Skip to main content
Log in

Biomechanical and transcriptional evidence that smooth muscle cell death drives an osteochondrogenic phenotype and severe proximal vascular disease in progeria

  • Original Paper
  • Published:
Biomechanics and Modeling in Mechanobiology Aims and scope Submit manuscript

Abstract

Hutchinson–Gilford Progeria Syndrome results in rapid aging and severe cardiovascular sequelae that accelerate near end-of-life. We found a progressive disease process in proximal elastic arteries that was less evident in distal muscular arteries. Changes in aortic structure and function were then associated with changes in transcriptomics assessed via both bulk and single cell RNA sequencing, which suggested a novel sequence of progressive aortic disease: adverse extracellular matrix remodeling followed by mechanical stress-induced smooth muscle cell death, leading a subset of remnant smooth muscle cells to an osteochondrogenic phenotype that results in an accumulation of proteoglycans that thickens the aortic wall and increases pulse wave velocity, with late calcification exacerbating these effects. Increased central artery pulse wave velocity is known to drive left ventricular diastolic dysfunction, the primary diagnosis in progeria children. It appears that mechanical stresses above ~ 80 kPa initiate this progressive aortic disease process, explaining why elastic lamellar structures that are organized early in development under low wall stresses appear to be nearly normal whereas other medial constituents worsen progressively in adulthood. Mitigating early mechanical stress-driven smooth muscle cell loss/phenotypic modulation promises to have important cardiovascular implications in progeria patients.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

Data availability

All data needed to evaluate the conclusions of the paper are in the paper and the Supplementary Materials. Additional information is available from the corresponding author upon reasonable request.

References

  • Bayer IM, Adamson SL, Langille BL (1999) Atrophic remodeling of the artery-cuffed artery. Arterioscler Thromb Vasc Biol 19:1499–1505

    Google Scholar 

  • Benedicto I, Dorado B, Andres V (2021) Molecular and cellular mechanisms driving cardiovascular disease in Hutchinson-Gilford progeria syndrome: lessons learned from animal models. Cells 10:1157

    Google Scholar 

  • Bibby JA, Agarwal D, Freiwald T, Kunz N, Merle NS, West EE, Singh P, Larochelle A, Chinian F, Mukherjee S, Afzali B, Kemper C, Zhang NR (2022) Systematic single-cell pathway analysis to characterize early T cell activation. Cell Rep 41:111697

    Google Scholar 

  • Boers JLV, Peeters EAG, Kuijpers JHJ et al (2004) Decreased mechanical stiffness in LMNA−/− cells is caused by defective nucleo-cytoskeletal integrity: implications for the development of laminopathies. Hum Mol Genet 13:2567–2580

    Google Scholar 

  • Boutouyrie P, Chowienczyk P, Humphrey JD, Mitchell GF (2021) Arterial Stiffness and Cardiovascular Risk in Hypertension. Circ Res 128:864–886

  • Briot A, Jaroszewicz A, Warren CM et al (2014) Repression of Sox9 by Jag1 is continuously required to suppress the default chondrogenic fate of vascular smooth muscle cells. Dev Cell 31:707–721

    Google Scholar 

  • Capell BC, Olive M, Erdos MR, Cao K, Faddah DA, Tavarez UL, Chen X (2008) A farnesyltransferase inhibitor prevents both the onset and late progression of cardiovascular disease in a progeria mouse model. Proc Natl Acad Sci USA 105:15902–15907

    Google Scholar 

  • Cavinato C, Murtada S-I, Rojas A, Humphrey JD (2021) Evolving structure-function relations during aortic maturation and aging revealed by multiphoton microscopy. Mechan Ageing Devel 196:111471

    Google Scholar 

  • Chambliss AB, Khatau SB, Erdenberger N, Robinson DK, Hodzic D, Longmore GD, Wirtz D (2013) The LINC-anchored actin cap connects the extracellular milieus to the nucleus for ultrafast mechanotransduction. Sci Rept 3:1087

    Google Scholar 

  • Clarke MCH, Littlewood TD, Figg N, Maguire JJ, Davenport AP, Goddard M, Bennett MR (2008) Chronic apoptosis of vascular smooth muscle cells accelerates atherosclerosis and promotes calcification and medial degeneration. Circ Res 102:1529–1538

    Google Scholar 

  • De Sandre-Giovannoli A, Bernard R, Cau P et al (2003) Lamin A truncation in Hutchinson-Gilford Progeria. Science 300:2055

    Article  Google Scholar 

  • Del Campo L, Sánchez-López A, Salaices M, von Kleeck RA, Expósito E, González-Gómez C, Cussó L, Guzmán-Martínez G, Ruiz-Cabello J, Desco M, Assoian RK, Briones AM, Andrés V (2019) Vascular smooth muscle cell-specific progerin expression in a mouse model of Hutchinson-Gilford progeria syndrome promotes arterial stiffness: therapeutic effect of dietary nitrite. Aging Cell 18:e12936

    Google Scholar 

  • Desai AS, Mitchell GF, Fang JC, Creager MA (2009) Central aortic stiffness is increased in patients with heart failure and preserved ejection fraction. J Card Fail 15:658–664

    Google Scholar 

  • Dolgalev I (2022). msigdbr: MSigDB Gene sets for multiple organisms in a tidy data format. R package version 7.5.1, https://CRAN.R-project.org/package=msigdbr

  • DuBose AJ, Lichtenstein ST, Petrash NM, Erdos MR, Gordon LB, Collins FS (2018) Everolimus rescues multiple cellular defects in laminopathy-patient fibroblasts. Proc Natl Acad Sci USA 115:4206–4211

    Google Scholar 

  • Durham AL, Speer MY, Scatena M, Giachelli CM, Shanahan CM (2018) Role of smooth muscle cells in vascular calcification: implications in atherosclerosis and arterial stiffness. Cardiovasc Res 114:590–600

    Google Scholar 

  • Eriksson M, Brown WT, Gordon LB et al (2003) Recurrent de novo point mutations in lamin A cause Hutchinson-Gilford progeria syndrome. Nature 423:293–298

    Google Scholar 

  • Estrada AC, Irons L, Rego BV, Li G, Tellides G, Humphrey JD (2021) Roles of mTOR in thoracic aortopathy understood by complex intracellular signaling interactions. PLoS Comput Biol 17:e1009683

  • Ferruzzi J, Madziva D, Caulk AW, Tellides G, Humphrey JD (2018) Compromised mechanical homeostasis in arterial aging and associated cardiovascular consequences. Biomech Model Mechanobiol 17:1281–1295

    Google Scholar 

  • Gerhard-Herman M, Smoot LB, Wake N, Kieran MW, Kleinman ME, Miller DT, Schwartzman A, Giobbie-Hurder A, Neuberg D, Gordon LB (2012) Mechanisms of premature vascular aging in children with Hutchison-Gilford progeria syndrome. Hypertension 59:92–97

    Google Scholar 

  • Gordon LB, Kleinman ME, Miller DT et al (2012) Clinical trial of a farnesyltransferase inhibitor in children with Hutchinson-Gilford progeria syndrome. Proc Natl Acad Sci USA 109:16666–16671

    Google Scholar 

  • Gordon LB, Massaro J, D’Agostino RB et al (2014) Impact of farnesylation inhibitors on survival in Hutchinson-Gilford progeria syndrome. Circulation 130:27–34

    Google Scholar 

  • Gordon LB, Shappell H, Massaro J, D’Agostino RB, Brazier J, Kleinman ME, Kieran MW (2018) Association of lonafarnib treatment vs no treatment with mortality rates in patients with Hutchison-Gilford progeria syndrome. JAMA 319:1687–1695

    Google Scholar 

  • Humphrey JD, Eberth JF, Dye WW, Gleason RL (2009) Fundamental role of axial stress in compensatory adaptations by arteries. J Biomech 42:1–8

  • Humphrey JD, Dufresne ER, Schwartz MA (2014) Mechanotransduction and extracellular matrix homeostasis. Nat Rev Mol Cell Biol 15:802–812

  • Humphrey JD, Tellides G (2019) Central artery stiffness and thoracic aortopathy. Am J Physiol 316:H169-182

    Google Scholar 

  • Johnson RC, Leopold JA, Loscalzo J (2006) Vascular calcification: pathobiological mechanisms and clinical implications. Circ Res 99:1044–1059

    Google Scholar 

  • Kelleher CM, McLean SE, Mecham RP (2004) Vascular extracellular matrix and aortic development. Curr Trends Develop Biol 62:153–188

    Google Scholar 

  • Kim PH, Luu J, Heizer P, et al. (2018) Disrupting the LINC complex in smooth muscle cells reduces aortic disease in a mouse model of Hutchinson-Gilford progeria syndrome. Sci Transl Med 10:eaat7163

  • Kim PH, Chen NY, Heizer PJ et al (2021) Nuclear membrane ruptures underlie the vascular pathology in a mouse model of Hutchinson-Gilford progeria syndrome. JCI Insight 6:e151515

    Google Scholar 

  • Kreienkamp R, Billon C, Bedia-Diaz G, Albert CJ, Toth Z, Butler AA, Gonzalo S (2019) Doubled lifespan and patient-like pathologies in progeria mice fed high-fat diet. Aging Cell 18:e12852

    Google Scholar 

  • Latorre M, Spronck B, Humphrey JD (2021) Complementary roles of mechanotransduction and inflammation in vascular homeostasis. Proc Math Phys Eng Sci 477:20200622

    MathSciNet  Google Scholar 

  • Laurent S, Boutouyrie P (2015) The structural factor of hypertension: large and small artery alterations. Circ Res 116:1007–1021

    Google Scholar 

  • Le VP, Cheng JK, Kim J et al (2015) Mechanical factors direct mouse aortic remodeling during early maturation. J R Soc Interface 12:20141350

    Google Scholar 

  • Lemire JM, Patis C, Gordon LB, Sandy JD, Toole BPM, Weiss AS, (2006) Aggrecan expression is substantially and abnormally upregulated in Hutchinson-Gilford progeria syndrome dermal fibroblasts. Mech Ageing Develop 127:660–669

  • Li G, Wang M, Caulk AW, Korneva A, Bersi MR, Wang G, Liu X, Mehta S, Geirsson A, Gulcher JR, Chittenden TW, Simons M, Humphrey JD, Tellides G (2020a) Chronic mTOR activation induces a degradative smooth muscle cell phenotype and aortopathy. J Clin Invest 130:1233–1251

  • Li Y, Sun Z, Zhang L, Yan J, Shao C, Jin L, Li L, Wang Z (2020b) Role of macrophages in the progression and regression of vascular calcification. Front Pharmacol 11:661

    Google Scholar 

  • Liu Y, Drozdov I, Shroff R, Beltran LE, Shanahan CM (2013) Prelamin A accelerates vascular calcification via activation of the DNA damage response and senescence-associated secretory phenotype in vascular smooth muscle cells. Circ Res 112:e99-109

    Google Scholar 

  • Liberzon A, Birger C, Thorvaldsdóttir H, Ghandi M, Mesirov JP, Tamayo P (2015) The molecular signatures database (MSigDB) hallmark gene set collection. Cell Syst 1(6):417–425

    Google Scholar 

  • Michel J-B (2003) Anoikis in the cardiovascular system: known and unknown extracellular mediators. Arterioscler Thromb Vasc Biol 23:2146–2154

    Google Scholar 

  • Murtada S-I, Kawamura Y, Caulk AW, Ahmadzadeh H, Mikush N, Zimmerman K, Kavanagh D, Weiss D, Latorre M, Zhuang ZW, Shadel GS, Braddock DT, Humphrey JD (2020) Paradoxical aortic stiffening and subsequent cardiac dysfunction in Hutchison-Gilford progeria. J R Soc Interface 17:20200066

    Google Scholar 

  • Murtada S-I, Kawamura Y, Li G, Schwartz MA, Tellides G, Humphrey JD (2021a) Developmental origins of mechanical homeostasis in the aorta. Dev Dyn 250:629–639

    Google Scholar 

  • Murtada S-I, Kawamura Y, Humphrey JD (2021b) Differential biomechanical responses of elastic and muscular arteries to angiotensin II-induced hypertension. J Biomech 119:110297

    Google Scholar 

  • Murtada S-I, Mikush N, Wang M, Ren P, Kawamura Y, Ramachandra AB, Braddock DT, Tellides G, Gordon LB, Humphrey JD (2023) Lonafarnib improves cardiovascular function and survival in a mouse model of Hutchinson-Gilford Progeria Syndrome. eLife 12:e82728.

  • Ngai D, Lino M, Bendeck MP (2018) Cell-matrix interactions and mataricine signaling in the pathogenesis of vascular calcification. Front Cardiovasc Med 5:174

    Google Scholar 

  • Olive M, Harten I, Mitchell R et al (2010) Cardiovascular pathology in Hutchinson-Gilford Progeria: correlation with the vascular pathology of aging. Arterioscler Thromb Vasc Biol 30:2301–2309

    Google Scholar 

  • Osmanagic-Myers S, Kiss A, Manakanatas C, Hamza O, Sedlmayer F, Szabo PL, Fischer I, Pichtinger P, Podesser BK, Eriksson M, Foisner R (2019) Endothelial progerin expression causes cardiovascular pathology through an impaired mechanoresponse. J Clin Invest 129:531–545

    Google Scholar 

  • Osorio FG, Navarro CL, Cadinanos J, et al (2011) Splicing-directed therapy in a new mouse model of human accelerated aging. Sci Transl Med 3: 106ra107

  • Prakash A, Gordon LB, Kleinman ME, Gurary EB, Massaro J, D’Agostino R, Kieran MW, Gerhard-Herman M, Smoot L (2018) Cardiac abnormalities in patients with Hutchison-Gilford progeria syndrome. JAMA Cardiol 3:326–334

    Google Scholar 

  • Purnomo E, Emoto N, Nugrahaningshi DAA et al (2013) Glycosaminoglycan overproduction in the aorta increases aortic calcification in murine chronic kidney disease. J Am Heart Assoc 2:e000405

    Google Scholar 

  • Roccabianca S, Bellini C, Humphrey JD (2014) Computational modeling suggests good, bad, and ugly roles of glycosaminoglycans in arterial mechanics and mechanobiology. J R Soc Interface 11:20140397

    Google Scholar 

  • Sage AP, Tintut Y, Demer LL (2010) Regulatory mechanisms in vascular calcification. Nat Rev Cardiol 7:528–536

    Google Scholar 

  • Sanchez-Lopez A, Espinos-Estevez C, Gonzalez-Gomez C, et al. (2021) Cardiovascular progerin suppression and lamin A restoration rescues Hutchinson-Gilford progeria syndrome. Circulation 144:1777–1794

  • Sazonova OV, Isenberg BC, Herrmann J et al (2015) Extracellular matrix presentation modulates vascular smooth muscle cell mechanotransduction. Matrix Biol 41:36–43

    Google Scholar 

  • Schroeder F, Polzer S, Slazansky M, Man V, Skacel P (2018) Predictive capabilities of various constitutive models for arterial tissue. J Mech Behav Biomed Matl 78:369–380

    Google Scholar 

  • Stehbens WE, Wakefield SJ, Gilbert-Barness E, Olson RE, Ackerman J (1999) Histological and ultrastructural features of atherosclerosis in progeria. Cardiovasc Pathol 8:29–39

    Google Scholar 

  • Stehbens WE, Delahunt B, Shozawa T, Gilbert-Barness E (2001) Smooth muscle cell depletion and collagen types in progeric arteries. Cardiovasc Path 10:133–136

    Google Scholar 

  • Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, Mesirov JP (2005) Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA 102(43):15545–15550

    Google Scholar 

  • Swift J, Ivanovska IL, Buxboim A, Harada T, Dingal PDP, Pinter J, Rehfeldt F (2013) Nuclear lamin-A scales with tissue stiffness and enhances matrix-directed differentiation. Science 341:1240104

    Google Scholar 

  • Tintut Y, Patel J, Territo M, Saini T, Parhami F, Demer LL (2002) Monocyte/macrophage regulation of vascular calcification in vitro. Circulation 105:650–655

    Google Scholar 

  • Townsend RR, Wilkinson IB, Schiffrin EL, et al. on behalf of the American Heart Association Council on Hypertension (2015) Recommendations for improving and standardizing vascular research on arterial stiffness. Hypertension 66:698–722

  • Varga R, Eriksson M, Erdos MR et al (2006) Progressive vascular smooth muscle cell defects in a mouse model of Hutchinson-Gilford progeria syndrome. Proc Natl Acad Sci USA 103:3250–3255

    Google Scholar 

  • Verstraeten VLRM, Ji JY, Cummings KS, Lee RT, Lammerding J (2008) Increased mechanosensitivity and nuclear stiffness in Hutchinson-Gilford progeria cells: effects of farnesyltransferase inhibitors. Aging Cell 7:383–393

    Google Scholar 

  • Villa-Bellosta R, Rivera-Torres J, Osorio FG, Acin-Perez R, Enriquez JA, Lopez-Otin C, Andres V (2013) Defective extracellular pyrophosphate metabolism promotes vascular calcification in a mouse model of Hutchinson-Gilford progeria syndrome that is ameliorated on pyrophosphate treatment. Circulation 127:3442–3451

    Google Scholar 

Download references

Acknowledgements

This work was supported by a grant from the US NIH (R01 HL105297, JDH) and the Masason Foundation (YK).

Author information

Authors and Affiliations

Authors

Contributions

Design of the project was contributed by SIM, YK, GT, JDH; Data collection was contributed by SIM, YK, CC, MW, ABR, BS; Data analysis and interpretation were contributed by SIM, YK, CC, DSL, GT, JDH. Writing and editing were contributed by SIM, YK, CC, GT, JDH.

Corresponding author

Correspondence to Jay D. Humphrey.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics statement

All live animal procedures were conducted in accordance with Federal Regulations and were approved by the Yale Institutional Animal Care and Use Committee prior to beginning the work.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 1167 kb)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Murtada, SI., Kawamura, Y., Cavinato, C. et al. Biomechanical and transcriptional evidence that smooth muscle cell death drives an osteochondrogenic phenotype and severe proximal vascular disease in progeria. Biomech Model Mechanobiol 22, 1333–1347 (2023). https://doi.org/10.1007/s10237-023-01722-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10237-023-01722-5

Keywords

Navigation