The cellular uncertainty in thoracic aortic dissections: the roles of METTL3 and NOTCH1 on m6A in human aortic smooth muscle cells
Editorial

The cellular uncertainty in thoracic aortic dissections: the roles of METTL3 and NOTCH1 on m6A in human aortic smooth muscle cells

Kelly Wright1^, Maryam Aboughdir2^, Amer Harky3^

1Department of Medicine, University of Liverpool, Liverpool, UK; 2Department of Medicine, St. George’s University of London, London, UK; 3Department of Cardiothoracic Surgery, Liverpool Heart and Chest Hospital, Liverpool, UK

^ORCID: Kelly Wright, 0000-0003-4087-6253; Maryam Aboughdir, 0000-0002-5050-6793; Amer Harky, 0000-0001-5507-5841.

Correspondence to: Amer Harky, MRCS, MSc. Department of Cardiothoracic Surgery, Liverpool Heart and Chest Hospital, Liverpool, UK. Email: aaharky@gmail.com.

Comment on: Yang J, Fang M, Yu C, et al. Human aortic smooth muscle cell regulation by METTL3 via upregulation of m6A NOTCH1 modification and inhibition of NOTCH1. Ann Transl Med 2023. doi: 10.21037/atm-22-1203.


Keywords: Thoracic aortic dissections (TADs); methyltransferase-like-3 (METTL3); NOTCH1; aorta; smooth muscle


Submitted Jan 25, 2023. Accepted for publication Feb 01, 2023. Published online Feb 09, 2023.

doi: 10.21037/atm-23-375


Thoracic aortic dissection (TAD) is a potentially life-threatening pathology with a high mortality rate, which warrants continual development for effective management approaches. TAD is defined by the progressive separation of the thoracic aortic wall layers (1,2). Since the anatomical extent of TAD has different clinical implications and treatment approaches, TAD is classified according to the areas involved into Stanford type A dissections which involves the ascending aorta up to the origin of left subclavian artery, whereas Stanford type B dissections starts from beyond left subclavian artery (3). Stanford type A dissections are more common and dangerous, accounting for approximately two-thirds of TADs (1,4).

Despite extensive investigation pertaining to TAD pathophysiology, the cellular and molecular mechanisms of TAD are inadequately understood. Human aortic smooth muscle cells (HASMCs) have contractile and synthetic phenotypes, both of which are characterised by the expression of different marker proteins. The contractile phenotype is maintained in healthy HASMCs to regulate vascular tone. However, in abnormal proliferation or degeneration of HASMCs, pathological remodelling occurs causing HASMC conversion from contractile to synthetic phenotype.

A pathological process in TAD development is the degeneration of HASMCs; and the association between TAD development and phenotypic HASMC conversion has been reported (5). RNA modifications may therefore play a role in the pathogenesis of TAD. N6-methyladenosine (m6A) is the most common endogenous epigenetic modification on messenger RNA (mRNA) because it can be modified and recognised by enzymes and binding proteins to regulate biological functions (6). Methyltransferase-like-3 (METTL3) is involved in m6A formation; and NOTCH1 signalling is involved in cell proliferation, differentiation, and apoptosis (7-10). METTL3 was reported to regulate the progression of abdominal aortic aneurysms through m6A processing (11). Another study reported that reduced METTL3 expression in urinary tumour cells was associated with upregulation of the NOTCH1 apoptosis pathway (12). However, the role of METTL3 and NOTCH1 on m6A modification in TAD has not been established.

A recent study by Yang et al. found that METTL3 was responsible for the m6A modification of NOTCH1 mRNA, which when overexpressed in vitro resulted in increased mRNA degradation and subsequent promotion of the synthetic HASMC phenotype associated with the progression of TAD (13). This was demonstrated through changes in expression of METTL3, NOTCH1, α-smooth muscle actin (α-SMA) and smooth muscle protein 22-α (SM22α) mRNA via quantitative real-time polymerase chain reaction (qRT-PCR) and relative protein levels via western blot. Given that α-SMA and SM22α are markers of contractile smooth muscle cells (SMCs), their decreased levels of expression with decreased NOTCH1 indicate that the HASMCs had dedifferentiated and subsequently changed phenotype.

To date, studies have identified that the conversion of the contractile SMC to the synthetic phenotype underpins the pathogenesis of most vascular pathologies. Increased METTL3 levels have previously been associated with atherosclerotic plaques, abdominal aortic aneurysm development and the presence of synthetic SMCs (11,14-16). Moreover, given that a study by Qin et al identified that METTL3 increased proliferation of PASMCs in vivo and in vitro via hypermethylation and resultant degradation of phosphatase and tensin homolog (PTEN) mRNA, it should not be surprising that similar results have been concluded regarding NOTCH1 (14).

Nevertheless, these disease processes are multifactorial and must also consider cell-to-cell interactions, endothelial dysfunction and the regulation of gene expression via the application of shear forces induced by blood flow. Consequently, evidence surrounding the relationship between METTL3, NOTCH1 and SMC phenotype is somewhat paradoxical. A number of ApoE(−/−) mouse model studies agree that increased (rather than decreased) NOTCH1 expression induces conversion of the contractile SMC to the synthetic phenotype, thus contributing to the progression of diseases such as atherosclerosis, aortic aneurysm and aortic dissection (17,18). Undoubtedly, the key distinction between these studies is the use of in vivo models rather than a two-dimensional in vitro model; in vitro models are simpler and enable tight control of the physical and chemical environment but often fail to replicate the in vivo conditions of that cell type.

Three-dimensional cultures are becoming more popular and offer a compromise between two-dimensional in vitro models and mouse models. These enable cell cultures to increase overall cell to cell contact, interact with extracellular matrices, promote differentiation and tissue organisation, as well as enable the development of multi-layered models that could closely replicate the vascular anatomy. HUASMCs in a three-dimensional model have been shown to increase expression of differentiation markers including α-SMA, induce quiescence and amplify transforming growth factor-β (TGF-β) expression, down-regulating downstream expression nuclear factor-kappaB (NF-κB) activity (19). Consequently, provided that NF-κB promotes NOTCH1 signalling, it would be expected that NOTCH1 activity would be low in contractile SMCs and that increased NOTCH1 expression would correlate to an increase in synthetic phenotype.

Moreover, the aorta undergoes approximately 10% stretch under physiological conditions. Less than 10% stretch on SMCs have been shown to inhibit NOTCH signalling and promote the contractile SMC phenotype (20). On the other hand, most aortic dissections occur as a result of high blood pressure, with acute hypertension inducing stretching over 20% resulting in the up-regulation of inflammatory markers such as IL-1β, IL-6, VCAM-1 and ICAM-1, which are controlled by NF-κB. Thus, it could be anticipated that increased stretch in hypertension could also promote the expression of NOTCH1.

Additionally, it was previously noted that SMCs were either of a contractile or synthetic phenotype; yet a recent review by Yap et al identified six distinct sub-populations of vascular SMCs via examination of the genotype of SMCs within different vascular pathologies (21). The synthetic SMC phenotype could be divided into mesenchymal-, fibroblast-, macrophage-, osteogenic- and adipocyte-like vascular SMCs. It would therefore be interesting to examine the genotype of the SMCs in aortic dissections to confer whether the subtype(s) correlated to the synthetic SMCs in the study by Yang et al. (13). If the SMCs correspond, the results could demonstrate that whilst increased METTL3 expression is associated with promoting the synthetic SMC phenotype, it is not dependant on the augmentation of NOTCH1 and alternative mechanisms may be responsible, such as those involved in mechano-transduction.

Conclusively, whilst the mechanism by which METTL3 contributes to synthetic transformation of SMCs is yet to be confirmed owing to contradictory evidence regarding its relation to NOTCH1 expression, global reduction of METTL3 expression may prevent the progression of several critical vascular diseases and have a widespread impact on mortality rates and healthcare costs attributed to cardiovascular disease. Despite this, given that METTL3 is responsible for mRNA processing, translations efficiency, editing and stability in fundamental biological processes, it may be difficult to specifically augment METTL3 expression in SMCs to reduce progression of TAD without inducing some off-target effects (22). The severity of these implications would be difficult to determine prior to in vivo testing but evidence suggests that further analysis would be warranted to perceive probability of success.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, Annals of Translational Medicine. The article did not undergo external peer review.

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://atm.amegroups.com/article/view/10.21037/atm-23-375/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. LeMaire SA, Russell L. Epidemiology of thoracic aortic dissection. Nat Rev Cardiol 2011;8:103-13. [Crossref] [PubMed]
  2. Takada M, Yamagishi K, Tamakoshi A, et al. Height and Mortality from Aortic Aneurysm and Dissection. J Atheroscler Thromb 2022;29:1166-75. [Crossref] [PubMed]
  3. Daily PO, Trueblood HW, Stinson EB, et al. Management of Acute Aortic Dissections. Ann Thorac Surg 1970;10:237-47. [Crossref] [PubMed]
  4. Nienaber CA, Fattori R, Mehta RH, et al. Gender-related differences in acute aortic dissection. Circulation 2004;109:3014-21. [Crossref] [PubMed]
  5. Pan L, Bai P, Weng X, et al. Legumain Is an Endogenous Modulator of Integrin αvβ3 Triggering Vascular Degeneration, Dissection, and Rupture. Circulation 2022;145:659-74. [Crossref] [PubMed]
  6. He Y, Xing J, Wang S, et al. Increased m6A methylation level is associated with the progression of human abdominal aortic aneurysm. Ann Transl Med 2019;7:797. [Crossref] [PubMed]
  7. Liu S, Zhuo L, Wang J, et al. METTL3 plays multiple functions in biological processes. Am J Cancer Res 2020;10:1631-46.
  8. Xia T, Wu X, Cao M, et al. The RNA m6A methyltransferase METTL3 promotes pancreatic cancer cell proliferation and invasion. Pathol Res Pract 2019;215:152666. [Crossref] [PubMed]
  9. Kopan R. Notch signaling. Cold Spring Harb Perspect Biol 2012;4:a011213. [Crossref] [PubMed]
  10. Zeronian MR, Klykov O, Portell I, de Montserrat J, et al. Notch-Jagged signaling complex defined by an interaction mosaic. Proc Natl Acad Sci U S A 2021;118:e2102502118. [Crossref] [PubMed]
  11. Zhong L, He X, Song H, et al. METTL3 Induces AAA Development and Progression by Modulating N6-Methyladenosine-Dependent Primary miR34a Processing. Mol Ther Nucleic Acids 2020;21:394-411. [Crossref] [PubMed]
  12. Tao Z, Zhao Y, Chen X. Role of methyltransferase-like enzyme 3 and methyltransferase-like enzyme 14 in urological cancers. PeerJ 2020;8:e9589. [Crossref] [PubMed]
  13. Yang J, Fang M, Yu C, et al. Human aortic smooth muscle cell regulation by METTL3 via upregulation of m6A NOTCH1 modification and inhibition of NOTCH1. Ann Transl Med 2023; [Crossref]
  14. Qin Y, Qiao Y, Li L, et al. The m(6)A methyltransferase METTL3 promotes hypoxic pulmonary arterial hypertension. Life Sci 2021;274:119366. [Crossref] [PubMed]
  15. Zhou Y, Jiang R, Jiang Y, et al. Exploration of N6-Methyladenosine Profiles of mRNAs and the Function of METTL3 in Atherosclerosis. Cells 2022;11:2980. [Crossref] [PubMed]
  16. Zhao Y, Xia A, Li C, et al. Methyltransferase like 3-mediated N6-methylatidin methylation inhibits vascular smooth muscle cells phenotype switching via promoting phosphatidylinositol 3-kinase mRNA decay. Front Cardiovasc Med 2022;9:913039. [Crossref] [PubMed]
  17. Li Y, Takeshita K, Liu PY, et al. Smooth muscle Notch1 mediates neointimal formation after vascular injury. Circulation 2009;119:2686-92. [Crossref] [PubMed]
  18. Sachdeva J, Mahajan A, Cheng J, et al. Smooth muscle cell-specific Notch1 haploinsufficiency restricts the progression of abdominal aortic aneurysm by modulating CTGF expression. PLoS One 2017;12:e0178538. [Crossref] [PubMed]
  19. Jäger MA, De La Torre C, Arnold C, et al. Assembly of vascular smooth muscle cells in 3D aggregates provokes cellular quiescence. Exp Cell Res 2020;388:111782. [Crossref] [PubMed]
  20. Jensen LF, Bentzon JF, Albarrán-Juárez J. The Phenotypic Responses of Vascular Smooth Muscle Cells Exposed to Mechanical Cues. Cells 2021;10:2209. [Crossref] [PubMed]
  21. Yap C, Mieremet A, de Vries CJM, et al. Six Shades of Vascular Smooth Muscle Cells Illuminated by KLF4 (Krüppel-Like Factor 4). Arterioscler Thromb Vasc Biol 2021;41:2693-707. [Crossref] [PubMed]
  22. METTL3 Protein Expression Summary. [cited 2023 Mar 20]. (The Human Protein Atlast). Available onine: https://www.proteinatlas.org/ENSG00000165819-METTL3
Cite this article as: Wright K, Aboughdir M, Harky A. The cellular uncertainty in thoracic aortic dissections: the roles of METTL3 and NOTCH1 on m6A in human aortic smooth muscle cells. Ann Transl Med 2023;11(4):162. doi: 10.21037/atm-23-375

Download Citation