Asialo-rhuEPO: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , , ,

Asialo-rhuEPO can be prepared by enzymatic removal of sialic acid residues from rhuEPOM (asialo-rhuEPOE) or by expressing human EPO gene in glycoengineered transgenic plants (asialo-rhuEPOP). Both types of asialo-rhuEPO, like rhuEPOM, displayed excellent neuroprotective effects by regulating multiple cellular pathways in cerebral I/R animal models.

  • multimodal neuroprotectant
  • erythropoietin
  • hematopoietic activity

1. Asialo-rhuEPO Structures and Properties

EPO lacking terminal sialic acid residues is known as asialo-rhuEPO, which contains neutral galactose residues as new terminal sugars instead of acidic sialic acid residues. Removal of sialic acid residues does not impact protein folding [1], but has shown to dramatically alter protein charge, binding capacity to the homodimeric (EPOR)2, in vitro and in vivo hematopoietic activities [2][3], and circulatory half-life [4][5]. In contrast to sialylated EPO with acidic pI, asialo-rhuEPO is a basic protein with a pI of 8.5 and a short circulatory half-life of ~2.5 min [5]. It has been reported to bind to the EPOR four times faster than its sialylated form in vitro [6]. Most importantly, asialo-rhuEPO was demonstrated to lack in vivo hematopoietic activity (non-erythropoietic) even at very high doses [5], and could cross the BBB to display excellent neuroprotective effects [5][7][8][9][10][11].

2. Methods of Asialo-rhuEPO Production

Currently, there are two methods used to produce asialo-rhuEPO. It is commonly prepared by enzymatic removal of sialic acid residues (desialylation) from rhuEPOM [1][5]. Researchers designate this enzymatically prepared one as asialo-rhuEPOE. Plants have been successfully glycoengineered to produce asialo-rhuEPO [12][13][14][15], which is designated as asialo-rhuEPOP.

2.1. Enzymatic Method

Asialo-rhuEPO in small quantities for basic research was produced by the enzymatic method. In this method, asialo-rhuEPOE was prepared by desialylation of rhuEPOM with commercially available enzymes called neuraminidases (also known as sialidases) [1][5][6]. Neuraminidases catalyze the hydrolysis of α2,3-, α2-6-, α2-8-, and α2-9-linked Neu5Ac (a type of sialic acid typically present on mammalian glycoproteins) from glycoproteins. Although this is a simple and straightforward method to obtain asialo-rhuEPO without the loss of biological activity, it is not an economically viable method for large-scale production because of the high cost (~4000 USD/mg protein) of rhuEPOM [16]. In addition, neuraminidases for the above purpose are unavailable in bulk quantities for large-scale desialylation of rhuEPOM. A mammalian cell-based expression system is also not available to directly express asialo-rhuEPO. Hence, the neuroprotective properties of asialo-rhuEPOE could not be translated into clinical practice. Alternative methods to produce asialo-rhuEPO inexpensively were therefore sought to realize its full therapeutic potential.

2.2. Plant-Based Expression Method

Plants have been used as an inexpensive expression system to produce asialo-rhuEPO because they lack sialylation capacity (as they lack the entire enzymatic machinery necessary for the synthesis and transfer of sialic acid residues to glycoproteins) but have the ability to synthesize similar complex biantennary N-glycans like mammalian cells [17][18][19]. Moreover, transgenic plants expressing wild-type or chimeric human GalT can sufficiently add galactose residues on the N-glycans of produced glycoproteins [20][21]. The other advantages of using a plant-based expression system are low production cost, lack of human pathogen contamination, and ease of scaling up in production [18][22]. Researchers produced asialo-rhuEPO in tobacco plants by stably co-expressing human EPO and β1,4-galactosyltransferase (GalT) genes [13][14][15], while Parson and co-workers produced it in moss [12].

3. Unique Properties of Asialo-rhuEPOP

3.1. Asialo-rhuEPOP Carries Plant-Specific Biantennary N-Glycans

Asialo-rhuEPOP accumulates as 28–30 kD bands representing different glycoforms in transgenic tobacco plants [14]. It is 162 amino acids long because of the proteolytic removal of the extreme C-terminal region Thr163-Arg166 [23]. This lost region is not important for the biological activity of EPO [24]. Asialo-rhuEPOP is a basic protein with a theoretical pI of 8.75. All three N-glycosylation sites in asialo-rhuEPOP are occupied with N-glycan chains each bearing terminal mammalian-type β1,4-galactose residues [14][15]. The proportion of β1,4-galactose residues on asialo-rhuEPOP was high (72%) when chimeric GalT was co-expressed with EPO [15], whereas it was low (8%) in the case of co-expressing wild-type GalT [14]. The N-glycan chains in asialo-rhuEPOP are slightly different from that in asialo-rhuEPOE. Asialo-rhuEPOP carries biantennary N-glycans containing plant-specific β1,2-xylose and α1,3-fucose residues, while those N-glycans in asialo-rhuEPOE lack β1,2-xylose but contain a core α1,6-fucose instead of α1,3-fucose. The presence of biantennary N-glycans with plant-specific β1,2-xylose and α1,3-fucose residues on asialo-rhuEPOP showed no impact on in vitro (EPOR)2 binding, since asialo-rhuEPOP displayed similar affinity for the (EPOR)2 as rhuEPOM [13]. Furthermore, it provided a similar level of protection to the brain as rhuEPOM after I/R injury [10], suggesting that the biantennary N-glycans bearing plant-specific sugars have no impact on its in vivo neuroprotective activity.

3.2. Asialo-rhuEPOP Is Non-Erythropoietic and Non-Immunogenic

RhuEPOM doses used for neuroprotection are typically higher (~40 µg/kg bw in a rodent stroke model and 4–36 µg/kg bw in clinical trials) than that used to improve hemoglobin (Hb) levels in anemia patients (~0.8 µg/kg bw) [25]. Therefore, rhuEPOM or any of its derivatives at high doses can increase RBC levels and pose a thrombosis risk if they possess hematopoietic activity. Researchers confirmed a lack of erythropoietic activity in asialo-rhuEPOP by repeated IV injection (twice a week for 5 weeks) in female BALB/c mice at a dose of 44 µg/kg bw (neuroprotective dose), which is 55 times higher than the dose of rhuEPOM typically used to stimulate RBC production. At this high dose, asialo-rhuEPOP showed no increase in Hb concentrations, while rhuEPOM significantly increased Hb concentrations as early as a week after two injections [10]. Consistent with this observation, at the end of the 5-week period, the RBC count in mice receiving asialo-rhuEPOP was 7.8 × 106/mm3, similar to the saline group (7.5 × 106/mm3), while that in mice administered rhuEPOM was 11 × 106/mm3, corresponding to a ~47% increase [10]. These results confirmed that asialo-rhuEPOP is also non-erythropoietic, like asialo-rhuEPOE.
Concerning the plant-specific sugars on asialo-rhuEPOP, there is an ongoing debate whether plant-specific sugars on therapeutic glycoproteins are immunogenic [26][27][28][29]. The immunogenicity of plant-specific sugars on asialo-rhuEPOP was investigated by immunizing BALB/c mice with 44 µg/kg bw (neuroprotective dose) and 88 µg/kg bw protein, along with the same doses of rhuEPOM as a negative control and horse radish peroxidase as a positive control. Reseasrchers detected no antibodies against plant-specific sugars in the sera of mice immunized with asialo-rhuEPOP, indicating that it is non-immunogenic even at high doses [10]. These results suggested that asialo-rhuEPOP is safe for use in clinical practice.

4. Neuroprotective Effects of Asialo-rhuEPO

Asialo-rhuEPO is a nonerythropoietic EPO derivative that has been proven to have neuroprotective functions in various studies (Table 1) [5][9][10][11]. Erbayraktar et al. [5] demonstrated that asialo-rhuEPOE is non-erythropoietic and neuroprotective in animal models of cerebral ischemia, spinal cord compression, and sciatic nerve crush. It has also been shown to attenuate neuronal cell death in the hippocampal CA1 region after transient forebrain ischemia [9]. In addition, asialo-rhuEPOE was reported to improve motor behavior and reduce motoneuron loss in the cervical spinal cord of wobbler mice, an animal model of amyotrophic lateral sclerosis, without affecting hematocrit values [11]. Despite its non-erythropoietic nature and excellent neuroprotective effects, no clinical trials have been conducted for ischemic stroke treatment or for other organ injury treatment, likely due to its limited availability and high cost. To circumvent this problem, researchers produced asialo-rhuEPOP in tobacco plants. Researchers describe the neuroprotective properties of asialo-rhuEPOP and discuss its potential as a multimodal drug for ischemic stroke treatment.
Table 1. In vitro and in vivo neuroprotective effects of asialo-rhuEPO.
Author/Year Cell Line/Animal Treatment/Model Outcome
Erbayraktar et al. [5]/2003 PC-12 cells Nerve growth factor (NGF) absence-triggered cell death 34% protection
Erbayraktar et al. [5]/2003 P-19 cells Hypoxia for 15 h 43% protection
Mennini et al. [11]/2006 Motoneuron culture Kainate-induced cell death Increased survival rate by 57%
Ishii et al. [30]/2012 PC-12 cells Nerve growth factor (NGF) absence-triggered cell death No observed protection
Kittur et al. [14]/2013 N2A cells Staurosporine-induced cell death 44% protection
Erbayraktar et al. [5]/2003 Sprague Dawley male rats MCAO model Reduced infarct volume by ~50%
Erbayraktar et al. [5]/2003 Sprague Dawley male rats Spinal cord compression Restricted injury with better neuron survival and motor score
Erbayraktar et al. [5]/2003 Sprague Dawley male rats Sciatic nerve crush model Reduced functional loss and improved motor testing score
Wang et al. [7]/2004 Wistar rat pups (7 days old) Hypoxia–ischemia model Reduced infarct volume by 52%
Grasso et al. [31]/2006 Sprague Dawley rats Spinal cord compression Significantly recovered affected motor function
Mennini et al. [11]/2006 Homozygous wobbler mice Amyotrophic lateral sclerosis model carrying a mutation of Vps54 gene Improved motor behavior and reduced inflammation
Price et al. [8]/2010 Sprague Dawley male rats MCAO model Significantly reduced infarct volume with reduced cell death
Yamashita et al. [9]/2010 Mongolian male gerbils Occlusion of the
common carotid arteries
Improved learning and memory function with better neuron survival
Ishii et al. [30]/2012 Wistar male rats MCAO model Significantly reduced cerebral I/R injury
He et al. [10]/2022 BALB/c male mice MCAO model Significant decreased neurological deficits, infarction volume, and edema volume with better neuron survival

4.1. In Vitro and In Vivo Neuroprotective Effects of Asialo-rhuEPOP

Researchers evaluated the in vitro neuroprotective effect of asialo-rhuEPOP by studying its ability to protect neuronal-like cells (N2A) against staurosporine (STS)-induced apoptosis. The results showed that simultaneous treatment of N2A cells with 1 µM STS and 20 IU/mL asialo-rhuEPOP or rhuEPOM (a positive control) resulted in lower cytotoxicity (47% and 66%, respectively) while treatment with 1 µM STS alone caused 84% cytotoxicity [14]. These results suggest that asialo-rhuEPOP is not only neuroprotective but also more effective (~2-fold) than rhuEPOM in vitro.
The in vivo neuroprotective effect of asialo-rhuEPOP was evaluated using a mouse model of I/R injury and compared with rhuEPOM. Researchers used a dose of 44 µg/kg bw because asialo-rhuEPOE has been found to be neuroprotective at this dose in a mouse model of I/R injury [5]. Following IV administration of asialo-rhuEPOP (rhuEPOM as well) immediately at the restoration of blood flow after 1 h occlusion, researchers observed a significant decrease in neurological deficits (from 3.1 in I/R–saline group to 1.8–1.9 in asialo-rhuEPOP and rhuEPOM groups), cerebral infarction volume (from 33.0% in I/R–saline group to 15.0% in asialo-rhuEPOP and rhuEPOM groups), and edema volume (from ~30% in I/R–saline group to 14% in asialo-rhuEPOP and rhuEPOM groups) [10]. Consistent with these observations, HE and Nissl staining of brain sections showed lesser cellular damage and higher neuron density in the asialo-rhuEPOP-treated group than in the I/R–saline group. Immunostaining for NeuN, a marker commonly used to assess the functional state of neurons, also revealed a higher number of NeuN-positive cells in the asialo-rhuEPOP-treated group than that in the I/R–saline group [10]. The results demonstrated that asialo-rhuEPOP is neuroprotective and equipotent to rhuEPOM in reducing brain damage induced by I/R injury.

4.2. Neuroprotective Mechanism of Asialo-rhuEPOP

Asialo-rhuEPOE has been shown to exert excellent neuroprotection against cerebral I/R injury [5][7][8][9][11][30], but its neuroprotective mechanism has not been dissected yet. Regarding asialo-rhuEPOP, the studies suggested that it protects the brain from I/R injury by restoring mitochondria fusion–fission-related proteins, preventing I/R injury-induced mitophagy and autophagy markers, and inhibiting apoptosis [10]. Mitochondria are both a source and target of I/R injury and cell death, and their dysfunction occurring via fission and fusion imbalance is considered one of the hallmarks of I/R-induced neuronal cell death [32][33]. Fission regulates the amounts of mitochondria and removes damaged mitochondria, whereas fusion maintains normal mitochondrial activity by complementing damaged mitochondrial contents with the components of healthy mitochondria [33][34]. I/R injury and other cerebral insults promote fission, leading to disturbed mitochondrial dynamics and compromised mitochondrial functions, thereby promoting the release of pro-apoptotic factors, such as cyt c [32][33][35]. The western blotting and immunofluorescence studies showed that the induced fission-related proteins dynamin-related protein 1 (p-Drp1) and Drp1 receptor fission 1 protein (Fis1) in the brain of the I/R-saline group were significantly restored in both asialo-rhuEPOP- and rhuEPOM-treated groups [10]. Similarly, the fusion-related proteins mitofusin 1 and 2 (Mfn1 and Mfn2) and optic atrophy protein 1 (OPA1), whose levels were reduced in the I/R-saline group, were significantly restored in asialo-rhuEPOP and rhuEPOM groups [10]. These results suggested that, like rhuEPOM, asialo-rhuEPOP can maintain mitochondrial fission–fusion balance under I/R conditions.
The results also showed that asialo-rhuEPOP treatment and rhuEPOM as well can regulate mitophagy-related markers. Mitochondrial fission is followed by mitophagy to remove damaged organelles [36][37]. In mammalian cells, the PINK1 (PTEN-induced putative kinase protein 1) and parkin (an E3 ubiquitin ligase PARK2) cooperatively sense cellular stress and mediate the removal of damaged mitochondria [36][37]. The Western blotting results showed that asialo-rhuEPOP (also rhuEPOM) treatment was able to restore both PINK1 and PARK2 levels that were elevated by I/R injury [10], consistent with lower mitochondrial fission anticipated from the restoration of fission-related proteins in EPO-treated groups. In addition to mitophagy, the Western blotting and immunofluorescence results of general autophagy markers (LC3B, p62, and Beclin1) revealed that their increased levels in the I/R–saline group were reinstated back in asialo-rhuEPOP and rhuEPOM groups to the levels similar to the sham group [10]. Furthermore, investigation of apoptotic markers and TUNEL staining revealed that asialo-rhuEPOP-treatment (also rhuEPOM) significantly attenuated I/R-induced Bax/Bcl2 ratio, cyt c release, and caspase 3 caspase cleavage, and reduced TUNEL-positive cells by 50% compared to the I/R–saline control, indicating that both asialo-rhuEPOP and rhuEPOM have similar suppressive effects on I/R injury-induced apoptosis [10]. Together, these results suggested that asialo-rhuEPOP- and rhuEPOM-treatment can restore I/R injury-affected mitochondrial fission–fusion-related proteins and mitophagy- and autophagy-related markers, leading to anti-apoptotic effects and cell survival.
Regarding signaling pathways responsible for displaying the neuroprotective effect of asialo-rhuEPOP, the Western blotting results showed a significant increase in phosphorylation of STAT5, PI3K, AKT, and ERK1/2 in asialo-rhuEPOP- and rhuEPOM-treated groups compared with the I/R–saline group [10]. These results suggest that, like rhuEPOM, the neuroprotective effects of asialo-rhuEPOP are mediated through the activation of STAT5, PI3K/AKT, and MAPK/ERK1/2 signaling pathways as reported previously by others for rhuEPOM [38][39][40][41][42]. These findings also indicate that asialo-rhuEPOP, like rhuEPOM, exhibits pleiotropic effects through these pathways. However, at this time, it remains unknown what the relative contribution of each of these asialo-rhuEPOP-induced signaling pathways is to the regulation of mitochondrial fission–fusion, mitophagy, autophagy, and apoptosis to display neuroprotective effects. In addition, the receptor responsible for transducing asialo-rhuEPOP tissue-protective signal remains to be identified.

This entry is adapted from the peer-reviewed paper 10.3390/ph16040610

References

  1. Narhi, L.; Arakawa, T.; Aoki, K.; Elmore, R.; Rohde, M.; Boone, T.; Strickland, T. The effect of carbohydrate on the structure and stability of erythropoietin. J. Biol. Chem. 1991, 266, 23022–23026.
  2. Lukowsky, W.A.; Painter, R.H. Studies on the Role of Sialic Acid in the Physical and Biological Properties of Erythropoietin. Can. J. Biochem. 1972, 50, 909–917.
  3. Goldwasser, E.; Kung, C.K.-H.; Eliason, J. On the Mechanism of Erythropoietin-induced Differentiation. J. Biol. Chem. 1974, 249, 4202–4206.
  4. Jelkmann, W. Erythropoietin: Structure, control of production, and function. Physiol. Rev. 1992, 72, 449–489.
  5. Erbayraktar, S.; Grasso, G.; Sfacteria, A.; Xie, Q.-W.; Coleman, T.; Kreilgaard, M.; Torup, L.; Sager, T.; Erbayraktar, Z.; Gokmen, N.; et al. Asialoerythropoietin is a nonerythropoietic cytokine with broad neuroprotective activity in vivo. Proc. Natl. Acad. Sci. USA 2003, 100, 6741–6746.
  6. Imai, N.; Higuchi, M.; Kawamura, A.; Tomonoh, K.; Oh-Eda, M.; Fujiwara, M.; Shimonaka, Y.; Ochi, N. Physicochemical and biological characterization of asialoerythropoietin. Suppressive effects of sialic acid in the expression of biological activity of human erythropoietin in vitro. JBIC J. Biol. Inorg. Chem. 1990, 194, 457–462.
  7. Wang, X.; Zhu, C.; Wang, X.; Gerwien, J.G.; Schrattenholz, A.; Sandberg, M.; Leist, M.; Blomgren, K. The nonerythropoietic asialoerythropoietin protects against neonatal hypoxia-ischemia as potently as erythropoietin. J. Neurochem. 2004, 91, 900–910.
  8. Price, C.D.; Yang, Z.; Karlnoski, R.; Kumar, D.; Chaparro, R.; Camporesi, E.M. Effect of continuous infusion of asialoerythropoietin on short-term changes in infarct volume, penumbra apoptosis and behaviour following middle cerebral artery occlusion in rats. Clin. Exp. Pharmacol. Physiol. 2010, 37, 185–192.
  9. Yamashita, T.; Nonoguchi, N.; Ikemoto, T.; Miyatake, S.-I.; Kuroiwa, T. Asialoerythropoietin attenuates neuronal cell death in the hippocampal CA1 region after transient forebrain ischemia in a gerbil model. Neurol. Res. 2010, 32, 957–962.
  10. He, M.; Kittur, F.S.; Hung, C.-Y.; Zhang, J.; Jing, L.; Sane, D.C.; Li, P.A.; Xie, J. A Novel Plant-Produced Asialo-rhuEPO Protects Brain from Ischemic Damage Without Erythropoietic Action. Transl. Stroke Res. 2021, 13, 338–354.
  11. Mennini, T.; De Paola, M.; Bigini, P.; Mastrotto, C.; Fumagalli, E.; Barbera, S.; Mengozzi, M.; Viviani, B.; Corsini, E.; Marinovich, M.; et al. Nonhematopoietic Erythropoietin Derivatives Prevent Motoneuron Degeneration In Vitro and In Vivo. Mol. Med. 2006, 12, 153–160.
  12. Parsons, J.; Altmann, F.; Arrenberg, C.K.; Koprivova, A.; Beike, A.K.; Stemmer, C.; Gorr, G.; Reski, R.; Decker, E.L. Moss-based production of asialo-erythropoietin devoid of Lewis A and other plant-typical carbohydrate determinants. Plant Biotechnol. J. 2012, 10, 851–861.
  13. Kittur, F.S.; Hung, C.-Y.; Darlington, D.E.; Sane, D.C.; Xie, J. N-Glycosylation engineering of tobacco plants to produce asialoerythropoietin. Plant Cell Rep. 2012, 31, 1233–1243.
  14. Kittur, F.S.; Bah, M.; Archer-Hartmann, S.; Hung, C.-Y.; Azadi, P.; Ishihara, M.; Sane, D.C.; Xie, J. Cytoprotective Effect of Recombinant Human Erythropoietin Produced in Transgenic Tobacco Plants. PLoS ONE 2013, 8, e76468.
  15. Kittur, F.S.; Hung, C.-Y.; Zhu, C.; Shajahan, A.; Azadi, P.; Thomas, M.D.; Pearce, J.L.; Gruber, C.; Kallolimath, S.; Xie, J. Glycoengineering tobacco plants to stably express recombinant human erythropoietin with different N-glycan profiles. Int. J. Biol. Macromol. 2020, 157, 158–169.
  16. Weise, A.; Altmann, F.; Rodriguez-Franco, M.; Sjoberg, E.R.; Bäumer, W.; Launhardt, H.; Kietzmann, M.; Gorr, G. High-level expression of secreted complex glycosylated recombinant human erythropoietin in the Physcomitrella Delta-fuc-t Delta-xyl-t mutant. Plant Biotechnol. J. 2007, 5, 389–401.
  17. Wee, E.G.-T.; Sherrier, D.J.; Prime, T.A.; DuPree, P. Targeting of Active Sialyltransferase to the Plant Golgi Apparatus. Plant Cell 1998, 10, 1759–1768.
  18. Ma, J.K.-C.; Drake, P.M.W.; Christou, P. The production of recombinant pharmaceutical proteins in plants. Nat. Rev. Genet. 2003, 4, 794–805.
  19. Gomord, V.; Faye, L. Posttranslational modification of therapeutic proteins in plants. Curr. Opin. Plant Biol. 2004, 7, 171–181.
  20. Palacpac, N.Q.; Yoshida, S.; Sakai, H.; Kimura, Y.; Fujiyama, K.; Yoshida, T.; Seki, T. Stable expression of human β1,4-galactosyltransferase in plant cells modifies N-linked glycosylation patterns. Proc. Natl. Acad. Sci. USA 1999, 96, 4692–4697.
  21. Bakker, H.; Rouwendal, G.J.A.; Karnoup, A.S.; Florack, D.E.A.; Stoopen, G.M.; Helsper, J.P.F.G.; van Ree, R.; van Die, I.; Bosch, D. An antibody produced in tobacco expressing a hybrid β-1,4-galactosyltransferase is essentially devoid of plant carbohydrate epitopes. Proc. Natl. Acad. Sci. USA 2006, 103, 7577–7582.
  22. Daniell, H.; Streatfield, S.J.; Wycoff, K. Medical molecular farming: Production of antibodies, biopharmaceuticals and edible vaccines in plants. Trends Plant Sci. 2001, 6, 219–226.
  23. Kittur, F.S.; Lalgondar, M.; Hung, C.-Y.; Sane, D.C.; Xie, J. C-Terminally fused affinity Strep-tag II is removed by proteolysis from recombinant human erythropoietin expressed in transgenic tobacco plants. Plant Cell Rep. 2014, 34, 507–516.
  24. Wen, D.; Boissel, J.; Showers, M.; Ruch, B.; Bunn, H. Erythropoietin structure-function relationships. Identification of functionally important domains. J. Biol. Chem. 1994, 269, 22839–22846.
  25. Brines, M.; Cerami, A. Erythropoietin-mediated tissue protection: Reducing collateral damage from the primary injury response. J. Intern. Med. 2008, 264, 405–432.
  26. Chargelegue, D.; Vine, N.D.; Van Dolleweerd, C.J.; Drake, P.M.; Ma, J.K.-C. A murine monoclonal antibody produced in transgenic plants with plant-specific glycans is not immunogenic in mice. Transgenic Res. 2000, 9, 187–194.
  27. Jin, C.; Altmann, F.; Strasser, R.; Mach, L.; Schähs, M.; Kunert, R.; Rademacher, T.; Glössl, J.; Steinkellner, H. A plant-derived human monoclonal antibody induces an anti-carbohydrate immune response in rabbits. Glycobiology 2007, 18, 235–241.
  28. Ward, B.J.; Landry, N.; Trépanier, S.; Mercier, G.; Dargis, M.; Couture, M.; D’aoust, M.-A.; Vézina, L.-P. Human antibody response to N-glycans present on plant-made influenza virus-like particle (VLP) vaccines. Vaccine 2014, 32, 6098–6106.
  29. Wang, X.; Jiang, D.; Shi, J.; Yang, D. Expression of α-1,6-fucosyltransferase (FUT8) in rice grain and immunogenicity evaluation of plant-specific glycans. J. Biotechnol. 2017, 242, 111–121.
  30. Ishii, T.; Asai, T.; Oyama, D.; Fukuta, T.; Yasuda, N.; Shimizu, K.; Minamino, T.; Oku, N. Amelioration of cerebral ischemia–reperfusion injury based on liposomal drug delivery system with asialo-erythropoietin. J. Control. Release 2012, 160, 81–87.
  31. Grasso, G.; Sfacteria, A.; Erbayraktar, S.; Passalacqua, M.; Meli, F.; Gokmen, N.; Yilmaz, O.; La Torre, D.; Buemi, M.; Iacopino, D.G.; et al. Amelioration of spinal cord compressive injury by pharmacological preconditioning with erythropoietin and a nonerythropoietic erythropoietin derivative. J. Neurosurg. Spine 2006, 4, 310–318.
  32. Cheung, E.C.C.; McBride, H.M.; Slack, R.S. Mitochondrial dynamics in the regulation of neuronal cell death. Apoptosis 2007, 12, 979–992.
  33. Balog, J.; Mehta, S.L.; Vemuganti, R. Mitochondrial fission and fusion in secondary brain damage after CNS insults. J. Cereb. Blood Flow Metab. 2016, 36, 2022–2033.
  34. Otera, H.; Mihara, K. Mitochondrial Dynamics: Functional Link with Apoptosis. Int. J. Cell Biol. 2012, 2012, 1–10.
  35. Kumar, R.; Bukowski, M.J.; Wider, J.M.; Reynolds, C.A.; Calo, L.; Lepore, B.; Tousignant, R.; Jones, M.; Przyklenk, K.; Sanderson, T.H. Mitochondrial dynamics following global cerebral ischemia. Mol. Cell. Neurosci. 2016, 76, 68–75.
  36. Burman, J.L.; Pickles, S.; Wang, C.; Sekine, S.; Vargas, J.N.S.; Zhang, Z.; Youle, A.M.; Nezich, C.L.; Wu, X.; Hammer, J.A.; et al. Mitochondrial fission facilitates the selective mitophagy of protein aggregates. J. Cell Biol. 2017, 216, 3231–3247.
  37. Ma, K.; Chen, G.; Li, W.; Kepp, O.; Zhu, Y.; Chen, Q. Mitophagy, Mitochondrial Homeostasis, and Cell Fate. Front. Cell Dev. Biol. 2020, 8, 467.
  38. Sirén, A.-L.; Knerlich, F.; Poser, W.; Gleiter, C.H.; Brück, W.; Ehrenreich, H. Erythropoietin and erythropoietin receptor in human ischemic/hypoxic brain. Acta Neuropathol. 2001, 101, 271–276.
  39. Digicaylioglu, M.; Garden, G.; Timberlake, S.; Fletcher, L.; Lipton, S.A. Acute neuroprotective synergy of erythropoietin and insulin-like growth factor I. Proc. Natl. Acad. Sci. USA 2004, 101, 9855–9860.
  40. Kilic, E.; Kilic, U.; Soliz, J.; Bassetti, C.L.; Gassmann, M.; Hermann, D.M. Brain-derived erythropoietin protects from focal cerebral ischemia by dual activation of ERK-1/-2 and Akt pathways. FASEB J. 2005, 19, 2026–2028.
  41. Um, M.; Lodish, H.F. Antiapoptotic Effects of Erythropoietin in Differentiated Neuroblastoma SH-SY5Y Cells Require Activation of Both the STAT5 and AKT Signaling Pathways. J. Biol. Chem. 2006, 281, 5648–5656.
  42. Sergio, C.-M.; Rolando, C.-A. Erythropoietin regulates signaling pathways associated with neuroprotective events. Exp. Brain Res. 2022, 240, 1303–1315.
More
This entry is offline, you can click here to edit this entry!
ScholarVision Creations