochondrial function [15]. To determine the functional actions of p53 within the process of VSMC calcification, we employed RNA interference to suppress p53 expression in VSMCs (2.5-fold decrease, p < 0.001; Figure 5A; p < 0.05; Figure 5B,C). In addition, p53 knockdown significantly reduced p21 mRNA (2.4-fold decrease, p < 0.01; Figure 5A) and protein expression (3.1-fold decrease, p < 0.05; Figure 5B,D). Furthermore, p53 knockdown reduced cellular senescence-associated β-galactosidase activity (2.6-fold decrease, p < 0.001; Figure 5E,F) and calcium deposition compared to scrambled siRNA-treated cells cultured in calcification medium (8.1-fold decrease, p < 0.01; Figure 5G), with an associated reduction in the protein expression of the key osteogenic molecules OCN (1.4-fold decrease, p < 0.001; Figure 5H,I) and TNAP (1.6-fold decrease, p < 0.05; Figure 5H,J).

2.5. Knockdown of p53 Reverses Mitochondrial Dysfunction

Next, we examined whether p53 is essential for mitochondrial elongation and dysfunction during arterial calcification. Mitochondria within p53 knockdown VSMCs cultured in calcification medium showed reduced mitochondrial ROS (p < 0.05; Figure 6A) and enhanced mitochondrial membrane potential (p < 0.01; Figure 6B). Furthermore, mitochondria within p53 knockdown mito-QC VSMCs showed reduced mitochondrial length (3.4-fold decrease, p < 0.001; Figure 6C,D) with a concomitant increase in mitophagy index (3.1-fold increase, p < 0.05; Figure 6C,E) indicative of restored mitophagy, compared to mitochondria within scramble control cells. Altered mitochondrial length was also confirmed with TOM20 staining (Figure S2).
Consistent with these changes, immunoblotting studies revealed reduced expression of OPA1 (4.1-fold decrease, p < 0.05; Figure 7A,B) and increased expression of DRP1 (2.6-fold increase, p < 0.01; Figure 7A,C) in whole cell lysates produced from p53 knockdown VSMCs cultured in calcification medium versus scrambled control cells. Increased expression of DRP1 was also observed in the enriched mitochondrial fraction isolated from calcified VSMCs, conversely, the phosphorylation of cytoplasmic DRP1 Ser637 was significantly reduced (3.20-fold decrease, p < 0.001; Figure 7E,F) following p53 knockdown (Figure 7E) (enrichment confirmed by TOM20 expression, Figure 7D).
In order to confirm our observation that p53 is essential for mitochondrial elongation and dysfunction during arterial calcification, we overexpressed p53 in VSMCs and subsequently calcified the cells for 7 days (Figure 8). Overexpression of p53 protein was confirmed by immunoblotting (2.2-fold increase, p < 0.05; Figure 8A,B). Overexpression of p53 induced an increase in the protein expression of p21 (2.6-fold increase, p < 0.05; Figure 8A,C) and a decrease in DRP1expression (2.1-fold decrease, p < 0.05; Figure 8A,E), with no further significant change in OPA1 expression (Figure 8A,D). A concomitant increase in mitochondrial length (2.1-fold increase, p < 0.001; Figure 8F,G) and VSMC calcification (3.7-fold increase, p < 0.001; Figure 8H) was also observed.
In conclusion, our data suggest that suppression of p53, a critical regulator of senescence, exerts protective effects against pathological calcification and restores mitochondrial dynamics by restoring DRP1-mediated fission required for mitophagic signalling in VSMCs.

3. Discussion

This study highlights the crucial role of mitochondrial function and cellular senescence in arterial calcification. Here, we provide novel functional evidence to suggest that p53 regulates this pathological process.
Cellular senescence is a critical mechanism that contributes to arterial calcification, a critical feature of vascular pathology in the aging population. Indeed, senescent VSMCs have been shown to calcify more readily than young, early passage VSMCs [29]. Typified as the cell cycle arrest of cells undergoing mitosis, cells activate p53 and p21 as final effectors for the senescence process [30]. Our data reveal that under calcifying culture conditions, murine VSMCs upregulate the expression of senescence markers, including p53, p21 and SA-β-gal, supporting and extending previous work in human cells and demonstrating that cellular senescence impacts vascular aging and arterial calcification [31]. Human VSMCs in culture can form multi-cellular nodules that spontaneously calcify and are positive for senescence-associated β-galactosidase [32]. Histological analysis of calcified arteries isolated from children on dialysis has revealed that VSMCs accumulate the senescence marker p21 [12]. Moreover, VSMCs cultured in vitro from these arteries underwent senescence at earlier passages than VSMCs from age-matched controls. Furthermore, VSMCs cultured from human calcified atherosclerotic plaques show a senescent phenotype [33], and it has been proposed that the senescence VSMCs could be the potential cause of plaque rupture [34].
Recent reports have shown that alterations in mitochondrial shape, size and function are important characteristics of cellular senescence and accelerate the senescent phenotype [25,35]. Furthermore, published data from our laboratory have demonstrated that calcified VSMCs show impaired respiratory status with reduced potential for ATP synthesis and reduced oxygen consumption rate [17]. Here, we highlight that under calcifying conditions, VSMCs have elongated mitochondria that avoid mitochondrial fission, which is required for their removal via mitophagy machinery. We propose that the elongated mitochondria evade mitophagy and sustain impaired mitochondria for a longer duration under calcifying conditions. This is further supported by our observation of reduced mitochondrial lysosomal delivery in calcified mito-QC VSMCs. Elongation may dilute the impaired respiratory status by equilibrating matrix metabolites and mitochondrial membrane components [36], thus exerting a protective action against cell death via apoptosis [37].
To understand the mechanisms underpinning mitochondrial elongation, we determined the expression levels of fusion and fission proteins, as increased mitochondrial fission is essential for apoptosis and mitophagy [37], whereas elongation (fusion) of mitochondria has been noted in cellular senescence models [25,38,39]. We show that mitochondrial elongation in calcified VSMCs is driven not only by the loss of DRP1, a key regulator of mitochondrial fission required for mitophagic degradation [40], but also enhanced mitochondrial fusion via the inhibitory phosphorylation of DRP1. Interestingly, our results are in contrast to previous data suggesting that DRP1 inhibition can diminish VSMC mitochondrial damage and calcification [41]; however, this previous work did not explore the regulation of DRP1 function by post-translational modifications. Further studies are now required to directly test the functional role of key regulators of mitochondrial function in VSMC calcification. These include the mitochondrial fusion/fission proteins OPA1, DRP1 and MFN1/2 [42], as well as PINK-1 and Parkin, which activate the clearance of damaged mitochondria [43].
Previous studies highlighting a critical role for the tumour suppressor p53 in mitochondrial function are supported by our findings that p53 knockdown in VSMCs leads to reduced mitochondrial length, restored mitophagy and enhanced membrane potential. Previously, p53 has been shown to promote mitochondrial respiration via cytochrome c oxidase 2 transcriptional activation [44]. Furthermore, p53 directly cooperates with the Bcl-2 protein family within the mitochondrial outer membrane during a genotoxic response [45]. Additionally, it has been shown that p53 directly modulates mitochondrial function in a human endometrial carcinoma cell-line, whereby infection with the p53 adenovirus induced a significant decrease in mitochondrial membrane potential and induced mitochondrial fusion preceding cellular senescence [15]. Indeed, it has been demonstrated that p53 activation and over-expression promote cellular senescence in a range of cells, including human bladder carcinoma cells and fibroblasts [15,46]. These findings are further supported by our observation of increased expression of the senescence regulator p21 following p53 overexpression in calcifying VSMCs. In addition, p16 ink4a has been reported to play an important role in proliferation arrest and cellular senescence [47]. However, in the present study, no changes in p16 ink4a expression levels were observed during VSMC calcification. These data likely reflect previous findings demonstrating that senescence induced by p16 ink4a is mostly regulated by environmental factors such as ionizing radiation, DNA-damaging agents and oncogene-mediated activation [48,49].
In contrast to our data, recent studies have reported reduced expression of p53 during VSMC calcification following treatment of cells with the putative cancer therapeutics, piperlongumine [50] and teniposide [51]. These divergent observations may reflect key differences in the species, passage status and culture conditions of the cells investigated. Furthermore, these findings likely reflect the activation of different signalling mechanisms, with piperlongumine modulating PTEN transcription [50] and teniposide regulating the miR-203-3p-BMP2 pathway [51]. Indeed, our data support novel findings challenging the commonly accepted paradigm of p53 having primarily pro-apoptotic functions in VSMCs [52].
A number of studies have revealed an important role for p53 overexpression in the induction of senescence and alteration of mitochondrial dynamics in a variety of cell types. It has been shown that p53 directly modulates the mitochondrial function in a human endometrial carcinoma cell-line, whereby infection with p53 adenovirus induced a significant decrease in mitochondrial membrane potential and induced mitochondrial fusion preceding cellular senescence [15,53]. Furthermore, it has been demonstrated that p53 activation and overexpression promote cellular senescence in a range of cells, including human bladder carcinoma cells and fibroblasts [15,46]. Future functional studies in non-calcified VSMCs would therefore be highly informative in clarifying whether calcification is a cause or consequence of altered p53 levels.
Our data further show that p53 induces mitochondrial elongation in VSMC calcification through a novel post-translational mechanism, mediating DRP1 Ser637 phosphorylation. Indeed, we observed reduced localisation of DRP1 in mitochondria, likely reflecting the previous reports that the phosphorylation status of Ser637 plays a dominant role in the subcellular localisation of DRP1 [54]. A comparable p53/DRP1-mediated mechanism has also been reported to induce cellular senescence in cancer cell lines [15], highlighting the importance of this pathway in different pathologies.
In summary, we demonstrate that VSMC calcification promotes notable mitochondrial elongation and cellular senescence via Drp1 phosphorylation. Furthermore, this work indicates that p53-induced mitochondrial fusion underpins cellular senescence by reducing mitochondrial function.

4. Materials and Methods

4.1. Maintenance of Mito-QC Mice

The generation and characterisation of the mito-QC mouse was previously described [22]. As described below, 2–3-month-old mito-QC mice were used for VSMC cultures. Mice were euthanised by CO2 followed by cervical dislocation. The mito-QC mouse work was approved by University of Edinburgh ethics committee.

4.2. VSMC Isolation, Culture and Calcification

Primary aortic VSMCs were obtained and calcification induced as described previously using protocols established by our laboratory [18]. Cells were cultured for up to 14 days, and the medium changed every two or three days. For all the experiments, n = 3 is representative of 3 independent experiments pooled from 8eight aortas. Calcium deposition was assessed by HCl leaching [18].

4.3. Transfection Assays

Cells were transfected with 80 pmol murine p53 (SC-29436; Santa Cruz Biotechnology, Dallas, TX, USA) or scrambled control siRNA (SC-37007; Santa Cruz Biotechnology) with siRNA transfection reagent (SC-29528; Santa Cruz Biotechnology) following the manufacturer’s protocol. For p53 overexpression, Trp53_OMu22609D_pcDNA3.1+/C-(K)-DYK, Clone ID:OMu22609D was transfected in VSMCs at a concentration of 2 μg/mL. Vector without Trp53 ORF was used as a control.

4.4. Cell Imaging

Cells were seeded in 24-well plates (5 × 104 cells per well) on glass cover slips and subsequently fixed with 3.7% paraformaldehyde (PFA) (pH 7.0) at 4 °C, permeabilised with 0.1% triton X100 (Sigma, St. Louis, MO, USA) and blocked with 2% goat before incubating with primary antibodies TOM20 (1:300; 42406; Cell Signaling Technology, Danvers, MA, USA) overnight. After washing, cells were incubated for 1 h in the dark with Alexa Fluor@488 anti-rabbit antibody (A11034; Life Technologies, Waltham, MA, USA). Hoechst staining was then performed (1:10,000; Sigma). Glass coverslips were mounted onto slides using Prolong®Gold Anti-Fade Reagent with DAPI (Life Technologies) and the fluorescence signal observed (Zeiss LSM 710; Oberkochen, Germany).

4.5. Senescence Associated β-Gal Staining

Senescence-associated β–galactosidase (SA β-gal) staining was undertaken according to the manufacturers’ instructions (Merk Millipore, Burlington, MA, USA). Briefly, cells were fixed (0.25% glutaraldehyde) and SA-β-gal staining (pH 6.0) was carried out. Images were captured using an Axiovert 25 (Zeiss) microscope. SA-β-gal positive cells were quantified using image analysis software (ImageJ, Bethesda, MD, USA).

4.6. MitoTracker Staining

Cells were stained with 100 nM MitoTracker Red CMXRos (Invitrogen, Waltham, MA, USA). Subsequently, cells were fixed in 4% PFA (pH7), images of the stained cells were captured (Zeiss LSM 710; Oberkochen, Germany) and staining quantified using Image J.

4.7. Measurement of ROS Levels

ROS production in cultured cells was determined using a commercially available mitochondrial ROS detection kit (Cayman Chemicals, Ann Arbor, MI, USA) with an excitation wavelength of 480–515 nm and emission wavelength at 560–600 nm.

4.8. Mitophagy Assay

Mito-QC VSMCs were fixed at 4 °C with 4% PFA (pH7). Images of the stained cells were captured (Zeiss LSM 710; Oberkochen, Germany). Red/mCherry puncta, indicative of mitophagy index, were counted (mitoQC counter, Fiji plugin) and organelle length measured using image analysis software (ImageJ, WI, USA).

4.9. Mitochondrial Length Measurement

Mitochondrial length measurement was quantified with image analysis software (ImageJ, WI, USA).

4.10. Analysis of Gene Expression

RNA was extracted and reverse transcribed, and specific cDNAs were quantified by real-time qPCR [5]. The following mouse primers (Sigma) were used: p53 Forward 5′-GTATTTCACCCTCAAGATCC, Reverse 5′-TGGGCATCCTTTAACTCTA; p21 Forward 5′-CCTGGTGATGTCCGACCTG, Reverse 5′-CCATGAGCGCATCGAATC.

4.11. Immunoblotting

Cells were lysed in radioimmunoprecipitation assay (RIPA) buffer containing Protease Inhibitor Cocktail (Thermo Fisher Scientific, Waltham, MA, USA) and total protein concentration measured using a commercially available BCA protein assay (Thermo Scientific). Mitochondrial fractions were isolated from cultured VSMCs using a commercial kit (89874; Thermo Scientific). Immunoblotting was performed as previously described [55]. Nitrocellulose membranes were incubated for up to 16 h at 4 °C with primary antibodies (1:1000 dilution in 5% milk in TBST), p53 (7F5; Cell Signaling Technology), p21 (2947; Cell Signaling Technology), TOM20 (DBT4N; Cell Signaling Technology), OPA1 (ab157457; Abcam, Cambridge, UK), DRP1 (ab184247; Abcam), DRP1-pS637 (BT-PHS00841; Bioassay Technology Laboratory, Birmingham, UK), TNAP (MAB 2909; R & D Systems, Minneapolis, MN, USA) and OCN (ab93876; Abcam). Membranes were then probed with HRP-conjugated goat anti-rabbit IgG (P0448; Dako) and goat anti-rat IgG (HAF005; Dako, Glostrup, Denmark) for 1 h. Membranes were developed using the GeneGenome system (Syngene, Frederick, MD, USA). Membranes were then washed and re-probed for mouse β-actin expression (1:10,000; A3845; Sigma). Semi-quantitative assessment of band intensity was performed using ImageJ image analysis software.

4.12. Statistical Analysis

All data are presented as mean ± SEM. Data were analysed by unpaired t-test or one-way analysis of variance (ANOVA) followed by Tukey’s range test. All statistical analysis was performed using GraphPad prism (CA, USA). Values of p < 0.05 were regarded as significant, and p values are shown as: * p < 0.05; ** p < 0.01; *** p < 0.001.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/ijms24021643/s1.

Author Contributions

Conceptualization, K.P. and V.E.M.; methodology, K.P., Q.-Y.T., I.L., J.-F.Z., R.K.S. and I.G.G.; software, K.P.; investigation, K.P. and Q.-Y.T.; resources, V.E.M., R.K.S. and I.G.G.; writing—original draft preparation, V.E.M. and K.P.; writing—review and editing, I.L., B.C., R.K.S. and I.G.G.; funding acquisition, V.E.M., R.K.S. and I.G.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Biotechnology and Biological Sciences Research Council (BBSRC) (BB/J004316/1; BBS/E/D/20221657; VEM), the Medical Research Council (MRC) (MC_UU_00018/2; IGG), The Wellcome Trust (210752/Z/18/Z; RKS), International Postdoc Grants (Swedish Research Council; 2019-06422; IL) and the British Heart Foundation Research Excellence Award Funding (VEM, KP, RS).

Institutional Review Board Statement

Ethics Committee Name: University of Edinburgh; Approval Code: P87539BCC; Approval Date: 19–24 September 2005.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available within the article or Supplementary Materials. For the purpose of open access, the authors have applied a Creative Commons Attribution (CC BY) licence to any author accepted manuscript version arising from this submission.

Acknowledgments

We thank Anna Raper for support with confocal microscopy.

Conflicts of Interest

IGG is a consultant for Mitobridge Inc. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Woodward, H.J.; Zhu, D.; Hadoke, P.W.F.; MacRae, V.E. Regulatory Role of Sex Hormones in Cardiovascular Calcification. Int. J. Mol. Sci. 2021, 22, 4620. [Google Scholar] [CrossRef] [PubMed]
  2. Zhu, D.; Mackenzie, N.C.; Farquharson, C.; Macrae, V.E. Mechanisms and clinical consequences of vascular calcification. Front. Endocrinol. 2012, 3, 95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Shroff, R.C.; Shanahan, C.M. The vascular biology of calcification. Semin. Dial. 2007, 20, 103–109. [Google Scholar] [CrossRef] [PubMed]
  4. Demer, L.L.; Tintut, Y. Vascular calcification: Pathobiology of a multifaceted disease. Circulation 2008, 117, 2938–2948. [Google Scholar] [CrossRef] [PubMed]
  5. Zhu, D.; Mackenzie, N.C.; Millan, J.L.; Farquharson, C.; MacRae, V.E. The appearance and modulation of osteocyte marker expression during calcification of vascular smooth muscle cells. PLoS ONE 2011, 6, e19595. [Google Scholar] [CrossRef] [Green Version]
  6. Speer, M.Y.; Chien, Y.C.; Quan, M.; Yang, H.Y.; Vali, H.; McKee, M.D.; Giachelli, C.M. Smooth muscle cells deficient in osteopontin have enhanced susceptibility to calcification in vitro. Cardiovasc. Res. 2005, 66, 324–333. [Google Scholar] [CrossRef] [Green Version]
  7. Speer, M.Y.; Yang, H.Y.; Brabb, T.; Leaf, E.; Look, A.; Lin, W.L.; Frutkin, A.; Dichek, D.; Giachelli, C.M. Smooth muscle cells give rise to osteochondrogenic precursors and chondrocytes in calcifying arteries. Circ. Res. 2009, 104, 733–741. [Google Scholar] [CrossRef]
  8. Back, M.; Aranyi, T.; Cancela, M.L.; Carracedo, M.; Conceicao, N.; Leftheriotis, G.; Macrae, V.; Martin, L.; Nitschke, Y.; Pasch, A.; et al. Endogenous Calcification Inhibitors in the Prevention of Vascular Calcification: A Consensus Statement From the COST Action EuroSoftCalcNet. Front. Cardiovasc. Med. 2018, 5, 196. [Google Scholar] [CrossRef]
  9. Troyano, N.; Nogal, M.D.; Mora, I.; Diaz-Naves, M.; Lopez-Carrillo, N.; Sosa, P.; Rodriguez-Puyol, D.; Olmos, G.; Ruiz-Torres, M.P. Hyperphosphatemia induces cellular senescence in human aorta smooth muscle cells through integrin linked kinase (ILK) up-regulation. Mech. Ageing Dev. 2015, 152, 43–55. [Google Scholar] [CrossRef]
  10. Mackenzie, N.C.; MacRae, V.E. The role of cellular senescence during vascular calcification: A key paradigm in aging research. Curr. Aging Sci. 2011, 4, 128–136. [Google Scholar] [CrossRef]
  11. Matthews, C.; Gorenne, I.; Scott, S.; Figg, N.; Kirkpatrick, P.; Ritchie, A.; Goddard, M.; Bennett, M. Vascular smooth muscle cells undergo telomere-based senescence in human atherosclerosis: Effects of telomerase and oxidative stress. Circ. Res. 2006, 99, 156–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Sanchis, P.; Ho, C.Y.; Liu, Y.; Beltran, L.E.; Ahmad, S.; Jacob, A.P.; Furmanik, M.; Laycock, J.; Long, D.A.; Shroff, R.; et al. Arterial "inflammaging" drives vascular calcification in children on dialysis. Kidney Int. 2019, 95, 958–972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Phadwal, K.; Vrahnas, C.; Ganley, I.G.; MacRae, V.E. Mitochondrial Dysfunction: Cause or Consequence of Vascular Calcification? Front. Cell Dev. Biol. 2021, 9, 611922. [Google Scholar] [CrossRef] [PubMed]
  14. Nishikawa, T.; Edelstein, D.; Du, X.L.; Yamagishi, S.; Matsumura, T.; Kaneda, Y.; Yorek, M.A.; Beebe, D.; Oates, P.J.; Hammes, H.P.; et al. Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature 2000, 404, 787–790. [Google Scholar] [CrossRef]
  15. Kim, Y.Y.; Um, J.H.; Yoon, J.H.; Lee, D.Y.; Lee, Y.J.; Kim, D.H.; Park, J.I.; Yun, J. p53 regulates mitochondrial dynamics by inhibiting Drp1 translocation into mitochondria during cellular senescence. FASEB J. 2020, 34, 2451–2464. [Google Scholar] [CrossRef] [Green Version]
  16. Ziegler, D.V.; Wiley, C.D.; Velarde, M.C. Mitochondrial effectors of cellular senescence: Beyond the free radical theory of aging. Aging Cell 2015, 14, 1–7. [Google Scholar] [CrossRef] [Green Version]
  17. Rashdan, N.A.; Sim, A.M.; Cui, L.; Phadwal, K.; Roberts, F.L.; Carter, R.; Ozdemir, D.D.; Hohenstein, P.; Hung, J.; Kaczynski, J.; et al. Osteocalcin Regulates Arterial Calcification Via Altered Wnt Signaling and Glucose Metabolism. J. Bone Miner. Res. 2020, 35, 357–367. [Google Scholar] [CrossRef]
  18. Zhu, D.; Rashdan, N.A.; Chapman, K.E.; Hadoke, P.W.; MacRae, V.E. A novel role for the mineralocorticoid receptor in glucocorticoid driven vascular calcification. Vascul. Pharmacol. 2016, 86, 87–93. [Google Scholar] [CrossRef] [Green Version]
  19. Zhu, D.; Mackenzie, N.C.; Shanahan, C.M.; Shroff, R.C.; Farquharson, C.; MacRae, V.E. BMP-9 regulates the osteoblastic differentiation and calcification of vascular smooth muscle cells through an ALK1 mediated pathway. J. Cell Mol. Med. 2015, 19, 165–174. [Google Scholar] [CrossRef]
  20. Palikaras, K.; Lionaki, E.; Tavernarakis, N. Mechanisms of mitophagy in cellular homeostasis, physiology and pathology. Nat. Cell Biol. 2018, 20, 1013–1022. [Google Scholar] [CrossRef]
  21. Rodger, C.E.; McWilliams, T.G.; Ganley, I.G. Mammalian mitophagy—from in vitro molecules to in vivo models. FEBS J. 2018, 285, 1185–1202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. McWilliams, T.G.; Prescott, A.R.; Allen, G.F.; Tamjar, J.; Munson, M.J.; Thomson, C.; Muqit, M.M.; Ganley, I.G. mito-QC illuminates mitophagy and mitochondrial architecture in vivo. J. Cell Biol. 2016, 214, 333–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ge, Y.; Shi, X.; Boopathy, S.; McDonald, J.; Smith, A.W.; Chao, L.H. Two forms of Opa1 cooperate to complete fusion of the mitochondrial inner-membrane. Elife 2020, 9, e50973. [Google Scholar] [CrossRef] [PubMed]
  24. Fonseca, T.B.; Sanchez-Guerrero, A.; Milosevic, I.; Raimundo, N. Mitochondrial fission requires DRP1 but not dynamins. Nature 2019, 570, E34–E42. [Google Scholar] [CrossRef]
  25. Yoon, Y.S.; Yoon, D.S.; Lim, I.K.; Yoon, S.H.; Chung, H.Y.; Rojo, M.; Malka, F.; Jou, M.J.; Martinou, J.C.; Yoon, G. Formation of elongated giant mitochondria in DFO-induced cellular senescence: Involvement of enhanced fusion process through modulation of Fis1. J. Cell Physiol. 2006, 209, 468–480. [Google Scholar] [CrossRef]
  26. Mai, S.; Klinkenberg, M.; Auburger, G.; Bereiter-Hahn, J.; Jendrach, M. Decreased expression of Drp1 and Fis1 mediates mitochondrial elongation in senescent cells and enhances resistance to oxidative stress through PINK1. J. Cell Sci. 2010, 123, 917–926. [Google Scholar] [CrossRef] [Green Version]
  27. Coleman, R.; Silbermann, M.; Gershon, D.; Reznick, A.Z. Giant mitochondria in the myocardium of aging and endurance-trained mice. Gerontology 1987, 33, 34–39. [Google Scholar] [CrossRef]
  28. Rufini, A.; Tucci, P.; Celardo, I.; Melino, G. Senescence and aging: The critical roles of p53. Oncogene 2013, 32, 5129–5143. [Google Scholar] [CrossRef]
  29. Nakano-Kurimoto, R.; Ikeda, K.; Uraoka, M.; Nakagawa, Y.; Yutaka, K.; Koide, M.; Takahashi, T.; Matoba, S.; Yamada, H.; Okigaki, M.; et al. Replicative senescence of vascular smooth muscle cells enhances the calcification through initiating the osteoblastic transition. Am. J. Physiol. Heart Circ. Physiol. 2009, 297, H1673–H1684. [Google Scholar] [CrossRef]
  30. Chen, J. The Cell-Cycle Arrest and Apoptotic Functions of p53 in Tumor Initiation and Progression. Cold Spring Harb Perspect Med. 2016, 6, a026104. [Google Scholar] [CrossRef]
  31. Baker, D.J.; Childs, B.G.; Durik, M.; Wijers, M.E.; Sieben, C.J.; Zhong, J.; Saltness, R.A.; Jeganathan, K.B.; Verzosa, G.C.; Pezeshki, A.; et al. Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature 2016, 530, 184–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Liu, Y.; Drozdov, I.; Shroff, R.; Beltran, L.E.; Shanahan, C.M. Prelamin A accelerates vascular calcification via activation of the DNA damage response and senescence-associated secretory phenotype in vascular smooth muscle cells. Circ. Res. 2013, 112, e99–e109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Bennett, M.R.; Macdonald, K.; Chan, S.W.; Boyle, J.J.; Weissberg, P.L. Cooperative interactions between RB and p53 regulate cell proliferation, cell senescence, and apoptosis in human vascular smooth muscle cells from atherosclerotic plaques. Circ. Res. 1998, 82, 704–712. [Google Scholar] [CrossRef] [PubMed]
  34. Gorenne, I.; Kavurma, M.; Scott, S.; Bennett, M. Vascular smooth muscle cell senescence in atherosclerosis. Cardiovasc. Res. 2006, 72, 9–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Lee, S.; Jeong, S.Y.; Lim, W.C.; Kim, S.; Park, Y.Y.; Sun, X.; Youle, R.J.; Cho, H. Mitochondrial fission and fusion mediators, hFis1 and OPA1, modulate cellular senescence. J. Biol. Chem. 2007, 282, 22977–22983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Busch, K.B.; Bereiter-Hahn, J.; Wittig, I.; Schagger, H.; Jendrach, M. Mitochondrial dynamics generate equal distribution but patchwork localization of respiratory Complex I. Mol. Membr. Biol. 2006, 23, 509–520. [Google Scholar] [CrossRef] [PubMed]
  37. Karbowski, M.; Arnoult, D.; Chen, H.; Chan, D.C.; Smith, C.L.; Youle, R.J. Quantitation of mitochondrial dynamics by photolabeling of individual organelles shows that mitochondrial fusion is blocked during the Bax activation phase of apoptosis. J. Cell Biol. 2004, 164, 493–499. [Google Scholar] [CrossRef] [Green Version]
  38. Yu, B.; Ma, J.; Li, J.; Wang, D.; Wang, Z.; Wang, S. Mitochondrial phosphatase PGAM5 modulates cellular senescence by regulating mitochondrial dynamics. Nat. Commun. 2020, 11, 2549. [Google Scholar] [CrossRef]
  39. Ren, X.; Chen, L.; Xie, J.; Zhang, Z.; Dong, G.; Liang, J.; Liu, L.; Zhou, H.; Luo, P. Resveratrol Ameliorates Mitochondrial Elongation via Drp1/Parkin/PINK1 Signaling in Senescent-Like Cardiomyocytes. Oxid. Med. Cell Longev. 2017, 2017, 4175353. [Google Scholar] [CrossRef] [Green Version]
  40. Twig, G.; Shirihai, O.S. The interplay between mitochondrial dynamics and mitophagy. Antioxid. Redox Signal. 2011, 14, 1939–1951. [Google Scholar] [CrossRef]
  41. Rogers, M.A.; Maldonado, N.; Hutcheson, J.D.; Goettsch, C.; Goto, S.; Yamada, I.; Faits, T.; Sesaki, H.; Aikawa, M.; Aikawa, E. Dynamin-Related Protein 1 Inhibition Attenuates Cardiovascular Calcification in the Presence of Oxidative Stress. Circ. Res. 2017, 121, 220–233. [Google Scholar] [CrossRef]
  42. Hall, A.R.; Burke, N.; Dongworth, R.K.; Hausenloy, D.J. Mitochondrial fusion and fission proteins: Novel therapeutic targets for combating cardiovascular disease. Br. J. Pharmacol. 2014, 171, 1890–1906. [Google Scholar] [CrossRef] [PubMed]
  43. Durcan, T.M.; Fon, E.A. The three ‘P’s of mitophagy: PARKIN, PINK1, and post-translational modifications. Genes Dev. 2015, 29, 989–999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Matoba, S.; Kang, J.G.; Patino, W.D.; Wragg, A.; Boehm, M.; Gavrilova, O.; Hurley, P.J.; Bunz, F.; Hwang, P.M. p53 regulates mitochondrial respiration. Science 2006, 312, 1650–1653. [Google Scholar] [CrossRef]
  45. Tomita, Y.; Marchenko, N.; Erster, S.; Nemajerova, A.; Dehner, A.; Klein, C.; Pan, H.; Kessler, H.; Pancoska, P.; Moll, U.M. WT p53, but not tumor-derived mutants, bind to Bcl2 via the DNA binding domain and induce mitochondrial permeabilization. J. Biol. Chem. 2006, 281, 8600–8606. [Google Scholar] [CrossRef] [Green Version]
  46. Sugrue, M.M.; Shin, D.Y.; Lee, S.W.; Aaronson, S.A. Wild-type p53 triggers a rapid senescence program in human tumor cells lacking functional p53. Proc. Natl. Acad. Sci. USA 1997, 94, 9648–9653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Rayess, H.; Wang, M.B.; Srivatsan, E.S. Cellular senescence and tumor suppressor gene p16. Int. J. Cancer 2012, 130, 1715–1725. [Google Scholar] [CrossRef] [Green Version]
  48. Meng, A.; Wang, Y.; Van Zant, G.; Zhou, D. Ionizing radiation and busulfan induce premature senescence in murine bone marrow hematopoietic cells. Cancer Res. 2003, 63, 5414–5419. [Google Scholar] [PubMed]
  49. Wang, Y.; Schulte, B.A.; LaRue, A.C.; Ogawa, M.; Zhou, D. Total body irradiation selectively induces murine hematopoietic stem cell senescence. Blood 2006, 107, 358–366. [Google Scholar] [CrossRef] [Green Version]
  50. Shi, W.; Lu, J.; Li, J.; Qiu, M.; Lu, Y.; Gu, J.; Kong, X.; Sun, W. Piperlongumine Attenuates High Calcium/Phosphate-Induced Arterial Calcification by Preserving P53/PTEN Signaling. Front. Cardiovasc. Med. 2020, 7, 625215. [Google Scholar] [CrossRef]
  51. Liu, L.; Zeng, P.; Yang, X.; Duan, Y.; Zhang, W.; Ma, C.; Zhang, X.; Yang, S.; Li, X.; Yang, J.; et al. Inhibition of Vascular Calcification. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 2382–2395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Popowich, D.A.; Vavra, A.K.; Walsh, C.P.; Bhikhapurwala, H.A.; Rossi, N.B.; Jiang, Q.; Aalami, O.O.; Kibbe, M.R. Regulation of reactive oxygen species by p53: Implications for nitric oxide-mediated apoptosis. Am J Physiol. Heart Circ. Physiol. 2010, 298, H2192–H2200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Kim, Y.Y.; Um, J.H.; Shin, D.J.; Jeong, D.J.; Hong, Y.B.; Yun, J. p53-mediated regulation of mitochondrial dynamics plays a pivotal role in the senescence of various normal cells as well as cancer cells. FASEB J. 2021, 35, e21319. [Google Scholar] [CrossRef] [PubMed]
  54. Cereghetti, G.M.; Stangherlin, A.; Martins de Brito, O.; Chang, C.R.; Blackstone, C.; Bernardi, P.; Scorrano, L. Dephosphorylation by calcineurin regulates translocation of Drp1 to mitochondria. Proc. Natl. Acad. Sci. USA 2008, 105, 15803–15808. [Google Scholar] [CrossRef] [Green Version]
  55. Phadwal, K.; Kurian, D.; Salamat, M.K.F.; MacRae, V.E.; Diack, A.B.; Manson, J.C. Spermine increases acetylation of tubulins and facilitates autophagic degradation of prion aggregates. Sci Rep. 2018, 8, 10004. [Google Scholar] [CrossRef]
Figure 1. Calcification of VSMCs in vitro. (A) Calcium content (mg/mg protein) of VSMCs (n = 6). (B) Representative image of immunoblotting and associated density quantification of (C) Osteocalcin (OCN) (n = 3) and (D) tissue-nonspecific alkaline phosphatase (TNAP) (n = 3) protein expression compared with ACTB (β-actin). Data shown as mean +/− S.E.M * p < 0.05; ** p < 0.01; *** p < 0.001 compared to control.
Figure 1. Calcification of VSMCs in vitro. (A) Calcium content (mg/mg protein) of VSMCs (n = 6). (B) Representative image of immunoblotting and associated density quantification of (C) Osteocalcin (OCN) (n = 3) and (D) tissue-nonspecific alkaline phosphatase (TNAP) (n = 3) protein expression compared with ACTB (β-actin). Data shown as mean +/− S.E.M * p < 0.05; ** p < 0.01; *** p < 0.001 compared to control.
Ijms 24 01643 g001
Figure 2. Mitochondrial elongation and dysfunction in calcified VSMCs. Quantification of (A) mitochondrial reactive oxygen species (ROS) (470/560 nm) and (B) mitochondrial membrane potential by mitotracker red CMXRos (mean intensity). (C) Representative confocal images of mito-QC VSMCs (white arrows show hyperfused mitochondria). Quantification of (D) mitochondrial length (µM) and (E) Mitophagy index. Data shown as mean +/− S.E.M. (n = 6) ** p < 0.01; *** p < 0.001; **** p < 0.0001 compared to control. Scale bars = 10 μm.
Figure 2. Mitochondrial elongation and dysfunction in calcified VSMCs. Quantification of (A) mitochondrial reactive oxygen species (ROS) (470/560 nm) and (B) mitochondrial membrane potential by mitotracker red CMXRos (mean intensity). (C) Representative confocal images of mito-QC VSMCs (white arrows show hyperfused mitochondria). Quantification of (D) mitochondrial length (µM) and (E) Mitophagy index. Data shown as mean +/− S.E.M. (n = 6) ** p < 0.01; *** p < 0.001; **** p < 0.0001 compared to control. Scale bars = 10 μm.
Ijms 24 01643 g002
Figure 3. Calcified VSMCs show modulated Optic Atrophy Protein 1 (OPA1) and Dynamin-related protein 1 (DRP1) protein expression (A) Representative immunoblotting of whole cell lysates and associated quantification of (B) OPA1 and (C) DRP1 expression compared with ACTB (β-actin). (D) Representative immunoblotting of mitochondrial and cytoplasmic fraction with TOM20 and ACTB (β-actin). (E) Representative immunoblotting of mitochondrial expression of DRP1 and cytoplasmic expression of DRP1– pS637 associated quantification of (F) ratio of cytoplasmic DRP1- pS637/mitochondrial DRP1. Data shown as mean +/− S.E.M (n = 3) * p < 0.05 compared to control.
Figure 3. Calcified VSMCs show modulated Optic Atrophy Protein 1 (OPA1) and Dynamin-related protein 1 (DRP1) protein expression (A) Representative immunoblotting of whole cell lysates and associated quantification of (B) OPA1 and (C) DRP1 expression compared with ACTB (β-actin). (D) Representative immunoblotting of mitochondrial and cytoplasmic fraction with TOM20 and ACTB (β-actin). (E) Representative immunoblotting of mitochondrial expression of DRP1 and cytoplasmic expression of DRP1– pS637 associated quantification of (F) ratio of cytoplasmic DRP1- pS637/mitochondrial DRP1. Data shown as mean +/− S.E.M (n = 3) * p < 0.05 compared to control.
Ijms 24 01643 g003
Figure 4. Increased senescence in calcified VSMCs (A) Fold change in mRNA expression of p53 and p21. (B) Representative immunoblots and associated quantification of (C) p53 and (D) p21 compared with ACTB (β-actin). (E) Representative images and (F) associated quantification of cells with p53 staining in the nucleus (G) Representative images and (H) associated quantification of SA β-gal staining (red arrows indicate positive staining). Data shown as mean +/− S.E.M (n = 3–6) * p < 0.05; *** p < 0.001 compared to control. Scale bars = 5 μm.
Figure 4. Increased senescence in calcified VSMCs (A) Fold change in mRNA expression of p53 and p21. (B) Representative immunoblots and associated quantification of (C) p53 and (D) p21 compared with ACTB (β-actin). (E) Representative images and (F) associated quantification of cells with p53 staining in the nucleus (G) Representative images and (H) associated quantification of SA β-gal staining (red arrows indicate positive staining). Data shown as mean +/− S.E.M (n = 3–6) * p < 0.05; *** p < 0.001 compared to control. Scale bars = 5 μm.
Ijms 24 01643 g004
Figure 5. Knockdown of p53 decreases VSMC calcification and senescence. VSMCs were transfected with either scrambled (SC) or p53 siRNA and cultured with high phosphate (3mM Pi) for up to 14 days. (A) Fold change in mRNA expression of p53 and p21. (B) Representative immunoblots and associated quantification of (C) p53 and (D) p21compared with ACTB (β-actin). (E) Representative images and (F) associated quantification of SA β-gal staining (red arrows indicate positive staining). (G) Calcium content (mg/mg protein) of cells. (H) Representative immunoblots and associated quantification of (I) Osteocalcin (OCN) and (J) tissue-nonspecific alkaline phosphatase (TNAP) compared with ACTB (β-actin). Data shown as mean +/− SEM (n = 3–6) * p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001 compared to control. Scale bars = 5 μm.
Figure 5. Knockdown of p53 decreases VSMC calcification and senescence. VSMCs were transfected with either scrambled (SC) or p53 siRNA and cultured with high phosphate (3mM Pi) for up to 14 days. (A) Fold change in mRNA expression of p53 and p21. (B) Representative immunoblots and associated quantification of (C) p53 and (D) p21compared with ACTB (β-actin). (E) Representative images and (F) associated quantification of SA β-gal staining (red arrows indicate positive staining). (G) Calcium content (mg/mg protein) of cells. (H) Representative immunoblots and associated quantification of (I) Osteocalcin (OCN) and (J) tissue-nonspecific alkaline phosphatase (TNAP) compared with ACTB (β-actin). Data shown as mean +/− SEM (n = 3–6) * p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001 compared to control. Scale bars = 5 μm.
Ijms 24 01643 g005
Figure 6. Knockdown of p53 in calcified VSMCs reduces mitochondrial length and improves mitochondrial function. VSMCs were transfected with either scrambled (SC) or p53 siRNA and cultured with high phosphate (3mM Pi) for up to 14 days. Quantification of (A) mitochondrial reactive oxygen species (ROS) (470/560 nm) and (B) mitochondrial membrane potential by mitotracker red CMXRos (mean intensity). (C) Representative confocal images of mito-QC VSMCs (white arrows show hyperfused mitochondria). Quantification of (D) mitochondrial length (µM) and (E) mitophagy index. Data shown as mean +/− S.E.M. (n = 3–6) *p < 0.05; ** p< 0.01; *** p < 0.001 compared to control. Scale bars = 10 μm.
Figure 6. Knockdown of p53 in calcified VSMCs reduces mitochondrial length and improves mitochondrial function. VSMCs were transfected with either scrambled (SC) or p53 siRNA and cultured with high phosphate (3mM Pi) for up to 14 days. Quantification of (A) mitochondrial reactive oxygen species (ROS) (470/560 nm) and (B) mitochondrial membrane potential by mitotracker red CMXRos (mean intensity). (C) Representative confocal images of mito-QC VSMCs (white arrows show hyperfused mitochondria). Quantification of (D) mitochondrial length (µM) and (E) mitophagy index. Data shown as mean +/− S.E.M. (n = 3–6) *p < 0.05; ** p< 0.01; *** p < 0.001 compared to control. Scale bars = 10 μm.
Ijms 24 01643 g006
Figure 7. Knockdown of p53 in calcified VSMCs reverses Optic Atrophy Protein 1 (OPA1) and Dynamin-related protein 1 (DRP1) protein expression. VSMCs were transfected with either scrambled (SC) or p53 siRNA and cultured with high phosphate (3mM Pi) for up to 14 days. (A) Representative immunoblotting of whole cell lysates and associated quantification of (B) OPA1 and (C) DRP1 expression compared with ACTB (β-actin). (D) Representative immunoblotting of the mitochondrial and cytoplasmic fractions with TOM20 and ACTB from scrambled (SC) and p53 SiRNA transfected VSMCs. (E) Representative immunoblotting of mitochondrial expression of DRP1 and cytoplasmic expression of DRP1 -pS637 and associated quantification of (F) ratio of cytoplasmic DRP1- pS637/mitochondrial DRP1. Data shown as mean +/− S.E.M. (n = 3–6) * p < 0.05; ** p < 0.01 compared to control.
Figure 7. Knockdown of p53 in calcified VSMCs reverses Optic Atrophy Protein 1 (OPA1) and Dynamin-related protein 1 (DRP1) protein expression. VSMCs were transfected with either scrambled (SC) or p53 siRNA and cultured with high phosphate (3mM Pi) for up to 14 days. (A) Representative immunoblotting of whole cell lysates and associated quantification of (B) OPA1 and (C) DRP1 expression compared with ACTB (β-actin). (D) Representative immunoblotting of the mitochondrial and cytoplasmic fractions with TOM20 and ACTB from scrambled (SC) and p53 SiRNA transfected VSMCs. (E) Representative immunoblotting of mitochondrial expression of DRP1 and cytoplasmic expression of DRP1 -pS637 and associated quantification of (F) ratio of cytoplasmic DRP1- pS637/mitochondrial DRP1. Data shown as mean +/− S.E.M. (n = 3–6) * p < 0.05; ** p < 0.01 compared to control.
Ijms 24 01643 g007
Figure 8. Overexpression of p53 enhances mitochondrial length, senescence and vascular calcification. VSMCs were transfected with either control or p53 vector (p53-OvEx) and cultured with high phosphate (3mM Pi) for up to 7 days. (A) Representative immunoblotting of whole cell lysates and associated quantification of (B) p53 and (C) p21 (D) Optic Atrophy Protein 1 (OPA1) expression compared with ACTB (β-actin) and (E) Dynamin-related protein 1 (DRP1). (F) Representative images of TOM20 staining (G) associated quantification of mitochondrial length. (H) Calcium content (mg/mg protein) of VSMCs. Data shown as mean +/− S.E.M. (n = 3–6) * p< 0.05; *** p < 0.001 compared to control.
Figure 8. Overexpression of p53 enhances mitochondrial length, senescence and vascular calcification. VSMCs were transfected with either control or p53 vector (p53-OvEx) and cultured with high phosphate (3mM Pi) for up to 7 days. (A) Representative immunoblotting of whole cell lysates and associated quantification of (B) p53 and (C) p21 (D) Optic Atrophy Protein 1 (OPA1) expression compared with ACTB (β-actin) and (E) Dynamin-related protein 1 (DRP1). (F) Representative images of TOM20 staining (G) associated quantification of mitochondrial length. (H) Calcium content (mg/mg protein) of VSMCs. Data shown as mean +/− S.E.M. (n = 3–6) * p< 0.05; *** p < 0.001 compared to control.
Ijms 24 01643 g008
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Phadwal, K.; Tang, Q.-Y.; Luijten, I.; Zhao, J.-F.; Corcoran, B.; Semple, R.K.; Ganley, I.G.; MacRae, V.E. p53 Regulates Mitochondrial Dynamics in Vascular Smooth Muscle Cell Calcification. Int. J. Mol. Sci. 2023, 24, 1643. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24021643

AMA Style

Phadwal K, Tang Q-Y, Luijten I, Zhao J-F, Corcoran B, Semple RK, Ganley IG, MacRae VE. p53 Regulates Mitochondrial Dynamics in Vascular Smooth Muscle Cell Calcification. International Journal of Molecular Sciences. 2023; 24(2):1643. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24021643

Chicago/Turabian Style

Phadwal, Kanchan, Qi-Yu Tang, Ineke Luijten, Jin-Feng Zhao, Brendan Corcoran, Robert K. Semple, Ian G. Ganley, and Vicky E. MacRae. 2023. "p53 Regulates Mitochondrial Dynamics in Vascular Smooth Muscle Cell Calcification" International Journal of Molecular Sciences 24, no. 2: 1643. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24021643

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop