Abstract
Vascular smooth muscle cells (VSMCs) have long been associated with phenotypic modulation/plasticity or dedifferentiation. Innovative technologies in cell lineage tracing, single cell RNA-sequencing, and human genomics have been integrated to gain unprecedented insights into the molecular reprogramming of VSMCs to other cell phenotypes in experimental and clinical atherosclerosis. The current thinking is an apparently small subset of contractile VSMCs undergoes a fate switch to transitional, multi-potential cells that can adopt plaque de-stabilizing (inflammation, ossification) or plaque-stabilizing (collagen matrix deposition) cell states. Several candidate mediators of such VSMC fate and state changes are coming to light with intriguing implications for understanding coronary artery disease risk and the development of new treatment modalities. Here, we briefly summarize some technical and conceptual advancements derived from two publications in Circulation and another in Nature Medicine that, collectively, illuminate new research directions to further explore the role of VSMCs in atherosclerotic disease.
Keywords: smooth muscle cells, atherosclerosis, genetics, genomics, differentiation
The concept of vascular smooth muscle cells (VSMCs) existing in a “transitional stage between smooth muscle and foam cells” of a human atheroma was first postulated in 1961 with the aid of an electron microscope.1 This proposition gained further support with the development of molecular probes to identify cell-restricted genes or their encoded proteins in human atherosclerosis2 and in cultured VSMCs undergoing transdifferentiation from a contractile VSMC state to a lipid-laden macrophage-like state.3 The emergence of lineage tracing reporters in mouse models of atherosclerosis strengthened the view of VSMC-to-macrophage transdifferentiation4, 5 and corroborated the decades-old theory6 of clonal expansion of a few VSMC-derived cells in the evolving atheroma.4, 7, 8 VSMC lineage tracing combined with single-cell RNA-sequencing (scRNA-seq) revealed transcriptomic heterogeneity of VSMCs in atherosclerosis,9 and Cre-loxP mouse models continually demonstrate an important role for VSMC-associated genes in atherogenesis.5, 10 These studies highlight the importance of innovative technologies in establishing phenotypic modulation of VSMCs from a contractile state to one resembling other cell types. Yet, while there is no doubt as to phenotypic plasticity of VSMCs in atherosclerosis,11 controversy lingers over the precise fate of VSMCs and their phenotypic state(s) in this chronic inflammatory disease.12, 13
Two recent papers in Circulation 14, 15 provide compelling proof for the 60-year-old idea of a “transitional stage”1 between contractile VSMCs of the media and VSMC-derived cell types that contribute to the complex milieu of an atherosclerotic lesion (athero-lesion). Armed with modern innovations in VSMC lineage tracing, scRNA-seq of mouse and human atherosclerotic vessels, and human genomics, both studies demonstrate a multi-potential fate of dedifferentiated VSMCs to cell types postulated to either stabilize or destabilize the evolving athero-lesion. The findings cement the importance of VSMC phenotypic modulation in experimental and clinical atherosclerosis and provide a trove of new research questions to explore. Here, we highlight the major findings of these reports and the integrative technologies used to conclude the presence of a multipotent VSMC-derived cell state in experimental and clinical atherosclerosis.
VSMC-Derived Transitional Cells in Atherosclerosis
Alencar et al14 utilized Apoe null mice carrying Myh11-CreERT2 with an inducible, fluorescently-tagged reporter that, upon tamoxifen (Tmx) administration, indelibly marks VSMCs in their native medial compartment of the vessel wall. Following a standard atherogenic diet, micro-dissected athero-lesions of the brachiocephalic artery (BCA), considered by some to be a surrogate for human coronary plaques,16 underwent cell sorting and scRNA-seq. The results disclosed seven different clusters of cells comprising previously contractile VSMCs. Further analysis of the top 100 genes in each cluster uncovered distinct molecular signatures, including inflammatory, extracellular matrix producing, and osteogenic states. Next, the authors repeated the same study in VSMC-restricted Krüppel-like factor 4 (Klf4) null mice and found a significant reduction in Galectin 3 (Lgals3) positive cells giving rise to cells of an osteogenic state and increases in VSMC-tagged cells exhibiting elevated expression of VSMC contractile markers, possibly plaque-stabilizing cells similar to the “fibromyocyte” of Wirka et al.17 These results are consistent with previous work showing reduced Lgals3+ VSMCs and lesion burden in VSMC Klf4 knockout mice 5 suggesting KLF4 acts as a molecular fate-switch, promoting the dedifferentiation of contractile VSMCs. KLF4 represses Myocardin,18, 19 “a component of a molecular switch for smooth muscle differentiation,”20 thereby affording a transcriptional mechanism for the fate switching of contractile VSMCs to a dedifferentiated transitional state.
Alencar et al14 also made use of a novel dual reporter mouse for unambiguous tracking of cell states emanating from dedifferentiated VSMCs in athero-lesions. The results of these elegant studies showed >60% of VSMCs in athero-lesions activate Lgals3 transcription. These so-called “pioneer cells” generate inflammatory, extracellular matrix, and osteogenic cell states in the athero-lesion. However, contrary to earlier findings,5 few formerly contractile VSMCs went on to a macrophage state despite the presence of Lgals3, typically thought of as a marker for macrophages. Thus, under an atherogenic regimen that promotes endoplasmic reticulum stress and activates unfolded protein response pathways,21 contractile VSMCs dedifferentiate to a transitional pioneer cell, fated for non-macrophage cellular states (Figure, left). Importantly, scRNA-seq of human carotid endarterectomy specimens revealed similar clusters of VSMC-derived pioneer cells that may give rise to an osteogenic (plaque destabilizing) cell state.14
Figure.
Diverse VSMC-derived cell states in atherosclerosis. The LGALS3+ transitional cell of the intima is derived from dedifferentiated VSMC, perhaps in a deterministic manner. These cells represent pioneer cells of Alencar et al14 or Stem/Endothelial/Macrophage (SEM) cells of Pan et al15 and likely are precursors to plaque-stabilizing fibromyocytes of Wirka et al.17 Depending on micro-environmental signaling cues (e.g., retinoid signaling) and expression level of key transcription factors (e.g., KLF4), transitional cells may differentiate into a number of cells types that lead to fibrous cap formation (e.g., ACTA2+/PHACTR1+ cells) and plaque stability or expansion of the athero-core (osteogenic and inflammatory cells) leading to plaque instability (red zone). Unresolved questions are indicated. See text for more details.
Meanwhile, Pan et al15 discovered a similar transitional cell in a temporal scRNA-seq analysis of lineage-marked VSMCs of atherosclerotic aorta in low density lipoprotein receptor (Ldlr) null mice. This transitional cell state increased progressively over the course of the atherogenic diet, giving rise to three macrophage-like cell clusters and one fibrochondrocyte cluster (Figure, right). Similar to Alencar et al,14 each of these clusters showed attenuated expression of VSMC contractile genes indicating a dedifferentiated VSMC state. Because a previous report, using the Apoe null model of atherosclerosis and a different lineage reporter, failed to show evidence for a macrophage-like state in lineage-tagged VSMCs of athero-lesions,17 Pan et al15 repeated their temporal study in the Apoe null background; the results revealed similar clusters of de-differentiated VSMCs exhibiting a macrophage-like state. The transitional cell state of Pan et al15 displayed expression of markers for stem cell (Ly6a), endothelial cell (Vcam), and monocyte/macrophage (Ly6c1) and so they named these cells SEM cells. The SEM cells were, like the pioneer cells of Alencar et al,14 Lgals3+ and multipotent (Figure, right).15 Consistent with results of Alencar et al,14 scRNA-seq data from human carotid and coronary artery lesions yielded a transcriptionally related SEM cell type indicating conservation of this transitional state independent of embryological origin of VSMCs.15
While the findings of Pan et al15 suggesting a VSMC-derived macrophage cell state align with previous in vivo lineage tracing studies,4, 5, 8, 22, 23 they differ from those of both Alencar et al14 and Wirka et al17 who showed little evidence of VSMC-derived macrophages in athero-lesions. Aside from variations in lineage reporter, dosing of Tmx, and vascular tissues analyzed (Table), there are caveats that must be considered such as the need for rigorous analysis of multiple markers of the macrophage lineage and combined lipid staining with lineage marker or cell state marker.17 Although VSMC-derived foam cell formation occurs in experimental models17, 24 and, as originally proposed by Geer et al,1 in human atherosclerosis,22 the molecular state of these cells remains controversial (Figure). Nevertheless, both studies in Circulation 14, 15 and the report of Wirka et al,17 provide incontrovertible molecular proof for complex VSMC transitions to plaque cell types in both experimental and human atherosclerosis (Figure). The relative importance of each VSMC-derived cellular state in the pathogenesis of atherosclerosis demands further probing given the fact that a number of patients with significant coronary artery disease (CAD) and its sequelae exhibit normal lipid profiles.25
Table.
Comparative methods and results of recent studies analyzing VSMC fate and state in atherosclerosis
Methods | Alencar et al14 | Pan et al15 | Wirka et al17 |
---|---|---|---|
Athero-lesion model | 10 or 18 week HFDa in Apoe−/− mice | 8, 16, 26 week HFD in Ldlr−/− and Apoe−/− mice | 8 and 16 week HFD in Apoe−/− mice |
Lineage reporter(s)b | SF-eYFP and SR-F-tdTomato-S-GFP | SF-ZsGreen1 | SF-tdTomato |
Cre driver mousec |
Myh11-CreERT2 Myh11-Cre(Dre)ERT2 Lgals3-SR-IRES-Cre |
Myh11-CreERT2 | Myh11-CreERT2 |
Tamoxifen schedule | 0.1 mg x 10 days, IP | 2 day diet | 0.2mg/g x 2 days, PO |
Mouse tissue for scRNA-seqd | Micro-dissected lesions of BCA | Whole BCA and ascending-thoracic aorta | Whole aortic root and ascending arch |
Human scRNA-seqd | Carotid endarterectomy | Carotid/coronary | Coronary |
scRNA-seq clustering | UMAP, 14 clusters | UMAP, 12 clusters | tSNE, 12 clusters |
De-differentiated VSMC | Pioneer cell | SEM cell | Modulated SMC |
Intimal VSMC-derived cell states | Chondro-osteogenic; ECM-rich (FM?); inflammatory | Monocyte/macrophage; fibrochondrocyte (FM?) | Fibromyocyte (FM) |
Protein analysis | CyTOF/IFM | metaVIPER/IFM | CITE-seq/IFM |
ChIP-seq | KLF4/OCT4 | na | TCF21 |
CAD GWAS | 88 KLF4/OCT4 TFBS near 64/163 CAD GWAS | 226 RA targets near 37 CAD GWAS | Neg TCF21 exp with 36 CAD SNPs |
VSMC KO | Oct4, Klf4 | na | Tcf21 |
Abbreviations: HFD, high fat diet; SF, stop flox; SR, stop rox; F, flox; S, stop; LCA, left carotid artery; BCA, brachiocephalic artery; UMAP, uniform manifold approximation and projection; tSNE, t-distributed stochastic neighbor embedding; SEM, stem cell, endothelial cell, monocyte/macrophage; CyTOF, cytometry by time of flight; VIPER, virtual inference of protein activity by enriched regulon; IFM, immunofluorescence microscopy; CITE-seq, cellular indexing of transcriptomes and epitopes by sequencing; ChIP-seq, chromatin immunoprecipitation sequencing; CAD, coronary artery disease; GWAS, genome wide association study; TFBS, transcription factor binding sites; RA, retinoic acid; Neg, negative; exp, expression; SNPs, single nucleotide polymorphisms; VSMC, vascular smooth muscle cell; KO, knockout; IP, intraperitoneal; PO, per os (oral gavage); na, not applicable.
Lineage reporters were activated by Myh11-CreERT2, except for the dual lineage reporter of Alencar et al; this reporter (abbreviated here as SR-F-tdTomato-S-GFP) was sequentially activated by a
Myh11-DreERT2 and Lgals3-SR-IRES-Cre.
Embryological origin of each mouse and human vascular tissue for scRNA-seq analysis is neural crest (ascending aorta, arch, LCA, and BCA); secondary heart field (aortic root); splanchnic mesoderm (thoracic aorta), and proepicardial organ (coronary).
Distinct Regulatory Mechanisms Govern the Fate of VSMC-Derived Transitional Cells in Atherosclerosis
Alencar et al14 performed bulk RNA-seq and ChIP-seq for KLF4 and OCT4 binding events in BCA lesions from high-fat diet fed Apoe null mice with Klf4 or Oct4 knocked out in VSMCs. Putative KLF4 target genes, reduced in the background of Klf4 null VSMCs, were elevated in Oct4 null VSMCs (e.g., inflammatory genes) whereas KLF4 targets elevated in Klf4 null VSMCs showed attenuated expression in Oct4 null VSMCs (e.g., myogenic genes). These opposing programs of gene expression are consistent with lesion pathology in the context of Klf4 null VSMCs (reduced athero-lesion) versus Oct4 null VSMCs (increased athero-lesion).5, 26 Remarkably, 88 of the KLF4 and OCT4 targets are the closest annotated gene in nearly 40% of genome wide association study (GWAS) loci implicated in CAD. As the ChIP-seq experiments were done in mice, it is unclear whether similarly positioned KLF4 and OCT4 binding sites exist in human genomes and if binding has any consequence for altered expression of CAD GWAS targets. Notably, the KLF4 gene is itself a CAD GWAS risk allele with the putative variant (rs944172) located ~265 kb upstream of the KLF4 transcription start site. The nature of this variant is unclear; however, it overlaps a weak AP1 binding site with some evidence for JUND binding (UCSC Genome Browser data). The recent Nobel Prize winning technology related to clustered regularly interspaced short palindromic repeats (CRISPR) genome editing27 will enable functional definition of KLF4 and OCT 4 binding sites in proximity to CAD risk alleles.
Pan et al15 employed a bioinformatics tool to gain insight into potential “master regulators” of the transitional cell. Meta Virtual Inference of Protein activity by Enriched Regulon (metaVIPER)28 uses differential gene expression between two conditions (VSMC and SEM states) to infer potential protein activities that explain differential transcript profiles. metaVIPER disclosed cellular retinoic acid binding protein 2 as a putative master regulator of the VSMC-SEM transition. Retinoids, such as all-trans retinoic acid (atRA), impact VSMC phenotype by inhibiting VSMC growth, migration, and lesion burden in experimental models of vascular disease, presumably through nuclear receptor-mediated effects on VSMC gene expression.29 Interestingly, one such nuclear receptor, RARB, was reduced in human athero-lesions and atRA inhibited the VSMC-SEM transition in vitro and in vivo (Figure, right).15 Moreover, atRA reduced intimal area in experimental athero-lesions and elevated the proportion of VSMC-tagged cells destined for the fibrous cap. Pan et al15 then turned to integrative genomics and found a number of down-regulated retinoid-response genes in unstable human athero-lesions. Of notable interest was the finding that some retinoid-responsive genes co-localized with CAD-associated risk variants. However, as with Alencar et al14, these associations will require follow up studies, including formal demonstration of retinoid receptor binding sites around CAD risk alleles and variant modeling/correcting using CRISPR or prime editing30 in human induced pluripotent stem cells. In addition, conserved regulatory variants in CAD GWAS risk alleles or retinoid receptor binding sites in mouse models of atherosclerosis could be genome edited as shown for other transcription factor binding sites.31, 32
In the study of Wirka et al, VSMC-restricted knockout of transcription factor 21 (Tcf21) attenuated VSMC phenotypic modulation and reduced the number of fibromyocytes at the fibrous cap.17 Consistent with the mouse data, expression of human TCF21, another CAD GWAS risk allele, correlated with VSMC phenotypic modulation in human athero-lesions as well as reduced CAD risk.17 Interestingly, the lead CAD GWAS variant (rs12190287) disrupts a microRNA binding site in the 3’ untranslated region of TCF21.33
Questions and Future Directions
The two recent studies in Circulation,14, 15 and an earlier one in Nature Medicine,17 integrated state-of-the-art tools in genetics and genomics to formally demonstrate VSMC fate switching in experimental and clinical atherosclerosis. All three studies, while subtly different in design and conclusion (Figure and Table), support a transitional cell type derived from previously contractile VSMCs and possessing a unique molecular signature. An additional study found a VSMC-derived transitional mesenchymal stem cell population in a model of human aneurysm formation.23 Together, these exciting findings engender new questions.
First, are a limited number of VSMCs pre-determined to migrate from the tunica media and fate switch to the pioneer or SEM cell state in the evolving intima or is the process stochastic? A recent Circulation paper suggests the former, as higher expressing NOTCH3 VSMCs appear to leave the media and clonally expand in pulmonary intimal lesions.34 Further characterization of the transcriptome, epigenome, and proteome of medial VSMCs will assist in distinguishing between a deterministic or stochastic basis for the migration and expansion of VSMC-derived transitional cells in vascular disease.7, 35-37 It is perhaps ironic that with all the controversy over whether or not stem cells reside in the medial layer of conduit arteries,38-40 the most abundant cell type in the vessel wall (VSMC) possesses the inherent potential to acquire so many different cell fates in vivo.4, 5, 14, 15, 17, 24, 41
Second, do pioneer/SEM cells clonally expand in athero-lesions? Although no formal proliferation assays were conducted in any of the three studies,14, 15, 17 previous lineage tracing experiments 7, 42 and a stem cell antigen 1 (Sca1) signature of pioneer/SEM cells 14, 15 would support their clonal expansion. In this context, a recent report concluded little contribution of baseline vascular Sca1 positive cells in the formation of athero-lesions.43 However, it remains to be formally shown whether the acquisition of a Sca1 positive state, as in VSMC-derived transitional cells,14, 15, 17 triggers their expansion and subsequent differentiation to other intimal cell types. A confetti reporter mouse study supports clonal expansion of VSMC-derived mesenchymal stem cells in a model of aneurysm formation.23 Perhaps the pioneer/SEM state represents re-expression of an earlier transitional state passed through during development of VSMCs in the embryo. It will be important to determine whether human VSMCs express genes with functions similar to Sca1 and if such factors drive the clonal expansion of human VSMCs.
Third, what is the complete molecular signature of VSMC-derived cell types in athero-lesions, especially with respect to long noncoding RNAs which, increasingly, play a role in atherogenesis?44 Addressing this question will be challenging given the constraints with current scRNA-seq library construction (polyadenylated RNA only) and the low abundance of most long noncoding RNAs. However, we can safely anticipate advances in next generation sequencing that will increase the depth of unbiased sequencing at the individual cell level.
Fourth, what are the signaling events in the local micro-environment that promote one SEM/pioneer cell-derived state over another? Put another way, what are the underlying mechanisms for Klf414 and Tcf2117 transcription or altered retinoid signaling15 under vascular pathological conditions? Insight into this potential "Achilles heel" could spawn the development of therapeutics to mitigate the number of plaque-destabilizing cells or enhance the number of plaque-stabilizing cells. In this context, conditions of reduced TGFβ signaling and elevated cholesterol in the vessel wall resulted in massive Klf4 induction, suggesting these micro-environmental perturbations are permissive for VSMC dedifferentiation and the emergence of a stem cell fate.23
Finally, are previously contractile VSMCs licensed to engulf lipid and release inflammatory mediators with a monocyte/macrophage-like phenotype in athero-lesions? Despite decades of research, this question remains to be firmly established.
From a clinical standpoint, one can imagine harnessing advances in imaging and nanotechnology to probe lesions for the presence and possible ablation of plaque destabilizing cells. Alternatively, it may be feasible to target the transitional cells directly to bias their fate towards plaque stabilizing cell types such as the fibromyocyte. If advances in biomedical innovations over the last 60 years are any indication, the next 60 years will surely unfold with unanticipated discoveries for precision-guided control of VSMC fate switching and cellular states in atherosclerosis.
Acknowledgments
Funding Sources: Research is supported by NIH grants HL147476 (to JMM); HL084312 (to EAF); and HL121877 and the Loie Power Robinson Stem Cell and Regenerative Medicine Fund (to MWM).
Non-standard Abbreviations and Acronyms
- atRA
all-trans retinoic acid
- BCA
Brachiocephalic artery
- CAD
Coronary artery disease
- ChIP-seq
Chromatin immunoprecipitation sequencing
- CRISPR
Clustered regularly interspaced short palindromic repeats
- GWAS
Genome wide association study
- KLF4
Krüppel like factor 4
- OCT4
Octamer-binding transcription factor 4
- Sca1
Stem cell antigen 1
- scRNA-seq
Single cell RNA-sequencing
- SEM
Stem cell, endothelial cell, monocyte/macrophage
- TCF21
Transcription factor 21
- Tmx
Tamoxifen
- t-SNE
t-distributed stochastic neighbor embedding
- UMAP
Uniform manifold approximation and projection
- VIPER
Virtual inference of protein activity by enriched regulon
- VSMC(s)
Vascular smooth muscle cell(s)
Footnotes
Disclosure Statement: None.
References
- 1.Geer JC, McGill H Jr. and Strong JP. The fine structure of human atherosclerotic lesions. American Journal of Pathology. 1961;38:263–287. [PMC free article] [PubMed] [Google Scholar]
- 2.Aqel NM, Ball RY, Waldmann H and Mitchinson MJ. Identification of macrophages and smooth muscle cells in human atherosclerosis using monoclonal antibodies. J Pathol. 1985;146:197–204. [DOI] [PubMed] [Google Scholar]
- 3.Rong JX, Shapiro M, Trogan E and Fisher EA. Transdifferentiation of mouse aortic smooth muscle cells to a macrophage-like state after cholesterol loading. Proceedings of the National Academy of Sciences of the United States of America. 2003;100:13531–13536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Feil S, Fehrenbacher B, Lukowski R, Essmann F, Schulze-Osthoff K, Schaller M and Feil R. Transdifferentiation of vascular smooth muscle cells to macrophage-like cells during atherogenesis. Circ Res. 2014;115:662–7. [DOI] [PubMed] [Google Scholar]
- 5.Shankman LS, Gomez D, Cherepanova OA, Salmon M, Alencar GF, Haskins RM, Swiatlowska P, Newman AA, Greene ES, Straub AC, Isakson B, Randolph GJ and Owens GK. KLF4-dependent phenotypic modulation of smooth muscle cells has a key role in atherosclerotic plaque pathogenesis. Nat Med. 2015;21:628–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Benditt EP and Benditt JM. Evidence for a monoclonal origin of human atherosclerotic plaques. Proceedings of the National Academy of Sciences of the United States of America. 1973;70:1753–1756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Chappell J, Harman JL, Narasimhan VM, Yu H, Foote K, Simons BD, Bennett MR and Jorgensen HF. Extensive proliferation of a subset of differentiated, yet plastic, medial vascular smooth muscle cells contributes to neointimal formation in mouse injury and atherosclerosis models. Circ Res. 2016;119:1313–1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Misra A, Feng Z, Chandran RR, Kabir I, Rotllan N, Aryal B, Sheikh AQ, Ding L, Qin L, Fernandez-Hernando C, Tellides G and Greif DM. Integrin beta3 regulates clonality and fate of smooth muscle-derived atherosclerotic plaque cells. Nat Commun. 2018;9:2073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Dobnikar L, Taylor AL, Chappell J, Oldach P, Harman JL, Oerton E, Dzierzak E, Bennett MR, Spivakov M and Jorgensen HF. Disease-relevant transcriptional signatures identified in individual smooth muscle cells from healthy mouse vessels. Nat Commun. 2018;9:4567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Li D, Chen A, Lan T, Zou Y, Zhao L, Yang P, Qu H, Wei L, Varghese Z, Moorhead JF, Chen Y and Ruan XZ. SCAP knockdown in vascular smooth muscle cells alleviates atherosclerosis plaque formation via up-regulating autophagy in ApoE(−/−) mice. FASEB J. 2019;33:3437–3450. [DOI] [PubMed] [Google Scholar]
- 11.Bennett MR, Sinha S and Owens GK. Vascular smooth muscle cells in atherosclerosis. Circ Res. 2016;118:692–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Owsiany KM, Alencar GF and Owens GK. Revealing the origins of foam cells in atherosclerotic lesions. Arterioscler Thromb Vasc Biol. 2019;39:836–838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Zernecke A, Winkels H, Cochain C, Williams JW, Wolf D, Soehnlein O, Robbins CS, Monaco C, Park I, McNamara CA, Binder CJ, Cybulsky MI, Scipione CA, Hedrick CC, Galkina EV, Kyaw T, Ghosheh Y, Dinh HQ and Ley K. Meta-analysis of leukocyte diversity in atherosclerotic mouse aortas. Circ Res. 2020;127:402–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Alencar GF, Owsiany KM, K S, Sukhavasi K, Mocci G, Nguyen A, Williams CM, Shamsuzzaman S, Mokry M, Henderson CA, Haskins R, Baylis RA, Finn AV, McNamara CA, Zunder ER, Venkata V, Pasterkamp G, Bjorkegren J, Bekiranov S and Owens GK. The stem cell pluripotency genes Klf4 and Oct4 regulate complex SMC phenotypic changes critical in late-stage atherosclerotic lesion pathogenesis. Circulation. 2020;142:2045–2059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Pan H, Xue C, Auerbach BJ, Fan J, Bashore AC, Cui J, Yang DY, Trignano SB, Liu W, Shi J, Ihuegbu CO, Bush EC, Worley J, Vlahos L, Laise P, Solomon RA, Connolly ES, Califano A, Sims PA, Zhang H, Li M and Reilly MP. Single-cell genomics reveals a novel cell state during smooth muscle cell phenotypic switching and potential therapeutic targets for atherosclerosis in mouse and human. Circulation. 2020;142:2060–2075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bond AR and Jackson CL. The fat-fed apolipoprotein E knockout mouse brachiocephalic artery in the study of atherosclerotic plaque rupture. J Biomed Biotechnol. 2011;2011:379069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wirka RC, Wagh D, Paik DT, Pjanic M, Nguyen T, Miller CL, Kundu R, Nagao M, Coller J, Koyano TK, Fong R, Woo YJ, Liu B, Montgomery SB, Wu JC, Zhu K, Chang R, Alamprese M, Tallquist MD, Kim JB and Quertermous T. Atheroprotective roles of smooth muscle cell phenotypic modulation and the TCF21 disease gene as revealed by single-cell analysis. Nat Med. 2019;25:1280–1289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Liu Y, Sinha S, McDonald OG, Shang Y, Hoofnagle MH and Owens GK. Kruppel-like factor 4 abrogates myocardin-induced activation of smooth muscle gene expression. Journal of Biological Chemistry. 2005;280:9719–9727. [DOI] [PubMed] [Google Scholar]
- 19.Turner EC, Huang CL, Govindarajan K and Caplice NM. Identification of a Klf4-dependent upstream repressor region mediating transcriptional regulation of the myocardin gene in human smooth muscle cells. Biochimica et biophysica acta. 2013;1829:1191–201. [DOI] [PubMed] [Google Scholar]
- 20.Chen J, Kitchen CM, Streb JW and Miano JM. Myocardin: a component of a molecular switch for smooth muscle differentiation. J Mol Cell Cardiol. 2002;34:1345–56. [DOI] [PubMed] [Google Scholar]
- 21.Chattopadhyay A, Kwartler CS, Kaw K, Li Y, Kaw A, Chen J, LeMaire SA, Shen YH and Milewicz DM. Cholesterol-Induced phenotypic modulation of smooth muscle cells to macrophage/fibroblast-like cells is driven by an unfolded protein response. Arterioscler Thromb Vasc Biol. 2021;41:302–316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Allahverdian S, Chehroudi AC, McManus BM, Abraham T and Francis GA. Contribution of intimal smooth muscle cells to cholesterol accumulation and macrophage-like cells in human atherosclerosis. Circulation. 2014;129:1551–1559. [DOI] [PubMed] [Google Scholar]
- 23.Chen PY, Qin L, Li G, Malagon-Lopez J, Wang Z, Bergaya S, Gujja S, Caulk AW, Murtada SI, Zhang X, Zhuang ZW, Rao DA, Wang G, Tobiasova Z, Jiang B, Montgomery RR, Sun L, Sun H, Fisher EA, Gulcher JR, Fernandez-Hernando C, Humphrey JD, Tellides G, Chittenden TW and Simons M. Smooth muscle cell reprogramming in aortic aneurysms. Cell Stem Cell. 2020;26:542–557 e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wang Y, Dubland JA, Allahverdian S, Asonye E, Sahin B, Jaw JE, Sin DD, Seidman MA, Leeper NJ and Francis GA. Smooth muscle cells contribute the majority of foam cells in ApoE (Apolipoprotein E)-deficient mouse atherosclerosis. Arterioscler Thromb Vasc Biol. 2019;39:876–887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Michos ED, McEvoy JW and Blumenthal RS. Lipid management for the prevention of atherosclerotic cardiovascular disease. N Engl J Med. 2019;381:1557–1567. [DOI] [PubMed] [Google Scholar]
- 26.Cherepanova OA, Gomez D, Shankman LS, Swiatlowska P, Williams J, Sarmento OF, Alencar GF, Hess DL, Bevard MH, Greene ES, Murgai M, Turner SD, Geng YJ, Bekiranov S, Connelly JJ, Tomilin A and Owens GK. Activation of the pluripotency factor OCT4 in smooth muscle cells is atheroprotective. Nat Med. 2016;22:657–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA and Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337:816–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ding H, Douglass EF Jr., Sonabend AM, Mela A, Bose S, Gonzalez C, Canoll PD, Sims PA, Alvarez MJ and Califano A. Quantitative assessment of protein activity in orphan tissues and single cells using the metaVIPER algorithm. Nat Commun. 2018;9:1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Miano JM and Berk BC. Retinoids: versatile biological response modifiers of vascular smooth muscle phenotype. Circ Res. 2000;87:355–62. [DOI] [PubMed] [Google Scholar]
- 30.Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, Chen PJ, Wilson C, Newby GA, Raguram A and Liu DR. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature. 2019;576:149–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Choi M, Lu YW, Zhao J, Wu M, Zhang W and Long X. Transcriptional control of a novel long noncoding RNA Mymsl in smooth muscle cells by a single Cis-element and its initial functional characterization in vessels. J Mol Cell Cardiol. 2020;138:147–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Gao P, Lyu Q, Ghanam AR, Lazzarotto CR, Newby GA, Zhang W, Choi M, Slivano OJ, Holden K, Walker JA 2nd, Kadina AP, Munroe RJ, Abratte CM, Schimenti JC, Liu DR, Tsai SQ, Long X and Miano JM. Prime editing in mice reveals the essentiality of a single base in driving tissue-specific gene expression. Genome Biol. 2021;22:83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Miller CL, Haas U, Diaz R, Leeper NJ, Kundu RK, Patlolla B, Assimes TL, Kaiser FJ, Perisic L, Hedin U, Maegdefessel L, Schunkert H, Erdmann J, Quertermous T and Sczakiel G. Coronary heart disease-associated variation in TCF21 disrupts a miR-224 binding site and miRNA-mediated regulation. PLoS Genet. 2014;10:e1004263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Steffes LC, Froistad AA, Andruska A, Boehm M, McGlynn M, Zhang F, Zhang W, Hou D, Tian X, Miquerol L, Nadeau K, Metzger RJ, Spiekerkoetter E and Kumar ME. A Notch3-marked subpopulation of vascular smooth muscle cells is the cell of origin for occlusive pulmonary vascular lesions. Circulation. 2020;142:1545–1561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Herring BP, Hoggatt AM, Burlak C and Offermanns S. Previously differentiated medial vascular smooth muscle cells contribute to neointima formation following vascular injury. Vascular Cell. 2014;6:21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Zhao G, Lu H, Chang Z, Zhao Y, Zhu T, Chang L, Guo Y, Garcia-Barrio MT, Chen YE and Zhang J. Single cell RNA sequencing reveals the cellular heterogeneity of aneurysmal infrarenal abdominal aorta. Cardiovasc Res. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Wu W, Wang C, Zang H, Qi L, Azhar M, Nagarkatti M, Nagarkatti P, Cai G, Weiser-Evans MCM and Cui T. Mature vascular smooth muscle cells, but not endothelial cells, serve as the major cellular source of intimal hyperplasia in vein grafts. Arterioscler Thromb Vasc Biol. 2020;40:1870–1890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Tang Z, Wang A, Yuan F, Yan Z, Liu B, Chu JS, Helms JA and Li S. Differentiation of multipotent vascular stem cells contributes to vascular diseases. Nature Communications. 2012;3:875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Nguyen AT, Gomez D, Bell RD, Campbell JH, Clowes AW, Gabbiani G, Giachelli CM, Parmacek MS, Raines EW, Rusch NJ, Speer MY, Sturek M, Thyberg J, Towler DA, Weiser-Evans MC, Yan C, Miano JM and Owens GK. Smooth muscle cell plasticity: fact or fiction? Circulation Research. 2013;112:17–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Tang Z, Wang A, Wang D and Li S. Smooth muscle cells: to be or not to be? Response to Nguyen et Al. Circ Res. 2013;112:23–6. [DOI] [PubMed] [Google Scholar]
- 41.Majesky MW, Horita H, Ostriker A, Lu S, Regan JN, Bagchi A, Dong XR, Poczobutt J, Nemenoff RA and Weiser-Evans MC. Differentiated smooth muscle cells generate a subpopulation of resident vascular progenitor cells in the adventitia regulated by Klf4. Circ Res. 2017;120:296–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Jacobsen K, Lund MB, Shim J, Gunnersen S, Fuchtbauer EM, Kjolby M, Carramolino L and Bentzon JF. Diverse cellular architecture of atherosclerotic plaque derives from clonal expansion of a few medial SMCs. JCI Insight. 2017;2:e95890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Wang H, Zhao H, Zhu H, Li Y, Tang J, Li Y and Zhou B. Sca1(+) cells minimally contribute to smooth muscle cells in atherosclerosis. Circ Res. 2021;128:133–135. [DOI] [PubMed] [Google Scholar]
- 44.Josefs T and Boon RA. The long non-coding road to atherosclerosis. Curr Atheroscler Rep. 2020;22:55. [DOI] [PMC free article] [PubMed] [Google Scholar]