Skip to main content

Advertisement

Log in

Pericytes: Properties, Functions and Applications in Tissue Engineering

  • Published:
Stem Cell Reviews and Reports Aims and scope Submit manuscript

Abstract

Mesenchymal stem cells (MSCs) are one of the most studied adult stem cells and in recent years. They have become attractive agents/cell source for cellular therapy and regenerative medicine applications. During investigations about their origin, researchers hypothesized that perivascular regions are the common anatomical regions where MSCs come from and perivascular cells like pericytes (PCs) (Rouget cells, mural cells) are in vivo counterparts of MSCs. Beside capillaries and microvessels as their most common locations, PCs are also found in large vessels (arteries and veins). They can be isolated from several tissues and organs particularly from retina and brain. There are different approaches about their isolation, characterization and culture but there has been no common protocol yet because of the lack of defined PC-specific marker. They make special contact with endothelial cells in the basement membrane and have very important functions in several tissues and organs. They participate in vascular development, stabilization, maturation, and remodeling, blood pressure control, endothelial cell proliferation and differentiation, contractility of vascular smooth muscle cells, wound healing, vasculogenesis and angiogenesis, long-term maintenance of hematopoietic stem cells in bone marrow niche. Their multipotential differentiation capacity and participation in many events in the body make PCs preferred cells in tissue engineering applications including 3D blood–brain barrier models, skeletal muscle constructs, bone tissue engineering and tissue-engineered vascular grafts.

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

Access this article

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

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Pittenger, M. F., et al. (1999). Multilineage potential of adult human mesenchymal stem cells. Science, 284(5411), 143–147.

    CAS  PubMed  Google Scholar 

  2. Friedenstein, A. J., Piatetzky, S., II, & Petrakova, K. V. (1966). Osteogenesis in transplants of bone marrow cells. Journal of Embryology and Experimental Morphology, 16(3), 381–390.

    CAS  PubMed  Google Scholar 

  3. Friedenstein, A. J., Chailakhjan, R. K., & Lalykina, K. S. (1970). The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell and Tissue Kinetics, 3(4), 393–403.

    CAS  PubMed  Google Scholar 

  4. Caplan, A. I. (1991). Mesenchymal stem cells. Journal of Orthopaedic Research, 9(5), 641–650.

    CAS  PubMed  Google Scholar 

  5. Zuk, P. A., et al. (2002). Human adipose tissue is a source of multipotent stem cells. Molecular Biology of the Cell, 13(12), 4279–4295.

    CAS  PubMed Central  PubMed  Google Scholar 

  6. Igura, K., et al. (2004). Isolation and characterization of mesenchymal progenitor cells from chorionic villi of human placenta. Cytotherapy, 6(6), 543–553.

    CAS  PubMed  Google Scholar 

  7. Rogers, I., & Casper, R. F. (2004). Umbilical cord blood stem cells. Best Practice & Research. Clinical Obstetrics & Gynaecology, 18(6), 893–908.

    Google Scholar 

  8. Kluth, S. M., et al. (2010). DLK-1 as a marker to distinguish unrestricted somatic stem cells and mesenchymal stromal cells in cord blood. Stem Cells and Development, 19(10), 1471–1483.

    CAS  PubMed  Google Scholar 

  9. Tsai, M. S., et al. (2004). Isolation of human multipotent mesenchymal stem cells from second-trimester amniotic fluid using a novel two-stage culture protocol. Human Reproduction, 19(6), 1450–1456.

    PubMed  Google Scholar 

  10. Asakura, A., Komaki, M., & Rudnicki, M. (2001). Muscle satellite cells are multipotential stem cells that exhibit myogenic, osteogenic, and adipogenic differentiation. Differentiation, 68(4–5), 245–253.

    CAS  PubMed  Google Scholar 

  11. Salingcarnboriboon, R., et al. (2003). Establishment of tendon-derived cell lines exhibiting pluripotent mesenchymal stem cell-like property. Experimental Cell Research, 287(2), 289–300.

    CAS  PubMed  Google Scholar 

  12. Shi, S., & Gronthos, S. (2003). Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp. Journal of Bone and Mineral Research, 18(4), 696–704.

    PubMed  Google Scholar 

  13. Seo, B. M., et al. (2004). Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet, 364(9429), 149–155.

    CAS  PubMed  Google Scholar 

  14. Toma, J. G., et al. (2001). Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nature Cell Biology, 3(9), 778–784.

    CAS  PubMed  Google Scholar 

  15. da Silva Meirelles, L., Chagastelles, P. C., & Nardi, N. B. (2006). Mesenchymal stem cells reside in virtually all post-natal organs and tissues. Journal of Cell Science, 119(Pt 11), 2204–2213.

    PubMed  Google Scholar 

  16. Crisan, M., et al. (2008). A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell, 3(3), 301–313.

    CAS  PubMed  Google Scholar 

  17. Dominici, M., et al. (2006). Minimal criteria for defining multipotent mesenchymal stromal cells. The international society for cellular therapy position statement. Cytotherapy, 8(4), 315–317.

    CAS  PubMed  Google Scholar 

  18. Kim, N., & Cho, S. G. (2013). Clinical applications of mesenchymal stem cells. Korean Journal of Internal Medicine, 28(4), 387–402.

    PubMed Central  PubMed  Google Scholar 

  19. Prasongchean, W., & Ferretti, P. (2012). Autologous stem cells for personalised medicine. New Biotechnology, 29(6), 641–650.

    CAS  PubMed  Google Scholar 

  20. Le Blanc, K., et al. (2008). Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet, 371(9624), 1579–1586.

    PubMed  Google Scholar 

  21. Wakitani, S., et al. (2007). Repair of articular cartilage defects in the patello-femoral joint with autologous bone marrow mesenchymal cell transplantation: three case reports involving nine defects in five knees. Journal of Tissue Engineering and Regenerative Medicine, 1(1), 74–79.

    PubMed  Google Scholar 

  22. Yang, Z., et al. (2010). A novel approach to transplanting bone marrow stem cells to repair human myocardial infarction: delivery via a noninfarct-relative artery. Cardiovascular Therapeutics, 28(6), 380–385.

    PubMed  Google Scholar 

  23. Liang, J., et al. (2010). Mesenchymal stem cell transplantation for diffuse alveolar hemorrhage in SLE. Nature Reviews Rheumatology, 6(8), 486–489.

    PubMed  Google Scholar 

  24. Kharaziha, P., et al. (2009). Improvement of liver function in liver cirrhosis patients after autologous mesenchymal stem cell injection: a phase I-II clinical trial. European Journal of Gastroenterology & Hepatology, 21(10), 1199–1205.

    CAS  Google Scholar 

  25. Le Blanc, K., et al. (2005). Fetal mesenchymal stem-cell engraftment in bone after in utero transplantation in a patient with severe osteogenesis imperfecta. Transplantation, 79(11), 1607–1614.

    PubMed  Google Scholar 

  26. Marion, N. W., & Mao, J. J. (2006). Mesenchymal stem cells and tissue engineering. Methods in Enzymology, 420, 339–361.

    CAS  PubMed Central  PubMed  Google Scholar 

  27. Covas, D. T., et al. (2008). Multipotent mesenchymal stromal cells obtained from diverse human tissues share functional properties and gene-expression profile with CD146+ perivascular cells and fibroblasts. Experimental Hematology, 36(5), 642–654.

    CAS  PubMed  Google Scholar 

  28. Murray, I. R., et al. (2014). Natural history of mesenchymal stem cells, from vessel walls to culture vessels. Cellular and Molecular Life Sciences, 71(8), 1353–1374.

    CAS  PubMed  Google Scholar 

  29. Crisan, M., et al. (2012). Perivascular cells for regenerative medicine. Journal of Cellular and Molecular Medicine, 16(12), 2851–2860.

    CAS  PubMed Central  PubMed  Google Scholar 

  30. Nees, S., et al. (2012). Isolation, bulk cultivation, and characterization of coronary microvascular pericytes: the second most frequent myocardial cell type in vitro. American Journal of Physiology. Heart and Circulatory Physiology, 302(1), H69–H84.

    CAS  PubMed  Google Scholar 

  31. Andreeva, E. R., et al. (1998). Continuous subendothelial network formed by pericyte-like cells in human vascular bed. Tissue & Cell, 30(1), 127–135.

    CAS  Google Scholar 

  32. Nees, S., Weiss, D. R., & Juchem, G. (2013). Focus on cardiac pericytes. Pflügers Archiv, 465(6), 779–787.

    CAS  PubMed  Google Scholar 

  33. Mills, S. J., Cowin, A. J., & Kaur, P. (2013). Pericytes, mesenchymal stem cells and the wound healing process. Cells, 2(3), 621–634.

    CAS  PubMed Central  PubMed  Google Scholar 

  34. Wagenseil, J. E., & Mecham, R. P. (2009). Vascular extracellular matrix and arterial mechanics. Physiological Reviews, 89(3), 957–989.

    CAS  PubMed Central  PubMed  Google Scholar 

  35. Gerhardt, H., & Betsholtz, C. (2003). Endothelial-pericyte interactions in angiogenesis. Cell and Tissue Research, 314(1), 15–23.

    PubMed  Google Scholar 

  36. Bergers, G., & Song, S. (2005). The role of pericytes in blood-vessel formation and maintenance. Neuro-Oncology, 7(4), 452–464.

    CAS  PubMed Central  PubMed  Google Scholar 

  37. Goumans, M. J., et al. (2002). Balancing the activation state of the endothelium via two distinct TGF-beta type I receptors. The EMBO Journal, 21(7), 1743–1753.

    CAS  PubMed Central  PubMed  Google Scholar 

  38. Sato, T. N., et al. (1995). Distinct roles of the receptor tyrosine kinases Tie-1 and Tie-2 in blood vessel formation. Nature, 376(6535), 70–74.

    CAS  PubMed  Google Scholar 

  39. Maisonpierre, P. C., et al. (1997). Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science, 277(5322), 55–60.

    CAS  PubMed  Google Scholar 

  40. Hoch, R. V., & Soriano, P. (2003). Roles of PDGF in animal development. Development, 130(20), 4769–4784.

    CAS  PubMed  Google Scholar 

  41. Paik, J. H., et al. (2004). Sphingosine 1-phosphate receptor regulation of N-cadherin mediates vascular stabilization. Genes and Development, 18(19), 2392–2403.

    CAS  PubMed Central  PubMed  Google Scholar 

  42. Pfister, F., et al. (2013). Pericytes in the eye. Pflügers Archiv, 465(6), 789–796.

    CAS  PubMed  Google Scholar 

  43. Sims, D. E. (2000). Diversity within pericytes. Clinical and Experimental Pharmacology and Physiology, 27(10), 842–846.

    CAS  PubMed  Google Scholar 

  44. Nehls, V., & Drenckhahn, D. (1993). The versatility of microvascular pericytes: from mesenchyme to smooth muscle? Histochemistry, 99(1), 1–12.

    CAS  PubMed  Google Scholar 

  45. Sundberg, C., et al. (1996). Pericytes as collagen-producing cells in excessive dermal scarring. Laboratory Investigation, 74(2), 452–466.

    CAS  PubMed  Google Scholar 

  46. Crisan, M., et al. (2009). Perivascular multipotent progenitor cells in human organs. Annals of the New York Academy of Sciences, 1176, 118–123.

    CAS  PubMed  Google Scholar 

  47. Park, T. S., et al. (2011). Placental perivascular cells for human muscle regeneration. Stem Cells and Development, 20(3), 451–463.

    CAS  PubMed Central  PubMed  Google Scholar 

  48. Armulik, A., Abramsson, A., & Betsholtz, C. (2005). Endothelial/pericyte interactions. Circulation Research, 97(6), 512–523.

    CAS  PubMed  Google Scholar 

  49. Rajkumar, V. S., et al. (2006). Platelet-derived growth factor-beta receptor activation is essential for fibroblast and pericyte recruitment during cutaneous wound healing. American Journal of Pathology, 169(6), 2254–2265.

    CAS  PubMed Central  PubMed  Google Scholar 

  50. Corselli, M., et al. (2010). Perivascular ancestors of adult multipotent stem cells. Arteriosclerosis, Thrombosis, and Vascular Biology, 30(6), 1104–1109.

    CAS  PubMed  Google Scholar 

  51. Davidoff, M. S., et al. (2009). The neuroendocrine Leydig cells and their stem cell progenitors, the pericytes. Advances in Anatomy Embryology and Cell Biology, 205, 1–107.

    Google Scholar 

  52. Stefanska, A., Peault, B., & Mullins, J. J. (2013). Renal pericytes: multifunctional cells of the kidneys. Pflügers Archiv, 465(6), 767–773.

    CAS  PubMed  Google Scholar 

  53. Maier, C. L., & Pober, J. S. (2011). Human placental pericytes poorly stimulate and actively regulate allogeneic CD4 T cell responses. Arteriosclerosis, Thrombosis, and Vascular Biology, 31(1), 183–189.

    CAS  PubMed Central  PubMed  Google Scholar 

  54. Balabanov, R., et al. (1996). CNS microvascular pericytes express macrophage-like function, cell surface integrin alpha M, and macrophage marker ED-2. Microvascular Research, 52(2), 127–142.

    CAS  PubMed  Google Scholar 

  55. Thomas, W. E. (1999). Brain macrophages: on the role of pericytes and perivascular cells. Brain Research. Brain Research Reviews, 31(1), 42–57.

    CAS  PubMed  Google Scholar 

  56. Ballabh, P., Braun, A., & Nedergaard, M. (2004). The blood–brain barrier: an overview: structure, regulation, and clinical implications. Neurobiology of Disease, 16(1), 1–13.

    CAS  PubMed  Google Scholar 

  57. Cleaver, O., & Melton, D. A. (2003). Endothelial signaling during development. Nature Medicine, 9(6), 661–668.

    CAS  PubMed  Google Scholar 

  58. Abbott, N. J. (2002). Astrocyte-endothelial interactions and blood–brain barrier permeability. Journal of Anatomy, 200(6), 629–638.

    CAS  PubMed Central  PubMed  Google Scholar 

  59. Sato, M., Suzuki, S., & Senoo, H. (2003). Hepatic stellate cells: unique characteristics in cell biology and phenotype. Cell Structure and Function, 28(2), 105–112.

    CAS  PubMed  Google Scholar 

  60. Knittel, T., et al. (1999). Expression and regulation of cell adhesion molecules by hepatic stellate cells (HSC) of rat liver: involvement of HSC in recruitment of inflammatory cells during hepatic tissue repair. American Journal of Pathology, 154(1), 153–167.

    CAS  PubMed Central  PubMed  Google Scholar 

  61. Betsholtz, C. (2004). Insight into the physiological functions of PDGF through genetic studies in mice. Cytokine and Growth Factor Reviews, 15(4), 215–228.

    CAS  PubMed  Google Scholar 

  62. Corselli, M., et al. (2013). Perivascular support of human hematopoietic stem/progenitor cells. Blood, 121(15), 2891–2901.

    CAS  PubMed Central  PubMed  Google Scholar 

  63. Distler, J. H., et al. (2003). Angiogenic and angiostatic factors in the molecular control of angiogenesis. Quarterly Journal of Nuclear Medicine, 47(3), 149–161.

    CAS  PubMed  Google Scholar 

  64. Lamagna, C., & Bergers, G. (2006). The bone marrow constitutes a reservoir of pericyte progenitors. Journal of Leukocyte Biology, 80(4), 677–681.

    CAS  PubMed  Google Scholar 

  65. Jain, R. K. (2003). Molecular regulation of vessel maturation. Nature Medicine, 9(6), 685–693.

    CAS  PubMed  Google Scholar 

  66. Armulik, A., Genove, G., & Betsholtz, C. (2011). Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Developmental Cell, 21(2), 193–215.

    CAS  PubMed  Google Scholar 

  67. Hirschi, K. K., & D’Amore, P. A. (1996). Pericytes in the microvasculature. Cardiovascular Research, 32(4), 687–698.

    CAS  PubMed  Google Scholar 

  68. Crisan, M. (2008). Purification and culture of human blood vessel-associated progenitor cells. Currents Protocols Stem Cell Biology, Chapter 2, Unit 2B 2 1–2B 2 13.

    Google Scholar 

  69. Mogensen, C., et al. (2011). Isolation and functional characterization of pericytes derived from hamster skeletal muscle. Acta Physiologica (Oxford, England), 201(4), 413–426.

    CAS  Google Scholar 

  70. Tsang, W. P., et al. (2013). CD146+ human umbilical cord perivascular cells maintain stemness under hypoxia and as a cell source for skeletal regeneration. PLoS One, 8(10), e76153.

    CAS  PubMed Central  PubMed  Google Scholar 

  71. Montemurro, T., et al. (2011). Differentiation and migration properties of human foetal umbilical cord perivascular cells: potential for lung repair. Journal of Cellular and Molecular Medicine, 15(4), 796–808.

    CAS  PubMed Central  PubMed  Google Scholar 

  72. Maier, C. L., et al. (2010). Explant outgrowth, propagation and characterization of human pericytes. Microcirculation, 17(5), 367–380.

    CAS  PubMed Central  PubMed  Google Scholar 

  73. Sarugaser, R., et al. (2005). Human umbilical cord perivascular (HUCPV) cells: a source of mesenchymal progenitors. Stem Cells, 23(2), 220–229.

    PubMed  Google Scholar 

  74. Schugar, R. C., et al. (2009). High harvest yield, high expansion, and phenotype stability of CD146 mesenchymal stromal cells from whole primitive human umbilical cord tissue. Journal of Biomedicine and Biotechnology, 2009, 789526.

    PubMed Central  PubMed  Google Scholar 

  75. Bryan, B. A., & D’Amore, P. A. (2008). Pericyte isolation and use in endothelial/pericyte coculture models. Methods in Enzymology, 443, 315–331.

    CAS  PubMed  Google Scholar 

  76. Nakagawa, S., et al. (2009). A new blood–brain barrier model using primary rat brain endothelial cells, pericytes and astrocytes. Neurochemistry International, 54(3–4), 253–263.

    CAS  PubMed  Google Scholar 

  77. Tigges, U., et al. (2012). A novel and simple method for culturing pericytes from mouse brain. Microvascular Research, 84(1), 74–80.

    CAS  PubMed Central  PubMed  Google Scholar 

  78. Neng, L., et al. (2013). Isolation and culture of endothelial cells, pericytes and perivascular resident macrophage-like melanocytes from the young mouse ear. Nature Protocols, 8(4), 709–720.

    PubMed Central  PubMed  Google Scholar 

  79. Yamagishi, S., et al. (2002). Pigment epithelium-derived factor protects cultured retinal pericytes from advanced glycation end product-induced injury through its antioxidative properties. Biochemical and Biophysical Research Communications, 296(4), 877–882.

    CAS  PubMed  Google Scholar 

  80. Blocki, A., et al. (2013). Not all MSCs can act as pericytes: functional in vitro assays to distinguish pericytes from other mesenchymal stem cells in angiogenesis. Stem Cells and Development, 22(17), 2347–2355.

    CAS  PubMed Central  PubMed  Google Scholar 

  81. Wanjare, M., Kusuma, S., & Gerecht, S. (2013). Perivascular cells in blood vessel regeneration. Biotechnology Journal, 8(4), 434–447.

    CAS  PubMed Central  PubMed  Google Scholar 

  82. Sacchetti, B., et al. (2007). Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell, 131(2), 324–336.

    CAS  PubMed  Google Scholar 

  83. Papetti, M., et al. (2003). FGF-2 antagonizes the TGF-beta1-mediated induction of pericyte alpha-smooth muscle actin expression: a role for myf-5 and Smad-mediated signaling pathways. Investigative Ophthalmology & Visual Science, 44(11), 4994–5005.

    Google Scholar 

  84. Wakui, S. (1992). Epidermal growth factor receptor at endothelial cell and pericyte interdigitation in human granulation tissue. Microvascular Research, 44(3), 255–262.

    CAS  PubMed  Google Scholar 

  85. Corselli, M., et al. (2013). Identification of perivascular mesenchymal stromal/stem cells by flow cytometry. Cytometry. Part A, 83(8), 714–720.

    Google Scholar 

  86. Langer, R., & Vacanti, J. P. (1993). Tissue engineering. Science, 260(5110), 920–926.

    CAS  PubMed  Google Scholar 

  87. Salgado, A. J., et al. (2013). Tissue engineering and regenerative medicine: past, present, and future. International Review of Neurobiology, 108, 1–33.

    CAS  PubMed  Google Scholar 

  88. Howard, D., et al. (2008). Tissue engineering: strategies, stem cells and scaffolds. Journal of Anatomy, 213(1), 66–72.

    CAS  PubMed Central  PubMed  Google Scholar 

  89. Mravic, M., et al. (2014). From pericytes to perivascular tumours: correlation between pathology, stem cell biology, and tissue engineering. International Orthopaedics, 38(9), 1819–1824.

    PubMed  Google Scholar 

  90. Tourovskaia, A., et al. (2014). Tissue-engineered microenvironment systems for modeling human vasculature. Experimental Biology and Medicine (Maywood, N.J.), 239(9), 1264–1271.

    CAS  Google Scholar 

  91. Fuoco, C., et al. (2014). 3D hydrogel environment rejuvenates aged pericytes for skeletal muscle tissue engineering. Frontiers in Physiology, 5, 203.

    PubMed Central  PubMed  Google Scholar 

  92. James, A. W., et al. (2012). An abundant perivascular source of stem cells for bone tissue engineering. Stem Cells Translation Medicine, 1(9), 673–684.

    CAS  Google Scholar 

  93. Askarinam, A., et al. (2013). Human perivascular stem cells show enhanced osteogenesis and vasculogenesis with Nel-like molecule I protein. Tissue Engineering Part A, 19(11–12), 1386–1397.

    CAS  PubMed Central  PubMed  Google Scholar 

  94. Morin, K. T., et al. (2013). Aligned human microvessels formed in 3D fibrin gel by constraint of gel contraction. Microvascular Research, 90, 12–22.

    CAS  PubMed  Google Scholar 

  95. Kim, S., et al. (2013). Engineering of functional, perfusable 3D microvascular networks on a chip. Lab on a Chip, 13(8), 1489–1500.

    CAS  PubMed  Google Scholar 

  96. Chen, C. W., et al. (2013). Human pericytes for ischemic heart repair. Stem Cells, 31(2), 305–316.

    CAS  PubMed Central  PubMed  Google Scholar 

  97. Andrejecsk, J. W., et al. (2013). Paracrine exchanges of molecular signals between alginate-encapsulated pericytes and freely suspended endothelial cells within a 3D protein gel. Biomaterials, 34(35), 8899–8908.

    CAS  PubMed  Google Scholar 

  98. He, W., et al. (2010). Pericyte-based human tissue engineered vascular grafts. Biomaterials, 31(32), 8235–8244.

    CAS  PubMed Central  PubMed  Google Scholar 

  99. Mendes, L. F., et al. (2012). Perivascular-like cells contribute to the stability of the vascular network of osteogenic tissue formed from cell sheet-based constructs. PLoS One, 7(7), e41051.

    CAS  PubMed Central  PubMed  Google Scholar 

  100. van der Meer, A. D., et al. (2013). Three-dimensional co-cultures of human endothelial cells and embryonic stem cell-derived pericytes inside a microfluidic device. Lab on a Chip, 13(18), 3562–3568.

    PubMed  Google Scholar 

  101. Anfuso, C. D., et al. (2012). Microcapillary-like structures prompted by phospholipase A2 activation in endothelial cells and pericytes co-cultures on a polyhydroxymethylsiloxane thin film. Biochimie, 94(9), 1860–1870.

    CAS  PubMed  Google Scholar 

  102. Saik, J. E., et al. (2011). Covalently immobilized platelet-derived growth factor-BB promotes angiogenesis in biomimetic poly(ethylene glycol) hydrogels. Acta Biomaterialia, 7(1), 133–143.

    CAS  PubMed Central  PubMed  Google Scholar 

  103. Sirabella, D., De Angelis, L., & Berghella, L. (2013). Sources for skeletal muscle repair: from satellite cells to reprogramming. Journal of Cachexia, Sarcopenia and Muscle, 4(2), 125–136.

    PubMed Central  PubMed  Google Scholar 

  104. James, A. W., et al. (2012). Perivascular stem cells: a prospectively purified mesenchymal stem cell population for bone tissue engineering. Stem Cells Translation Medicine, 1(6), 510–519.

    CAS  Google Scholar 

Download references

Conflict of Interest

The authors declare no potential conflicts of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Betül Çelebi-Saltik.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gökçinar-Yagci, B., Uçkan-Çetinkaya, D. & Çelebi-Saltik, B. Pericytes: Properties, Functions and Applications in Tissue Engineering. Stem Cell Rev and Rep 11, 549–559 (2015). https://doi.org/10.1007/s12015-015-9590-z

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12015-015-9590-z

Keywords

Navigation