You are on page 1of 7

REVIEW

CURRENT
OPINION Calcinosis: insights from other calcinoses
Sofia Serena Tsakali and Catherine M. Shanahan

Purpose of review
This review examines the current knowledge and recent developments in the field of vascular calcification
focusing on the emerging role of senescence and inflammation in driving this disorder and exploring the
overlap and relevance of these pathways to calcinosis in rheumatic disease.
Recent findings
Vascular calcification is an age-associated disorder. Recent studies have identified DNA damage, cellular
senescence and consequent inflammation as key drivers of vascular smooth muscle cell osteogenic change
and mineralization. Similar ageing and inflammatory factors are associated with calcinosis in rheumatic
disease and some are targets of experimental drugs currently undergoing clinical trials.
Downloaded from http://journals.lww.com/co-rheumatology by BhDMf5ePHKbH4TTImqenVLDYB5WEYn0wskXcqoz/SXyAF5R6ugCWjgQU64gm/qbGmgqSFjLEl7I= on 10/08/2020

Summary
Calcinosis in the vascular system and in rheumatic disease share similarities in terms of biomineralization
and cardiovascular outcomes. Although research into the role of senescence and inflammation has recently
been advanced in vascular calcification, little is known about the mechanistic role of inflammation in
calcinosis in rheumatic disease. This review explores whether lessons from one calcinosis can be
transferred and applied to the other to provide further insights and inform treatment strategies.
Keywords
calcinosis, inflammation, rheumatic disease, senescence, vascular calcification

INTRODUCTION (SSc), systemic lupus erythematosus (SLE) or rheu-


Calcinosis or dystrophic calcification is the abnor- matoid arthritis (RA) and other fibrosing syndromes,
mal deposition of insoluble calcium apatite in soft which are part of the CREST syndrome (calcinosis,
tissues. Vascular calcinosis is the most widespread Raynaud’s phenomenon, oesophageal dysmotility,
&

form of dystrophic calcification and is prevalent sclerodactyly and telangiectasia) [5,6,7 ]. In these
in ageing and accelerated in diabetes and chronic rheumatic diseases, calcification occurs in conjunc-
kidney disease (CKD) [1]. Calcification is an inde- tion with accelerated cardiovascular disease (CVD)
pendent risk factor for cardiovascular mortality and including atherosclerosis, pulmonary arterial hyper-
directly causal in cardiovascular events [2]. Vascular tension and elevated pulse wave velocity (PWV),
calcification can be divided into intimal or medial which can be a surrogate marker for vascular calcifi-
calcification depending on its anatomical location cation [8–11]. In these patients, calcinosis also man-
in the vessel wall. The mineral deposited varies in ifests on the surface of skin and facial areas as well as
shape and size and the type and location of mineral in pressure sites including the elbows and knees [12].
impacts on the damage inflicted on the vessel. Inti- These soft tissue mineral deposits vary in size and
mal calcification is associated with atherosclerosis shape and can form sheets that adhere to muscle,
and characterized by lipid accumulation, inflamma-
tory macrophages and fibrosis and the extent of
calcification present in coronary arteries strongly
correlates with the risk of myocardial infarction British Heart Foundation Centre for Research Excellence, School of
Cardiovascular Medicine and Sciences, James Black Centre, King’s
[3]. Medial calcification is associated with ageing,
College London, London, UK
diabetes and CKD and causes arterial stiffening,
Correspondence to Catherine M. Shanahan, British Heart Foundation
which in turn leads to hypertension, poor cardiac Centre of Research Excellence, School of Cardiovascular Medicine and
perfusion, eventually leading to cardiac remodelling Sciences, James Black Centre, King’s College London, 125Coldharbour
and heart failure [4]. Lane, London SE5 9 NU, UK. Tel: +44 2078485221;
Calcinosis of the vasculature is also regularly e-mail: cathy.shanahan@kcl.ac.uk
observed in patients who suffer from rheumatic Curr Opin Rheumatol 2020, 32:472–478
autoimmune diseases, such as systemic sclerosis DOI:10.1097/BOR.0000000000000746

www.co-rheumatology.com Volume 32  Number 6  November 2020

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.


Calcinosis: insights from other calcinoses Tsakali and Shanahan

damage and death, that together act to drive miner-


KEY POINTS alization. This includes phenotypic modulation from
 Calcinosis is prevalent in rheumatic diseases presenting a contractile cell to a more proliferative, mesenchy-
with risk factors common to vascular calcification mal-like cell, secretion of extracellular vesicles, loss of
including ageing and inflammation, however, the cause protective mineralization inhibitors and differentia-
of its pathogenesis remains unknown. tion into an osteochondrogenic phenotype to
orchestrate mineralization of the extracellular matrix
 Cellular senescence and inflammation are emerging as
mediators of vascular calcification whereas interleukin- [18]. Driving this VSMC osteochondrogenic conver-
6, in particular, is shown to be a common factor in sion is increased expression of transcription factors
both vascular and rheumatic calcinosis. that include the key bone and cartilage master reg-
ulators Runx2 and Sox9 resulting in the upregulation
 The treatment and management of calcinosis in
of their downstream targets. These targets include
rheumatic diseases remains an urgent need with current
drugs targeting and inhibiting key inflammatory mineralization regulators, such as bone sialoprotein
mediators or cellular ageing pathways potential and alkaline phosphatase as well as extracellular
new targets. matrix components, such as collagens and proteo-
glycans [2]. There is an array of environmental stress-
ors including mechanical and chemical stimuli
known to activate multiple signalling pathways
tendon and ligaments that result in significant pain that upregulate these transcription factors and
and limit motility [13,14]. impact calcification. Amongst the most potent are
Currently, the underlying mechanisms and oxidative stress driven by factors, such as hypergly-
etiopathogenesis of soft tissue calcification in these caemia and elevated calcium and phosphate concen-
rheumatic diseases remain underexplored. However, trations, which are prevalent in patients with CKD
the presence of vascular calcification and elevated on dialysis [15]. Recent studies have shown that these
cardiovascular risk means there may be mechanistic stressors can drive DNA damage and that DNA dam-
similarities with vascular calcification. age signalling can directly impact on pathways to
Ageing is the strongest risk factor for vascular osteochondrogenic differentiation, inflammation
calcification and calcification is accelerated in and calcification [19,20].
patients with CKD leading to the suggestion that
these patients manifest with premature vascular
ageing [15]. Importantly, cellular senescence is DNA damage, the senescence-associated
associated with the proinflammatory senescence- secretory phenotype and inflammation
associated secretory phenotype (SASP) whereas ath- The preservation of the human genome is of vital
erosclerosis itself is a chronic inflammatory condition importance for the healthy function of the human
[2,16]. Likewise, rheumatic conditions are inflamma- body and the conservation of life. In living organ-
tory autoimmune disorders that increase with preva- isms, the genome is constantly exposed to harmful
lence with age and calcinosis in soft tissues is also agents that induce DNA damage that, if left unre-
known to promote a strong local inflammatory paired can lead to the accumulation of senescent
response. This has led to the suggestion that sterile cells that impact health and instigate disease-
inflammation in association with tissue ageing may associated disorders [17]. In order to maintain
also be a key pathway driving dystrophic calcification genomic integrity and stability, cells respond to
&
in the CREST syndromes [7 ,17]. DNA damage by activating the DNA damage
This review will present recent evidence from response (DDR). This is a robust and highly efficient
vascular calcification that confirms these links sug- system of complex signalling pathways that act to
gesting that the accrual of persistent DNA damage repair the inflicted damage. In the case of failure
and subsequent cellular senescence-induced inflam- or inefficiency of any of these DDR pathways,
mation may be important for dystrophic calcifica- unrepairable or persistent DNA damage leads to
tion in all contexts. cellular ageing, apoptosis and/or senescence [21].
In response to strand breaks in the DNA, the first
protein to be recruited to the lesion site is the kinase
VASCULAR CALCIFICATION: PATHWAYS ataxia telangiectasia mutated (ATM), which is
TO PREMATURE AGEING responsible for the phosphorylation of gH2AX
Vascular calcification is an active process mediated histone [22]. This initial event acts to promote the
by vascular smooth muscle cells (VSMCs). These appropriate DNA repair factors and effectors
cells respond to environmental stressors and insti- depending on the type of DNA lesion. A frequent
gate a number of processes, in response to cellular form of stressor-induced damage is oxidative DNA

1040-8711 Copyright ß 2020 Wolters Kluwer Health, Inc. All rights reserved. www.co-rheumatology.com 473

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.


Raynaud phenomenon, scleroderma, overlap syndromes and other fibrosing syndromes

damage that triggers recruitment of the repair effec- reduced transcriptional and secreted levels of these
tor proteins poly[ADP-ribose] polymerases (PARPs) 1 SASP factors and reduced osteochondrogenic differ-
and 2, which conjugate the posttranslational modi- entiation and mineralization strongly suggesting a
fication poly[ADP-ribose] (PAR) to proteins in a causal link between pro-inflammatory factors and
process known as the PARylation [23]. This modifies vascular calcification [31]. Interestingly, previous
the signalling responses of target proteins to deter- studies had already identified NF-kB and Akt signal-
mine cell fate and enable cellular growth arrest in ling as promoters of VSMC osteochondrogenic dif-
order to facilitate repair, induce senescence or as a ferentiation and calcification in response to oxidative
last resolve proceed with apoptosis/cell death [24]. stress and both these pathways are also upstream of
Cells exit the cell cycle and enter senescence by SASP activation [37–40].
activating p53 and/or upregulating cyclin-depen- Inflammatory mediators, such as IL-6 and IL-8
dent kinases p21 (CDKN1A) and p16 (CDKN2A) increase in the circulation with age and are also
[25,26]. Senescent cells exhibit an inflammatory increased in patients with CKD and this has led to
phenotype and activate the SASP during which they the idea of ‘Inflammaging’ as causal in chronic age-
start to secrete a diverse selection of proteases and associated conditions, such as vascular calcification
pro-inflammatory factors, such as cytokines and [41]. However, studying ageing in elderly popula-
chemokines [27]. More importantly, the senescence tions is complicated because of the multiplicity of
cascade and subsequent inflammation is a down- factors that can potentially drive the increases in
stream event of DDR signalling and numerous stud- ageing indicators. Instead children with CKD on
ies have reported that VSMCs display DNA damage, dialysis who develop rapid vascular calcification
ageing and senescence markers at sites of calcifica- and show evidence of premature vascular ageing
&& &
tion (Fig. 1) [17,28 ,29 ,30]. make an ideal model to investigate how biological
ageing mechanisms may impact on disorder in vivo
[17,42]. Calcified vessels from children on dialysis
The DNA damage response and vascular showed an accumulation of prelamin A, the DNA
smooth muscle cell osteogenic signalling damage marker 8-oxo-dG and upregulation of senes-
&&
The first evidence that vascular calcification was cence factors p16 and p21 [28 ]. Moreover, cultured
associated with cellular ageing came from studies VSMCs from these vessels showed increased DNA
showing that the nuclear ageing biomarker prelamin damage and deficient DNA damage repair leading to
A, that is causal in the premature vascular ageing of early senescence when compared with healthy con-
patients with progeria, accumulated in calcifying trol VSMCs. In addition to the upregulation of p16
VSMCs both in vitro and in vivo, and this accumula- and p21 markers, VSMCs from children on dialysis
tion was independent of chronological age [31–33]. also showed elevated levels of pro-inflammatory
Mechanistic studies in vitro showed that prelamin A SASP factors IL-6 and IL-8, as well as the osteogenic
&&
acts to interfere with DNA damage repair driving marker BMP2 [28 ,43]. Interestingly, ATM inhibi-
persistent DNA damage and premature senescence tion impeded calcification and showed loss of SASP
[31,34]. Calcifying nodular VSMCs in vitro were also factor expression in response to calcifying media
found to accumulate prelamin A and to be positive supplemented with elevated levels of calcium and
&&
for the senescence markers senescence-associated phosphate [44 ,45]. In clinical studies with paedi-
galactosidase (SabG) and p16. Correlating with prel- atric CKD patients, circulating levels of both BMP2
amin A accumulation and senescence was increased and IL-6 positively correlated with pulse wave veloc-
osteogenic marker expression [35,36]. These obser- ity, which is indicative of vascular stiffening,
vations led to the hypothesis that DNA damage sig- and with coronary artery calcification measured
&&
nalling may be a driver of VSMC osteochondrogenic by computer tomography scanning [28 ,46]. Stud-
differentiation. Studies using inhibitors of ATM sig- ies in adult patients with CKD and calcification also
nalling showed that blocking the DDR could block indicated increased tissue indices of senescence
VSMC osteochondrogenic differentiation and calci- and increased circulating SASP factors, IL-6, IL-8
fication. Co-culture studies with mesenchymal pre- and BMP2, further suggesting that inflammation
cursor cells showed that senescent VSMCs could drive may be one of the drivers of calcification in this
&&
osteochondrogenic differentiation in a paracrine population [28 ,47–49].
manner [17]. Importantly, several of the SASP factors Further evidence for the role of DDR signalling
released by the senescent VSMCs, including BMP2 as in vascular calcification has come from studies
well as pro-inflammatory factors interleukin-6 (IL-6) examining PARP signalling, which is activated in
and interleukin-8 (IL-8), had previously been impli- response to oxidative DNA damage. Studies focusing
cated in driving VSMC osteochondrogenic differen- on animal models and human vessels showed that
&&
tiation and calcification [28 ,31]. Inhibition of ATM PAR was present at sites of calcification whereas

474 www.co-rheumatology.com Volume 32  Number 6  November 2020

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.


Calcinosis: insights from other calcinoses Tsakali and Shanahan

FIGURE 1. Persistent DNA damage promotes the senescence-associated secretory phenotype response in VSMCs. Oxidative
stress induces DNA damage that if unrepaired leads to activation of the SASP in VSMCs leading to the secretion of cytokines
and chemokines that further promote senescence and reinforce vascular osteochondrogenic change and calcification. Similar
pathways may also apply in rheumatic diseases. SASP, senescence associated secretory phenotype; VSMCs, vascular smooth
muscle cells.

1040-8711 Copyright ß 2020 Wolters Kluwer Health, Inc. All rights reserved. www.co-rheumatology.com 475

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.


Raynaud phenomenon, scleroderma, overlap syndromes and other fibrosing syndromes

PARP inhibitors impede calcification in both cellular Janus kinases and signal transducers and activators
&&
and animal models [44 ,50]. PAR is a posttransla- of transcription (JAK/STAT) pathway that mediates
tion modification normally produced in the gene transcription modulating immune, differenti-
nucleus. Chemically, it is a phosphate sugar struc- ation and growth [58,59]. IL-6-induced calcification
turally similar to mineralization regulators. Studies in mesenchymal stem cells derived from adipose
suggest that PAR, produced in response to oxidative tissues has been shown to be STAT3-dependent
stress, is released into the extracellular matrix dur- and can activate osteochondrogenic differentiation
ing the cell death and vesicle release that accom- via Runx2 modulation [60]. These studies showcase
&&
pany VSMC osteochondrogenic change [44 ,50]. that IL-6 may act as a potential link and driver of
Chemically PAR acts to gather and concentrate calcinoses in cell types in close apposition to calci-
calcium and can biomimetically mineralize both nosis in rheumatic diseases.
collagen and elastin. Thus, a by-product of the
DDR plays a direct role in the nucleation of mineral
within the extracellular matrix providing an expla- Targeting inflammation and senescence to
nation for why calcification might associate treat calcinosis
strongly with ageing as DNA damage accrues. Currently, there are no treatments for vascular cal-
cification and despite the evidence for the role of
inflammation and senescence in calcification, there
MECHANISMS OF CALCINOSIS IN have been few clinical studies examining how tar-
RHEUMATIC DISORDER geting these pathways might impact on disease.
Inflammation and senescence are also emerging as However, preclinical studies using PARP inhibitors
key features in rheumatic diseases suggesting and senolytics are suggesting that these may be
senescent VSMCs, fibroblasts or other mesenchy- effective targets for further therapy. Minocycline
mal-like cells, such as pericytes may account for at is an antibiotic that is a potent PARP2 inhibitor.
least some of the calcinosis observed in soft tissues. This drug was shown to reduce calcification in rats
A number of studies have shown that all these cell induced to calcify in response to CKD [36]. Interest-
types can undergo Runx2-mediated osteogenic dif- ingly, this drug has also been used in patients with
ferentiation in response to pathological damage cutaneous SSc and is potentially effective in the
[51–54]. control of calcinosis; however, further trials in larger
patient cohorts are now required to verify its effec-
&&
tiveness [44 ,61].
A role for inflammation and senescence in Senolytic approaches to remove senescent cells
calcinosis in rheumatic disease have been examined in preclinical animal models
Studies in vitro and in vivo have shown that cell of atherosclerosis and have been shown to extend
senescence is a feature of rheumatic diseases. Bone lifespan and delay vascular calcification [62–64].
marrow-derived mesenchymal stem cells (BM- Senolytic drugs have also been used to combat
MSCs) from patients with SSc, were positive for senescence and inflammation both in vitro and in
SabG, expressed high levels of p21 and p53 and animal models mimicking systemic autoimmune
showed increased mRNA and protein levels of IL-6 syndromes [65,66]. Fibroblast treatment with the
when cultured in vitro [55]. Similarly, synovial tissue senolytic drug fenofibrate normalized the levels of
and fibroblasts from old healthy donors or patients the SASP factors highlighting the link between pro-
with RA or osteoarthritis were positive for SabG inflammatory mediators and senescence in RA [56].
staining and also showed upregulation of p16, IL- However, whether senolytic drugs had an effect on
6 and IL-8 compared with healthy young donors. calcinosis was not addressed making this a crucial
Oxidative stress induced by H2O2 or TNFa enhanced question for further study.
senescence in these fibroblasts as well as increased Targeting inflammatory factors in vascular cal-
IL-6, IL-8, monocyte recruitment chemokine CCL2 cification has not yet been trialled. The Canakinu-
and matrix metallopeptidase 3 transcription and mab Antiinflammatory Thrombosis Outcome Study
protein expression [56]. Elevated circulating levels (CANTOS) used an antibody, canakinumab, to
of the procalcific inflammatory mediator IL-6 is also inhibit interleukin-1b to impede inflammation in
a consistent feature in rheumatic diseases with SSc patients with previous myocardial infarction and
patients also showing elevated levels in skin found reduced IL-6 levels after 12 months and
although it remains unclear whether it is in associa- reduced cardiovascular events; however, the effects
tion with calcinosis sites [49,57]. Multiple cell types on calcification were not examined [67]. The use of
including macrophages and fibroblasts secrete IL-6 specific inflammatory inhibitors has also been tri-
that binds to the IL-6 receptor and activates the alled in patients with rheumatic diseases. A Swedish

476 www.co-rheumatology.com Volume 32  Number 6  November 2020

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.


Calcinosis: insights from other calcinoses Tsakali and Shanahan

cohort of LSE patients treated with the monoclonal REFERENCES AND RECOMMENDED
antibody belimumab against the counterpart of READING
Papers of particular interest, published within the annual period of review, have
B-cell activating factor, showed reduced IL-6 serum been highlighted as:
levels during early stages, which correlated with & of special interest
&& of outstanding interest
favourable patient outcome including reduced
disease activity [68]. Tocilizumab (TCZ), an IL-6 1. Paloian NJ, Giachelli CM. A current understanding of vascular calcification in
inhibitor has been administrated to RA patients CKD. Am J Physiol Ren Physiol 2014; 307:F891–F900.
2. Durham AL, Speer MY, Scatena M, et al. Role of smooth muscle cells in
for over a decade, exhibiting evidence of efficacy vascular calcification: implications in atherosclerosis and arterial stiffness.
in the management of inflammatory symptoms Cardiovasc Res 2018; 114:590–600.
3. Pohle K, Ropers D, Mäffert R, et al. Coronary calcifications in young patients
[69]. Although the effect of drugs for RA treatment with first, unheralded myocardial infarction: a risk factor matched analysis by
on CVD risk remains unclear, a study comparing electron beam tomography. Heart 2003; 89:625–628.
4. Proudfoot D, Shanahan CM. Biology of calcification in vascular cells: intima
TCZ with other RA drugs showed that there was versus media. Herz 2001; 26:245–251.
no increased risk of CVD in these patients [70]. 5. Harigane K, Mochida Y, Ishii K, et al. Dystrophic calcinosis in a patient with
rheumatoid arthritis. Mod Rheumatol 2011; 21:85–88.
Importantly, treatment with a combination or 6. Duzgun N. Cutaneous calcinosis in a patient with limited scleroderma:
monotherapy of conventional synthetic or biolog- CREST syndrome. Eur J Rheumatol 2017; 4:305–306.
7. Hsu V, Varga J, Schlesinger N. Calcinosis in scleroderma made crystal clear.
ical disease-modifying antirheumatic drugs, such & Curr Opin Rheumatol 2019; 31:589–594.
as the immunosuppressant methotrexate or an This review summarized the current knowledge on the effects of rituximab on
calcinosis in patients with various rheumatic diseases.
antibody directed against B cells, rituximab, in 8. Cannarile F, Valentini V, Mirabelli G, et al. Cardiovascular disease in systemic
RA patients resulted in improved PWV and preven- sclerosis. Ann Transl Med 2015; 3:8.
9. Udachkina HV, Novikova DS, Popkova TV, et al. Calcification of coronary
tion of arterial stiffening progression [71]. TCZ and arteries in early rheumatoid arthritis prior to antirheumatic therapy. Rheumatol
rituximab in combination was also found to reduce Int 2018; 38:211–217.
10. Gunter S, Robinson C, Norton GR, et al. Cardiovascular risk factors and
PWV in RA patients [72]. However, there is no clear disease characteristics are consistently associated with arterial function in
data on whether any of these drugs impact on rheumatoid arthritis. J Rheumatol 2017; 44:1125–1133.
11. Paccou J, Brazier M, Mentaverri R, et al. Vascular calcification in rheumatoid
calcinosis in either the vasculature or soft tissues, arthritis: prevalence, pathophysiological aspects and potential targets. Ather-
so further trials are needed to address these specific osclerosis 2012; 224:283–290.
& 12. Valenzuela A, Chung L. Management of calcinosis associated with systemic
questions [7 ]. sclerosis. Curr Treat Options Rheumatol 2016; 2:85–96.
13. Freire V, Moser TP, Lepage-Saucier M. Radiological identification and ana-
lysis of soft tissue musculoskeletal calcifications. Insights Imaging 2018;
9:477–492.
CONCLUSION 14. Hwang ZA, Suh KJ, Chen D, et al. Imaging features of soft-tissue calcifications
and related diseases: a systematic approach. Korean J Radiol 2018;
Vascular calcification and calcinosis in systemic 19:1147–1160.
autoimmune diseases share features including pre- 15. Shroff R, Long DA, Shanahan C. Mechanistic insights into vascular calcifica-
tion in CKD. J Am Soc Nephrol 2013; 24:179–189.
mature cellular ageing, senescence and inflamma- 16. Gardner SE, Humphry M, Bennett MR, Clarke MCH. Senescent vascular
tion. Research in the field of vascular calcification is smooth muscle cells drive inflammation through an interleukin-1a-dependent
senescence-associated secretory phenotype. Arterioscler Thromb Vasc Biol
showing promising advances in better understand- 2015; 35:1963–1974.
ing the molecular mechanisms driving calcinosis, 17. Dai L, Schurgers LJ, Shiels PG, Stenvinkel P. Early vascular ageing in chronic
kidney disease: impact of inflammation, vitamin K, senescence and genomic
including the role of DDR signalling, and therefore, damage. Nephrol Dial Transplant 2020; 35(Suppl 2):ii31–ii37.
indicating novel targets for drug development. Even 18. Shanahan CM. Mechanisms of vascular calcification in CKD—evidence for
premature ageing? Nat Rev Nephrol 2013; 9:661–670.
though further investigation in calcinosis in rheu- 19. Pescatore LA, Gamarra LF, Liberman M. Multifaceted mechanisms of
matic disorders is clearly required, it is worth noting vascular calcification in aging. Arterioscler Thromb Vasc Biol 2019;
39:1307–1316.
that common inflammatory mediators, such as IL-6 20. Chung HY, Kim DH, Lee EK, et al. Redefining chronic inflammation in aging
and IL-8 could in part be driving these disorders. and age-related diseases: proposal of the senoinflammation concept. Aging
Dis 2019; 10:367–382.
Further dissection of the key procalcific inflamma- 21. Fumagalli M, d’Adda di Fagagna F. SASPense and DDRama in cancer and
tory mediators may provide the groundwork for ageing. Nat Cell Biol 2009; 11:921–923.
22. Turgeon MO, Perry NJS, Poulogiannis G. DNA damage, repair, and cancer
the development of novel treatments for these metabolism. Front Oncol 2018; 8:15.
devastating disorders. 23. Morales JC, Li L, Fattah FJ, et al. Review of poly (ADP-ribose) polymerase
(PARP) mechanisms of action and rationale for targeting in cancer and other
diseases. Crit Rev Eukaryot Gene Expr 2014; 24:15–28.
Acknowledgements 24. Schuhwerk H, Bruhn C, Siniuk K, et al. Kinetics of poly(ADP-ribosyl)ation, but
not PARP1 itself, determines the cell fate in response to DNA damage in vitro
None. and in vivo. Nucleic Acids Res 2017; 45:11174–11192.
25. Chen J, Huang X, Halicka D, et al. Contribution of p16INK4a and p21CIP1
pathways to induction of premature senescence of human endothelial cells:
Financial support and sponsorship permissive role of p53. Am J Physiol Hear Circ Physiol 2006;
290:H1575–H1586.
This work was supported by a British Heart Foundation 26. Miao SB, Xie XL, Yin YJ, et al. Accumulation of smooth muscle 22a protein
Project Grant (PG/17/37/33023). accelerates senescence of vascular smooth muscle cells via stabilization
of p53 in vitro and in vivo. Arterioscler Thromb Vasc Biol 2017;
37:1849–1859.
Conflicts of interest 27. Burton DGA, Matsubara H, Ikeda K. Pathophysiology of vascular calcification:
pivotal role of cellular senescence in vascular smooth muscle cells. Exp
There are no conflicts of interest. Gerontol 2010; 45:819–824.

1040-8711 Copyright ß 2020 Wolters Kluwer Health, Inc. All rights reserved. www.co-rheumatology.com 477

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.


Raynaud phenomenon, scleroderma, overlap syndromes and other fibrosing syndromes

28. Sanchis P, Ho CY, Liu Y, et al. Arterial ‘inflammaging’ drives vascular 49. Henaut L, Massy Z. New insights into the key role of interleukin 6 in vascular
&& calcification in children on dialysis. Kidney Int 2019; 95:958–972. calcification of chronic kidney disease. Nephrol Dial Transplant 2018;
This article highlights the link bewteen DNA damage and the pro-inflammatory 33:543–548.
SASP in vascular calcification providing direction for new strategies targeting DNA 50. Wang C, Xu W, An J, et al. Poly(ADP-ribose) polymerase 1 accelerates
damage signalling or senescence to prevent the disorder. vascular calcification by upregulating Runx2. Nat Commun 2019; 10:1203.
29. Bartoli-Leonard F, Wilkinson FL, Schiro A, et al. Loss of SIRT1 in diabetes 51. Guauque-Olarte S, Messika-Zeitoun D, Droit A, et al. Calcium signaling
& accelerates DNA damage-induced vascular calcification. Cardiovasc Res pathway genes RUNX2 and CACNA1C are associated with calcific aortic
2020; 1:1–14. valve disease. Circ Cardiovasc Genet 2015; 8:812–822.
This article provides evidence that DNA damage signalling is also prevalent in 52. Pillai ICL, Li S, Romay M, et al. Cardiac fibroblasts adopt osteogenic fates and
calcification in diabetes and implicates SIRT1 signalling as a key switch in can be targeted to attenuate pathological heart calcification. Cell Stem Cell
regulating smooth muscle cells osteogenic differentiation. 2017; 20:218.e5 –232.e5.
30. Yamada S, Tatsumoto N, Tokumoto M, et al. Phosphate binders prevent 53. Song R, Fullerton DA, Ao L, et al. Altered MicroRNA expression is responsible
phosphate-induced cellular senescence of vascular smooth muscle cells and for the pro-osteogenic phenotype of interstitial cells in calcified human aortic
vascular calcification in a modified, adenine-based uremic rat model. Calcif valves. J Am Heart Assoc 2017; 6:e005364.
Tissue Int 2015; 96:347–358. 54. Yu C, Li L, Xie F, et al. LncRNA TUG1 sponges miR-204-5p to promote
31. Liu Y, Drozdov I, Shroff R, et al. Prelamin A accelerates vascular calcification osteoblast differentiation through upregulating Runx2 in aortic valve calcifica-
via activation of the DNA damage response and senescence-associated tion. Cardiovasc Res 2018; 114:168–179.
secretory phenotype in vascular smooth muscle cells. Circ Res 2013; 55. Cipriani P, Di Benedetto P, Liakouli V, et al. Mesenchymal stem cells (MSCs)
112:e99–e109. from scleroderma patients (SSc) preserve their immunomodulatory properties
32. Hamczyk MR, del Campo L, Andrés V. Aging in the cardiovascular system: although senescent and normally induce T regulatory cells (Tregs) with a
lessons from hutchinson-gilford progeria syndrome. Annu Rev Physiol 2018; functional phenotype: implications for cellular-based therapy. Clin Exp Im-
80:27–48. munol 2013; 173:195–206.
33. Casasola A, Scalzo D, Nandakumar V, et al. Prelamin A processing, accu- 56. Del Rey MJ, Valı́n Á, Usategui A, et al. Senescent synovial fibroblasts
mulation and distribution in normal cells and laminopathy disorders. Nucleus accumulate prematurely in rheumatoid arthritis tissues and display an en-
2016; 7:84–102. hanced inflammatory phenotype. Immun Ageing 2019; 16:29.
34. Cobb AM, Larrieu D, Warren DT, et al. Prelamin A impairs 53BP1 nuclear 57. Choy EH, De Benedetti F, Takeuchi T, et al. Translating IL-6 biology into
entry by mislocalizing NUP153 and disrupting the Ran gradient. Aging Cell effective treatments. Nat Rev Rheumatol 2020; 16:335–345.
2016; 15:1039–1050. 58. Daniel E Johnson, Rachel A, Grandis JR. Targeting the IL-6/JAK/STAT3
35. Ragnauth CD, Warren DT, Liu Y, et al. Prelamin A acts to accelerate smooth signalling axis in cancer. Nat Rev Clin Oncol 2018; 15:234–248.
muscle cell senescence and is a novel biomarker of human vascular aging. 59. Fragoulis GE, Mcinnes IB, Siebert S; JAK-inhibitors. New players in the field of
Circulation 2010; 121:2200–2210. immune-mediated diseases, beyond rheumatoid arthritis. Rheumatology (Ox-
36. Duer M, Cobb AM, Shanahan CM. Arteriosclerosis, thrombosis, and vascular ford) 2019; 58:i43–i54.
biology DNA damage response. ATVB 2020; 40:1–10. 60. Fukuyo S, Yamaoka K, Sonomoto K, et al. IL-6-accelerated calcification by
37. Zhao G, Xu MJ, Zhao MM, et al. Activation of nuclear factor-kappa B induction of ROR2 in human adipose tissue-derived mesenchymal stem cells
accelerates vascular calcification by inhibiting ankylosis protein homolog is STAT3 dependent. Rheumatology (Oxford) 2014; 53:1282–1290.
expression. Kidney Int 2012; 82:34–44. 61. Robertson LP, Marshall RW, Hickling P. Treatment of cutaneous calcinosis in
38. Bent EH, Gilbert LA, Hemann MT. A senescence secretory switch mediated limited systemic sclerosis with minocycline. Ann Rheum Dis 2003;
by PI3K/AKT/mTOR activation controls chemoprotective endothelial secre- 62:267–269.
tory responses. Genes Dev 2016; 30:1811–1821. 62. Childs BG, Baker DJ, Wijshake T, et al. Senescent intimal foam cells are
39. Xie J, Lin J, Wei M, et al. Sustained Akt signaling in articular chondrocytes deleterious at all stages of atherosclerosis. Science 2016; 354:472–477.
causes osteoarthritis via oxidative stress-induced senescence in mice. Bone 63. Baker DJ, Wijshake T, Tchkonia T, et al. Clearance of p16 Ink4a-positive
Res 2019; 7:23. senescent cells delays ageing-associated disorders. Nature 2011;
40. Chien Y, Scuoppo C, Wang X, et al. Control of the senescence-associated 479:232–236.
secretory phenotype by NF-kB promotes senescence and enhances che- 64. Stojanović SD, Fiedler J, Bauersachs J, et al. Senescence-induced inflamma-
mosensitivity. Genes Dev 2011; 25:2125–2136. tion: an important player and key therapeutic target in atherosclerosis. Eur
41. Calder PC, Bosco N, Bourdet-Sicard R, et al. Health relevance of the Heart J 2020; ehz919.
modification of low grade inflammation in ageing (inflammageing) and the 65. Gao L, Slack M, McDavid A, et al. Cell senescence in lupus. Curr Rheumatol
role of nutrition. Ageing Res Rev 2017; 40:95–119. Rep 2019; 21:1.
42. Dai L, Qureshi AR, Witasp A, et al. Early vascular ageing and cellular 66. Gorgoulis V, Adams PD, Alimonti A, et al. Cellular senescence: defining a path
senescence in chronic kidney disease. Comput Struct Biotechnol J 2019; forward. Cell 2019; 179:813–827.
17:721–729. 67. Ridker PM, Everett BM, Thuren T, et al., CANTOS Trial Group. Antiinflamma-
43. Akchurin O, Patino E, Dalal V, et al. Interleukin-6 Contributes to the Devel- tory therapy with canakinumab for atherosclerotic disease. N Engl J Med
opment of Anemia in Juvenile CKD. Kidney Int Rep 2019; 4:470–483. 2017; 377:1119–1131.
44. M€ uller KH, Hayward R, Rajan R, et al. Poly(ADP-Ribose) links the DNA 68. Parodis I, Åkerström E, Sjöwall C, et al. Autoantibody and cytokine profiles
&& damage response and biomineralization. Cell Rep 2019; during treatment with belimumab in patients with systemic lupus erythema-
27:3124.e13–3138.e13. tosus. Int J Mol Sci 2020; 21:3463.
This article shows the detailed evidence implicating PARP signalling in vascular 69. Ogata A, Kato Y, Higa S, Yoshizaki K. IL-6 inhibitor for the treatment of
calcification demonstrating PAR can act to nucleate mineral and highlighting the rheumatoid arthritis: a comprehensive review. Mod Rheumatol 2019;
efficacy of the specific PARP2 inhibitor monocycline in inhibiting vascular calci- 29:258–267.
fication in vitro and in vivo. 70. Xie F, Yun H, Levitan EB, et al. Tocilizumab and the risk of cardiovascular
45. Paez-Ribes M, González-Gualda E, Doherty GJ, Muñoz-Espı́n D. Targeting disease: direct comparison among biologic disease-modifying antirheumatic
senescent cells in translational medicine. EMBO Mol Med 2019; 11:e10234. drugs for rheumatoid arthritis patients. Arthritis Care Res (Hoboken) 2019;
46. Wang Y, Osborne MT, Tung B, et al. Imaging cardiovascular calcification. J 71:1004–1018.
Am Heart Assoc 2018; 7:e008564. 71. Tam LH, Shang Q, Li EK, et al. Effect of treat-to-target strategies aiming at
47. Kamińska J, Stopiński M, Mucha K, et al. IL 6 but not TNF is linked to coronary remission of arterial stiffness in early rheumatoid arthritis: a randomized
artery calcification in patients with chronic kidney disease. Cytokine 2019; controlled study. J Rheumatol 2018; 45:1229–1239.
120:9–14. 72. Provan SA, Berg IJ, Hammer HB, et al. The impact of newer biological disease
48. Rysz J, Banach M, Cialkowska-Rysz A, et al. Blood serum levels of IL-2, IL-6, modifying antirheumatic drugs on cardiovascular risk factors: a 12-month
IL-8, TNF-alpha and IL-1beta in patients on maintenance hemodialysis. Cell longitudinal study in rheumatoid arthritis patients treated with rituximab,
Mol Immunol 2006; 3:151–154. abatacept and tociliziumab. PLoS One 2015; 10:e0130709.

478 www.co-rheumatology.com Volume 32  Number 6  November 2020

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.

You might also like