Neuroprotective effects of low-dose G-CSF plus meloxicam in a rat model of anterior ischemic optic neuropathy

Pei-Kang Liu, Yao-Tseng Wen, Wei Lin, Kishan Kapupara, Minghong Tai, Rong-Kung Tsai, Pei-Kang Liu, Yao-Tseng Wen, Wei Lin, Kishan Kapupara, Minghong Tai, Rong-Kung Tsai

Abstract

Non-arteritic anterior ischemic optic neuropathy (NAION) causes a sudden loss of vision and lacks effective treatment. Granulocyte colony-stimulating factor (G-CSF) provides neuroprotection against the experimental optic nerve injuries but also induce leukocytosis upon typical administration. We found synergetic neuroprotective effects of meloxicam and low dose G-CSF without leukocytosis in a rat model of anterior ischemic optic neuropathy (rAION). The WBC counts in the low-dose G-CSF-plus meloxicam-treated group were similar to the sham-operated group. Combination treatment of low-dose G-CSF plus meloxicam preserved RGCs survival and visual function, reduced RGC apoptosis and the macrophages infiltration, and promote more M2 phenotype of macrophage/microglial transition than the low-dose GCSF treatment or the meloxicam treatment. Moreover, the combination treatment induced higher serine/threonine kinase 1 (Akt1) expression. The combination treatment of low-dose G-CSF plus meloxicam lessened the leukocytotic side effect and provided neuroprotective effects via Akt1 activation in the rAION model. This approach provides crucial preclinical information for the development of alternative therapy in AION.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Analysis of WBC counts in the sham-operated rats, PBS-treated rats, meloxicam-treated rats, high-dose G-CSF-treated rats, low-dose G-CSF-treated rats, and low-dose G-CSF-plus-meloxicam-treated rats. Treatment with meloxicam, low-dose G-CSF, and low-dose G-CSF plus meloxicam did not induce leukocytosis in the rats, but treatment with high-dose G-CSF induced leukocytosis after a 5-day treatment. *p 

Figure 2

Evaluation of the visual function…

Figure 2

Evaluation of the visual function by using FVEPs in the rAION model. (…

Figure 2
Evaluation of the visual function by using FVEPs in the rAION model. (A) Representative FVEP tracings at four weeks after rAION induction in the sham group, PBS-treated group, meloxicam-treated group, low-dose G-CSF-treated group, and low-dose G-CSF-plus-meloxicam-treated group. (B) Bar charts demonstrate the P1-N2 amplitude. The values of amplitude are expressed as mean ± SD in each group (n = 12 in each group). The amplitudes of the P1-N2 waves in the combination-treated group were significantly higher than meloxicam-treated and low-dose G-CSF-treated groups, respectively. *p < 0.05.

Figure 3

Survival of RGCs in rAION-induced…

Figure 3

Survival of RGCs in rAION-induced rats with PBS treatment, meloxicam treatment, G-CSF treatment,…

Figure 3
Survival of RGCs in rAION-induced rats with PBS treatment, meloxicam treatment, G-CSF treatment, and G-CSF plus meloxicam treatment at 28 days after rAION induction. (A) A representative of flat-mounted central retinas and the morphometry of RGCs in each group through FluoroGold retrograde labeling at four weeks after rAION induction. (B) RGC density in the central retina in each group. Data are expressed as mean ± SD for each group (n = 12). The number of RGCs in the combination-treated group was 1.58- and 1.45-fold higher than in the meloxicam-treated and low-dose G-CSF-treated groups, respectively. *p < 0.05.

Figure 4

Analysis of RGC apoptosis in…

Figure 4

Analysis of RGC apoptosis in the RGC layer through TUNEL assay at four…

Figure 4
Analysis of RGC apoptosis in the RGC layer through TUNEL assay at four weeks after rAION induction. (A) Representative images of double-stained apoptotic cells in the RGC layers in each group. The apoptotic cells (TUNEL-positive cells) in green were stained with TUNEL staining, and the nuclei of the RGCs in blue were labeled with DAPI staining. (B) Quantification of TUNEL-positive cells per high-power field. Data are expressed as mean ± SD for each group (n = 6). Treatment with low-dose G-CSF plus meloxicam significantly reduced the number of apoptotic RGC by 3.6- and 2.5-fold compared with the meloxicam-treated and low-dose G-CSF-treated groups, respectively. *p < 0.05.

Figure 5

Immunohistochemistry (IHC) of ED1 in…

Figure 5

Immunohistochemistry (IHC) of ED1 in the optic nerve at four weeks after rAION…

Figure 5
Immunohistochemistry (IHC) of ED1 in the optic nerve at four weeks after rAION induction for evaluating the inflammatory infiltration of macrophages. (A) Representative images of ED1 staining in the longitudinal sections of the optic nerve. The ED1-positive cells in green were stained with FITC, and the nuclei in blue were labeled with DAPI. (B) Quantification of ED1-positive cells per high-power field. Data are expressed as mean ± SD in each group (n = 6). Macrophage recruitment was decreased by 6.75- and 4.1-fold in the combination-treated group compared with the meloxicam-treated and low-dose G-CSF-treated groups, respectively (C) Evaluation of M2 macrophage polarization at four weeks after rAION induction. Relative mRNA expression levels of the markers of M2 macrophages in the optic nerve are shown as histograms. Each value was normalized to CypA. The expression levels of Arg 1, CD206, and Fizz1 (markers of M2 macrophages) increased after treatment with low-dose G-CSF plus meloxicam compared with treatment with PBS-treated group, meloxicam alone, and low-dose G-CSF alone, respectively. *p < 0.05, **p < 0.01.

Figure 6

Immunoblots of the optic nerve.…

Figure 6

Immunoblots of the optic nerve. ( A ) Analysis of p-Akt1 expression by…

Figure 6
Immunoblots of the optic nerve. (A) Analysis of p-Akt1 expression by using Western blotting. (B) Quantification of the protein bands of p-Akt1. Each value was normalized to GAPDH. Data are expressed as mean ± SD in each group (n = 6 in each group). The combination treatment induced higher p-Akt1 expression than treatment with meloxicam or low-dose G-CSF in the rAION model. *p < 0.05.
Figure 2
Figure 2
Evaluation of the visual function by using FVEPs in the rAION model. (A) Representative FVEP tracings at four weeks after rAION induction in the sham group, PBS-treated group, meloxicam-treated group, low-dose G-CSF-treated group, and low-dose G-CSF-plus-meloxicam-treated group. (B) Bar charts demonstrate the P1-N2 amplitude. The values of amplitude are expressed as mean ± SD in each group (n = 12 in each group). The amplitudes of the P1-N2 waves in the combination-treated group were significantly higher than meloxicam-treated and low-dose G-CSF-treated groups, respectively. *p < 0.05.
Figure 3
Figure 3
Survival of RGCs in rAION-induced rats with PBS treatment, meloxicam treatment, G-CSF treatment, and G-CSF plus meloxicam treatment at 28 days after rAION induction. (A) A representative of flat-mounted central retinas and the morphometry of RGCs in each group through FluoroGold retrograde labeling at four weeks after rAION induction. (B) RGC density in the central retina in each group. Data are expressed as mean ± SD for each group (n = 12). The number of RGCs in the combination-treated group was 1.58- and 1.45-fold higher than in the meloxicam-treated and low-dose G-CSF-treated groups, respectively. *p < 0.05.
Figure 4
Figure 4
Analysis of RGC apoptosis in the RGC layer through TUNEL assay at four weeks after rAION induction. (A) Representative images of double-stained apoptotic cells in the RGC layers in each group. The apoptotic cells (TUNEL-positive cells) in green were stained with TUNEL staining, and the nuclei of the RGCs in blue were labeled with DAPI staining. (B) Quantification of TUNEL-positive cells per high-power field. Data are expressed as mean ± SD for each group (n = 6). Treatment with low-dose G-CSF plus meloxicam significantly reduced the number of apoptotic RGC by 3.6- and 2.5-fold compared with the meloxicam-treated and low-dose G-CSF-treated groups, respectively. *p < 0.05.
Figure 5
Figure 5
Immunohistochemistry (IHC) of ED1 in the optic nerve at four weeks after rAION induction for evaluating the inflammatory infiltration of macrophages. (A) Representative images of ED1 staining in the longitudinal sections of the optic nerve. The ED1-positive cells in green were stained with FITC, and the nuclei in blue were labeled with DAPI. (B) Quantification of ED1-positive cells per high-power field. Data are expressed as mean ± SD in each group (n = 6). Macrophage recruitment was decreased by 6.75- and 4.1-fold in the combination-treated group compared with the meloxicam-treated and low-dose G-CSF-treated groups, respectively (C) Evaluation of M2 macrophage polarization at four weeks after rAION induction. Relative mRNA expression levels of the markers of M2 macrophages in the optic nerve are shown as histograms. Each value was normalized to CypA. The expression levels of Arg 1, CD206, and Fizz1 (markers of M2 macrophages) increased after treatment with low-dose G-CSF plus meloxicam compared with treatment with PBS-treated group, meloxicam alone, and low-dose G-CSF alone, respectively. *p < 0.05, **p < 0.01.
Figure 6
Figure 6
Immunoblots of the optic nerve. (A) Analysis of p-Akt1 expression by using Western blotting. (B) Quantification of the protein bands of p-Akt1. Each value was normalized to GAPDH. Data are expressed as mean ± SD in each group (n = 6 in each group). The combination treatment induced higher p-Akt1 expression than treatment with meloxicam or low-dose G-CSF in the rAION model. *p < 0.05.

References

    1. Johnson LN, Arnold AC. Incidence of nonarteritic and arteritic anterior ischemic optic neuropathy. Population-based study in the state of Missouri and Los Angeles County, California. J Neuroophthalmol. 1994;14:38–44. doi: 10.1097/00041327-199403000-00011.
    1. Hattenhauer MG, Leavitt JA, Hodge DO, Grill R, Gray DT. Incidence of nonarteritic anterior ischemic optic neuropathy. Am J Ophthalmol. 1997;123:103–107. doi: 10.1016/S0002-9394(14)70999-7.
    1. Lee YC, Wang JH, Huang TL, Tsai RK. Increased risk of stroke in patients with nonarteritic anterior ischemic optic neuropathy: a nationwide retrospective cohort study. Am J Ophthalmol. 2016;170:183–189. doi: 10.1016/j.ajo.2016.08.006.
    1. Levin LA, Louhab A. Apoptosis of retinal ganglion cells in anterior ischemic optic neuropathy. Arch Ophthalmol. 1996;114:488–491. doi: 10.1001/archopht.1996.01100130484027.
    1. Frampton JE, Lee CR, Faulds D. Filgrastim. A review of its pharmacological properties and therapeutic efficacy in neutropenia. Drugs. 1994;48:731–760. doi: 10.2165/00003495-199448050-00007.
    1. Demetri GD, Griffin JD. Granulocyte colony-stimulating factor and its receptor. Blood. 1991;78:2791–2808. doi: 10.1182/blood.V78.11.2791.bloodjournal78112791.
    1. Weaver CH, et al. Syngeneic transplantation with peripheral blood mononuclear cells collected after the administration of recombinant human granulocyte colony-stimulating factor. Blood. 1993;82:1981–1984. doi: 10.1182/blood.V82.7.1981.1981.
    1. Grigg AP, et al. Optimizing dose and scheduling of filgrastim (granulocyte colony-stimulating factor) for mobilization and collection of peripheral blood progenitor cells in normal volunteers. Blood. 1995;86:4437–4445. doi: 10.1182/blood.V86.12.4437.bloodjournal86124437.
    1. Chang CH, Huang TL, Huang SP, Tsai RK. Neuroprotective effects of recombinant human granulocyte colony-stimulating factor (G-CSF) in a rat model of anterior ischemic optic neuropathy (rAION) Exp Eye Res. 2014;118:109–116. doi: 10.1016/j.exer.2013.11.012.
    1. Wen YT, Huang TL, Huang SP, Chang CH, Tsai RK. Early applications of granulocyte colony-stimulating factor (G-CSF) can stabilize the blood-optic-nerve barrier and ameliorate inflammation in a rat model of anterior ischemic optic neuropathy (rAION) Dis Model Mech. 2016;9:1193–1202. doi: 10.1242/dmm.025999.
    1. Tsai RK, Chang CH, Wang HZ. Neuroprotective effects of recombinant human granulocyte colony-stimulating factor (G-CSF) in neurodegeneration after optic nerve crush in rats. Exp Eye Res. 2008;87:242–250. doi: 10.1016/j.exer.2008.06.004.
    1. Tsai RK, Chang CH, Sheu MM, Huang ZL. Anti-apoptotic effects of human granulocyte colony-stimulating factor (G-CSF) on retinal ganglion cells after optic nerve crush are PI3K/AKT-dependent. Exp Eye Res. 2010;90:537–545. doi: 10.1016/j.exer.2010.01.004.
    1. McCullough J, Clay M, Herr G, Smith J, Stroncek D. Effects of granulocyte-colony-stimulating factor on potential normal granulocyte donors. Transfusion. 1999;39:1136–1140. doi: 10.1046/j.1537-2995.1999.39101136.x.
    1. Barner A. Review of clinical trials and benefit/risk ratio of meloxicam. Scand J Rheumatol Suppl. 1996;102:29–37. doi: 10.3109/03009749609097228.
    1. Noble, S. & Balfour, J. A. Meloxicam. Drugs51, 424–430; discussion 431–432 (1996).
    1. Engelhardt G. Pharmacology of meloxicam, a new non-steroidal anti-inflammatory drug with an improved safety profile through preferential inhibition of COX-2. Br J Rheumatol. 1996;35(Suppl 1):4–12. doi: 10.1093/rheumatology/35.suppl_1.4.
    1. Mills CD. M1 and M2 Macrophages: Oracles of Health and Disease. Crit Rev Immunol. 2012;32:463–488. doi: 10.1615/CritRevImmunol.v32.i6.10.
    1. Georgiou T, et al. Neuroprotective Effects of Omega-3 Polyunsaturated Fatty Acids in a Rat Model of Anterior Ischemic Optic Neuropathy. Invest Ophthalmol Vis Sci. 2017;58:1603–1611. doi: 10.1167/iovs.16-20979.
    1. Bernstein SL, Guo Y, Kelman SE, Flower RW, Johnson MA. Functional and cellular responses in a novel rodent model of anterior ischemic optic neuropathy. Investigative ophthalmology & visual science. 2003;44:4153–4162. doi: 10.1167/iovs.03-0274.
    1. Slater BJ, Mehrabian Z, Guo Y, Hunter A, Bernstein SL. Rodent anterior ischemic optic neuropathy (rAION) induces regional retinal ganglion cell apoptosis with a unique temporal pattern. Investigative ophthalmology & visual science. 2008;49:3671–3676. doi: 10.1167/iovs.07-0504.
    1. Furst DE. Meloxicam: selective COX-2 inhibition in clinical practice. Semin Arthritis Rheum. 1997;26:21–27. doi: 10.1016/S0049-0172(97)80049-2.
    1. Monteiro BP, et al. Analgesic efficacy of an oral transmucosal spray formulation of meloxicam alone or in combination with tramadol in cats with naturally occurring osteoarthritis. Veterinary anaesthesia and analgesia. 2016;43:643–651. doi: 10.1111/vaa.12360.
    1. Tiurin VP, Rakov AL. [Efficacy and intolerance of meloxicam in injection and tablet form] Voen Med Zh. 2003;324:54–58.
    1. Tsvetkova ES. [Meloxicam: intramuscular administration in rheumatology] Ter Arkh. 2003;75:96–97.
    1. Park JH, et al. Meloxicam inhibits fipronil-induced apoptosis via modulation of the oxidative stress and inflammatory response in SH-SY5Y cells. J Appl Toxicol. 2016;36:10–23. doi: 10.1002/jat.3136.
    1. Tasaki Y, et al. Meloxicam protects cell damage from 1-methyl-4-phenyl pyridinium toxicity via the phosphatidylinositol 3-kinase/Akt pathway in human dopaminergic neuroblastoma SH-SY5Y cells. Brain Res. 2010;1344:25–33. doi: 10.1016/j.brainres.2010.04.085.
    1. Levkovitch-Verbin H. Retinal ganglion cell apoptotic pathway in glaucoma: Initiating and downstream mechanisms. Prog Brain Res. 2015;220:37–57. doi: 10.1016/bs.pbr.2015.05.005.
    1. Li HB, et al. Long Non-Coding RNA-MALAT1 Mediates Retinal Ganglion Cell Apoptosis Through the PI3K/Akt Signaling Pathway in Rats with Glaucoma. Cell Physiol Biochem. 2017;43:2117–2132. doi: 10.1159/000484231.
    1. Nie XG, et al. Downregulation of microRNA-149 in retinal ganglion cells suppresses apoptosis through activation of the PI3K/Akt signaling pathway in mice with glaucoma. American journal of physiology. Cell physiology. 2018;315:C839–c849. doi: 10.1152/ajpcell.00324.2017.
    1. Bernstein SL, Johnson MA, Miller NR. Nonarteritic anterior ischemic optic neuropathy (NAION) and its experimental models. Prog Retin Eye Res. 2011;30:167–187. doi: 10.1016/j.preteyeres.2011.02.003.
    1. Li L, et al. G-CSF attenuates neuroinflammation and stabilizes the blood-brain barrier via the PI3K/Akt/GSK-3beta signaling pathway following neonatal hypoxia-ischemia in rats. Exp Neurol. 2015;272:135–144. doi: 10.1016/j.expneurol.2014.12.020.
    1. Ghorbani M, et al. G-CSF administration attenuates brain injury in rats following carbon monoxide poisoning via different mechanisms. Environ Toxicol. 2017;32:37–47. doi: 10.1002/tox.22210.
    1. Wei X, et al. Granulocyte Colony-Stimulating Factor Attenuates Blood-Brain Barrier Damage and Improves Cognitive Function in Spontaneously Hypertensive Rats. CNS Neurol Disord Drug Targets. 2017;16:781–788. doi: 10.2174/1871527316666170207155730.
    1. Hesketh, M., Sahin, K. B., West, Z. E. & Murray, R. Z. Macrophage Phenotypes Regulate Scar Formation and Chronic Wound Healing. Int J Mol Sci18, 10.3390/ijms18071545 (2017).
    1. Hofer M, et al. Meloxicam, an inhibitor of cyclooxygenase-2, increases the level of serum G-CSF and might be usable as an auxiliary means in G-CSF therapy. Physiol Res. 2008;57:307–310. doi: 10.33549/physiolres.931237.
    1. Vergadi E, Ieronymaki E, Lyroni K, Vaporidi K, Tsatsanis C. Akt Signaling Pathway in Macrophage Activation and M1/M2 Polarization. J Immunol. 2017;198:1006–1014. doi: 10.4049/jimmunol.1601515.
    1. Liu R, et al. Negative Immune Regulator TIPE2 Promotes M2 Macrophage Differentiation through the Activation of PI3K-AKT Signaling Pathway. PloS one. 2017;12:e0170666. doi: 10.1371/journal.pone.0170666.
    1. Arranz A, et al. Akt1 and Akt2 protein kinases differentially contribute to macrophage polarization. Proc Natl Acad Sci USA. 2012;109:9517–9522. doi: 10.1073/pnas.1119038109.

Source: PubMed

3
Tilaa