Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Technical Report
  • Published:

Generation and comparative analysis of an Itga8-CreERT2 mouse with preferential activity in vascular smooth muscle cells

An Author Correction to this article was published on 09 June 2023

This article has been updated

Abstract

All current smooth muscle cell (SMC) Cre mice similarly recombine floxed alleles in vascular and visceral SMCs. Here we present an Itga8-CreERT2 knock-in mouse and compare its activity with a Myh11-CreERT2 mouse. Both Cre drivers demonstrate equivalent recombination in vascular SMCs. However, Myh11-CreERT2 mice, but not Itga8-CreERT2 mice, display high activity in visceral SMC-containing tissues such as intestine, show early tamoxifen-independent activity and produce high levels of CreERT2 protein. Whereas Myh11-CreERT2-mediated knockout of serum response factor (Srf) causes a lethal intestinal phenotype precluding analysis of the vasculature, loss of Srf with Itga8-CreERT2 (SrfItga8) yields viable mice with no evidence of intestinal pathology. Male and female SrfItga8 mice exhibit vascular contractile incompetence, and angiotensin II causes elevated blood pressure in wild-type, but not SrfItga8, male mice. These findings establish the Itga8-CreERT2 mouse as an alternative to existing SMC Cre mice for unfettered phenotyping of vascular SMCs after selective gene loss.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Comparative recombination activity of Itga8-CreERT2 versus Myh11-CreERT2 in adult tissues.
Fig. 2: Distinguishing features of the Myh11-CreERT2 mouse.
Fig. 3: Recombination efficiency of Itga8-CreERT2 versus Myh11-CreERT2.
Fig. 4: Chronic blood pressure measurements in Itga8-CreERT2-mediated Srf knockout male mice.
Fig. 5: Chronic blood pressure measurements in Itga8-CreERT2-mediated Srf knockout female mice.
Fig. 6: Vascular contractile activity in Itga8-CreERT2-mediated Srf knockout mice.
Fig. 7: Bulk RNA-seq of aortas from Itga8-CreERT2-mediated Srf knockout mice.

Similar content being viewed by others

Data availability

RNA-seq data are available through the Gene Expression Omnibus (GSE138824 and GSE199244). Long-read data (informative reads only) are available through the National Center of Biotechnology Informationʼs Sequence Read Archive database, which can be downloaded from www.ncbi.nlm.nih.gov/sra, under BioProject number PRJNA825511. Original source data are included in the Supplementary Materials. Updates to Supplementary Table 1 are available upon reasonable request.

Change history

References

  1. Sauer, B. Inducible gene targeting in mice using the Cre/lox system. Methods 14, 381–392 (1998).

    Article  CAS  PubMed  Google Scholar 

  2. Gu, H., Marth, J. D., Orban, P. C., Mossmann, H. & Rajewsky, K. Deletion of a DNA polymerase beta gene segment in T cells using cell type-specific gene targeting. Science 265, 103–106 (1994).

    Article  CAS  PubMed  Google Scholar 

  3. Feil, R. et al. Ligand-activated site-specific recombination in mice. Proc. Natl Acad. Sci. USA 93, 10887–10890 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Majesky, M. W. Developmental basis of vascular smooth muscle diversity. Arter. Thromb. Vasc. Biol. 27, 1248–1258 (2007).

    Article  CAS  Google Scholar 

  5. Tasian, G., Cunha, G. & Baskin, L. Smooth muscle differentiation and patterning in the urinary bladder. Differentiation 80, 106–117 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Somlyo, A. P. & Somlyo, A. V. Signal transduction and regulation in smooth muscle. Nature 372, 231–236 (1994).

    Article  CAS  PubMed  Google Scholar 

  7. Kuriyama, H., Kitamura, K., Itoh, T. & Inoue, R. Physiological features of visceral smooth muscle cells, with special reference to receptors and ion channels. Physiol. Rev. 78, 811–920 (1998).

    Article  CAS  PubMed  Google Scholar 

  8. Owens, G. K., Kumar, M. S. & Wamhoff, B. R. Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol. Rev. 84, 767–801 (2004).

    Article  CAS  PubMed  Google Scholar 

  9. Chakraborty, R. et al. Promoters to study vascular smooth muscle: mistaken identity? Arter. Thromb. Vasc. Biol. 39, 603–612 (2019).

    Article  CAS  Google Scholar 

  10. Angstenberger, M. et al. Severe intestinal obstruction on induced smooth muscle-specific ablation of the transcription factor SRF in adult mice. Gastroenterology 133, 1948–1959 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. Mericskay, M. et al. Inducible mouse model of chronic intestinal pseudo-obstruction by smooth muscle-specific inactivation of the SRF gene. Gastroenterology 133, 1960–1970 (2007).

    Article  CAS  PubMed  Google Scholar 

  12. He, W. Q. et al. Myosin light chain kinase is central to smooth muscle contraction and required for gastrointestinal motility in mice. Gastroenterology 135, 610–620 (2008).

    Article  CAS  PubMed  Google Scholar 

  13. Albinsson, S. et al. Smooth muscle miRNAs are critical for post-natal regulation of blood pressure and vascular function. PLoS ONE 6, e18869 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Huang, J. et al. Myocardin is required for maintenance of vascular and visceral smooth muscle homeostasis during postnatal development. Proc. Natl Acad. Sci. USA 112, 4447–4452 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Morano, I. et al. Smooth-muscle contraction without smooth-muscle myosin. Nat. Cell Biol. 2, 371–375 (2000).

    Article  CAS  PubMed  Google Scholar 

  16. Niessen, P. et al. Smoothelin-A is essential for functional intestinal smooth muscle contractility in mice. Gastroenterology 129, 1592–1601 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. Halim, D. et al. Loss of LMOD1 impairs smooth muscle cytocontractility and causes megacystis microcolon intestinal hypoperistalsis syndrome in humans and mice. Proc. Natl Acad. Sci. USA 114, E2739–E2747 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Li, S., Wang, D. Z., Richardson, J. A. & Olson, E. N. The serum response factor coactivator myocardin is required for vascular smooth muscle development. Proc. Natl Acad. Sci. USA 100, 9366–9370 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Guzzardo, P. M. et al. A small cassette enables conditional gene inactivation by CRISPR/Cas9. Sci Rep. 7, 16770 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Horii, T. et al. Efficient generation of conditional knockout mice via sequential introduction of lox sites. Sci. Rep. 7, 7891 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149–157 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gurumurthy, C. B. et al. Reproducibility of CRISPR–Cas9 methods for generation of conditional mouse alleles: a multi-center evaluation. Genome Biol. 20, 171 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Bossy, B., Bossy-Wetzel, E. & Reichardt, L. F. Characterization of the integrin a8 subunit: a new integrin b1-associated subunit, which is prominently expressed on axons and on cells in contact with basal laminae in chick embryos. EMBO J. 10, 2375–2385 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Schnapp, L. M., Breuss, J. M., Ramos, D. M., Sheppard, D. & Pytela, R. Sequence and tissue distribution of the human integrin a8 subunit: a b1-associated a subunit expressed in smooth muscle cells. J. Cell Sci. 108, 537–544 (1995).

    Article  CAS  PubMed  Google Scholar 

  25. Hartner, A., Schocklmann, H., Prols, F., Muller, U. & Sterzel, R. B. α8 integrin in glomerular mesangial cells and in experimental glomerulonephritis. Kidney Int. 56, 1468–1480 (1999).

    Article  CAS  PubMed  Google Scholar 

  26. Chen, J., Maltby, K. M. & Miano, J. M. A novel retinoid-response gene set in vascular smooth muscle cells. Biochem. Biophys. Res. Commun. 281, 475–482 (2001).

    Article  CAS  PubMed  Google Scholar 

  27. Zargham, R. & Thibault, G. α8β1 integrin expression in the rat carotid artery: involvement in smooth muscle cell migration and neointima formation. Cardiovascular Res. 65, 813–822 (2005).

    Article  CAS  Google Scholar 

  28. Zargham, R., Touyz, R. M. & Thibault, G. α8 integrin overexpression in de-differentiated vascular smooth muscle cells attenuates migratory activity and restores the characteristics of the differentiated phenotype. Atherosclerosis 195, 303–312 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Kitchen, C. M., Cowan, S. L., Long, X. & Miano, J. M. Expression and promoter analysis of a highly restricted integrin alpha gene in vascular smooth muscle. Gene 513, 82–89 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Muller, U. et al. Integrin α8β1 is critically important for epithelial-mesenchymal interactions during kidney morphogenesis. Cell 88, 603–613 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Shen, Z. et al. Smooth muscle protein 22 alpha-Cre is expressed in myeloid cells in mice. Biochem. Biophys. Res. Commun. 422, 639–642 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wirth, A. et al. G12-G13–LARG–mediated signaling in vascular smooth muscle is required for salt-induced hypertension. Nat. Med. 14, 64–68 (2008).

    Article  CAS  PubMed  Google Scholar 

  33. Miano, J. M. et al. Restricted inactivation of serum response factor to the cardiovascular system. Proc. Natl Acad. Sci. USA 101, 17132–17137 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Alvarez-Aznar, A. et al. Tamoxifen-independent recombination of reporter genes limits lineage tracing and mosaic analysis using CreERT2 lines. Transgenic Res. 29, 53–68 (2020).

    Article  CAS  PubMed  Google Scholar 

  35. Vanlandewijck, M. et al. A molecular atlas of cell types and zonation in the brain vasculature. Nature 554, 475–480 (2018).

  36. Shimshek, D. R. et al. Codon-improved Cre recombinase (iCre) expression in the mouse. Genesis 32, 19–26 (2002).

    Article  CAS  PubMed  Google Scholar 

  37. Nicholls, P. K., Bellott, D. W., Cho, T. J., Pyntikova, T. & Page, D. C. Locating and characterizing a transgene integration site by nanopore sequencing. G3 (Bethesda) 9, 1481–1486 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Xue, B., Pamidimukkala, J. & Hay, M. Sex differences in the development of angiotensin II-induced hypertension in conscious mice. Am. J. Physiol. Heart. Circ. Physiol. 288, H2177–H2184 (2005).

    Article  CAS  PubMed  Google Scholar 

  39. Park, C. et al. Loss of serum response factor induces microRNA-mediated apoptosis in intestinal smooth muscle cells. Cell Death Dis. 6, e2011 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Retailleau, K. et al. Selective involvement of serum response factor in pressure-induced myogenic tone in resistance arteries. Arter. Thromb. Vasc. Biol. 33, 339–346 (2013).

    Article  CAS  Google Scholar 

  41. Galmiche, G. et al. Inactivation of serum response factor contributes to decrease vascular muscular tone and arterial stiffness in mice. Circ. Res. 112, 1035–1045 (2013).

    Article  CAS  PubMed  Google Scholar 

  42. Herring, B. P., Hoggatt, A. M., Gupta, A. & Wo, J. M. Gastroparesis is associated with decreased FOXF1 and FOXF2 in humans, and loss of FOXF1 and FOXF2 results in gastroparesis in mice. Neurogastroenterol Motil. 31, e13528 (2019).

    Article  PubMed  Google Scholar 

  43. Rashbrook, V. S., Brash, J. T. & Ruhrberg, C. Cre toxicity in mouse models of cardiovascular physiology and disease. Nat. Cardiovasc. Res. (in the press).

  44. Matthaei, K. I. Genetically manipulated mice: a powerful tool with unsuspected caveats. J. Physiol. 582, 481–488 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Perry, M. N. et al. Annotated expression and activity data for murine recombinase alleles and transgenes: the CrePortal resource. Mamm Genome 33, 55–65 (2022).

    Article  CAS  PubMed  Google Scholar 

  46. Buelow, B. & Scharenberg, A. M. Characterization of parameters required for effective use of tamoxifen-regulated recombination. PLoS ONE 3, e3264 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Sandlesh, P., Juang, T., Safina, A., Higgins, M. J. & Gurova, K. V. Uncovering the fine print of the CreERT2-LoxP system while generating a conditional knockout mouse model of Ssrp1 gene. PLoS ONE 13, e0199785 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Meinke, G., Bohm, A., Hauber, J., Pisabarro, M. T. & Buchholz, F. Cre recombinase and other tyrosine recombinases. Chem. Rev. 116, 12785–12820 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Clayton, J. A. Applying the new SABV (sex as a biological variable) policy to research and clinical care. Physiol. Behav. 187, 2–5 (2018).

    Article  CAS  PubMed  Google Scholar 

  50. Liao, M. et al. An X-linked Myh11-CreERT2 mouse line resulting from Y to X chromosome-translocation of the Cre allele. Genesis 55, 10.1002/dvg.23054 (2017).

  51. Morgan, A. P., Bell, T. A., Crowley, J. J. & Pardo-Manuel de Villena, F. Instability of the pseudoautosomal boundary in house mice. Genetics 212, 469–487 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Goodwin, L. O. et al. Large-scale discovery of mouse transgenic integration sites reveals frequent structural variation and insertional mutagenesis. Genome Res. 29, 494–505 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Zargham, R., Pepin, J. & Thibault, G. α8β1 integrin is up-regulated in the neointima concomitant with late luminal loss after balloon injury. Cardiovasc. Pathol. 16, 212–220 (2007).

    Article  CAS  PubMed  Google Scholar 

  54. Fuchs, H. et al. Mouse phenotyping. Methods 53, 120–135 (2011).

    Article  CAS  PubMed  Google Scholar 

  55. Nishimichi, N. et al. Induced hepatic stellate cell integrin, α8β1, enhances cellular contractility and TGFβ activity in liver fibrosis. J. Pathol. 253, 366–373 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Corliss, B. A. et al. Myh11 lineage corneal endothelial cells and ASCs populate corneal endothelium. Invest. Ophthalmol. Vis. Sci. 60, 5095–5103 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Armulik, A., Abramsson, A. & Betsholtz, C. Endothelial/pericyte interactions. Circ. Res. 97, 512–523 (2005).

    Article  CAS  PubMed  Google Scholar 

  58. Hess, D. L. et al. Perivascular cell-specific knockout of the stem cell pluripotency gene Oct4 inhibits angiogenesis. Nat. Commun. 10, 967 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Orlich, M. M. et al. Mural cell SRF controls pericyte migration, vessel patterning and blood flow. Circ. Res. 131, 308–327 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Miano, J. M. Serum response factor: toggling between disparate programs of gene expression. J. Mol. Cell. Cardiol. 35, 577–593 (2003).

    Article  CAS  PubMed  Google Scholar 

  61. George, S. H. et al. Developmental and adult phenotyping directly from mutant embryonic stem cells. Proc. Natl Acad. Sci. USA 104, 4455–4460 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  63. Preibisch, S., Saalfeld, S. & Tomancak, P. Globally optimal stitching of tiled 3D microscopic image acquisitions. Bioinformatics 25, 1463–1465 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ho Sui, S. J., Fulton, D. L., Arenillas, D. J., Kwon, A. T. & Wasserman, W. W. oPOSSUM: integrated tools for analysis of regulatory motif over-representation. Nucleic Acids Res. 35, W245–W252 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Gao, P. et al. Prime editing in mice reveals the essentiality of a single base in driving tissue-specific gene expression. Genome Biol. 22, 83 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Faulkner, J. L. et al. Lack of suppression of aldosterone production leads to salt-sensitive hypertension in female but not male Balb/C mice. Hypertension 72, 1397–1406 (2018).

    Article  CAS  PubMed  Google Scholar 

  67. Wu, J. et al. Inflammation and mechanical stretch promote aortic stiffening in hypertension through activation of p38 mitogen-activated protein kinase. Circ. Res. 114, 616–625 (2014).

    Article  CAS  PubMed  Google Scholar 

  68. Bryant, W. B. et al. CRISPR-LRS for mapping transgenes in the mouse genome. Preprint at https://www.biorxiv.org/content/10.1101/2022.01.05.475144v1 (2022).

Download references

Acknowledgements

We thank members of the University of Rochester Medical Center Genomics Core for carrying out the RNA-seq experiments and generating Fig. 2e, and we thank the Electron Microscopy and Histology Core at Augusta University for performing the immunogold studies and generating Fig. 1b. This work was supported by American Heart Association postdoctoral grants 17POST3360938 to Q.L. and CDA858380 to J.L.F. and by National Institutes of Health grants R00HL146948 to J.F.L.; HL155265 to E.B.C.; HL122578 to M.J.D.; HL122686 and HL139794 to X.L.; HL138987, HL136224 and HL147476 to J.M.M.; EY026614 to L.G.; and R00-HL143198 to S.D.Z.

Author information

Authors and Affiliations

Authors

Contributions

G.D.W. helped develop the figures and performed western blotting studies in Extended Data Figs. 2e, 4d and 8a,b and Figs. 2b and 3g,h; bulk RNA-seq studies in Fig. 2e and Supplementary Table 2; and Fig. 3a,b. G.D.W. also assisted with the blood pressure studies of Figs. 4 and 5 and with performing the myography studies of Fig. 6. J.L.F. performed and/or analyzed vascular function and blood pressure data of Figs. 4, 5 and 6a–c and wrote the Methods subsections related to these experiments. J.D. performed the studies in Fig. 3c,d and Extended Data Fig. 10 and conducted tamoxifen injections for studies of blood pressure and myography measurements. A.R.G. provided foundational work and helped generate Supplementary Fig. 1. P.G. assisted with western blotting studies of Extended Data Fig. 2a,b. A.C.Y. generated the data for Supplementary Fig. 2. O.J.S. performed all histology, immunostaining, confocal immunofluorescence and bright-field microscopy and generated all of the images, save lymphatic vessels. M.J.D. and S.D.Z. planned and performed experiments in Fig. 3e,f and Extended Data Figs. 6 and 7; they also contributed to the writing and editing of the Methods and Results sections. C.K.C., A.C.Y. and S.H.G. did all breeding and genotyping of mice. C.K.C. provided images to Extended Data Fig. 8c and Supplementary Fig. 4a. C.T.B. helped perform the telemetry experiments of Figs. 4 and 5. T.C.K. isolated aortas and helped run myography experiments in Fig. 6. W.B.B. prepared and analyzed the Sanger sequencing of the Cre in Myh11-CreERT2 of Supplementary Fig. 3 and the long-read sequence mapping of Myh11-CreERT2 in Fig. 2c,d. A.A. helped with the interpretation of Myh11-CreERT2 mapping and the design of Fig. 2d. A.K. contributed to western blotting studies of Fig. 2b. X.L. contributed to data in Fig. 2a and provided intellectual input on study design and interpretation. L.G. and X.X. designed and generated the Itga8-CreERT2 mouse and developed Extended Data Fig. 1a. E.B.C. helped with the acquisition and interpretation of the blood pressure and vascular reactivity data. Q.R.L. performed studies in Extended Data Figs. 2a–d, 3 and 9b–g, Supplementary Fig. 4b,d and Fig. 7; he also provided intellectual input throughout the study and finalized the figures. J.M.M. conceived and supervised the entire project, analyzed all data, developed Extended Data Fig. 1b and Supplementary Fig. 5, assembled original western blots in Extended Data Figs. 4a–c and 5 and Supplementary Fig. 6 and outlined, wrote and edited the manuscript.

Corresponding authors

Correspondence to Qing R. Lyu or Joseph M. Miano.

Ethics declarations

Competing interests

A.A. is a co-founder of Tango Therapeutics, Azkarra Therapeutics, Ovibio Corporation and Kytarro; a member of the board of Cytomx and Cambridge Science Corporation; a member of the scientific advisory board of Genentech, GLAdiator, Circle, Bluestar, Earli, Ambagon, Phoenix Molecular Designs and Trial Library; a consultant for SPARC, ProLynx and GlaxoSmithKline; receives grant or research support from SPARC and AstraZeneca; and holds patents on the use of PARP inhibitors held jointly with AstraZeneca from which he has benefited financially (and may do so in the future). The patents are unrelated to any aspect of the current study. All other authors declare no competing interests.

Peer review

Peer review information

Nature Cardiovascular Research thanks Kathleen Martin, Brenda Lilly and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Strategy and sequence validation of Itga8-CreERT2.

a, Partial Itga8 locus, overlapping long noncoding RNA, and general strategy for mouse ESC targeting. The 22 amino acid P2A self-cleaving cassette (GSGATNFSLLKQAGDVEENPGP) allows for concurrent detection of the CreERT2 fusion and the mCherry reporter; however, mCherry fluorescence was not easily detected and therefore not pursued in this study. Small horizontal arrows (p1-p4) denote primers for PCR genotyping of mice. b, Nucleotide sequence of targeted knock-in cassette following Flippase-mediated removal of the Neo gene. Colors correspond to the schematic in a. Bases highlighted in black boxes represent codon-optimized substitutions from original sequences. HR, homologous recombination.

Extended Data Fig. 2 ITGA8 protein expression in heterozygous Itga8-CreERT2 mice.

a, Western blots of ITGA8 protein in two independent experiments performed by two independent investigators with quantitative data at right. n = 6 independent male aortas for each condition. b, qRT-PCR analysis of Itga8 mRNA and c, the antisense Ak086420 long noncoding RNA in wild type versus Itga8-CreERT2 heterozygous aorta. n = 6 male aortas per genotype. d, qRT-PCR of cytosolic versus nuclear Actb and Ak086420 expression in wild type aorta (n = 4 male aortas). Error bars represent the mean ± standard deviation; p-values determined by two-tailed, unpaired Student’s t-test. e, Western blot of ITGA8 and CreERT2 in aorta of seven week-old male and female Itga8-CreERT2 heterozygous mice (n = 3 independent mice of each sex). Molecular weight markers at right here and below are in kilodaltons.

Source data

Extended Data Fig. 3 Recombination activity of different SMC Cre mice in myeloid cells.

a, Representative flow cytometry of GFP labeled cells from wild type C57BL/6J mice (i); mT/mG reporter mice (ii); and mT/mG in Sm22-Cre (iii); tamoxifen-treated male Myh11-CreERT2 (iv); tamoxifen-treated male Itga8-CreERT2 (v); tamoxifen-treated female Itga8-CreERT2 (vi) mice. Quantitative data for GFP + circulating cells in upper left quadrant Q1 (vii) and upper right quadrant Q2 (viii) are shown for each Cre driver line (n = 3 male mice per Cre line, save the Itga8CreERT2 line which represent male, n = 2, and female, n = 2, samples pooled for the graphs shown). The horizontal axis was set to the same threshold for all panels according to the apparent two populations of cells in the positive control (Sm22-Cre-mT/mG). The vertical axis divides two apparent populations of different sized cells in the Sm22-Cre-mT/mG strain. Note, as we did not gate for any surface markers in this study, we cannot label any quadrant as to cell type. Source of cells was from circulation cleared of red blood cells. Error bars represent the mean ± standard deviation. One-way ANOVA and posthoc testing revealed indicated p-values. See Supplementary Fig. 5 for more details. b, Bone marrow aspirates of 10-week-old male Itga8-CreERT2 (i, ii) and Myh11-CreERT2 (iii, iv) mice (n = 2 mice/genotype) carrying mT/mG reporter and treated with tamoxifen. Arrows indicate multi-nucleated megakaryocytes. Scale bars are 20 μm.

Source data

Extended Data Fig. 4 Comparative Cre activity in adult male and female blood vessels.

a, The GFP signal demonstrates recombination of the mT/mG reporter in medial SMCs of both the aorta and vena cava, whereas the red stained endothelium indicates the absence of recombination. Scale bar is 33 μm for aorta and 100 μm for vena cava. The aorta and vena cava of Itga8-CreERT2 was from a female mouse. b, Activity of each CreERT2 driver in indicated segments of male mouse aorta. Scale bars are 20 μm for all panels. Lu, lumen of vessel wall. c, Tamoxifen-treated 54-week-old female (i-iii) and male (iv-vi) mouse thoracic aorta (i, iv), brain (ii, v), and heart (iii, vi). Each panel is representative of at least two independent male or female mice. Scale bars are all 20 μm. d, Western blot of ITGA8 protein in 10-week-old versus 54-week-old female mouse aorta (n = 3 independent mice per time point).

Source data

Extended Data Fig. 5 Comparative Cre activity in adult mouse tissues.

a, GFP signal restricted to VSMCs in blood vessels (arrows) of each indicated tissue type. b, GFP signal in VSMCs and non-VSMCs of indicated tissue types. White arrowheads, blood vessels of kidney and thymus; white arrows, glomeruli of kidney; yellow arrows, sinusoids of liver. Itga8-CreERT2-mediated GFP signal was also present in myoepithelial cells of mammary gland and thecal cells around mature follicle (white asterisk) of ovary. All images were processed the same except for the Testis panel under Itga8-CreERT2, which was uniformly enhanced to bring out more of the signal that otherwise would be too dark to visualize. Scale bar in a is 100 μm for all images; scale bars in b are 50 μm for kidney and thymus and 20 μm for liver, mammary gland, and ovary. Data are representative of at least two independent male or female mice analyzed over the course of five years in two independent labs.

Extended Data Fig. 6 Leaky CreERT2 activity in aged mice.

Sections of aorta from 24-week-old or 54-week-old male Itga8-CreERT2 and 54-week-old Myh11-CreERT2 mice. The brightness in the 54-week Itga8-CreERT2 image was uniformly enhanced to better appreciate the GFP signal. Scale bars are 20 μm for all panels. Images are representative of two independent mice.

Extended Data Fig. 7 Quantitative activity of Myh11-CreERT2 versus Itga8-CreERT2 in popliteal blood vessels.

Representative 2D maximum projections from confocal imaging of live GFP and tdTomato fluorescence popliteal artery (a, b) and vein (c, d) from Myh11-CreERT2 versus Itga8-CreERT2 with quantitation of each in panels e and f, respectively. n = 4 male mice per genotype. Scale bars represent 50 μm (a, b) and 100 μm (c, d). Error bars in panels e-f represented by mean ± standard deviation; p-values determined by two-tailed, unpaired Student’s t-test.

Source data

Extended Data Fig. 8 Intestinal phenotype in Myh11-CreERT2 versus Itga8-CreERT2 mediated knockout of serum response factor (Srf).

a, Western blot showing lack of effect of tamoxifen on SRF in wild type aorta (n = 3 independent male mice per treatment). b, Quantitation of panel a represented by mean ± standard deviation; p-value determined by two-tailed, unpaired Student’s t-test. c, Anatomy of abdominal cavity in the indicated CreERT2 driver mice used for Srf inactivation. The two SrfMyh11 images were from mice 14 days following tamoxifen administration, whereas the SrfItga8 image was from a mouse 8 weeks after tamoxifen administration. d, Oil (panels i-iii) and Tamoxifen (panels iv-vi) treated SrfMyh11 mice with dissected gross intestine (i, iv), H&E stained intestine (ii, v), and immunostaining for SRF (green) and ACTA2 (red) in intestine (iii, vi). Scale bar is 100 μm for ii, iii, v, and vi or 1 mm for i and iv. Studies of panel d are representative of at least two independent male or female mice analyzed over the course of five years in two independent labs.

Source data

Extended Data Fig. 9 Itga8-CreERT2 mediated inactivation of Srf in adult mouse tissues.

a, Immunofluorescence confocal microscopy of sections of carotid artery (i, iv), bladder (ii, v), and intestine (iii, vi) from tamoxifen-treated male mice carrying homofloxed Srf alleles in the absence (i-iii) or presence (iv-vi) of Itga8-CreERT2 (abbreviated Cre). Sections were stained with antibodies to ACTA2 (red), SRF (green), and DAPI. Arrows and arrowheads point to blood vessels and visceral SMCs, respectively. Scale bar is 20 μm for all panels. Data from each panel are representative of at least two independent mice. b, SRF positive VSMCs were counted in sections of carotid arteries from Tam-administered homozygous floxed Srf mice without Cre (HoF + Tam, n = 6 male mice per condition) or homozygous floxed Srf mice with Cre (HoF + Cre+Tam, n = 5 male mice per condition). c, Same quantitative measures as in panel b only from intestine (n = 3 mice per condition). Western blots of SRF in aorta (d) and bladder (e) of indicated genotypes, all treated with the same schedule of tamoxifen. Corresponding quantitative data are shown for mouse aorta (f) and bladder (g) (n = 3 mice per genotype). HeF, heterozygous floxed Srf; HoF, homozygous floxed Srf. Error bars are mean ± standard deviation. Student unpaired, one-tailed t-test was applied in b and c, and one-way ANOVA with Tukey’s t-test for f and g to reveal indicated p-values in each graph.

Source data

Extended Data Fig. 10 SRF and contractile protein expression in mesenteric artery of SrfItga8 conditional knockout mice.

a, Confocal immunofluorescence microscopy of SRF and DAPI alone (i, iii) and merged with ACTA2 (ii, iv) in mesenteric arteries of Oil control (i, ii) versus tamoxifen-treated (iii, iv) homozygous SrfItga8 mice; scale bars are 20 μm. Results are representative of two independent mice for each condition. Western blots of SRF target, MYH11, in Oil versus Tam-induced SrfItga8 homozygous male (b) and female (c) mesenteric arteries. n = 3 independent animals for each condition. Error bars represented by mean ± standard deviation; p-values determined by two-tailed, unpaired Student’s t-test.

Source data

Supplementary information

Supplementary Information

Supplementary Figures 1-6, Supplementary Figure Legends

Reporting Summary

Supplementary Table

Supplementary Tables 1–6. Supplementary Table 1. Floxed alleles and associated SMC Cre drivers. Supplementary Table 2. Differentially expressed genes in aorta of wild-type versus Cre-containing mice. Supplementary Table 3. Mouse models. Supplementary Table 4. Oligonucleotide primers and single guide RNAs for mapping Myh11-Cre transgene. Supplementary Table 5. Antibodies. Supplementary Table 6. Cell lines.

Supplementary Data 1

Source data for Supplementary Fig. 4

Source data

Source Data Fig. 2

Unprocessed western blot

Source Data Fig. 2

Raw data and statistics

Source Data Fig. 3

Unprocessed western blot

Source Data Fig. 3

Raw data and statistics

Source Data Fig. 4

Raw data and statistics

Source Data Fig. 5

Raw data and statistics

Source Data Fig. 6

Raw data and statistics

Source Data Extended Data Fig. 2

Unprocessed western blot

Source Data Extended Data Fig. 2

Raw data and statistics

Source Data Extended Data Fig. 3

Raw data and statistics

Source Data Extended Data Fig. 4

Unprocessed western blot

Source Data Extended Data Fig. 7

Raw data and statistics

Source Data Extended Data Fig. 8

Unprocessed western blot

Source Data Extended Data Fig. 8

Raw data and statistics

Source Data Extended Data Fig. 9

Unprocessed western blot

Source Data Extended Data Fig. 9

Raw data and statistics

Source Data Extended Data Fig. 10

Unprocessed western blot

Source Data Extended Data Fig. 10

Raw data and statistics

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

Warthi, G., Faulkner, J.L., Doja, J. et al. Generation and comparative analysis of an Itga8-CreERT2 mouse with preferential activity in vascular smooth muscle cells. Nat Cardiovasc Res 1, 1084–1100 (2022). https://doi.org/10.1038/s44161-022-00162-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s44161-022-00162-1

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing