CC BY-NC-ND 4.0 · Thromb Haemost 2022; 122(10): 1814-1826
DOI: 10.1055/s-0042-1755329
Atherosclerosis and Ischaemic Disease

Protein Tyrosine Phosphatase 1B Deficiency in Vascular Smooth Muscle Cells Promotes Perivascular Fibrosis following Arterial Injury

Rajinikanth Gogiraju*
1   Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
,
Sogol Gachkar*
1   Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
,
David Velmeden*
1   Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
,
Magdalena L. Bochenek
1   Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
2   Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
,
Konstantinos Zifkos
2   Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
,
Astrid Hubert
1   Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
,
Thomas Münzel
1   Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
3   German Center for Cardiovascular Research (DZHK), Rhine-Main Site, Mainz, Germany
,
Stefan Offermanns
4   Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
5   Centre for Molecular Medicine, Medical Faculty, JW Goethe University Frankfurt, Frankfurt, Germany
6   Cardiopulmonary Institute (CPI), Frankfurt, Germany
7   German Center for Cardiovascular Research (DZHK e.V.), Rhine-Main Site, Frankfurt and Bad Nauheim, Germany
,
1   Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
3   German Center for Cardiovascular Research (DZHK), Rhine-Main Site, Mainz, Germany
› Author Affiliations
Funding This study was supported by grants from the Deutsche Forschungsgemeinschaft (SCHA 808/15-1 to K.S.) and the German Center for Cardiovascular Research (DZHK e.V.; Doktoranden-Stipendium to D.V.). Results shown in this study are part of the medical thesis of D.V.


Abstract

Background Smooth muscle cell (SMC) phenotype switching plays a central role during vascular remodeling. Growth factor receptors are negatively regulated by protein tyrosine phosphatases (PTPs), including its prototype PTP1B. Here, we examine how reduction of PTP1B in SMCs affects the vascular remodeling response to injury.

Methods Mice with inducible PTP1B deletion in SMCs (SMC.PTP1B-KO) were generated by crossing mice expressing Cre.ERT2 recombinase under the Myh11 promoter with PTP1Bflox/flox mice and subjected to FeCl3 carotid artery injury.

Results Genetic deletion of PTP1B in SMCs resulted in adventitia enlargement, perivascular SMA+ and PDGFRβ+ myofibroblast expansion, and collagen accumulation following vascular injury. Lineage tracing confirmed the appearance of Myh11-Cre reporter cells in the remodeling adventitia, and SCA1+ CD45- vascular progenitor cells increased. Elevated mRNA expression of transforming growth factor β (TGFβ) signaling components or enzymes involved in extracellular matrix remodeling and TGFβ liberation was seen in injured SMC.PTP1B-KO mouse carotid arteries, and mRNA transcript levels of contractile SMC marker genes were reduced already at baseline. Mechanistically, Cre recombinase (mice) or siRNA (cells)-mediated downregulation of PTP1B or inhibition of ERK1/2 signaling in SMCs resulted in nuclear accumulation of KLF4, a central transcriptional repressor of SMC differentiation, whereas phosphorylation and nuclear translocation of SMAD2 and SMAD3 were reduced. SMAD2 siRNA transfection increased protein levels of PDGFRβ and MYH10 while reducing ERK1/2 phosphorylation, thus phenocopying genetic PTP1B deletion.

Conclusion Chronic reduction of PTP1B in SMCs promotes dedifferentiation, perivascular fibrosis, and adverse remodeling following vascular injury by mechanisms involving an ERK1/2 phosphorylation-driven shift from SMAD2 to KLF4-regulated gene transcription.

* These authors share the first authorship.


Supplementary Material



Publication History

Received: 15 March 2022

Accepted: 25 June 2022

Article published online:
08 September 2022

© 2022. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Shi N, Mei X, Chen SY. Smooth muscle cells in vascular remodeling. Arterioscler Thromb Vasc Biol 2019; 39 (12) e247-e252
  • 2 Bennett MR, Sinha S, Owens GK. Vascular smooth muscle cells in atherosclerosis. Circ Res 2016; 118 (04) 692-702
  • 3 Yang M, Haase AD, Huang FK. et al. Dephosphorylation of tyrosine 393 in argonaute 2 by protein tyrosine phosphatase 1B regulates gene silencing in oncogenic RAS-induced senescence. Mol Cell 2014; 55 (05) 782-790
  • 4 Jäger M, Hubert A, Gogiraju R, Bochenek ML, Münzel T, Schäfer K. Inducible knockdown of endothelial protein tyrosine phosphatase-1B promotes neointima formation in obese mice by enhancing endothelial senescence. Antioxid Redox Signal 2019; 30 (07) 927-944
  • 5 Shimizu H, Shiota M, Yamada N. et al. Low M(r) protein tyrosine phosphatase inhibits growth and migration of vascular smooth muscle cells induced by platelet-derived growth factor. Biochem Biophys Res Commun 2001; 289 (02) 602-607
  • 6 Wright MB, Seifert RA, Bowen-Pope DF. Protein-tyrosine phosphatases in the vessel wall: differential expression after acute arterial injury. Arterioscler Thromb Vasc Biol 2000; 20 (05) 1189-1198
  • 7 Chang Y, Zhuang D, Zhang C, Hassid A. Increase of PTP levels in vascular injury and in cultured aortic smooth muscle cells treated with specific growth factors. Am J Physiol Heart Circ Physiol 2004; 287 (05) H2201-H2208
  • 8 Chang Y, Ceacareanu B, Zhuang D. et al. Counter-regulatory function of protein tyrosine phosphatase 1B in platelet-derived growth factor- or fibroblast growth factor-induced motility and proliferation of cultured smooth muscle cells and in neointima formation. Arterioscler Thromb Vasc Biol 2006; 26 (03) 501-507
  • 9 Golomb G, Fishbein I, Banai S. et al. Controlled delivery of a tyrphostin inhibits intimal hyperplasia in a rat carotid artery injury model. Atherosclerosis 1996; 125 (02) 171-182
  • 10 Majesky MW, Dong XR, Hoglund V, Mahoney Jr WM, Daum G. The adventitia: a dynamic interface containing resident progenitor cells. Arterioscler Thromb Vasc Biol 2011; 31 (07) 1530-1539
  • 11 Chappell J, Harman JL, Narasimhan VM. et al. 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 (12) 1313-1323
  • 12 Mourani PM, Garl PJ, Wenzlau JM, Carpenter TC, Stenmark KR, Weiser-Evans MC. Unique, highly proliferative growth phenotype expressed by embryonic and neointimal smooth muscle cells is driven by constitutive Akt, mTOR, and p70S6K signaling and is actively repressed by PTEN. Circulation 2004; 109 (10) 1299-1306
  • 13 Nemenoff RA, Simpson PA, Furgeson SB. et al. Targeted deletion of PTEN in smooth muscle cells results in vascular remodeling and recruitment of progenitor cells through induction of stromal cell-derived factor-1alpha. Circ Res 2008; 102 (09) 1036-1045
  • 14 Horita H, Wysoczynski CL, Walker LA. et al. Nuclear PTEN functions as an essential regulator of SRF-dependent transcription to control smooth muscle differentiation. Nat Commun 2016; 7: 10830
  • 15 Bence KK, Delibegovic M, Xue B. et al. Neuronal PTP1B regulates body weight, adiposity and leptin action. Nat Med 2006; 12 (08) 917-924
  • 16 Wirth A, Benyó Z, Lukasova M. et al. G12-G13-LARG-mediated signaling in vascular smooth muscle is required for salt-induced hypertension. Nat Med 2008; 14 (01) 64-68
  • 17 Chakraborty R, Saddouk FZ, Carrao AC, Krause DS, Greif DM, Martin KA. Promoters to study vascular smooth muscle. Arterioscler Thromb Vasc Biol 2019; 39 (04) 603-612
  • 18 Hubert A, Bochenek ML, Schütz E, Gogiraju R, Münzel T, Schäfer K. Selective deletion of leptin signaling in endothelial cells enhances neointima formation and phenocopies the vascular effects of diet-induced obesity in mice. Arterioscler Thromb Vasc Biol 2017; 37 (09) 1683-1697
  • 19 De Gasperi R, Rocher AB, Sosa MA. et al. The IRG mouse: a two-color fluorescent reporter for assessing Cre-mediated recombination and imaging complex cellular relationships in situ. Genesis 2008; 46 (06) 308-317
  • 20 Konstantinides S, Schäfer K, Thinnes T, Loskutoff DJ. Plasminogen activator inhibitor-1 and its cofactor vitronectin stabilize arterial thrombi after vascular injury in mice. Circulation 2001; 103 (04) 576-583
  • 21 Zhuang D, Pu Q, Ceacareanu B, Chang Y, Dixit M, Hassid A. Chronic insulin treatment amplifies PDGF-induced motility in differentiated aortic smooth muscle cells by suppressing the expression and function of PTP1B. Am J Physiol Heart Circ Physiol 2008; 295 (01) H163-H173
  • 22 Majesky MW, Horita H, Ostriker A. et al. Differentiated smooth muscle cells generate a subpopulation of resident vascular progenitor cells in the adventitia regulated by Klf4. Circ Res 2017; 120 (02) 296-311
  • 23 Bulut GB, Alencar GF, Owsiany KM. et al. KLF4 (Kruppel-Like Factor 4)-dependent perivascular plasticity contributes to adipose tissue inflammation. Arterioscler Thromb Vasc Biol 2021; 41 (01) 284-301
  • 24 Hu Y, Zhang Z, Torsney E. et al. Abundant progenitor cells in the adventitia contribute to atherosclerosis of vein grafts in ApoE-deficient mice. J Clin Invest 2004; 113 (09) 1258-1265
  • 25 Passman JN, Dong XR, Wu SP. et al. A sonic hedgehog signaling domain in the arterial adventitia supports resident Sca1+ smooth muscle progenitor cells. Proc Natl Acad Sci U S A 2008; 105 (27) 9349-9354
  • 26 Liu Y, Sinha S, McDonald OG, Shang Y, Hoofnagle MH, Owens GK. Kruppel-like factor 4 abrogates myocardin-induced activation of smooth muscle gene expression. J Biol Chem 2005; 280 (10) 9719-9727
  • 27 Dhaliwal NK, Abatti LE, Mitchell JA. KLF4 protein stability regulated by interaction with pluripotency transcription factors overrides transcriptional control. Genes Dev 2019; 33 (15–16): 1069-1082
  • 28 Dhaliwal NK, Miri K, Davidson S, Tamim El Jarkass H, Mitchell JA. KLF4 nuclear export requires ERK activation and initiates exit from naive pluripotency. Stem Cell Reports 2018; 10 (04) 1308-1323
  • 29 Li HX, Han M, Bernier M. et al. Krüppel-like factor 4 promotes differentiation by transforming growth factor-beta receptor-mediated Smad and p38 MAPK signaling in vascular smooth muscle cells. J Biol Chem 2010; 285 (23) 17846-17856
  • 30 Aoki J, Tanabe K. Mechanisms of drug-eluting stent restenosis. Cardiovasc Interv Ther 2021; 36 (01) 23-29
  • 31 Markova B, Herrlich P, Rönnstrand L, Böhmer FD. Identification of protein tyrosine phosphatases associating with the PDGF receptor. Biochemistry 2003; 42 (09) 2691-2699
  • 32 Thyberg J, Palmberg L, Nilsson J, Ksiazek T, Sjölund M. Phenotype modulation in primary cultures of arterial smooth muscle cells. On the role of platelet-derived growth factor. Differentiation 1983; 25 (02) 156-167
  • 33 Holycross BJ, Blank RS, Thompson MM, Peach MJ, Owens GK. Platelet-derived growth factor-BB-induced suppression of smooth muscle cell differentiation. Circ Res 1992; 71 (06) 1525-1532
  • 34 Ray HC, Corliss BA, Bruce AC. et al. Myh11+ microvascular mural cells and derived mesenchymal stem cells promote retinal fibrosis. Sci Rep 2020; 10 (01) 15808
  • 35 Liao M, Zhou J, Wang F. et al. An X-linked Myh11-CreERT2 mouse line resulting from Y to X chromosome-translocation of the Cre allele. Genesis 2017; 55 (09) 55
  • 36 Xiao Q, Zeng L, Zhang Z, Hu Y, Xu Q. Stem cell-derived Sca-1+ progenitors differentiate into smooth muscle cells, which is mediated by collagen IV-integrin alpha1/beta1/alphav and PDGF receptor pathways. Am J Physiol Cell Physiol 2007; 292 (01) C342-C352
  • 37 Miano JM, Cserjesi P, Ligon KL, Periasamy M, Olson EN. Smooth muscle myosin heavy chain exclusively marks the smooth muscle lineage during mouse embryogenesis. Circ Res 1994; 75 (05) 803-812
  • 38 Tigges U, Komatsu M, Stallcup WB. Adventitial pericyte progenitor/mesenchymal stem cells participate in the restenotic response to arterial injury. J Vasc Res 2013; 50 (02) 134-144
  • 39 Marais R, Wynne J, Treisman R. The SRF accessory protein Elk-1 contains a growth factor-regulated transcriptional activation domain. Cell 1993; 73 (02) 381-393
  • 40 Gille H, Kortenjann M, Thomae O. et al. ERK phosphorylation potentiates Elk-1-mediated ternary complex formation and transactivation. EMBO J 1995; 14 (05) 951-962
  • 41 Yoshida T, Hayashi M. Role of Krüppel-like factor 4 and its binding proteins in vascular disease. J Atheroscler Thromb 2014; 21 (05) 402-413
  • 42 Shankman LS, Gomez D, Cherepanova OA. et al. KLF4-dependent phenotypic modulation of smooth muscle cells has a key role in atherosclerotic plaque pathogenesis. Nat Med 2015; 21 (06) 628-637
  • 43 Wang T-M, Chen K-C, Hsu P-Y. et al. microRNA let-7g suppresses PDGF-induced conversion of vascular smooth muscle cell into the synthetic phenotype. J Cell Mol Med 2017; 21 (12) 3592-3601
  • 44 Kim MO, Kim SH, Cho YY. et al. ERK1 and ERK2 regulate embryonic stem cell self-renewal through phosphorylation of Klf4. Nat Struct Mol Biol 2012; 19 (03) 283-290
  • 45 Wang Z, Wang DZ, Hockemeyer D, McAnally J, Nordheim A, Olson EN. Myocardin and ternary complex factors compete for SRF to control smooth muscle gene expression. Nature 2004; 428 (6979): 185-189
  • 46 Malak PN, Dannenmann B, Hirth A, Rothfuss OC, Schulze-Osthoff K. Novel AKT phosphorylation sites identified in the pluripotency factors OCT4, SOX2 and KLF4. Cell Cycle 2015; 14 (23) 3748-3754
  • 47 He M, Zheng B, Zhang Y. et al. KLF4 mediates the link between TGF-β1-induced gene transcription and H3 acetylation in vascular smooth muscle cells. FASEB J 2015; 29 (09) 4059-4070
  • 48 Meng XM, Huang XR, Chung AC. et al. Smad2 protects against TGF-beta/Smad3-mediated renal fibrosis. J Am Soc Nephrol 2010; 21 (09) 1477-1487
  • 49 Frangogiannis N. Transforming growth factor-β in tissue fibrosis. J Exp Med 2020; 217 (03) e20190103
  • 50 Nemoto S, Matsumoto T, Taguchi K, Kobayashi T. Relationships among protein tyrosine phosphatase 1B, angiotensin II, and insulin-mediated aortic responses in type 2 diabetic Goto-Kakizaki rats. Atherosclerosis 2014; 233 (01) 64-71
  • 51 Texakalidis P, Tzoumas A, Giannopoulos S. et al. Risk factors for restenosis after carotid revascularization: a meta-analysis of hazard ratios. World Neurosurg 2019; 125: 414-424
  • 52 Londhe AD, Bergeron A, Curley SM. et al. Regulation of PTP1B activation through disruption of redox-complex formation. Nat Chem Biol 2020; 16 (02) 122-125
  • 53 Zierold S, Buschmann K, Gachkar S. et al. Brain-derived neurotrophic factor expression and signaling in different perivascular adipose tissue depots of patients with coronary artery disease. J Am Heart Assoc 2021; 10 (06) e018322