Endothelial stem cell

Endothelial stem cells (ESCs) are one of three types of stem cells found in bone marrow. They are multipotent, which describes the ability to give rise to many cell types, whereas a pluripotent stem cell can give rise to all types. ESCs have the characteristic properties of a stem cell: self-renewal and differentiation. These parent stem cells, ESCs, give rise to progenitor cells, which are intermediate stem cells that lose potency. Progenitor stem cells are committed to differentiating along a particular cell developmental pathway. ESCs will eventually produce endothelial cells (ECs), which create the thin-walled endothelium that lines the inner surface of blood vessels and lymphatic vessels.

Endothelial stem cell
CD34+ endothelial cell among a population of bovine aortic endothelial cells
Details
LocationBone marrow
Identifiers
LatinCellula endothelialis praecursoria
THH2.00.01.0.00003
Anatomical terms of microanatomy

Development

ECs were first thought to arise from extraembryonic tissues because blood vessels were observed in the avian and mammalian embryos. However, after histological analysis, it was seen that ECs were in the embryo. This meant that blood vessels come from an intraembryonic source, the mesoderm.[1]

Role of insulin-like growth factors in endothelium differentiation

ECs derived from stem cells are the beginning of vasculogenesis.[2] Vasculogenesis is the new production of a vascular network from mesodermal progenitor cells. This can be distinguished from angiogenesis, which is the creation of new capillaries from vessels that already exist through the process of splitting or sprouting.[3] This can occur "in vitro" in embryoid bodies (EB) derived from embryonic stem cells; this process in EB is similar to "in vivo" vasculogenesis. Important signaling factors for vasculogenesis are TGF-β, BMP4, and VEGF, all of which promote pluripotent stem cells to differentiate into mesoderm, endothelial progenitor cells, and then into mature endothelium.[2]

It is well established that insulin-like growth factor (IGF) signaling is important for cell responses such as mitogenesis, cell growth, proliferation, angiogenesis, and differentiation. IGF1 and IGF2 increase the production of ECs in EB. A method that IGF employs to increase vasculogenesis is upregulation of VEGF. Not only is VEGF critical for mesoderm cells to become an EC, but also for EPCs to differentiate into mature endothelium. Understanding this process can lead to further research in vascular regeneration.[2]

Function

Self-renewal and differentiation

Stem cells have the unique ability make identical copies of themselves. This property maintains unspecialized and undifferentiated cells within the body. Differentiation is the process by which a cell becomes more specialized. For stem cells, this usually occurs through several stages, where a cell proliferates giving rise to daughter cells that are further specialized.[4] For example, an endothelial progenitor cell (EPC) is more specialized than an ESC, and an EC is more specialized than an EPC. The further specialized a cell is, the more differentiated it is and as a result it is considered to be more committed to a certain cellular lineage.[4]

Blood vessel formation

Blood vessels are made of a thin layer of ECs. As part of the circulatory system, blood vessels play a critical role in transporting blood throughout the body. Consequently, ECs have unique functions such as fluid filtration, homeostasis and hormone trafficking. ECs are the most differentiated form of an ESC. Formation of new blood vessels occurs by two different processes: vasculogenesis and angiogenesis.[5] The former requires differentiation of endothelial cells from hemangioblasts and then the further organization into a primary capillary network. The latter occurs when new vessels are built from preexisting blood vessels.[5]

Markers

The vascular system is made up of two parts: 1) Blood vasculature 2) Lymphatic vessels

Both parts consist of ECs that show differential expression of various genes. A study showed that ectopic expression of Prox-1 in blood vascular ECs (BECs) induced one-third of LEC specific gene expression. Prox-1is a homeobox transcription factor found in lymphatic ECs (LECs). For example, specific mRNAs such as VEGFR-3 and p57Kip2 were expressed by the BEC that was induced to express Prox-1.[6]

Lymphatic-specific vascular endothelial growth factors VEGF-C and VEGF-D function as ligands for the vascular endothelial growth factor receptor 3 (VEGFR-3). The ligand-receptor interaction is essential for normal development of lymphatic tissues.[7]

Tal1 gene is specifically found in the vascular endothelium and developing brain.[5] This gene encodes the basic helix-loop-helix structure and functions as a transcription factor. Embryos lacking Tal1 fail to develop past embryonic day 9.5. However, the study found that Tal1 is actually required for vascular remodeling of the capillary network, rather than early endothelial development itself.[7]

Fetal liver kinase-1 (Flk-1) is a cell surface receptor protein that is commonly used as a marker for ESCs and EPCs.[4]

CD34 is another marker that can be found on the surface of ESCs and EPCs. It is characteristic of hematopoietic stem cells, as well as muscle stem cells.[4]

Role in formation of vascular system

The two lineages arising from the EPC and the hematopoietic progenitor cell (HPC) form the blood circulatory system. Hematopoietic stem cells can of course undergo self-renewal, and are multipotent cells that give rise to erythrocytes (red blood cells), megakaryocytes/platelets, mast cells, T-lymphocytes, B-lymphocytes, dendritic cells, natural killer cells, monocyte/macrophage, and granulocytes.[8] A study found that in the beginning stages of mouse embryogenesis, commencing at embryonic day 7.5, HPCs are produced close to the emerging vascular system. In the yolk sac’s blood islands, HPCs and EC lineages emerge from the extraembryonic mesoderm in near unison. This creates a formation in which early erythrocytes are enveloped by angioblasts, and together they give rise to mature ECs. This observation gave rise to the hypothesis that the two lineages come from the same precursor, termed hemangioblast.[7] Even though there is evidence that corroborates a hemangioblast, the isolation and exact location in the embryo has been difficult to pinpoint. Some researchers have found that cells with hemangioblast properties have been located in the posterior end of the primitive streak during gastrulation.[1]

In 1917, Florence Sabin first observer of blood vessels and red blood cells in the yolk sac of chick embryos occur in close proximity and time.[9] Then, in 1932, Murray detected the same event and created the term "hemangioblast" for what Sabin had seen.[10]

Further evidence to corroborate hemangioblasts come from the expression of various genes such as CD34 and Tie2 by both lineages. The fact that this expression was seen in both EC and HPC lineages led researchers to propose a common origin. However, endothelial markers like Flk1/VEGFR-2 are exclusive to ECs but stop HPCs from progressing into an EC. It is accepted that VEGFR-2+ cells are a common precursor for HPCs and ECs. If the Vegfr3 gene is deleted then both HPC and EC differentiation comes to a halt in embryos. VEGF promotes angioblast differentiation; whereas, VEGFR-1 stops the hemangioblast from becoming an EC. In addition, basic fibroblast growth factor FGF-2 is also involved in promoting angioblasts from the mesoderm. After angioblasts commit to becoming an EC, the angioblasts gather and rearrange to assemble in a tube similar to a capillary. Angioblasts can travel during the formation of the circulatory system to configure the branches to allow for directional blood flow. Pericytes and smooth muscle cells encircle ECs when they are differentiating into arterial or venous arrangements. Surrounding the ECs creates a brace to help stabilize the vessels known as the pericellular basal lamina. It is suggested pericytes and smooth muscle cells come from neural crest cells and the surrounding mesenchyme.[7]

Role in recovery

ESCs and EPCs eventually differentiate into ECs. The endothelium secretes soluble factors to regulate vasodilatation and to preserve homeostasis.[11] When there is any dysfunction in the endothelium, the body aims to repair the damage. Resident ESCs can generate mature ECs that replace the damaged ones.[12] However, the intermediate progenitor cell cannot always generate functional ECs. This is because some of the differentiated cells may just have angiogenic properties.[12]

Studies have shown that when vascular trauma occurs, EPCs and circulating endothelial progenitors (CEPs) are attracted to the site due to the release of specific chemokines.[13] CEPs are derived from EPCs within the bone marrow, and the bone marrow is a reservoir of stem and progenitor cells. These cell types accelerate the healing process and prevent further complications such as hypoxia by gathering the cellular materials to reconstruct the endothelium.[13]

Endothelium dysfunction is a prototypical characteristic of vascular disease, common in patients with autoimmune diseases such as systemic lupus erythematosus.[14] Further, there is an inverse relationship between age and levels of EPCs. With a decline in EPCs the body loses its ability to repair the endothelium.[12]

The use of stem cells for treatment has become a growing interest in the scientific community. Distinguishing between an ESC and its intermediate progenitor is nearly impossible,[4] so research is now being done broadly on EPCs. One study showed that brief exposure to sevoflurane promoted growth and proliferation of EPCs.[15] Sevoflurane is used in general anesthesia, but this finding shows the potential to induce endothelial progenitors. Using stem cells for cell replacement therapies is known as "regenerative medicine", which is a booming field that is now working on transplanting cells as opposed to bigger tissues or organs.[15]

Clinical significance

Role in cancer

Understanding more about ESCs is important in cancer research. Tumours induce angiogenesis, which is the formation of new blood vessels. These cancerous cells do this by secreting factors such as VEGF and by reducing the amount of PGK, an anti-VEGF enzyme. The result is an uncontrolled production of beta-catenin, which regulates cell growth and cell mobility. With uncontrolled beta-catenin, the cell loses its adhesive properties. As ECs get packed together to create the lining for a new blood vessel, a single cancer cell is able to travel through the vessel to a distant site. If that cancer cell implants itself and begins forming a new tumour, the cancer has metastasized.[16]

Research

Stem cells have always been a huge interest for scientists due to their unique properties that make them unlike any other cell in the body. Generally, the idea boils down to harnessing the power of plasticity and the ability to go from an unspecialized cell to a highly specialized differentiated cell. ESCs play an incredibly important role in establishing the vascular network that is vital for a functional circulatory system. Consequently, EPCs are under study to determine the potential for treatment of ischemic heart disease.[17] Scientists are still trying to find a way to definitely distinguish the stem cell from the progenitor. In the case of endothelial cells, it is even difficult to distinguish a mature EC from an EPC. However, because of the multipotency of the ESC, the discoveries made about EPCs will parallel or understate the powers of the ESC.[17]

Animal models

There are a number of models used to study vasculogenesis. Avian embryos, Xenopus laevis embryos, are both fair models. However, zebrafish and mouse embryos have widespread use for easily observed development of vascular systems, and the recognition of key parts of molecular regulation when ECs differentiate.[1]

See also

References

  1. Ferguson JW, Kelley RW, Patterson C (2005). "Mechanisms of endothelial differentiation in embryonic vasculogenesis". Journal of the American Heart Association. 25 (11): 2246–2254. doi:10.1161/01.atv.0000183609.55154.44.
  2. Piecewicz SM, Pandey A, Roy B, Xiang SH, Zetter BR, Sengupta S (2012). "Insulin-like growth factors promote vasculogenesis in embryonic stem cells". PLoS ONE. 7 (17): e32191. Bibcode:2012PLoSO...732191P. doi:10.1371/journal.pone.0032191. PMC 3283730. PMID 22363814.
  3. Kovacic JC, Moore J, Herbert A, Ma D, Boehm M, Graham RM (2008). "Endothelial Progenitor Cells, Angioblasts, and Angiogenesis- Old terms Reconsidered from a new current perspective". Trends in Cardiovascular Medicine. 18 (2): 45–51. doi:10.1016/j.tcm.2007.12.002. PMID 18308194.
  4. Bethesda MD. (6 April 2009). "Stem Cell Basics". In Stem Cell Information. National Institutes of Health, U.S. Department of Health and Human Services. Archived from the original on 31 March 2012. Retrieved 6 March 2012.
  5. Gehling U, Ergun S, Schumacher U, Wagener C, Pantel K, Otte M, Schuch G, Schafhausen P, Mende T, Kilic N, Kluge K, Schafer B, Hossfeld D, Fiedler W (2000). "In vitro differentiation of endothelial cells from AC133-positive progenitor cells". Blood. 95 (10): 3106–3112. PMID 10807776.
  6. Petrova TV, Makinen T, Makela TP, Saarela J, Virtanen I, Ferrell RE, Finegold DN, Kerjaschki D, Y, a-Herttuala S, Alitalo K (2002). "Lymphatic endothelial reprogramming of vascular endothelial cells by the Prox-1 homeobox transcription factor". EMBO Journal. 21 (17): 4593–4599. doi:10.1093/emboj/cdf470. PMC 125413. PMID 12198161.
  7. Kubo H, Alitalo K (2003). "The bloody fate of endothelial stem cells". Genes & Development. 17 (3): 322–329. doi:10.1101/gad.1071203. PMID 12569121.
  8. Seita J, Weissman IL (2010). "Hematopoietic stem cell: self-renewal versus differentiation". Systems Biology and Medicine. 2 (6): 640–653. doi:10.1002/wsbm.86. PMC 2950323. PMID 20890962.
  9. Sabin F. (1917). "Preliminary note on the differentiation of angioblasts and the method by which they produce blood-vessels, blood-plasma and red blood-cells as seen in the living chick". The Anatomical Record. 13 (4): 199–204. doi:10.1002/ar.1090130403.
  10. Murray PDF. (1932). "The development in vitro of the blood of the early chick embryo". Proceedings of the Royal Society of London. Series B, Containing Papers of a Biological Character. 111 (773): 497–521. Bibcode:1932RSPSB.111..497M. doi:10.1098/rspb.1932.0070.
  11. Cheek D, Graulty R, Bryant S (2002). "Meet the multitasking endothelium". Nursing Made Incredibly Easy!. 6 (4): 18–25. doi:10.1097/01.nme.0000324934.19114.e0.
  12. Siddique A, Shantsila E, Lip G, Varma C (2010). "Endothelial progenitor cells: what use for the cardiologist?". Journal Angiogenesis Research. 2 (6): 6. doi:10.1186/2040-2384-2-6. PMC 2834645. PMID 20298532.
  13. Rafil S, Lyden D (2003). "Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration". Nature Medicine. 9 (6): 702–12. doi:10.1038/nm0603-702. PMID 12778169.
  14. Deanfield J, Donald A, Ferri C, Giannattasio C, Halcox J, Halligan S, Lerman A, Mancia G, Oliver JJ, Pessina AC, Rizzoni D, Rossi GP, Salvetti A, Schiffrin EL, Taddei S, Webb DJ (2005). "Endothelial function and dysfunction. Part I: Methodological issues for assessment in the different vascular beds: a statement by the Working Group on Endothelin and Endothelial Factors of the European Society of Hypertension". Journal of Hypertension. 23 (1): 7–17. doi:10.1097/00004872-200501000-00004. PMID 15643116.
  15. Lucchinetti E, Zeisberger SM, Baruscotti I, Wacker J, Feng J, Dubey R, Zisch AH, Zaugg M (2009). "Stem cell-like human endothelial progenitors show enhanced colony-forming capacity after brief sevofluorane exposure: preconditioning of angiogenic cells by volatile anesthetics". Anesthesia & Analgesia. 109 (4): 1117–26. doi:10.1213/ane.0b013e3181b5a277. PMID 19762739.
  16. Enzyme eliminated by cancer cells holds promise for cancer treatment
  17. Fan CL, Li Y, Gao PJ, Liu JJ, Zhang XJ, Zhu DL (2003). "Differentiation of endothelial progenitor cells from human umbilical cord blood CD 34+ cells in vitro". Acta Pharmacologica Sinica. 24 (3): 212–218.
  18. Kovina, MV; Krasheninnikov, ME; Dyuzheva, TG; Danilevsky, MI; Klabukov, ID; Balyasin, MV; Chivilgina, OK; Lyundup, AV (2018). "Human endometrial stem cells: High-yield isolation and characterization". Cytotherapy. 20 (3): 361–374. doi:10.1016/j.jcyt.2017.12.012. ISSN 1477-2566. PMID 29397307.
This article is issued from Wikipedia. The text is licensed under Creative Commons - Attribution - Sharealike. Additional terms may apply for the media files.