Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-qsmjn Total loading time: 0 Render date: 2024-04-19T23:58:24.567Z Has data issue: false hasContentIssue false

35 - Stem cells for vascular engineering

from Part V - Animal models and clinical applications

Published online by Cambridge University Press:  05 February 2015

Y. Eugene Chen
Affiliation:
University of Michigan
Changqing Xie
Affiliation:
University of Michigan
Bo Yang
Affiliation:
University of Michigan
Peter X. Ma
Affiliation:
University of Michigan, Ann Arbor
Get access

Summary

Introduction

Normal blood vessels consist of three layers, including the tunica intima, the tunica media, and the tunica adventitia [1]. Each structural layer consists of distinct cell and matrix types. A monolayer of endothelial cells (ECs) lines the lumen of blood vessels to provide a continuous, selectively permeable, hemo-compatible blood-contacting surface. Meanwhile, ECs play a key role in various physiological and pathological processes including blood supply, metabolic homeostasis, immune cell trafficking, and inflammation [2]. Vascular smooth muscle cells (VSMCs) and pericytes cover the outside of the endothelium, protect the fragile channels from rupture, and contribute to the contraction and relaxation of the vessels [3]. The vascular wall extracellular matrix (ECM) is composed of structural proteins such as collagen and elastin and adhesion proteins such as fibronectin and laminin that determine mechanical strength, cell response, and ultimately hierarchical tissue organization [4]. An intact and functioning vasculature is crucial in order to maintain homeostasis and provides necessary nutrients and oxygen exchange to all parts of the body.

Diseases that affect the integrity of blood vessels lead to serious and often deadly outcomes. Vascular diseases are the major causes of morbidity around the world [5]. At present, approximately 12 million people in the USA are affected by peripheral vascular disease, but only approximately one in four of them has been diagnosed and is receiving treatment [6]. EC and SMC pathology has been implicated in various vascular diseases [7–10]. Current important therapeutic options for vascular disease incorporate the surgical implantation of stents or grafts and greatly reconstruct impaired vascular function to drain downstream tissues and organs. However, the implanted grafts may incompletely recover the functional integrity of the vasculature. In addition, these therapeutic methods neither provide long-lasting solutions nor prevent damage to downstream tissues and organs [11]. In this scenario, cell-based engineered vessel grafts may offer the opportunity to permanently and effectively treat many vascular diseases [11]. The regeneration of some or all of the vessel layers and their original properties may provide potentially functional vascular grafts. The use of autologous bypass grafts, including saphenous vein, internal mammary artery, and radial artery bypass grafts, remains an important therapeutic option for the treatment of coronary artery disease. However, many patients do not have a vessel suitable for use because of concomitant vascular disease, amputation, or previous harvest, and hence artificial grafts must also be used [12].

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2014

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Humphrey, J. D. and Na, S. 2002. Elastodynamics and arterial wall stress. Ann. Biomed. Eng., 30(4), 509–23.CrossRefGoogle ScholarPubMed
Danese, S., Dejana, E. and Fiocchi, C. 2007. Immune regulation by microvascular endothelial cells: directing innate and adaptive immunity, coagulation, and inflammation. J. Immunol., 178(10), 6017–22.CrossRefGoogle Scholar
Jain, R. K. 2003. Molecular regulation of vessel maturation. Nature Med., 9(6), 685–93.CrossRefGoogle ScholarPubMed
Ruoslahti, E. and Engvall, E. 1997. Integrins and vascular extracellular matrix assembly. J. Clin. Invest., 99(6), 1149–52.CrossRefGoogle ScholarPubMed
Roger, V. L., Go, A. S., Lloyd-Jones, D. M. et al. 2011. Heart disease and stroke statistics – 2011 update: a report from the American Heart Association. Circulation, 123(4), e18–209.CrossRefGoogle ScholarPubMed
Hirsch, A. T., Criqui, M. H., Treat-Jacobson, D. et al. 2001. Peripheral arterial disease detection, awareness, and treatment in primary care. JAMA, 286(11), 1317–24.CrossRefGoogle ScholarPubMed
Cai, H. and Harrison, D. G. 2000. Endothelial dysfunction in cardiovascular diseases: the role of oxidant stress. Circ. Res., 87(10), 840–4.CrossRefGoogle ScholarPubMed
Celermajer, D. S., Sorensen, K. E., Gooch, V. M. et al. 1992. Non-invasive detection of endothelial dysfunction in children and adults at risk of atherosclerosis. Lancet, 340(8828), 1111–15.CrossRefGoogle ScholarPubMed
Owens, G. K., Kumar, M. S. and Wamhoff, B. R. 2004. Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol. Rev., 84(3), 767–801.CrossRefGoogle ScholarPubMed
Ross, R. 1999. Atherosclerosis – an inflammatory disease. N. Engl. J. Med., 340(2), 115–26.CrossRefGoogle ScholarPubMed
Sun, G. and Gerecht, S. 2009. Vascular regeneration: engineering the stem cell microenvironment. Regen. Med., 4(3), 435–47.CrossRefGoogle ScholarPubMed
Arrigoni, C., Camozzi, D. and Remuzzi, A. 2006. Vascular tissue engineering. Cell Transplant., 15(Suppl. 1), S119–25.CrossRefGoogle ScholarPubMed
Langer, R. and Vacanti, J. P. 1993. Tissue engineering. Science, 260(5110), 920–6.CrossRefGoogle ScholarPubMed
Dahl, S. L., Kypson, A. P., Lawson, J. H. et al. 2011. Readily available tissue-engineered vascular grafts. Sci. Transl. Med., 3(68), 68–9.CrossRefGoogle ScholarPubMed
Owens, G. K. 1995. Regulation of differentiation of vascular smooth muscle cells. Physiol. Rev., 75(3), 487–517.CrossRefGoogle ScholarPubMed
Kadner, A., Hoerstrup, S. P., Zund, G. et al. 2002. A new source for cardiovascular tissue engineering: human bone marrow stromal cells. Eur. J. Cardiothorac. Surg., 21(6), 1055–60.CrossRefGoogle ScholarPubMed
Lavik, E. and Langer, R. 2004. Tissue engineering: current state and perspectives. Appl. Microbiol. Biotechnol., 65(1), 1–8.CrossRefGoogle Scholar
Pankajakshan, D. and Agrawal, D. K. 2010. Scaffolds in tissue engineering of blood vessels. Can. J. Physiol. Pharmacol., 88(9), 855–73.CrossRefGoogle ScholarPubMed
2011. Stem cell basics: what are the similarities and differences between embryonic and adult stem cells? In Stem Cell Information. Bethesda, MD: National Institutes of Health, US Department of Health and Human Services, .
Risau, W., Sariola, H., Zerwes, H. G. et al. 1988. Vasculogenesis and angiogenesis in embryonic-stem-cell-derived embryoid bodies. Development, 102(3), 471–8.Google ScholarPubMed
Rafii, S. and Lyden, D. 2003. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nature Med., 9(6), 702–12.CrossRefGoogle ScholarPubMed
Choi, K., Kennedy, M., Kazarov, A., Papadimitriou, J. C. and Keller, G. 1998. A common precursor for hematopoietic and endothelial cells. Development, 125(4), 725–32.Google Scholar
Vittet, D., Prandini, M. H., Berthier, R. et al. 1996. Embryonic stem cells differentiate in vitro to endothelial cells through successive maturation steps. Blood, 88(9), 3424–31.Google ScholarPubMed
Choi, K., Chung, Y. S. and Zhang, W. J. 2005. Hematopoietic and endothelial development of mouse embryonic stem cells in culture. Methods Molec. Med., 105, 359–68.Google ScholarPubMed
Levenberg, S., Golub, J. S., Amit, M., Itskovitz-Eldor, J. and Langer, R. 2002. Endothelial cells derived from human embryonic stem cells. Proc. Nat. Acad. Sci. USA, 99(7), 4391–6.CrossRefGoogle Scholar
Gerecht-Nir, S., Dazard, J. E., Golan-Mashiach, M. et al. 2005. Vascular gene expression and phenotypic correlation during differentiation of human embryonic stem cells. Dev. Dyn., 232(2), 487–97.CrossRefGoogle ScholarPubMed
Wang, L., Li, L., Shojaei, F. et al. 2004. Endothelial and hematopoietic cell fate of human embryonic stem cells originates from primitive endothelium with hemangioblastic properties. Immunity, 21(1), 31–41.CrossRefGoogle ScholarPubMed
Ferreira, L. S., Gerecht, S., Shieh, H. F. et al. 2007. Vascular progenitor cells isolated from human embryonic stem cells give rise to endothelial and smooth muscle like cells and form vascular networks in vivo. Circ. Res., 101(3), 286–94.CrossRefGoogle ScholarPubMed
Lu, S. J., Ivanova, Y., Feng, Q., Luo, C. and Lanza, R. 2009. Hemangioblasts from human embryonic stem cells generate multilayered blood vessels with functional smooth muscle cells. Regen. Med., 4(1), 37–47.CrossRefGoogle ScholarPubMed
Cho, S. W., Moon, S. H., Lee, S. H. et al. 2007. Improvement of postnatal neovascularization by human embryonic stem cell derived endothelial-like cell transplantation in a mouse model of hindlimb ischemia. Circulation, 116(21), 2409–19.CrossRefGoogle Scholar
Nourse, M. B., Halpin, D. E., Scatena, M. et al. 2010. VEGF induces differentiation of functional endothelium from human embryonic stem cells: implications for tissue engineering. Arterioscler. Thromb. Vasc. Biol., 30(1), 80–9.CrossRefGoogle ScholarPubMed
James, D., Nam, H. S., Seandel, M. et al. 2010. Expansion and maintenance of human embryonic stem cell-derived endothelial cells by TGFβ inhibition is Id1 dependent. Nature Biotechnol., 28(2), 161–6.CrossRefGoogle ScholarPubMed
Kaufman, D. S., Hanson, E. T., Lewis, R. L., Auerbach, R. and Thomson, J. A. 2001. Hematopoietic colony-forming cells derived from human embryonic stem cells. Proc. Nat. Acad. Sci. USA, 98(19), 10716–21.CrossRefGoogle ScholarPubMed
Vodyanik, M. A., Bork, J. A., Thomson, J. A. and Slukvin, I. I. 2005. Human embryonic stem cell-derived CD34+ cells: efficient production in the coculture with OP9 stromal cells and analysis of lymphohematopoietic potential. Blood, 105(2), 617–26.CrossRefGoogle ScholarPubMed
Wang, Z. Z., Au, P., Chen, T. et al. 2007. Endothelial cells derived from human embryonic stem cells form durable blood vessels in vivo. Nature Biotechnol., 25(3), 317–18.CrossRefGoogle ScholarPubMed
Zambidis, E. T., Peault, B., Park, T. S., Bunz, F. and Civin, C. I. 2005. Hematopoietic differentiation of human embryonic stem cells progresses through sequential hematoendothelial, primitive, and definitive stages resembling human yolk sac development. Blood, 106(3), 860–70.CrossRefGoogle ScholarPubMed
Vodyanik, M. A. and Slukvin, I. I. 2007. Hematoendothelial differentiation of human embryonic stem cells. In Current Protocols in Cell Biology, Chapter 23, Unit 23.6.
Sone, M., Itoh, H., Yamahara, K. et al. 2007. Pathway for differentiation of human embryonic stem cells to vascular cell components and their potential for vascular regeneration. Arterioscler. Thromb. Vasc. Biol., 27(10), 2127–34.CrossRefGoogle ScholarPubMed
Hill, K. L., Obrtlikova, P., Alvarez, D. F. et al. 2010. Human embryonic stem cell-derived vascular progenitor cells capable of endothelial and smooth muscle cell function. Exp. Hematol., 38(3), 246–57.CrossRefGoogle ScholarPubMed
Bai, H., Gao, Y., Arzigian, M. et al. 2010. BMP4 regulates vascular progenitor development in human embryonic stem cells through a Smad-dependent pathway. J. Cell Biochem., 109(2), 363–74.Google ScholarPubMed
Gerecht-Nir, S., Ziskind, A., Cohen, S. and Itskovitz-Eldor, J. 2003. Human embryonic stem cells as an in vitro model for human vascular development and the induction of vascular differentiation. Lab. Invest., 83(12), 1811–20.CrossRefGoogle Scholar
Goldman, O., Feraud, O., Boyer-Di Ponio, J. et al. 2009. A boost of BMP4 accelerates the commitment of human embryonic stem cells to the endothelial lineage. Stem Cells, 27(8), 1750–9.CrossRefGoogle ScholarPubMed
Woll, P. S., Morris, J. K., Painschab, M. S. et al. 2008. Wnt signaling promotes hematoendothelial cell development from human embryonic stem cells. Blood, 111(1), 122–31.CrossRefGoogle ScholarPubMed
Vo, E., Hanjaya-Putra, D., Zha, Y., Kusuma, S. and Gerecht, S. 2010. Smooth-muscle-like cells derived from human embryonic stem cells support and augment cord-like structures in vitro. Stem Cell Rev., 6(2), 237–47.CrossRefGoogle ScholarPubMed
Huang, H., Zhao, X., Chen, L. et al. 2006. Differentiation of human embryonic stem cells into smooth muscle cells in adherent monolayer culture. Biochem. Biophys. Res. Commun., 351(2), 321–7.CrossRefGoogle ScholarPubMed
Xie, C. Q., Zhang, J., Villacorta, L. et al. 2007. A highly efficient method to differentiate smooth muscle cells from human embryonic stem cells. Arterioscler. Thromb. Vasc. Biol., 27(12), e311–12.CrossRefGoogle ScholarPubMed
Cheung, C., Bernardo, A. S., Trotter, M. W., Pedersen, R. A. and Sinha, S. 2012. Generation of human vascular smooth muscle subtypes provides insight into embryological origin-dependent disease susceptibility. Nature Biotechnol., 30(2), 165–73.CrossRefGoogle ScholarPubMed
Takahashi, K., Tanabe, K., Ohnuki, M. et al. 2007. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell, 131(5), 861–72.CrossRefGoogle ScholarPubMed
Takahashi, K. and Yamanaka, S. 2006. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell, 126(4), 663–76.CrossRefGoogle ScholarPubMed
Dimos, J. T., Rodolfa, K. T., Niakan, K. K. et al. 2008. Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science, 321(5893), 1218–21.CrossRefGoogle ScholarPubMed
Park, I. H., Arora, N., Huo, H. et al. 2008. Disease-specific induced pluripotent stem cells. Cell, 134(5), 877–86.CrossRefGoogle ScholarPubMed
Soldner, F., Laganiere, J., Cheng, A. W. et al. 2011. Generation of isogenic pluripotent stem cells differing exclusively at two early onset Parkinson point mutations. Cell, 146(2), 318–31.CrossRefGoogle ScholarPubMed
Choi, K. D., Yu, J., Smuga-Otto, K. et al. 2009. Hematopoietic and endothelial differentiation of human induced pluripotent stem cells. Stem Cells, 27(3), 559–67.CrossRefGoogle ScholarPubMed
Feng, Q., Lu, S. J., Klimanskaya, I. et al. 2010. Hemangioblastic derivatives from human induced pluripotent stem cells exhibit limited expansion and early senescence. Stem Cells, 28(4), 704–12.CrossRefGoogle ScholarPubMed
Homma, K., Sone, M., Taura, D. et al. 2010. Sirt1 plays an important role in mediating greater functionality of human ES/iPS-derived vascular endothelial cells. Atherosclerosis, 212(1), 42–7.CrossRefGoogle ScholarPubMed
Taura, D., Sone, M., Homma, K. et al. 2009. Induction and isolation of vascular cells from human induced pluripotent stem cells – brief report. Arterioscler. Thromb. Vasc. Biol., 29(7), 1100–3.CrossRefGoogle ScholarPubMed
Rufaihah, A. J., Huang, N. F., Jame, S. et al. 2011. Endothelial cells derived from human iPSCs increase capillary density and improve perfusion in a mouse model of peripheral arterial disease. Arterioscler. Thromb. Vasc. Biol., 31(11), e72–9.CrossRefGoogle Scholar
Xie, C. Q., Huang, H., Wei, S. et al. 2009. A comparison of murine smooth muscle cells generated from embryonic versus induced pluripotent stem cells. Stem Cells Dev., 18(5), 741–8.CrossRefGoogle ScholarPubMed
Lee, T. H., Song, S. H., Kim, K. L. et al. 2010. Functional recapitulation of smooth muscle cells via induced pluripotent stem cells from human aortic smooth muscle cells. Circ Res., 106(1), 120–8.CrossRefGoogle ScholarPubMed
Xie, C., Hu, J., Ma, H. et al. 2011. Three-dimensional growth of iPS cell-derived smooth muscle cells on nanofibrous scaffolds. Biomaterials, 32(19), 4369–75.CrossRefGoogle ScholarPubMed
Korbling, M. and Estrov, Z. 2003. Adult stem cells for tissue repair – a new therapeutic concept?N. Engl. J. Med., 349(6), 570–82.CrossRefGoogle ScholarPubMed
Bianco, P., Riminucci, M., Gronthos, S. and Robey, P. G. 2001. Bone marrow stromal stem cells: nature, biology, and potential applications. Stem Cells, 19(3), 180–92.CrossRefGoogle ScholarPubMed
Asahara, T., Murohara, T., Sullivan, A. et al. 1997. Isolation of putative progenitor endothelial cells for angiogenesis. Science, 275(5302), 964–7.CrossRefGoogle ScholarPubMed
Jackson, K. A., Majka, S. M., Wang, H. et al. 2001. Regeneration of ischemic cardiac muscle and vascular endothelium by adult stem cells. J. Clin. Invest., 107(11), 1395–402.CrossRefGoogle ScholarPubMed
Kamihata, H., Matsubara, H., Nishiue, T. et al. 2001. Implantation of bone marrow mononuclear cells into ischemic myocardium enhances collateral perfusion and regional function via side supply of angioblasts, angiogenic ligands, and cytokines. Circulation, 104(9), 1046–52.CrossRefGoogle ScholarPubMed
Kocher, A. A., Schuster, M. D., Szabolcs, M. J. et al. 2001. Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nature Med., 7(4), 430–6.CrossRefGoogle ScholarPubMed
Orlic, D., Kajstura, J., Chimenti, S. et al. 2001. Bone marrow cells regenerate infarcted myocardium. Nature, 410(6829), 701–5.CrossRefGoogle ScholarPubMed
Orlic, D., Kajstura, J., Chimenti, S. et al. 2001. Mobilized bone marrow cells repair the infarcted heart, improving function and survival. Proc. Nat. Acad. Sci. USA, 98(18), 10344–9.CrossRefGoogle Scholar
Reyes, M., Dudek, A., Jahagirdar, B. et al. 2002. Origin of endothelial progenitors in human postnatal bone marrow. J. Clin. Invest., 109(3), 337–46.CrossRefGoogle ScholarPubMed
Shintani, S., Murohara, T., Ikeda, H. et al. 2001. Augmentation of postnatal neovascularization with autologous bone marrow transplantation. Circulation, 103(6), 897–903.CrossRefGoogle ScholarPubMed
Takahashi, T., Kalka, C., Masuda, H. et al. 1999. Ischemia- and cytokine-induced mobilization of bone marrow-derived endothelial progenitor cells for neovascularization. Nature Med., 5(4), 434–8.CrossRefGoogle ScholarPubMed
Iba, O., Matsubara, H., Nozawa, Y. et al. 2002. Angiogenesis by implantation of peripheral blood mononuclear cells and platelets into ischemic limbs. Circulation, 106(15), 2019–25.CrossRefGoogle ScholarPubMed
Pesce, M., Orlandi, A., Iachininoto, M. G. et al. 2003. Myoendothelial differentiation of human umbilical cord blood-derived stem cells in ischemic limb tissues. Circ. Res., 93(5), e51–62.CrossRefGoogle ScholarPubMed
Beltrami, A. P., Barlucchi, L., Torella, D. et al. 2003. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell, 114(6), 763–76.CrossRefGoogle ScholarPubMed
Campagnolo, P., Cesselli, D., Al Haj Zen, A. et al. 2010. Human adult vena saphena contains perivascular progenitor cells endowed with clonogenic and proangiogenic potential. Circulation, 121(15), 1735–45.CrossRefGoogle ScholarPubMed
Werner, N., Junk, S., Laufs, U. et al. 2003. Intravenous transfusion of endothelial progenitor cells reduces neointima formation after vascular injury. Circ. Res., 93(2), e17–24.CrossRefGoogle ScholarPubMed
Lin, Y., Chen, X., Yan, Z. et al. 2006. Multilineage differentiation of adipose-derived stromal cells from GFP transgenic mice. Molec. Cell Biochem., 285(1–2), 69–78.CrossRefGoogle Scholar
Planat-Benard, V., Silvestre, J. S., Cousin, B. et al. 2004. Plasticity of human adipose lineage cells toward endothelial cells: physiological and therapeutic perspectives. Circulation, 109(5), 656–63.CrossRefGoogle ScholarPubMed
Friedenstein, A. J., Piatetzky, S. and Petrakova, K. V. 1966. Osteogenesis in transplants of bone marrow cells. J. Embryol. Exp. Morphol., 16(3), 381–90.Google Scholar
Kern, S., Eichler, H., Stoeve, J., Kluter, H. and Bieback, K. 2006. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells, 24(5), 1294–301.CrossRefGoogle ScholarPubMed
Pasquinelli, G., Pacilli, A., Alviano, F. et al. 2010. Multidistrict human mesenchymal vascular cells: pluripotency and stemness characteristics. Cytotherapy, 12(3), 275–87.CrossRefGoogle ScholarPubMed
Pasquinelli, G., Tazzari, P. L., Vaselli, C. et al. 2007. Thoracic aortas from multiorgan donors are suitable for obtaining resident angiogenic mesenchymal stromal cells. Stem Cells, 25(7), 1627–34.CrossRefGoogle ScholarPubMed
Liu, J. Y., Peng, H. F., Gopinath, S., Tian, J. and Andreadis, S. T. 2010. Derivation of functional smooth muscle cells from multipotent human hair follicle mesenchymal stem cells. Tissue Eng. Part A, 16(8), 2553–64.CrossRefGoogle ScholarPubMed
Parolini, O., Alviano, F., Bagnara, G. P. et al. 2008. Concise review: isolation and characterization of cells from human term placenta: outcome of the First International Workshop on Placenta Derived Stem Cells. Stem Cells, 26(2), 300–11.CrossRefGoogle ScholarPubMed
Erices, A., Conget, P. and Minguell, J. J. 2000. Mesenchymal progenitor cells in human umbilical cord blood. Br. J. Haematol., 109(1), 235–42.CrossRefGoogle ScholarPubMed
Goodwin, H. S., Bicknese, A. R., Chien, S. N. et al. 2001. Multilineage differentiation activity by cells isolated from umbilical cord blood: expression of bone, fat, and neural markers. Biol. Blood Marrow Transplant., 7(11), 581–8.CrossRefGoogle ScholarPubMed
Mareschi, K., Biasin, E., Piacibello, W. et al. 2001. Isolation of human mesenchymal stem cells: bone marrow versus umbilical cord blood. Haematologica, 86(10), 1099–100.Google ScholarPubMed
Lee, O. K., Kuo, T. K., Chen, W. M. et al. 2004. Isolation of multipotent mesenchymal stem cells from umbilical cord blood. Blood, 103(5), 1669–75.CrossRefGoogle ScholarPubMed
Zuk, P. A., Zhu, M., Mizuno, H. et al. 2001. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng., 7(2), 211–28.CrossRefGoogle ScholarPubMed
Covas, D. T., Siufi, J. L., Silva, A. R. and Orellana, M. D. 2003. Isolation and culture of umbilical vein mesenchymal stem cells. Braz. J. Med. Biol. Res., 36(9), 1179–83.CrossRefGoogle ScholarPubMed
Romanov, Y. A., Svintsitskaya, V. A. and Smirnov, V. N. 2003. Searching for alternative sources of postnatal human mesenchymal stem cells: candidate MSC-like cells from umbilical cord. Stem Cells, 21(1), 105–10.CrossRefGoogle ScholarPubMed
Fernandez, M., Simon, V., Herrera, G. et al. 1997. Detection of stromal cells in peripheral blood progenitor cell collections from breast cancer patients. Bone Marrow Transplant., 20(4), 265–71.CrossRefGoogle ScholarPubMed
Kuramochi, Y., Fukazawa, R., Migita, M. et al. 2003. Cardiomyocyte regeneration from circulating bone marrow cells in mice. Pediatr. Res., 54(3), 319–25.CrossRefGoogle ScholarPubMed
Zvaifler, N. J., Marinova-Mutafchieva, L., Adams, G. et al. 2000. Mesenchymal precursor cells in the blood of normal individuals. Arthritis Res., 2(6), 477–88.CrossRefGoogle ScholarPubMed
Davani, S., Marandin, A., Mersin, N. et al. 2003. Mesenchymal progenitor cells differentiate into an endothelial phenotype, enhance vascular density, and improve heart function in a rat cellular cardiomyoplasty model. Circulation, 108(Suppl. 1), II253–8.CrossRefGoogle Scholar
Gojo, S., Gojo, N., Takeda, Y. et al. 2003. In vivo cardiovasculogenesis by direct injection of isolated adult mesenchymal stem cells. Exp. Cell. Res., 288(1), 51–9.CrossRefGoogle ScholarPubMed
Oswald, J., Boxberger, S., Jorgensen, B. et al. 2004. Mesenchymal stem cells can be differentiated into endothelial cells in vitro. Stem Cells, 22(3), 377–84.CrossRefGoogle ScholarPubMed
Dong, J. D., Gu, Y. Q., Li, C. M. et al. 2009. Response of mesenchymal stem cells to shear stress in tissue-engineered vascular grafts. Acta Pharmacol. Sin., 30(5), 530–6.CrossRefGoogle ScholarPubMed
Shin’oka, T., Matsumura, G., Hibino, N. et al. 2005. Midterm clinical result of tissue-engineered vascular autografts seeded with autologous bone marrow cells. J. Thorac. Cardiovasc. Surg., 129(6), 1330–8.CrossRefGoogle ScholarPubMed
Galmiche, M. C., Koteliansky, V. E., Briere, J., Herve, P. and Charbord, P. 1993. Stromal cells from human long-term marrow cultures are mesenchymal cells that differentiate following a vascular smooth muscle differentiation pathway. Blood, 82(1), 66–76.Google ScholarPubMed
Seruya, M., Shah, A., Pedrotty, D. et al. 2004. Clonal population of adult stem cells: life span and differentiation potential. Cell Transplant., 13(2), 93–101.CrossRefGoogle ScholarPubMed
Kinner, B., Zaleskas, J. M. and Spector, M. 2002. Regulation of smooth muscle actin expression and contraction in adult human mesenchymal stem cells. Exp. Cell Res., 278(1), 72–83.CrossRefGoogle ScholarPubMed
Fierro, F. A., Kalomoiris, S., Sondergaard, C. S. and Nolta, J. A. 2011. Effects on proliferation and differentiation of multipotent bone marrow stromal cells engineered to express growth factors for combined cell and gene therapy. Stem Cells, 29(11), 1727–37.CrossRefGoogle ScholarPubMed
Ball, S. G., Shuttleworth, A. C. and Kielty, C. M. 2004. Direct cell contact influences bone marrow mesenchymal stem cell fate. Int. J. Biochem. Cell Biol., 36(4), 714–27.CrossRefGoogle ScholarPubMed
Hamilton, D. W., Maul, T. M. and Vorp, D. A. 2004. Characterization of the response of bone marrow-derived progenitor cells to cyclic strain: implications for vascular tissue-engineering applications. Tissue Eng., 10(3–4), 361–9.CrossRefGoogle ScholarPubMed
Gong, Z. and Niklason, L. E. 2008. Small-diameter human vessel wall engineered from bone marrow-derived mesenchymal stem cells (hMSCs). FASEB J., 22(6), 1635–48.CrossRefGoogle Scholar
Cho, S. W., Lim, S. H., Kim, I. K. et al. 2005. Small-diameter blood vessels engineered with bone marrow-derived cells. Ann. Surg., 241(3), 506–15.CrossRefGoogle ScholarPubMed
Zhao, Y., Zhang, S., Zhou, J. et al. 2010. The development of a tissue-engineered artery using decellularized scaffold and autologous ovine mesenchymal stem cells. Biomaterials, 31(2), 296–307.CrossRefGoogle ScholarPubMed
Caplan, A. I. 2009. Why are MSCs therapeutic? New data: new insight. J. Pathol., 217(2), 318–24.CrossRefGoogle ScholarPubMed
Peichev, M., Naiyer, A. J., Pereira, D. et al. 2000. Expression of VEGFR-2 and AC133 by circulating human CD34(+) cells identifies a population of functional endothelial precursors. Blood, 95(3), 952–8.Google Scholar
Schmid, M. C. and Varner, J. A. 2009. Circulating endothelial progenitor cells. Methods Molec. Biol., 467, 139–55.CrossRefGoogle ScholarPubMed
Miranville, A., Heeschen, C., Sengenes, C. et al. 2004. Improvement of postnatal neovascularization by human adipose tissue-derived stem cells. Circulation, 110(3), 349–55.CrossRefGoogle ScholarPubMed
Xue, S., Zhang, H. T., Zhang, P. et al. 2010. Functional endothelial progenitor cells derived from adipose tissue show beneficial effect on cell therapy of traumatic brain injury. Neurosci. Lett., 473(3), 186–91.CrossRefGoogle ScholarPubMed
Ingram, D. A., Mead, L. E., Moore, D. B. et al. 2005. Vessel wall-derived endothelial cells rapidly proliferate because they contain a complete hierarchy of endothelial progenitor cells. Blood, 105(7), 2783–6.CrossRefGoogle ScholarPubMed
Murohara, T., Ikeda, H., Duan, J. et al. 2000. Transplanted cord blood-derived endothelial precursor cells augment postnatal neovascularization. J. Clin. Invest., 105(11), 1527–36.CrossRefGoogle ScholarPubMed
Heeschen, C., Aicher, A., Lehmann, R. et al. 2003. Erythropoietin is a potent physiologic stimulus for endothelial progenitor cell mobilization. Blood, 102(4), 1340–6.CrossRefGoogle ScholarPubMed
Strehlow, K., Werner, N., Berweiler, J. et al. 2003. Estrogen increases bone marrow-derived endothelial progenitor cell production and diminishes neointima formation. Circulation, 107(24), 3059–65.CrossRefGoogle ScholarPubMed
Cherqui, S., Kurian, S. M., Schussler, O. et al. 2006. Isolation and angiogenesis by endothelial progenitors in the fetal liver. Stem Cells, 24(1), 44–54.CrossRefGoogle ScholarPubMed
Kordes, C., Sawitza, I., Muller-Marbach, A. et al. 2007. CD133+ hepatic stellate cells are progenitor cells. Biochem. Biophys. Res. Commun., 352(2), 410–17.CrossRefGoogle ScholarPubMed
Messina, E., De Angelis, L., Frati, G. et al. 2004. Isolation and expansion of adult cardiac stem cells from human and murine heart. Circ. Res., 95(9), 911–21.CrossRefGoogle ScholarPubMed
Adams, V., Lenk, K., Linke, A. et al. 2004. Increase of circulating endothelial progenitor cells in patients with coronary artery disease after exercise-induced ischemia. Arterioscler. Thromb. Vasc. Biol., 24(4), 684–90.CrossRefGoogle ScholarPubMed
Vasa, M., Fichtlscherer, S., Adler, K. et al. 2001. Increase in circulating endothelial progenitor cells by statin therapy in patients with stable coronary artery disease. Circulation, 103(24), 2885–90.CrossRefGoogle ScholarPubMed
Vasa, M., Fichtlscherer, S., Aicher, A. et al. 2001. Number and migratory activity of circulating endothelial progenitor cells inversely correlate with risk factors for coronary artery disease. Circ. Res., 89(1), E1–7.CrossRefGoogle ScholarPubMed
Al Attar, N., Carrion, C., Ghostine, S. et al. 2003. Long-term (1 year) functional and histological results of autologous skeletal muscle cells transplantation in rat. Cardiovasc. Res., 58(1), 142–8.CrossRefGoogle ScholarPubMed
Werner, N., Priller, J., Laufs, U. et al. 2002. Bone marrow-derived progenitor cells modulate vascular reendothelialization and neointimal formation: effect of 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibition. Arterioscler. Thromb. Vasc. Biol., 22(10), 1567–72.CrossRefGoogle ScholarPubMed
Bahlmann, F. H., De Groot, K., Spandau, J. M. et al. 2004. Erythropoietin regulates endothelial progenitor cells. Blood, 103(3), 921–6.CrossRefGoogle ScholarPubMed
Masuda, H. and Asahara, T. 2003. Post-natal endothelial progenitor cells for neovascularization in tissue regeneration. Cardiovasc. Res., 58(2), 390–8.CrossRefGoogle ScholarPubMed
Khakoo, A. Y. and Finkel, T. 2005. Endothelial progenitor cells. Ann. Rev. Med., 56, 79–101.CrossRefGoogle ScholarPubMed
Kalka, C., Masuda, H., Takahashi, T. et al. 2000. Transplantation of ex vivo expanded endothelial progenitor cells for therapeutic neovascularization. Proc. Nat. Acad. Sci. USA, 97(7), 3422–7.CrossRefGoogle ScholarPubMed
Tateishi-Yuyama, E., Matsubara, H., Murohara, T. et al. 2002. Therapeutic angiogenesis for patients with limb ischaemia by autologous transplantation of bone-marrow cells: a pilot study and a randomised controlled trial. Lancet, 360(9331), 427–35.CrossRefGoogle Scholar
Asahara, T., Masuda, H., Takahashi, T. et al. 1999. Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ. Res., 85(3), 221–8.CrossRefGoogle ScholarPubMed
Bhattacharya, V., McSweeney, P. A., Shi, Q. et al. 2000. Enhanced endothelialization and microvessel formation in polyester grafts seeded with CD34(+) bone marrow cells. Blood, 95(2), 581–5.Google ScholarPubMed
Shi, Q., Rafii, S., Wu, M. H. et al. 1998. Evidence for circulating bone marrow-derived endothelial cells. Blood, 92(2), 362–7.Google ScholarPubMed
Barrilleaux, B., Phinney, D. G., Prockop, D. J. and O’Connor, K. C. 2006. Ex vivo engineering of living tissues with adult stem cells. Tissue Eng., 12(11), 3007–19.CrossRefGoogle ScholarPubMed
Isenberg, B. C., Williams, C. and Tranquillo, R. T. 2006. Small-diameter artificial arteries engineered in vitro. Circ. Res., 98(1), 25–35.CrossRefGoogle ScholarPubMed
Sales, K. M., Salacinski, H. J., Alobaid, N. et al. 2005. Advancing vascular tissue engineering: the role of stem cell technology. Trends Biotechnol., 23(9), 461–7.CrossRefGoogle ScholarPubMed
Wijelath, E. S., Rahman, S., Murray, J. et al. 2004. Fibronectin promotes VEGF-induced CD34 cell differentiation into endothelial cells. J. Vasc. Surg., 39(3), 655–60.CrossRefGoogle ScholarPubMed
Kanayasu-Toyoda, T., Yamaguchi, T., Oshizawa, T. and Hayakawa, T. 2003. CD31 (PECAM-1)-bright cells derived from AC133-positive cells in human peripheral blood as endothelial-precursor cells. J. Cell Physiol., 195(1), 119–29.CrossRefGoogle ScholarPubMed
Asahara, T., Takahashi, T., Masuda, H. et al. 1999. VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. EMBO J., 18(14), 3964–72.CrossRefGoogle ScholarPubMed
Gehling, U. M., Ergun, S., Schumacher, U. et al. 2000. In vitro differentiation of endothelial cells from AC133-positive progenitor cells. Blood, 95(10), 3106–12.Google ScholarPubMed
Rehman, J., Li, J., Orschell, C. M. and March, K. L. 2003. Peripheral blood “endothelial progenitor cells” are derived from monocyte/macrophages and secrete angiogenic growth factors. Circulation, 107(8), 1164–9.CrossRefGoogle ScholarPubMed
Yamamoto, K., Takahashi, T., Asahara, T. et al. 2003. Proliferation, differentiation, and tube formation by endothelial progenitor cells in response to shear stress. J. Appl. Physiol., 95(5), 2081–8.CrossRefGoogle ScholarPubMed
Dvorin, E. L., Wylie-Sears, J., Kaushal, S., Martin, D. P. and Bischoff, J. 2003. Quantitative evaluation of endothelial progenitors and cardiac valve endothelial cells: proliferation and differentiation on poly-glycolic acid/poly-4-hydroxybutyrate scaffold in response to vascular endothelial growth factor and transforming growth factor beta1. Tissue Eng., 9(3), 487–93.CrossRefGoogle ScholarPubMed
Matsumura, G., Miyagawa-Tomita, S., Shin’oka, T., Ikada, Y. and Kurosawa, H. 2003. First evidence that bone marrow cells contribute to the construction of tissue-engineered vascular autografts in vivo. Circulation, 108(14), 1729–34.CrossRefGoogle ScholarPubMed
Wu, X., Rabkin-Aikawa, E., Guleserian, K. J. et al. 2004. Tissue-engineered microvessels on three-dimensional biodegradable scaffolds using human endothelial progenitor cells. Am. J. Physiol. Heart Circ. Physiol., 287(2), H480–7.CrossRefGoogle ScholarPubMed
Aper, T., Schmidt, A., Duchrow, M. and Bruch, H. P. 2007. Autologous blood vessels engineered from peripheral blood sample. Eur. J. Vasc. Endovasc. Surg., 33(1), 33–9.CrossRefGoogle ScholarPubMed
Schmidt, D., Breymann, C., Weber, A. et al. 2004. Umbilical cord blood derived endothelial progenitor cells for tissue engineering of vascular grafts. Ann. Thorac. Surg., 78(6), 2094–8.CrossRefGoogle ScholarPubMed
Kaushal, S., Amiel, G. E., Guleserian, K. J. et al. 2001. Functional small-diameter neovessels created using endothelial progenitor cells expanded ex vivo. Nature Med., 7(9), 1035–40.CrossRefGoogle ScholarPubMed
Li, X., Tjwa, M., Moons, L. et al. 2005. Revascularization of ischemic tissues by PDGF-CC via effects on endothelial cells and their progenitors. J. Clin. Invest., 115(1), 118–27.CrossRefGoogle ScholarPubMed
Miyata, T., Iizasa, H., Sai, Y. et al. 2005. Platelet-derived growth factor-BB (PDGF-BB) induces differentiation of bone marrow endothelial progenitor cell-derived cell line TR-BME2 into mural cells, and changes the phenotype. J. Cell Physiol., 204(3), 948–55.CrossRefGoogle ScholarPubMed
Diez, M., Musri, M. M., Ferrer, E., Barbera, J. A. and Peinado, V. I. 2010. Endothelial progenitor cells undergo an endothelial-to-mesenchymal transition-like process mediated by TGFβRI. Cardiovasc. Res., 88(3), 502–11.CrossRefGoogle ScholarPubMed
Diez, M., Barbera, J. A., Ferrer, E. et al. 2007. Plasticity of CD133+ cells: role in pulmonary vascular remodeling. Cardiovasc. Res., 76(3), 517–27.CrossRefGoogle ScholarPubMed
Katz, A. J., Tholpady, A., Tholpady, S. S., Shang, H. and Ogle, R. C. 2005. Cell surface and transcriptional characterization of human adipose-derived adherent stromal (hADAS) cells. Stem Cells, 23(3), 412–23.CrossRefGoogle ScholarPubMed
Zuk, P. A., Zhu, M., Ashjian, P. et al. 2000. Human adipose tissue is a source of multipotent stem cells. Molec. Biol. Cell, 13(12), 4279–95.CrossRefGoogle Scholar
Gronthos, S., Franklin, D. M., Leddy, H. A. et al. 2001. Surface protein characterization of human adipose tissue-derived stromal cells. J. Cell Physiol., 189(1), 54–63.CrossRefGoogle ScholarPubMed
Musina, R. A., Bekchanova, E. S. and Sukhikh, G. T. 2005. Comparison of mesenchymal stem cells obtained from different human tissues. Bull. Exp. Biol. Med., 139(4), 504–9.CrossRefGoogle ScholarPubMed
Romanov, Y. A., Darevskaya, A. N., Merzlikina, N. V. and Buravkova, L. B. 2005. Mesenchymal stem cells from human bone marrow and adipose tissue: isolation, characterization, and differentiation potentialities. Bull. Exp. Biol. Med., 140(1), 138–43.CrossRefGoogle ScholarPubMed
Cao, Y., Sun, Z., Liao, L. et al. 2005. Human adipose tissue-derived stem cells differentiate into endothelial cells in vitro and improve postnatal neovascularization in vivo. Biochem. Biophys. Res. Commun., 332(2), 370–9.CrossRefGoogle ScholarPubMed
Martinez-Estrada, O. M., Munoz-Santos, Y., Julve, J., Reina, M. and Vilaro, S. 2005. Human adipose tissue as a source of Flk-1+ cells: new method of differentiation and expansion. Cardiovasc. Res., 65(2), 328–33.CrossRefGoogle Scholar
Rodriguez, L. V., Alfonso, Z., Zhang, R. et al. 2006. Clonogenic multipotent stem cells in human adipose tissue differentiate into functional smooth muscle cells. Proc. Nat. Acad. Sci. USA, 103(32), 12167–72.CrossRefGoogle ScholarPubMed
Kim, Y. M., Jeon, E. S., Kim, M. R. et al. 2008. Angiotensin II-induced differentiation of adipose tissue-derived mesenchymal stem cells to smooth muscle-like cells. Int. J. Biochem. Cell Biol., 40(11), 2482–91.CrossRefGoogle ScholarPubMed
Wang, C., Yin, S., Cen, L. et al. 2010. Differentiation of adipose-derived stem cells into contractile smooth muscle cells induced by transforming growth factor-β1 and bone morphogenetic protein-4. Tissue Eng. Part A, 16(4), 1201–13.CrossRefGoogle ScholarPubMed
Harris, L. J., Abdollahi, H., Zhang, P. et al. 2011. Differentiation of adult stem cells into smooth muscle for vascular tissue engineering. J. Surg. Res., 168(2), 306–14.CrossRefGoogle ScholarPubMed
Wang, C., Cen, L., Yin, S. et al. 2010. A small diameter elastic blood vessel wall prepared under pulsatile conditions from polyglycolic acid mesh and smooth muscle cells differentiated from adipose-derived stem cells. Biomaterials, 31(4), 621–30.CrossRefGoogle ScholarPubMed
Huang, X. and Saint-Jeannet, J. P. 2004. Induction of the neural crest and the opportunities of life on the edge. Dev. Biol., 275(1), 1–11.CrossRefGoogle Scholar
Shakhova, O. and Sommer, L. 2010. Neural crest-derived stem cells. In The Stem Cell Research Community, editors. StemBook, , .
Wurmser, A. E., Nakashima, K., Summers, R. G. et al. 2004. Cell fusion-independent differentiation of neural stem cells to the endothelial lineage. Nature, 430(6997), 350–6.CrossRefGoogle ScholarPubMed
Majesky, M. W. 2007. Developmental basis of vascular smooth muscle diversity. Arterioscler. Thromb. Vasc. Biol., 27(6), 1248–58.CrossRefGoogle ScholarPubMed
Xie, C., Ritchie, R. P., Huang, H., Zhang, J. and Chen, Y. E. 2011. Smooth muscle cell differentiation in vitro: models and underlying molecular mechanisms. Arterioscler. Thromb. Vasc. Biol., 31(7), 1485–94.CrossRefGoogle ScholarPubMed
Fernandes, K. J., McKenzie, I. A., Mill, P. et al. 2004. A dermal niche for multipotent adult skin-derived precursor cells. Nature Cell Biol., 6(11), 1082–93.CrossRefGoogle ScholarPubMed
Fujiwara, H., Ferreira, M., Donati, G. et al. 2011. The basement membrane of hair follicle stem cells is a muscle cell niche. Cell, 144(4), 577–89.CrossRefGoogle ScholarPubMed
Steinbach, S. K., El-Mounayri, O., DaCosta, R. S. et al. 2011. Directed differentiation of skin-derived precursors into functional vascular smooth muscle cells. Arterioscler. Thromb. Vasc. Biol., 31(12), 2938–48.CrossRefGoogle ScholarPubMed
De Coppi, P., Bartsch, G., Siddiqui, M. M. et al. 2007. Isolation of amniotic stem cell lines with potential for therapy. Nature Biotechnol., 25(1), 100–6.CrossRefGoogle ScholarPubMed
Perin, L., Sedrakyan, S., Da Sacco, S. and De Filippo, R. 2008. Characterization of human amniotic fluid stem cells and their pluripotential capability. Methods Cell Biol., 86, 85–99.CrossRefGoogle ScholarPubMed
Zhang, P., Baxter, J., Vinod, K., Tulenko, T. N. and Di Muzio, P. J. 2009. Endothelial differentiation of amniotic fluid-derived stem cells: synergism of biochemical and shear force stimuli. Stem Cells Dev., 18(9), 1299–308.CrossRefGoogle ScholarPubMed
Simper, D., Stalboerger, P. G., Panetta, C. J., Wang, S. and Caplice, N. M. 2002. Smooth muscle progenitor cells in human blood. Circulation, 106(10), 1199–204.CrossRefGoogle ScholarPubMed
Aird, W. C. 2007. Phenotypic heterogeneity of the endothelium: II. Representative vascular beds. Circ. Res., 100(2), 174–90.CrossRefGoogle ScholarPubMed
Thorgeirsson, G. and Robertson, A. L. 1978. The vascular endothelium – pathobiologic significance. Am. J. Pathol., 93(3), 803–48.Google ScholarPubMed
Nagy, Z., Peters, H. and Huttner, I. 1984. Fracture faces of cell junctions in cerebral endothelium during normal and hyperosmotic conditions. Lab. Invest., 50(3), 313–22.Google ScholarPubMed
Wolburg, H., Neuhaus, J., Kniesel, U. et al. 1994. Modulation of tight junction structure in blood–brain barrier endothelial cells. Effects of tissue culture, second messengers and cocultured astrocytes. J. Cell Sci., 107(Part 5), 1347–57.Google ScholarPubMed
Risau, W. 1995. Differentiation of endothelium. FASEB J., 9(10), 926–33.CrossRefGoogle ScholarPubMed
Aird, W. C. 2003. Endothelial cell heterogeneity. Crit. Care Med., 31(4 Suppl.), S221–30.CrossRefGoogle ScholarPubMed
Craig, L. E., Spelman, J. P., Strandberg, J. D. and Zink, M. C. 1998. Endothelial cells from diverse tissues exhibit differences in growth and morphology. Microvasc. Res., 55(1), 65–76.CrossRefGoogle ScholarPubMed
Garlanda, C. and Dejana, E. 1997. Heterogeneity of endothelial cells. Specific markers. Arterioscler. Thromb. Vasc. Biol., 17(7), 1193–202.CrossRefGoogle ScholarPubMed
Atkins, G. B., Jain, M. K. and Hamik, A. 2011. Endothelial differentiation: molecular mechanisms of specification and heterogeneity. Arterioscler. Thromb. Vasc. Biol., 31(7), 1476–84.CrossRefGoogle ScholarPubMed
Ferguson, J. E., Kelley, R. W. and Patterson, C. 2005. Mechanisms of endothelial differentiation in embryonic vasculogenesis. Arterioscler. Thromb. Vasc. Biol., 25(11), 2246–54.CrossRefGoogle ScholarPubMed
Riha, G. M., Lin, P. H., Lumsden, A. B., Yao, Q. and Chen, C. 2005. Application of stem cells for vascular tissue engineering. Tissue Eng., 11(9–10), 1535–52.CrossRefGoogle ScholarPubMed
Hu, J., Sun, X., Ma, H. et al. 2010. Porous nanofibrous PLLA scaffolds for vascular tissue engineering. Biomaterials, 31(31), 7971–7.CrossRefGoogle ScholarPubMed
Wei, G., Jin, Q., Giannobile, W. V. and Ma, P. X. 2007. The enhancement of osteogenesis by nano-fibrous scaffolds incorporating rhBMP-7 nanospheres. Biomaterials, 28(12), 2087–96.CrossRefGoogle ScholarPubMed
Wu, S. M. and Hochedlinger, K. 2011. Harnessing the potential of induced pluripotent stem cells for regenerative medicine. Nature Cell Biol., 13(5), 497–505.CrossRefGoogle ScholarPubMed
Liu, G. H., Suzuki, K., Qu, J. et al. 2011. Targeted gene correction of laminopathy-associated LMNA mutations in patient-specific iPSCs. Cell Stem Cell, 8(6), 688–94.CrossRefGoogle ScholarPubMed
Pontikoglou, C., Delorme, B. and Charbord, P. 2008. Human bone marrow native mesenchymal stem cells. Regen. Med., 3(5), 731–41.CrossRefGoogle ScholarPubMed
Miller-Kasprzak, E. and Jagodziński, P. P. 2007. Endothelial progenitor cells as a new agent contributing to vascular repair. Arch. Immunol. Ther. Exp. (Warszawa), 55(4), 247–59.CrossRefGoogle ScholarPubMed
Yoder, M. C. 2010. Is endothelium the origin of endothelial progenitor cells?Arterioscler. Thromb. Vasc. Biol., 30(6), 1094–103.CrossRefGoogle ScholarPubMed
Ratajczak, M. Z. 2008. Phenotypic and functional characterization of hematopoietic stem cells. Curr. Opin. Hematol., 15(4), 293–300.CrossRefGoogle ScholarPubMed
Di Nardo, P., Forte, G., Ahluwalia, A. and Minieri, M. 2010. Cardiac progenitor cells: potency and control. J. Cell Physiol., 224(3), 590–600.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×