Open Access

Role of hypoxia‑mediated cellular prion protein functional change in stem cells and potential application in angiogenesis (Review)

  • Authors:
    • Seung Pil Yun
    • Yong‑Seok Han
    • Jun Hee Lee
    • Yeo Min Yoon
    • Chul Won Yun
    • Peter Rhee
    • Sang Hun Lee
  • View Affiliations

  • Published online on: August 29, 2017     https://doi.org/10.3892/mmr.2017.7387
  • Pages: 5747-5751
  • Copyright: © Yun et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cellular prion protein (PrPC) can replace other pivotal molecules due to its interaction with several partners in performing a variety of important biological functions that may differ between embryonic and mature stem cells. Recent studies have revealed major advances in elucidating the putative role of PrPC in the regulation of stem cells and its application in stem cell therapy. What is special about PrPC is that its expression may be regulated by hypoxia‑inducible factor (HIF)‑1α, which is the transcriptional factor of cellular response to hypoxia. Hypoxic conditions have been known to drive cellular responses that can enhance cell survival, differentiation and angiogenesis through adaptive processes. Our group recently reported hypoxia‑enhanced vascular repair of endothelial colony‑forming cells on ischemic injury. Hypoxia‑induced AKT/signal transducer and activator of transcription 3 phosphorylation eventually increases neovasculogenesis. In stem cell biology, hypoxia promotes the expression of growth factors. According to other studies, aspects of tissue regeneration and cell function are influenced by hypoxia, which serves an essential role in stem cell HIF‑1α signaling. All these data suggest the possibility that hypoxia‑mediated PrPC serves an important role in angiogenesis. Therefore, the present review summarizes the characteristics of PrPC, which is produced by HIF‑1α in hypoxia, as it relates to angiogenesis.

Introduction

Prions are neuro-degenerative disease-causing agents, that are responsible for changing cellular prion protein (PrPC) to the infectious β-structure-rich insoluble conformer (PrPSC) in the neurons of the brain and spinal cord, as in Creutzfeldt-Jakob disease in humans and Bovine Spongiform Encephalopathy in animals (1). PrPC is known for its involvement in regenerative processes including adhesion, proliferation, differentiation and angiogenesis. According to Stella et al (2), muscles with low PrPC grow slowly compared with wild-type muscles, suggesting that PrPC serves a role in tissue recovery and/or regeneration. For these reasons, recent research has focused on obtaining more conclusive information about the functional role of PrPC in tissue regeneration. Additionally, regulating PrPC expression by hypoxia has become an important topic (3). Hypoxia occurs when blood oxygen concentrations are insufficient and long periods of hypoxia can induce cell death. However, temporary or short periods of exposure to hypoxic conditions actually enhances cell survival by increasing hypoxia-inducible factor-1 (HIF-1), composed of α- and β-subunits, in addition to other transcription factors (46). During hypoxia, an alteration in HIF-1 expression is essential for metabolic adaptation (7,8), as HIF-1α is associated with angiogenesis and growth factors, glucose uptake, and metabolism (8). Therefore, the present review focuses on the association between HIF-1α and PrPC in stem cells. It will also examine how HIF-1α-mediated PrPC expression can serve a role in angiogenesis.

The effect of hypoxia-preconditioning in cultured stem cells

According to previous studies, under hypoxic conditions, aged mesenchymal stem cells (MSCs) increase the secretion of angiogenic and anti-apoptotic related growth factors including vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF)-2, human growth factor (HGF) and insulin growth factor-1, resulting in enhanced angiogenic properties (912). To demonstrate the effect of growth factor secretion in MSCs under hypoxic conditions, a recent study transplanted hypoxia-conditioned stem cell media into rats with traumatic brain injury and demonstrated excellent rescue effects when compared to animals transplanted with normoxia-conditioned media (13). To observe the effect of restorative neurological function Chang et al (13) transplanted media from hypoxia-treated bone marrow (BM)-MSCs into rats with brain injury rat model and demonstrated that it was more efficient compared with normoxia conditioned medium. Chang et al (13) also demonstrated that the neuroprotective effect of hypoxia-conditioned media involved the generation of VEGF and HGF, which are associated with the inducement of endogenous neurogenesis. In another study, the therapeutic activity of MSCs under hypoxia or normoxia was compared in a massive hepatectomy rat model. In vitro, the levels of VEGF in MSCs under hypoxia were markedly higher than normoxia condition. In vivo, MSCs under hypoxia significantly elevated the expression of cyclin D1, proliferating cell nuclear antigen-positive hepatocytes, the liver weight/body weight ratio and survival when compared with normoxia. Notably, the therapeutic effect of hypoxia was negated by anti-VEGF antibody-induced blockade of VEGF in vivo (14). Increasing the activity of matrix metalloproteinase-2 also had a therapeutic effect that was associated with the protection of cardiomyocytes via the inhibition of caspase-3, transforming growth factor β1 and the upregulation of B-cell lymphoma 2 apoptosis regulator/Bcl-2 associated protein X apoptosis regulator ratio (15). According to Lee et al (16), the proliferation and migration of mouse embryonic stem (ES) cells increases upon activation of fibronectin-integrin β1 production through HIF-1α and phosphoinositide 3-kinase/Akt pathways under conditions of hypoxia. Additionally, mouse ES cells that have undergone hypoxic preconditioning exhibit HIF-1α-, mitogen-activated protein kinase- and nuclear factor κB-stimulated interleukin-6 production (17). Hypoxia preconditioning also facilitates the functional bioactivities of endothelial progenitor cells by mediating the regulation of the signal transducer and activator of transcription 3 (STAT3)-B-cell CLL/lymphoma 3 (BCL3) axis. Therefore, expansion and functional bioactivities of endothelial progenitor cells (EPCs) through modulation of the hypoxia-induced STAT3-BCL3 axis can be triggered by a hypoxic preconditioned ex vivo expansion protocol. It has been suggested that hypoxia preconditioning of EPCs may offer a therapeutic strategy for accelerated neovasculogenesis in ischemic diseases (18). In summary, the hypoxic conditioning of cultured stem cells can result in increased production and secretion of trophic factors, augmentation of angiogenic effects and enhanced anti-apoptotic activity from conditioned cells compared with normoxic conditioned culture.

PrPC expression is increased under hypoxic conditions

Oxygen is an indispensable element required for biological energy (19). Thus, it is not surprising that a lack of oxygen causes cell damage (20). Oxygen concentrations within the vascular system that supplies mammals with oxygen vary: The heart and arteries have oxygen concentrations that range from 10–14% (21); however, the majority of tissues contain <5% oxygen, while bone marrow and the thymus contain <1% oxygen (2224). At the cellular level, microenvironment changes are important for cell function. For example, EPC proliferation and cell functions have been demonstrated to be enhanced in hypoxic cultures (18). Jeong et al (25) revealed that hypoxia can protect neurons from PrP fragment-induced apoptosis and can increase PrPC expression, suggesting that HIF-1α mediates PrPC expression. PrPC is generated in the early stages of embryogenesis (26,27) and exists in high levels in neurons of the brain and spinal cord (28). However, glial cells of the central nervous system, and a number of peripheral cell types in adults, possess lower levels of PrPC (29,30). The majority of PrPC molecules lie on the cell surface and are attached to the lipid bilayer through a C-terminal, glycosyl-phosphatidylinositol anchor (31).

HIF-1α regulates PrPC

Under hypoxic conditions, HIF, a protein with a basic helix loop helix-Per/ARNT/Sim structure (32), regulates the expression of various target genes (3335). HIF can be categorized into several types according to its subunits, which comprise an O2-regulated α-subunit (i.e., HIF-1α, −2α or −3α) and a constitutively expressed β-subunit of the Aryl hydrocarbon nuclear translocator (ARNT) family (i.e., ARNT, ARNT2 or ARNT3). Under hypoxic conditions, HIF is inactivated by HIF-a degradation via von Hippel-Lindau E3 ubiquitin ligase (36); however, HIF can still function as a transcription factor by binding HIF-1α and ARNT, consequently increasing the expression of cell growth, proliferation and pro-angiogenesis factors. This is particularly the case for HIF-regulated pro-angiogenic genes including TEK receptor tyrosine kinase, monocyte chemoattractant protein-1, VEGF, basic FGF, angiopoietin (ANGPT)1, ANGPT2 and platelet-derived growth factor (37). HIF-regulated pro-angiogenic factors initiate the HIF-specific angiogenic program by increasing propagation, adhesion, tube formation, migration, vascular permeability and endothelial cell proliferation (38,39). HIF broadly targets pro-angiogenic genes and comprehensively regulates angiogenesis. Thus, HIF is often termed a ‘master-regulator of angiogenesis’. As previously mentioned, PrPC also is regulated by HIF, and PrPC expression is increased under hypoxic conditions. Park et al (3) demonstrated that the effects of HIF-1α and PrPC on neuronal cell death are prion peptide-induced. In hypoxic conditions, neurons are protected from PrP-induced cell death via the activation of p65 and HIF-1α and subsequent inactivation of p21 and p53 signals. Deferoxamine-elevated HIF-1α has similar effects to the hypoxia-mediated inhibition of neuronal cell death under normoxic conditions. Furthermore, knockdown of HIF-1α leads to the downregulation of PrPC expression under hypoxic conditions.

Anti-oxidative effect of PrPC

Numerous enzymes have copper or zinc as essential cofactors, as in the case of cytochrome c oxidase, tyrosinase, various metalloproteinases and Cu/Zn superoxide dismutase 1 (SOD1) (40,41). It has been demonstrated that PrPC has an antioxidant effect relative to the level of copper, and that the level of this effect does not significantly vary between recombinant and tissue-purified PrPC, although the molecular mechanism of the antioxidant properties exhibited by PrPC remains to be elucidated (42,43). Nonetheless, it has been indicated that the decrease in oxidative stress is mediated by the interaction of copper and PrPC (44). Therefore, it is suggested that PrPC has a similar effect to the function of antioxidant enzymes including SOD1 (45). However, changes in expression levels of PrPC do not induce changes in the activation levels of SOD1 (46). PrPC knockout cells are more sensitive to copper toxicity by oxidative stress when compared with wild-type cells (47). Similarly, cerebellar cells obtained from PrPC null mice are more vulnerable to oxidative stress than wild type cells (45,47). The deletion of octapeptide repeats within PrPC inhibits the antioxidant properties of PrPC (42). Indeed, PrPC null mice are more sensitive to acute seizures (48). Therefore, it appears that the status of anti-oxidative defense in PrPC null mice serves as an important factor in determining their lower thresholds of damage when reflecting the severity of injury and clinical pathology (4951). Furthermore, in the skeletal muscles, heart and liver of PrPC null mice, its absence greatly increases protein and lipid oxidation, leading to a lower catalase activity (50). Thus, being reduced in free Cu/Zn, via SOD and glutathione reductase in oxidative stress, PrPC may have influence in the resistance against oxidative stress. Sauer et al (52) demonstrated that overexpression of PrPC completely inhibits reactive oxygen species generation, even with increased activation treatment with adenosine triphosphate. This is in accordance with the hypothesis that PrPC may have a function in protecting against oxidative stress as a free radical scavenger or a molecular sensor (52).

Role of PrPC in endothelial cells under angiogenesis

Endothelial cells express and present PrPC on their surface (53). As resting vascular endothelial cells exhibit minimal or no PrPC in vivo, normal resting endothelial cells of the umbilical cord and adult blood vessels (aorta, saphenous vein and normal transplant endothelial cells) did not produce detectable quantities of PrPC (54). PrPC is expressed in endothelial cells of the blood capillaries in the intestinal wall of the digestive tract and in renal capillaries (55). Another study noted a sudden increase in expression of PrPC on the surface of endothelial cells, astrocytes and neurons in penumbra regions in a rat model of cerebral ischemia (56). Endothelial cells can express PrPC and release it through the cell membrane, as a soluble protein and as a form bound to microparticles, while vascular endothelium may be an origin for PrPC released within the blood (53,57,58). PrPC has been demonstrated to be a component of caveolae, which are the lipid raft of flask-shaped membrane invaginations in endothelial cells that take part in signal transduction associated with cell survival, differentiation and angiogenesis (59). Another study suggested that caveolae have functions in angiogenesis, as implied by the involvement of caveolae in VEGF signaling in the endothelium (60). This signaling mechanism confirms a key function for caveolae, and possibly PrPC, in the regulation of angiogenesis (59). Satoh et al (61) identified that disruption of the PrPC gene results in abnormal regulation of genes important for cell proliferation, differentiation and survival, including Ras and Rac signaling pathways connected to angiogenesis. During development, neonatal brain endothelial cells temporarily express PrPC transcripts, indicating a role in central nervous system angiogenesis and blood-brain barrier maturation (62,63). PrPC expression may be regulated by various growth factors through protein-protein interactions with normal protease sensitive PrPC (52,64,65).

The function of PrPC in tissue regeneration

Muscle regeneration and its association with PrPC has been investigated in a cardiotoxin-induced injury animal model (2). Adult stem cells have the ability to regenerate specific tissues, recapitulating mechanisms observed during morphogenesis (17). Experiments conducted by Stella et al (2) indicated that cardiotoxin-degenerated skeletal muscles release tumor necrosis factor-α (TNF-α), which is affected by PrPC, a factor that is involved in both muscle differentiation and downstream signaling pathways. Thus, in vivo morphogenesis of adult injured muscle tissue can be influenced by PrPC. Their data also support the possibility that the activity of TNF-α converting enzyme (TACE), which hydrolyzes TNF-α from its precursor, is modulated by PrPC. Prospective in vitro studies may investigate this hypothesis and elucidate whether the two proteins (PrPC and TACE) interact directly or indirectly. The current review has provided a number of examples of the interaction of PrPC with extracellular proteins or neuronal membranes, e.g., its interaction with β-secretase 1, which is glycosaminoglycan-mediated, attenuates β-secretase cleavage of the amyloid precursor protein (66). Furthermore, the binding of PrPC to the N-Methyl-D-aspartic acid receptor 2D subunit attenuates glutamate-induced Ca2+ influx (67). Lastly, the results also substantiated that in muscle tissue, the Akt signaling pathway and the regulation of p38 by PrPC have specific physiological significance. This also suggested that PrPC serves a significant role in the regeneration process, specifically in the proliferation and differentiation of myogenic precursor cells (2).

Conclusion

Recent studies have clearly established hypoxia and HIF-1α as master regulators of stem cell growth factors. In hypoxia-pretreated stem cells, HIF-1α mainly controls angiogenesis and tissue regeneration factors, including HGF, VEGF and FGF. Hypoxia has also been revealed to increase the expression of the prion protein and growth factors involved in the function of cells and PrPC has been shown to be regulated by HIF-1α. Stella et al (2) have suggested that PrPC is involved in muscle differentiation and that it influences the morphogenesis of adult injured tissue in vivo. Additionally, regulation of the p38 and Akt signaling pathways by PrPC has clear physiologic importance in tissue in vivo, in addition to the promotion of tissue regeneration. In conclusion, the present review summarized the essential roles of PrPC and HIF-1α in the promotion of tissue regeneration and in the function of stem cells.

Acknowledgements

The present study was supported by a National Research Foundation (NRF) grant funded by the Ministry of Education (grant no. 2016R1D1A3B01007727) and a grant from the Korean Health Technology R&D Project, Ministry of Health and Welfare, Republic of Korea (grant no. HI14C2253). The funders had no role in the study design, data collection or analysis, the decision to publish, or the preparation of the manuscript.

References

1 

Prusiner SB: Prions. Proc Natl Acad Sci USA. 95:pp. 13363–13383. 1998; View Article : Google Scholar : PubMed/NCBI

2 

Stella R, Massimino ML, Sandri M, Sorgato MC and Bertoli A: Cellular prion protein promotes regeneration of adult muscle tissue. Mol Cell Biol. 30:4864–4876. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Park JY, Jeong JK, Lee JH, Moon JH, Kim SW, Lee YJ and Park SY: Induction of cellular prion protein (PrPc) under hypoxia inhibits apoptosis caused by TRAIL treatment. Oncotarget. 6:5342–5353. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Adams JM, Difazio LT, Rolandelli RH, Luján JJ, Haskó G, Csóka B, Selmeczy Z and Németh ZH: HIF-1: A key mediator in hypoxia. Acta Physiol Hung. 96:19–28. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Benzi G, Gorini A, Arnaboldi R, Ghigini B and Villa RF: Age-related changes by hypoxia and TRH analogue on synaptic ATPase activities. Neurobiol Aging. 15:409–417. 1994. View Article : Google Scholar : PubMed/NCBI

6 

Li L, Zhang X, Yang D, Luo G, Chen S and Le W: Hypoxia increases Abeta generation by altering beta- and gamma-cleavage of APP. Neurobiol Aging. 30:1091–1098. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Nakayama K: Cellular signal transduction of the hypoxia response. J Biochem. 146:757–765. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Ryan HE, Lo J and Johnson RS: HIF-1 alpha is required for solid tumor formation and embryonic vascularization. EMBO J. 17:3005–3015. 1998. View Article : Google Scholar : PubMed/NCBI

9 

Efimenko A, Starostina E, Kalinina N and Stolzing A: Angiogenic properties of aged adipose derived mesenchymal stem cells after hypoxic conditioning. J Transl Med. 9:102011. View Article : Google Scholar : PubMed/NCBI

10 

Han YS, Lee JH, Yoon YM, Yun CW, Noh H and Lee SH: Hypoxia-induced expression of cellular prion protein improves the therapeutic potential of mesenchymal stem cells. Cell Death Dis. 7:e23952016. View Article : Google Scholar : PubMed/NCBI

11 

Yuan X, Wang X, Chen C, Zhou J and Han M: Bone mesenchymal stem cells ameliorate ischemia/reperfusion-induced damage in renal epithelial cells via microRNA-223. Stem Cell Res Ther. 8:1462017. View Article : Google Scholar : PubMed/NCBI

12 

He J, Cai Y, Luo LM and Liu HB: Hypoxic adipose mesenchymal stem cells derived conditioned medium protects myocardial infarct in rat. Eur Rev Med Pharmacol Sci. 19:4397–4406. 2015.PubMed/NCBI

13 

Chang CP, Chio CC, Cheong CU, Chao CM, Cheng BC and Lin MT: Hypoxic preconditioning enhances the therapeutic potential of the secretome from cultured human mesenchymal stem cells in experimental traumatic brain injury. Clin Sci (Lond). 124:165–176. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Yu J, Yin S, Zhang W, Gao F, Liu Y, Chen Z, Zhang M, He J and Zheng S: Hypoxia preconditioned bone marrow mesenchymal stem cells promote liver regeneration in a rat massive hepatectomy model. Stem Cell Res Ther. 4:832013. View Article : Google Scholar : PubMed/NCBI

15 

Li JH, Zhang N and Wang JA: Improved anti-apoptotic and anti-remodeling potency of bone marrow mesenchymal stem cells by anoxic pre-conditioning in diabetic cardiomyopathy. J Endocrinol Invest. 31:103–110. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Lee SH, Lee YJ and Han HJ: Role of hypoxia-induced fibronectin-integrin β1 expression in embryonic stem cell proliferation and migration: Involvement of PI3K/Akt and FAK. J Cell Physiol. 226:484–493. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Lee SH, Lee YJ and Han HJ: Effect of arachidonic acid on hypoxia-induced IL-6 production in mouse ES cells: Involvement of MAPKs, NF-kappaB, and HIF-1alpha. J Cell Physiol. 222:574–585. 2010.PubMed/NCBI

18 

Lee SH, Lee JH, Han YS, Ryu JM, Yoon YM and Han HJ: Hypoxia accelerates vascular repair of endothelial colony-forming cells on ischemic injury via STAT3-BCL3 axis. Stem Cell Res Ther. 6:1392015. View Article : Google Scholar : PubMed/NCBI

19 

Semenza GL: Life with oxygen. Science. 318:62–64. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Saikumar P, Dong Z, Patel Y, Hall K, Hopfer U, Weinberg JM and Venkatachalam MA: Role of hypoxia-induced Bax translocation and cytochrome c release in reoxygenation injury. Oncogene. 17:3401–3415. 1998. View Article : Google Scholar : PubMed/NCBI

21 

Carreau A, El Hafny-Rahbi B, Matejuk A, Grillon C and Kieda C: Why is the partial oxygen pressure of human tissues a crucial parameter? Small molecules and hypoxia. J Cell Mol Med. 15:1239–1253. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Parmar K, Mauch P, Vergilio JA, Sackstein R and Down JD: Distribution of hematopoietic stem cells in the bone marrow according to regional hypoxia. Proc Natl Acad Sci USA. 104:5431–5436. 2007; View Article : Google Scholar : PubMed/NCBI

23 

Braun RD, Lanzen JL, Snyder SA and Dewhirst MW: Comparison of tumor and normal tissue oxygen tension measurements using OxyLite or microelectrodes in rodents. Am J Physiol Heart Circ Physiol. 280:H2533–H2544. 2001.PubMed/NCBI

24 

Stockwell RA: Morphometry of cytoplasmic components of mammalian articular chondrocytes and corneal keratocytes: Species and zonal variations of mitochondria in relation to nutrition. J Anat. 175:251–261. 1991.PubMed/NCBI

25 

Jeong JK, Seo JS, Moon MH, Lee YJ, Seol JW and Park SY: Hypoxia-inducible factor-1 α regulates prion protein expression to protect against neuron cell damage. Neurobiol Aging. 33:1006–e1006, e1–10. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Peralta OA, Huckle WR and Eyestone WH: Expression and knockdown of cellular prion protein (PrPC) in differentiating mouse embryonic stem cells. Differentiation. 81:68–77. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Manson J, West JD, Thomson V, McBride P, Kaufman MH and Hope J: The prion protein gene: A role in mouse embryogenesis? Development. 115:117–122. 1992.PubMed/NCBI

28 

Harris DA, Lele P and Snider WD: Localization of the mRNA for a chicken prion protein by in situ hybridization. Proc Natl Acad Sci USA. 90:4309–4313. 1993; View Article : Google Scholar : PubMed/NCBI

29 

Brown KL, Ritchie DL, McBride PA and Bruce ME: Detection of PrP in extraneural tissues. Microsc Res Tech. 50:40–45. 2000. View Article : Google Scholar : PubMed/NCBI

30 

Ford MJ, Burton LJ, Morris RJ and Hall SM: Selective expression of prion protein in peripheral tissues of the adult mouse. Neuroscience. 113:177–192. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Moser M, Colello RJ, Pott U and Oesch B: Developmental expression of the prion protein gene in glial cells. Neuron. 14:509–517. 1995. View Article : Google Scholar : PubMed/NCBI

32 

Kewley RJ, Whitelaw ML and Chapman-Smith A: The mammalian basic helix-loop-helix/PAS family of transcriptional regulators. Int J Biochem Cell Biol. 36:189–204. 2004. View Article : Google Scholar : PubMed/NCBI

33 

Semenza GL and Wang GL: A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol Cell Biol. 12:5447–5454. 1992. View Article : Google Scholar : PubMed/NCBI

34 

Wang GL and Semenza GL: Characterization of hypoxia-inducible factor 1 and regulation of DNA binding activity by hypoxia. J Biol Chem. 268:21513–21518. 1993.PubMed/NCBI

35 

Wang GL, Jiang BH, Rue EA and Semenza GL: Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci USA. 92:5510–5514. 1995; View Article : Google Scholar : PubMed/NCBI

36 

Kaelin WG Jr and Ratcliffe PJ: Oxygen sensing by metazoans: The central role of the HIF hydroxylase pathway. Mol Cell. 30:393–402. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Fong GH: Mechanisms of adaptive angiogenesis to tissue hypoxia. Angiogenesis. 11:121–140. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Matsui J, Wakabayashi T, Asada M, Yoshimatsu K and Okada M: Stem cell factor/c-kit signaling promotes the survival, migration, and capillary tube formation of human umbilical vein endothelial cells. J Biol Chem. 279:18600–18607. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Tang N, Wang L, Esko J, Giordano FJ, Huang Y, Gerber HP, Ferrara N and Johnson RS: Loss of HIF-1alpha in endothelial cells disrupts a hypoxia-driven VEGF autocrine loop necessary for tumorigenesis. Cancer Cell. 6:485–495. 2004. View Article : Google Scholar : PubMed/NCBI

40 

Tapiero H, Townsend DM and Tew KD: Trace elements in human physiology and pathology. Copper. Biomed Pharmacother. 57:386–398. 2003. View Article : Google Scholar : PubMed/NCBI

41 

Tapiero H and Tew KD: Trace elements in human physiology and pathology: Zinc and metallothioneins. Biomed Pharmacother. 57:399–411. 2003. View Article : Google Scholar : PubMed/NCBI

42 

Brown DR, Wong BS, Hafiz F, Clive C, Haswell SJ and Jones IM: Normal prion protein has an activity like that of superoxide dismutase. Biochem J 344 Pt. 1:1–5. 1999. View Article : Google Scholar

43 

Brown DR, Clive C and Haswell SJ: Antioxidant activity related to copper binding of native prion protein. J Neurochem. 76:69–76. 2001. View Article : Google Scholar : PubMed/NCBI

44 

Vassallo N and Herms J: Cellular prion protein function in copper homeostasis and redox signalling at the synapse. J Neurochem. 86:538–544. 2003. View Article : Google Scholar : PubMed/NCBI

45 

Brown DR and Besinger A: Prion protein expression and superoxide dismutase activity. Biochem J. 334:423–429. 1998. View Article : Google Scholar : PubMed/NCBI

46 

Waggoner DJ, Drisaldi B, Bartnikas TB, Casareno RL, Prohaska JR, Gitlin JD and Harris DA: Brain copper content and cuproenzyme activity do not vary with prion protein expression level. J Biol Chem. 275:7455–7458. 2000. View Article : Google Scholar : PubMed/NCBI

47 

Brown DR, Schulz-Schaeffer WJ, Schmidt B and Kretzschmar HA: Prion protein-deficient cells show altered response to oxidative stress due to decreased SOD-1 activity. Exp Neurol. 146:104–112. 1997. View Article : Google Scholar : PubMed/NCBI

48 

Walz R, Amaral OB, Rockenbach IC, Roesler R, Izquierdo I, Cavalheiro EA, Martins VR and Brentani RR: Increased sensitivity to seizures in mice lacking cellular prion protein. Epilepsia. 40:1679–1682. 1999. View Article : Google Scholar : PubMed/NCBI

49 

Dal-Pizzol F, Klamt F, Vianna MM, Schröder N, Quevedo J, Benfato MS, Moreira JC and Walz R: Lipid peroxidation in hippocampus early and late after status epilepticus induced by pilocarpine or kainic acid in Wistar rats. Neurosci Lett. 291:179–182. 2000. View Article : Google Scholar : PubMed/NCBI

50 

Klamt F, Dal-Pizzol F, da Frota ML Conte Jr, Walz R, Andrades ME, da Silva EG, Brentani RR, Izquierdo I and Moreira JC Fonseca: Imbalance of antioxidant defense in mice lacking cellular prion protein. Free Radic Biol Med. 30:1137–1144. 2001. View Article : Google Scholar : PubMed/NCBI

51 

Pereira GS, Walz R, Bonan CD, Battastini AM, Izquierdo I, Martins VR, Brentani RR and Sarkis JJ: Changes in cortical and hippocampal ectonucleotidase activities in mice lacking cellular prion protein. Neurosci Lett. 301:72–74. 2001. View Article : Google Scholar : PubMed/NCBI

52 

Sauer H, Wefer K, Vetrugno V, Pocchiari M, Gissel C, Sachinidis A, Hescheler J and Wartenberg M: Regulation of intrinsic prion protein by growth factors and TNF-alpha: The role of intracellular reactive oxygen species. Free Radic Biol Med. 35:586–594. 2003. View Article : Google Scholar : PubMed/NCBI

53 

Starke R, Drummond O, MacGregor I, Biggerstaff J, Gale R, Camilleri R, Mackie I, Machin S and Harrison P: The expression of prion protein by endothelial cells: A source of the plasma form of prion protein? Br J Haematol. 119:863–873. 2002. View Article : Google Scholar : PubMed/NCBI

54 

Sivakumaran M: The expression of prion protein (PrPc) by endothelial cells: An in vitro culture-induced artefactual phenomenon? Br J Haematol. 121:673–674. 2003. View Article : Google Scholar : PubMed/NCBI

55 

Lemaire-Vieille C, Schulze T, Podevin-Dimster V, Follet J, Bailly Y, Blanquet-Grossard F, Decavel JP, Heinen E and Cesbron JY: Epithelial and endothelial expression of the green fluorescent protein reporter gene under the control of bovine prion protein (PrP) gene regulatory sequences in transgenic mice. Proc Natl Acad Sci USA. 97:5422–5427. 2000; View Article : Google Scholar : PubMed/NCBI

56 

Shyu WC, Lin SZ, Chiang MF, Ding DC, Li KW, Chen SF, Yang HI and Li H: Overexpression of PrPC by adenovirus-mediated gene targeting reduces ischemic injury in a stroke rat model. J Neurosci. 25:8967–8977. 2005. View Article : Google Scholar : PubMed/NCBI

57 

Simak J, Holada K, D'Agnillo F, Janota J and Vostal JG: Cellular prion protein is expressed on endothelial cells and is released during apoptosis on membrane microparticles found in human plasma. Transfusion. 42:334–342. 2002. View Article : Google Scholar : PubMed/NCBI

58 

Starke R, Harrison P, Gale R, Mackie I, Drummond O, MacGregor I and Machin S: Endothelial cells express normal cellular prion protein. Br J Haematol. 123:372–373. 2003. View Article : Google Scholar : PubMed/NCBI

59 

Massimino ML, Griffoni C, Spisni E, Toni M and Tomasi V: Involvement of caveolae and caveolae-like domains in signalling, cell survival and angiogenesis. Cell Signal. 14:93–98. 2002. View Article : Google Scholar : PubMed/NCBI

60 

Feng Y, Venema VJ, Venema RC, Tsai N, Behzadian MA and Caldwell RB: VEGF-induced permeability increase is mediated by caveolae. Invest Ophthalmol Vis Sci. 40:157–167. 1999.PubMed/NCBI

61 

Satoh J, Kuroda Y and Katamine S: Gene expression profile in prion protein-deficient fibroblasts in culture. Am J Pathol. 157:59–68. 2000. View Article : Google Scholar : PubMed/NCBI

62 

Li A, Sakaguchi S, Shigematsu K, Atarashi R, Roy BC, Nakaoke R, Arima K, Okimura N, Kopacek J and Katamine S: Physiological expression of the gene for PrP-like protein, PrPLP/Dpl, by brain endothelial cells and its ectopic expression in neurons of PrP-deficient mice ataxic due to Purkinje cell degeneration. Am J Pathol. 157:1447–1452. 2000. View Article : Google Scholar : PubMed/NCBI

63 

Adle-Biassette H, Verney C, Peoc'h K, Dauge MC, Razavi F, Choudat L, Gressens P, Budka H and Henin D: Immunohistochemical expression of prion protein (PrPC) in the human forebrain during development. J Neuropathol Exp Neurol. 65:698–706. 2006. View Article : Google Scholar : PubMed/NCBI

64 

Kuwahara C, Kubosaki A, Nishimura T, Nasu Y, Nakamura Y, Saeki K, Matsumoto Y and Onodera T: Enhanced expression of cellular prion protein gene by insulin or nerve growth factor in immortalized mouse neuronal precursor cell lines. Biochem Biophys Res Commun. 268:763–766. 2000. View Article : Google Scholar : PubMed/NCBI

65 

West DC, Rees CG, Duchesne L, Patey SJ, Terry CJ, Turnbull JE, Delehedde M, Heegaard CW, Allain F, Vanpouille C, et al: Interactions of multiple heparin binding growth factors with neuropilin-1 and potentiation of the activity of fibroblast growth factor-2. J Biol Chem. 280:13457–13464. 2005. View Article : Google Scholar : PubMed/NCBI

66 

Parkin ET, Watt NT, Hussain I, Eckman EA, Eckman CB, Manson JC, Baybutt HN, Turner AJ and Hooper NM: Cellular prion protein regulates beta-secretase cleavage of the Alzheimer's amyloid precursor protein. Proc Natl Acad Sci USA. 104:11062–11067. 2007; View Article : Google Scholar : PubMed/NCBI

67 

Khosravani H, Zhang Y, Tsutsui S, Hameed S, Altier C, Hamid J, Chen L, Villemaire M, Ali Z, Jirik FR and Zamponi GW: Prion protein attenuates excitotoxicity by inhibiting NMDA receptors. J Gen Physiol. 131:i52008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 16 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yun SP, Han YS, Lee JH, Yoon YM, Yun CW, Rhee P and Lee SH: Role of hypoxia‑mediated cellular prion protein functional change in stem cells and potential application in angiogenesis (Review). Mol Med Rep 16: 5747-5751, 2017
APA
Yun, S.P., Han, Y., Lee, J.H., Yoon, Y.M., Yun, C.W., Rhee, P., & Lee, S.H. (2017). Role of hypoxia‑mediated cellular prion protein functional change in stem cells and potential application in angiogenesis (Review). Molecular Medicine Reports, 16, 5747-5751. https://doi.org/10.3892/mmr.2017.7387
MLA
Yun, S. P., Han, Y., Lee, J. H., Yoon, Y. M., Yun, C. W., Rhee, P., Lee, S. H."Role of hypoxia‑mediated cellular prion protein functional change in stem cells and potential application in angiogenesis (Review)". Molecular Medicine Reports 16.5 (2017): 5747-5751.
Chicago
Yun, S. P., Han, Y., Lee, J. H., Yoon, Y. M., Yun, C. W., Rhee, P., Lee, S. H."Role of hypoxia‑mediated cellular prion protein functional change in stem cells and potential application in angiogenesis (Review)". Molecular Medicine Reports 16, no. 5 (2017): 5747-5751. https://doi.org/10.3892/mmr.2017.7387