<<

Mol. Cells 2014; 37(8): 613-619 http://dx.doi.org/10.14348/molcells.2014.0155 Molecules

and Cells http://molcells.org

Established in 1990

Oncomodulin/Truncated Protamine-Mediated Nogo-66 Small Interference RNA Delivery Promotes Regeneration in Retinal Ganglion Cells

Zhili Cui, Jun Kang1, Dan Hu, Jian Zhou, and Yusheng Wang*

The optic nerve often suffers regenerative failure after in- proteins retained the functions of both OM and tp, and that jury, leading to serious visual impairment such as glauco- OM/tp-NgR siRNA might potentially be used for the treat- ma. The main inhibitory factors, including Nogo-A, oligo- ment of optic nerve injury. dendrocyte myelin glycoprotein, and myelin-associated glycoprotein, exert their inhibitory effects on axonal growth through the same receptor, the Nogo-66 receptor (NgR). INTRODUCTION (OM), a calcium-binding protein with a mole- cular weight of an ~12 kDa, which is secreted from acti- The optic nerve, composed of from retinal ganglion cells vated , has been demonstrated to have high (RGC), often suffers regenerative failure after injury, leading to and specific affinity for retinal ganglion cells (RGC) and serious visual impairment (Frade et al., 1999). However, there is promote greater axonal regeneration than other known still a lack of effective methods for clinical treatment. The optic polypeptide growth factors. Protamine has been reported to nerve belongs to the (CNS) and the effectively deliver small interference RNA (siRNA) into cells. major obstacles to the regeneration of CNS neurons are a defi- Accordingly, a fusion protein of OM and truncated prota- ciency of neurotropic factors and the enrichment of myelin- mine (tp) may be used as a vehicle for the delivery of NgR associated inhibitory molecules (Spencer and Filbin, 2004). Un- siRNA into RGC for gene therapy. To test this hypothesis, like the peripheral nervous system, myelin of the CNS is made we constructed OM and tp fusion protein (OM/tp) expres- up of oligodendroglial cells, which mainly secrete neuronal sion vectors. Using the indirect immunofluorescence labe- growth inhibitors and inhibit the axonal growth of the CNS ling method, OM/tp fusion proteins were found to have a (Schachner, 1994). high affinity for RGC. The gel shift assay showed that the After CNS injury, neurons are in an extracellular environment OM/tp fusion proteins retained the capacity to bind to DNA. full of inhibitory factors. To date, three myelin-associated inhibito- Using OM/tp fusion proteins as a delivery tool, the siRNA of ry molecules of the CNS have been characterized: Nogo-A NgR was effectively transfected into cells and significantly (Chen et al., 2000; GrandPre et al., 2000), oligodendrocyte mye- down-regulated NgR expression levels. More importantly, lin glycoprotein (Pham-Dinh et al., 1994), and myelin-associated OM/tp-NgR siRNA dramatically promoted axonal growth of glycoprotein (McKerracher et al., 1994; Mukhopadhyay et al., RGC compared with the application of OM/tp recombinant 1994); all of these exert their effects through the same receptor, protein or NgR siRNA alone in vitro. In addition, OM/tp-NgR the Nogo-66 receptor (NgR) (Wang et al., 2002a). A previous siRNA highly elevated intracellular cyclic adenosine1mono- study has demonstrated that inhibiting the function of NgR by an phosphate (cAMP) levels and inhibited activation of the Ras antagonist relieves endogenous inhibitory activity and homolog gene family, member A (RhoA). Taken together, promotes axonal growth, implying that NgR plays a central role our data demonstrated that the recombinant OM/tp fusion in the inhibition of axon regeneration in the CNS after injury (GrandPre et al., 2002). Disturbing the intravitreal environment has been found to activate RGC to regenerate axonal growth Department of Ophthalmology, Xijing Hospital, Fourth Military Medical (Berry et al., 1996; Fischer et al., 2001; Leon et al., 2000; Lorber University, Xi'an710032, China, 1Department of Ophthalmology, No. 451 et al., 2005), which has been suggested to be associated with Hospital of PLA, Xi'an 710054, China activation (Yin et al., 2003). It is intriguing that pro- *Correspondence: [email protected] teins secreted by macrophages show a potent effect on axon

regeneration and that this effect is enhanced by elevating intra- Received 9 June, 2014; revised 6 July, 2014; accepted 7 July, 2014; published online 18 August, 2014 cellular cyclic adenosine monophosphate (cAMP) levels (Li et al., 2003). Later, it was demonstrated that among macrophage- Keywords: axon regeneration, Nogo-66 receptor, oncomodulin, retinal derived factors, oncomodulin (OM) is the principal mediator of ganglion cells optic nerve regeneration (Yin et al., 2006). OM is a calcium-

eISSN: 0219-1032 The Korean Society for Molecular and Cellular Biology. All rights reserved. This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/. 613 Mol. Cells http://molcells.org

OM/tp-NgR siRNA Promotes Axonal Regeneration Zhili Cui et al.

binding protein of 108 amino acids, with a molecular mass of sites, respectively. The fragments were obtained and the se- 11.7 kDa (MacManus et al., 1983). OM was originally identified quences were verified, and then subcloned into pET30a plasmid in hepatocellular carcinoma of the rat in the late 1970s, and was to generate the recombinant plasmid pET30a-OM/tp with restric- also found to be expressed in rat fibrosarcoma and mouse sar- tion sites. coma (MacManus, 1979). OM has been reported to have a low The recombinant plasmid pET30a-OM/tp was introduced into expression level in the retina, but is highly expressed upon sti- Escherichia coli Rosetta host cells. Isopropyl--D-thiogalacto- mulation of (Berry et al, 1996). OM shows high and pyranoside (0.1 mM IPTG; Sangon, China) was added and in- specific affinity for its receptor on RGC (Meyer-Franke et al., cubated for 6 h at 35°C to induce target protein expression. Af- 1998). terwards, bacteria were harvested and lysed in phosphate- In the present study, we speculated that OM activation com- buffered saline (PBS) containing 0.2% Triton X-100. The com- bined with the inhibition of neuron inhibitory signals might show a plexes were sonicated, and centrifuged at 15,000  g for 30 min more potent stimulatory effect on axonal growth. Truncated pro- at 4°C. The supernatant, containing recombinant proteins, was tamine (tp), a short type of protamine consisting of 22 amino collected and the fusion proteins were purified using His-Bind acids, contains many basic amino acids and is capable of bind- resin (Ni2+-resin; Novagen, Germany) according to standard ing to nucleic acids (Inoh et al., 2013). Thus, we introduced tp as protocols. The purified proteins were analyzed by 12.5% sodium an agent for the delivery of OM protein and NgR small interfe- dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) rence RNA (siRNA). The direct effect of OM/tp-NgR siRNA on and further verified by Western blot analysis using 6×His rabbit axonal growth in RGC was investigated in vitro. monoclonal ( Technology, USA).

MATERIALS AND METHODS Isolation and culture of rat primary RGC Rat RGC were isolated and cultured from rats, as previously Animals reported (Barres et al., 1988; Winzeler and Wang, 2013) with Eight-week-old Fisher 344 rats (Vital River, China) were housed some modifications. Briefly, neonatal rats at postnatal days 2-3 in clean polypropylene cages at a controlled temperature of 22  were euthanized and disinfected in alcohol (75%). The approx- 2°C and a relative humidity of 55  5% with a 12/12 h light/dark imate retinas were separated from the enucleated eyeballs and cycle. The rats had free access to food and water throughout the treated with papain followed by dissociated into a single cell study. All experimental animal procedures were approved by the suspension. Non-neuronal cells including endothelial cells, ma- Institutional Animal Care and Use Committee of the Fourth Mili- crophages and microglia, and were depleted by using Bandeira tary University (China). All efforts were made to minimize animal simplifolica lectin I and/or anti-macrophage antibody coated- suffering. dishes. RGC were positively purified by using anti-Thy-1 antibo- dies coated-dishes. Then, the Thy-1 positive RGC were col- Isolation and culture of rat macrophages lected and cultured in DMEM containing trypsin (0.25%) for 30 Rats were euthanized by intraperitoneal injection of pentobarbit- min. After digestion, the RGC were washed three times with al sodium (100 mg/kg; Merck KGaA, Germany). Peritoneal ma- DMEM and then transferred into fresh medium containing B27 crophages were obtained by sterile lavage of the abdomen of supplement (1:50; Invitrogen, USA) and penicillin/streptomycin the rats with Hanks’ balanced salt solution. The lavage fluid was (0.2 mg/ml), and maintained at 5% CO2 at 37°C in an incubator. removed from the abdomen and centrifuged at 1,000  g for 8 min. The supernatant was discarded and the cell pellet was Flow cytometry assay collected. Cells were resuspended in Dulbecco’s modified eagle The 6× His monoclonal antibody (Cell Signaling Technology) medium (DMEM) supplemented with 10% fetal bovine serum was dialyzed three times with crosslinking reaction solution (0.05 and 1% penicillin/streptomycin, and cultured at 37°C in 5% CO2. mM NaHCO3) at 4°C. Fluorescein isothiocyanate (FITC) was dissolved in dimethylsulfoxide to a concentration of 1 mg/ml and Cloning, expression, and identification of OM/tp recombi- gently added to the mAb solution at the ratio of 1 mg:150 g nant proteins before being incubated for 8 h at 4°C in the dark. Then, FITC- Macrophages were treated with zymosan (1.25 mg/ml; Sangon, conjugated mAbs were dialyzed and purified by Sephadex G-50 China) and incubated for 8 h at 37°C in 5% CO2. Thereafter, (Seebio, China). A total of 100 mg OM/tp fusion protein was total RNA of macrophages was extracted with TRIzol reagent added and incubated with RGC for 12 h. Thereafter, cells were (Invitrogen, USA) and 5 g of total RNA was reverse-transcribed collected and washed three times in ice-cold PBS and resus- into cDNA using Moloney Murine Leukemia Virus (M-MLV) re- pended in PBS. FITC-conjugated mAbs were added and incu- verse transcriptase (Clontech, USA). The cDNAs were used as bated with RGC for 20 min, followed by two washes with ice-cold templates for cloning of the OM gene. The OM gene was ampli- PBS. Then, the cells were analyzed by flow cytometry assay. fied by polymerase chain reaction (PCR) using the OM forward primer (5-ATAGGATCCATGAGCATCACGGACATCCTGAGC Gel shift assay G-3) and OM reverse primer (5-CCGAAGCTTCGAGAGTGCA The plasmid pET30a (1 g) was mixed with different amounts of CCATTTCCTGGAATTCA-3), which contained NdeI and XhoI OM/tp fusion protein (0, 2, 4, and 8 g) in NaCl solution (0.2 M) restriction sites, respectively. The DNA fragments of OM were at room temperature for 30 min. Thereafter, the mobility of the obtained and verified by gene sequencing (BGI, China). Subse- OM-tp-DNA complex was analyzed by electrophoresis on aga- quently, the purified OM DNA fragments were used as templates rose gels (1%). for cloning of OM/tp using the OM/tp forward primer (5- ATAGGATCCATGAGCATCACGGACATCCTGAGCG-3) and Preparation of OM/tp-siRNA reverse primer (5-CTAAAGCTTAATGCTCCGCC TCCTTCGT- According to the gene sequence of rat NgR, two siRNA targeting CTGCGACTCTTTGTCTCTGGCGGTAATATCTGCTCCGGCT NgR (NgR-1 siRNA, sense: 3-AUUUCCAACAGACGCCGGCC- CTGGCTGCGAAGTGCACCATTTCCTGGAATTCA-3; the se- U-5 and antisense: 3-GCCGGCGUCUGUUGGAAAUGC-5; quence for tp is underlined), containing NdeI and XhoI restriction NgR-2 siRNA, sense: 3-AACAACCUGGCCUCCUGCGGG-5

614 Mol. Cells http://molcells.org

OM/tp-NgR siRNA Promotes Axonal Regeneration Zhili Cui et al.

and antisense: 3-CGCAGGAGGCCAGGUUGUUCC-5) were A designed. Meanwhile, nonspecific sequences (sense: 3-UUCU- CCGAACGUGUCACGUUU-5 and antisense: 3’-ACGUGACA- CGUUCGGAGAAUU-5) were used as a control. The siRNA was synthesized by Shanghai GenePharma Co. Ltd (China). To prepare OM/tp-siRNA, the siRNA and fusion proteins were mixed at a molar ratio of 1:6 (Wen et al., 2007) in 500 l of DMEM and incubated for 30 min at room temperature. Then, the mixture was added to the RGC and incubated for the indicated B times.

Neurite outgrowth assays RGC were fixed with paraformaldehyde (4%) and stained with growth-associated protein 43 (GAP-43) mAbs. Neurite out- growth was measured in images taken by the Olympus FV300 laser scanning confocal microscope (Japan). Approximately 60 cells were randomly selected in each group and the average axon length of the cells was measured using the Image-Pro Plus 6.0 software (Media Cybernetics Inc., USA). Data are expressed as mean  standard deviation (SD) from three independent ex- periments.

Western blot analysis A total of 20  was separated by 12% SDS-PAGE fol- lowed by electro-blotting onto a nitrocellulose membrane (Amer- sham, UK). Then, the membrane was incubated with 2% non-fat dry milk in Tris-buffered saline to block nonspecific binding at room temperature for 1 h. Next, the membrane was incubated with primary (rabbit monoclonal RhoA antibody (1:500), Santa Cruz Biotechnology, USA; rabbit polyclonal NgR Fig. 1. Expression and characterization of OM/tp fusion protein. (A) antibody (1:600), Abcam, UK; 6× His rabbit monoclonal antibody Schematic structure of pET30a-OM/tp recombinant plasmid. (B) (1:800), Cell Signaling Technology) and diluted in the blocking SDS-PAGE and Western blot analysis of OM/tp recombinant protein. buffer overnight at 4°C. Subsequently, the membrane was incu- M, protein markers; lane 1, total bacterial protein without induction; bated with horseradish peroxidase (HRP)-conjugated goat anti- lane 2, total bacterial protein after induction by IPTG (0.1 mM); lane 3, rabbit immunoglobulin G (Boster Corporation, China) diluted the supernatant from IPTG-induced total bacterial protein; lane 4, 1:1000 in the blocking buffer for 1 h. Finally, the HRP substrate lysates of inclusion bodies from IPTG-induced total bacterial protein; 4-chloro-1-naphthol (4-CN) was used for protein visualization. lane 5, purified OM/tp recombinant protein; lane 6, Western blot analysis of purified OM/tp recombinant protein. Measurements of active RhoA and cAMP levels The activity of RhoA was measured according to a previously described method (Noren et al., 2000). Briefly, cells were har- vested and washed with ice-cold 4-(2-hydroxyethyl)-1- A p value of less than 0.05 was considered statistically signifi- piperazineethanesulfonic acid (HEPES)-buffered saline (pH 7.4). cant. All statistical analyses were performed using SPSS version Then, cells were lysed in Buffer A, containing Tris (50 mM, pH 11.5 (SPSS Inc., USA). 7.6), NaCl (500 mM), MgCl2 (10 mM), orthovanadate (100 M), deoxycholate (0.5%), Triton X-100 (1%), and SDS (0.1%), sup- RESULTS plemented with inhibitors (100 M). After centrifugation, supernatants were collected and divided into two equal samples. Expression and characterization of OM/tp fusion protein One sample was incubated with glutathione S-transferase To generate recombinant OM/tp proteins, the DNA fragment of (GST)-Rho-binding domain (RBD) beads followed by washing OM/tp was amplified and subcloned into pET30a plasmid with three times with ice-cold Buffer B containing Tris (50 mM, pH NdeI and XhoI restriction sites to obtain pET30a recombinant 7.6), NaCl (150 mM), MgCl2 (0.5 mM), orthovanadate (100 M), plasmid (Fig. 1A). The pET30a-OM/tp recombinant plasmids and Triton X-100 (1%), supplemented with protease inhibitors. were then transformed into E. coli Rosetta host cells, and the The bound fraction, containing active GTP-RhoA, was collected expression of recombinant OM/tp proteins was induced by IPTG. for further analysis. The other sample of whole cell lysate, with As expected, the results showed that OM/tp fusion proteins had an equal volume, was used for total RhoA analysis. Protein le- an approximate molecular mass of 21 kDa and were mainly vels of total RhoA and active RhoA were analyzed by Western present in the supernatants (Fig. 1B, lanes 1-4). The OM/tp fu- blotting. Intracellular cAMP levels were assessed by an EIA sion proteins were further purified by Ni2+-resin affinity chroma- cAMP ELISA (Assay Designs, USA), according to the manu- tography and were detected by anti-6× His antibody (Fig. 1B, facturer’s instructions. lanes 5 and 6). These results indicated that OM/tp fusion pro- teins were successfully generated in a soluble expression man- Statistical analysis ner. Data are presented as mean  SD. Comparisons between two groups and among multiple groups were performed by Student t-test and one-way analysis of variance (ANOVA), respectively.

http://molcells.org Mol. Cells 615

OM/tp-NgR siRNA Promotes Axonal Regeneration Zhili Cui et al.

A B C

Fig. 2. Flow cytometry assay to determine the binding capacity of OM/tp to RGC. Cells were treated with PBS (A), recombinant OM (B), or re- combinant OM/tp (C). FITC-conjugated 6× His mAbs were added and the fluorescence intensity was determined by flow cytometry assay.

A Fig. 3. Assessment of efficiency of OM/tp in delivering nucleic acid. (A) The DNA-binding ability of recombinant OM/tp was determined by gel shift assay. Different amounts (0, 2, 4, and 8 g) of OM/tp were incubated with pET30a plasmid (1 g) for 30 min and then the mixtures B were analyzed by agarose gel electrophoresis. qRT-PCR (B) and Western blotting (C) were used to analyze the knockdown efficiency of NgR-1 and NgR-2 siRNA. Nonspecific siRNA (Non) was used as control. *P < 0.05 vs Non or C NgR-2 siRNA group. qRT-PCR (D) and West- ern blot (E) were used to analyze the capacity of OM/tp to deliver siRNA to the target gene. Cells were treated with recombinant OM/tp, NgR-1 siRNA without lipofectamine, NgR-1 siRNA mixed with lipofectamine, OM/tp-NgR-1 siRNA, and OM/tp-nonspecific siRNA, respec- tively. *p < 0.05 vs OM/tp, NgR-1, and OM/tp-

Non groups. #p > 0.05 denotes no significant D difference.

E

Recombinant OM/tp protein highly binds to RGC showed 94.3% positive cells (Fig. 2B). Similarly, recombinant To determine whether the recombinant OM/tp retained the ca- OM/tp-treated cells showed 90.1% positive cells (Fig. 2C). The pacity to target RGC, RGC were treated with OM/tp fusion pro- data implied that the OM/tp fusion protein retained the capacity tein. FITC-conjugated 6× His mAbs were added to the cells and of the OM protein to bind to RGC through a . fluorescence was measured by flow cytometry. Compared with the control group treated with PBS, which showed 0.4% positive cells (Fig. 2A), the cells treated with His-tagged recombinant OM

616 Mol. Cells http://molcells.org

OM/tp-NgR siRNA Promotes Axonal Regeneration Zhili Cui et al.

A B C

D E F

Fig. 4. Effect of OM/tp-NgR siRNA on axon regeneration of RGC. RGC were pre-incubated with forskolin (100 M) for 6 h. Then the cells were treated with PBS (A), OM/tp (B), NgR siRNA (C), OM/tp-NgR siRNA (D), or OM/tp-nonspecific siRNA (E) and incubated for 72 h. Cells were immunostained with GAP-43 antibody and observed by laser scanning confocal microscopy. (F) The average axon length of the cells was measured by Image-Pro Plus 6.0 software. *p < 0.05 and **p < 0.01.

OM/tp-NgR siRNA significantly knocks-down NgR expres- forskolin and then treated with OM/tp, NgR siRNA, OM/tp-NgR sion in RGC siRNA, and OM/tp-nonspecific siRNA. Three days later, the Protamine is capable of binding to DNA and carrying DNA length of the axons was measured. Compared with the control through the into to the cell interior (Junghans et group, which was treated with PBS (30.52  5.48 m), the aver- al., 2000). To investigate whether the generated recombinant age axonal length was longer in cells treated with OM/tp recom- OM/tp could bind to DNA, pET30a plasmid was mixed with dif- binant proteins (68.27  8.36 m) and in cells treated with NgR ferent amounts (0, 2, 4, and 8 g) of OM/tp, and the mobility was siRNA without OM/tp recombinant proteins (51.35  6.41 m). analyzed by agarose gel electrophoresis. The results showed As expected, in the OM/tp-NgR siRNA-treated group, RGC that plasmid DNA mixed with OM/tp moved slowly in a dose- showed a potent increase in the average axonal length (101.38 dependent manner, implying the formation of a DNA/OM/tp  15.70 m) compared with the control group (p < 0.01), OM/tp complex (Fig. 3A). group (p < 0.05), and NgR siRNA group (p < 0.05). However, Next, we synthesized two siRNA, NgR-1 and NgR-2, which RGC treated with OM/tp-nonspecific siRNA showed an average targeted the NgR gene sequence. NgR-1 and NgR-2 siRNA axonal length of 66.56  10.45 m, which was similar to that of were then transfected into RGC to determine their knockdown the OM/tp recombinant group but shorter than that of the OM/tp- efficiency. Compared with nonspecific siRNA-treated cells, NgR- NgR siRNA group (p < 0.05). The data demonstrated that 1 siRNA-treated cells showed a significant decrease of NgR OM/tp-NgR siRNA had a potent stimulatory effect on axonal mRNA (Fig. 3B). The decrease of NgR protein was confirmed by growth in RGC (Figs. 4A-4F). Western blot analysis (Fig. 3C). However, NgR-2 siRNA had no effect on NgR expression. The data verified that NgR-1 siRNA OM/tp-NgR siRNA markedly increases axon regeneration specifically targeted the NgR gene. signals To further determine whether OM/tp carrying siRNA was capa- To further explore the effect of OM/tp-NgR siRNA on axon ble of passing through the cell membrane to knockdown the target regeneration signals, we first measured the changes in intra- gene, recombinant OM/tp was mixed with siRNA, and was then cellular cAMP levels upon treatment with OM/tp, NgR siRNA, transfected into cells. The results showed that NgR mRNA ex- OM/tp-NgR siRNA, and OM/tp-nonspecific siRNA. Intracellular pression was significantly decreased in the NgR-1 siRNA mixed cAMP levels were increased in OM/tp- and NgR siRNA-treated with lipofectamine group and OM/tp-NgR-1 siRNA group com- groups, which were 16.25  1.32 pmol/L and 12.68  1.17 pared with the OM/tp group, NgR-1 siRNA without lipofectamine pmol/L, respectively, compared with the levels in the control group, and OM/tp-nonspecific siRNA group (Fig. 3D). There was group (7.24  1.52 pmol/L). The OM/tp-NgR siRNA-treated no significant difference between the NgR-1 siRNA with lipofecta- group showed higher levels of cAMP (20.43  1.91 pmol/L) mine group and the OM/tp-NgR-1 siRNA group, implying that compared with the control group (p < 0.01), OM/tp group (p < OM/tp was an efficient agent for siRNA delivery. These results 0.05), and NgR siRNA group (p < 0.05). However, the OM/tp- were further confirmed by Western blot analysis (Fig. 3E). nonspecific siRNA-treated group showed lower levels of cAMP (15.86  1.86 pmol/L) than the OM/tp-NgR siRNA-treated OM/tp-NgR siRNA significantly promotes axonal growth in group (Fig. 5A). Next, we assessed the activity of RhoA. Simi- RGC larly, the activation of RhoA was significantly inhibited in the To investigate whether OM/tp-NgR siRNA was able to stimulate OM/tp-NgR siRNA group compared with the other groups (Figs. RGC to extend axons in culture, RGC were pre-incubated with 5B and 5C).

http://molcells.org Mol. Cells 617

OM/tp-NgR siRNA Promotes Axonal Regeneration Zhili Cui et al.

B

C A

Fig. 5. Effect of OM/tp-NgR siRNA on axon regeneration signals. (A) Detection of intracellular levels of cAMP. (B) Measurement of active RhoA and total RhoA by Western blot analysis (C). Protein levels were analyzed by Image-Pro Plus 6.0 software and relative RhoA activity is pre- sented as active RhoA normalized to total RhoA. *p < 0.05 and **p < 0.01.

DISCUSSION nificantly increased the average length of axons compared with the application of OM/tp recombinant protein or NgR siRNA Here, we studied for the first time the effect of tp carrying OM alone. These results implied that OM/tp-NgR siRNA had a po- and NgR siRNA on axonal growth in RGC. We found that tent stimulatory effect on axonal growth in RGC. OM/tp-NgR siRNA efficiently promoted axonal growth in RGC in Most recently, , the first responders of the innate vitro. OM had a stimulatory effect on axonal growth and NgR immune system, were demonstrated to express OM and mar- siRNA-mediated NgR gene silencing blocked the transduction of kedly stimulate optic nerve regeneration (Kurimoto et al., 2013). inhibitory signals. The data further implied that OM plays an important role in By expressing recombinant DNA, we successfully obtained a axonal growth. However, the mechanism underlying the regula- fusion protein of OM/tp. We then showed that the OM/tp recom- tion of neuronal growth and regeneration by OM remains poorly binant protein retained the capacity of OM protein to bind to understood. Yin et al. (2006) proposed that the effect of OM in RGC through a cell surface receptor. Yin et al. (2006) described promoting greater axon outgrowth is associated with a calmodu- that the OM protein binds strongly to RGC in the presence of lin-dependent II-dependent pathway. Neurotrophic fac- elevated intracellular cAMP stimulated by forskolin. They further tors overcome the inhibitory effects of myelin by elevating cAMP suggested that OM shows high affinity and specific binding to levels (Fischer et al., 2004). It is reported that OM exerts its ef- RGC, which are cAMP-dependent. However, the receptor for fects in a cAMP-dependent manner and can further elevate OM has not been identified to date. It has been proposed that intracellular cAMP levels (Yin et al., 2006). cAMP induces the receptor to translocate from the cytosol to the Inhibitory signals transduced into cells through NgR activates cell surface, which is necessary for OM binding (Meyer-Franke RhoA, which causes myosin depolymerization and et al., 1998). collapse, and ultimately leads to axon regeneration failure [15]. Protamine is now taken as an ideal agent for the delivery of Thus, blocking the interaction of NgR with inhibitory molecules DNA into cells, as it can condense DNA into compact particles, can suppress the activation of RhoA. Also, disrupting the interac- accelerating DNA uptake and increasing transfection efficiency tion of NgR with the p75 receptor complex blocked the transduc- (You et al., 1999). Furthermore, protamine is also capable of tion of inhibitory signals into the interior of responding neurons binding to siRNA to form a protein/siRNA complex, delivering (Wang et al., 2002b). These reports suggest that NgR plays a siRNA into various types of cells. Additionally, the protamine- central role in the regulation of inhibitory . based systemic delivery is now considered to be a safe and Intracellular cAMP has been suggested to activate protein ki- reliable method for siRNA delivery (Choi et al., 2010; Peer et al., nase A, which induces the phosphorylation and inactivation of 2007). Here, we found that OM/tp recombinant protein carrying RhoA (Ellezam et al., 2002). Here, we found that OM activation NgR siRNA efficiently silenced NgR gene expression in RGC. combined with NgR knockdown significantly elevated cAMP The data suggest that our OM/tp fusion proteins not only bound levels and inhibited RhoA activation in RGC compared with the to RGC with high affinity, but were also capable of delivering application of OM/tp recombinant protein or NgR siRNA alone. siRNA to knockdown the target gene. To further investigate the Nogo-A, one of the myelin-associated inhibitory factors which application of OM/tp-NgR siRNA as an effective drug for axon directly interacts with NgR, is also found in the central and peri- regeneration, we evaluated the effect of OM/tp-NgR siRNA on pheral neurons during development (Caroni and Schwab, 1988; RGC in vitro. Because forskolin promoted interaction of OM with Chen et al., 2000). Nogo-A is also expressed in the neonatal rat its receptor (Yin et al., 2006), we pretreated RGC with forskolin visual system during development (Huo et al., 2011). Huo et al. and then added OM/tp-NgR siRNA to investigate its effect on (2013) demonstrated that Nogo-A was highly expressed in RGC axonal growth. The results showed that OM/tp-NgR siRNA sig- in which knockdown of Nogo-A by siRNA significantly promoted

618 Mol. Cells http://molcells.org

OM/tp-NgR siRNA Promotes Axonal Regeneration Zhili Cui et al.

axonal length of RGC. Therefore, the ligands of NgR such as Junghans, M., Kreuter, J., and Zimmer, A. (2000). Antisense delivery Nogo-A can be expressed in RGC and may exert an inhibitory using protamine-oligonucleotide particles. Nucleic Acids Res. 28, effect on axonal growth through NgR. Accordingly, we demon- E45. Kurimoto, T., Yin, Y., Habboub, G., Gilbert, H.Y., Li, Y., Nakao, S., strated that the knockdown of NgR alone by siRNA promoted Hafezi-Moghadam, A., and Benowitz, L.I. (2013). Neutrophils ex- axonal growth of RGC. OM exerted its effect by interacting with press oncomodulin and promote optic nerve regeneration. J. its receptor on RGC to promote axonal growth. In addition, NgR Neurosci. 33, 14816-14824. siRNA specifically inhibited the transduction of inhibitory signals. Leon, S., Yin, Y., Nguyen, J., Irwin, N., and Benowitz, L.I. (2000). Taken together, OM/tp-NgR siRNA dramatically promoted Lens injury stimulates axon regeneration in the mature rat optic nerve. J. Neurosci. 20, 4615-4626. axonal growth in RGC. It appears that OM combined with NgR Li, Y., Irwin, N., Yin, Y., Lanser, M., and Benowitz, L.I. (2003). Axon knockdown can enhance the stimulatory effect on axonal growth. regeneration in goldfish and rat retinal ganglion cells: differential Thus, the OM/tp-NgR siRNA designed provided novel insight for responsiveness to carbohydrates and cAMP. J. Neurosci. 23, the potential of gene therapy of optic nerve injury. As OM/tp-NgR 7830-7838. siRNA might potentially be used for clinical therapy, we are in- Lorber, B., Berry, M., and Logan, A. (2005). Lens injury stimulates terested in further evaluating its safety and biological activity in adult mouse retinal ganglion cell axon regeneration via both ma- crophage- and lens-derived factors. Eur. J. Neurosci. 21, 2029- vivo. Further investigation of OM/tp-NgR siRNA in animal mod- 2034. els will be carried out in our laboratory. MacManus, J.P. (1979). Occurrence of a low-molecular-weight cal- cium-binding protein in neoplastic liver. Cancer Res. 39, 3000- ACKNOWLEDGMENTS 3005. This study was supported by grants from the National Natural MacManus, J.P., Watson, D.C., and Yaguchi, M. (1983). The com- Science Foundation of China (No. 30700930). plete amino acid sequence of oncomodulin--a -like calcium-binding protein from Morris hepatoma 5123tc. Eur. J. Bi- ochem. 136, 9-17. REFERENCES McKerracher, L., David, S., Jackson, D.L., Kottis, V., Dunn, R.J., and Braun, P.E. (1994). Identification of myelin-associated glycoprote- Barres, B.A., Silverstein, B.E., Corey, D.P., and Chun, L.L. (1988). in as a major myelin-derived inhibitor of neurite growth. Neuron Immunological, morphological, and electrophysiological variation 13, 805-811. among retinal ganglion cells purified by panning. Neuron 1, 791- Meyer-Franke, A., Wilkinson, G.A., Kruttgen, A., Hu M., Munro, E., 803. Hanson, M.G., Jr., Reichardt, L.F., and Barres, B.A. (1998). De- Berry, M., Carlile, J., and Hunter, A. (1996). Peripheral nerve ex- polarization and cAMP elevation rapidly recruit TrkB to the plas- plants grafted into the vitreous body of the eye promote the rege- ma membrane of CNS neurons. Neuron 21, 681-693. neration of retinal ganglion cell axons severed in the optic nerve. Mukhopadhyay, G., Doherty, P., Walsh, F.S., Crocker, P.R., and J. Neurocytol. 25, 147-170. Filbin, M.T. (1994). A novel role for myelin-associated glycoprote- Caroni, P., and Schwab, M.E. (1988). Antibody against myelin- in as an inhibitor of axonal regeneration. Neuron 13, 757-767. associated inhibitor of neurite growth neutralizes nonpermissive Noren, N.K., Liu, B.P., Burridge, K., and Kreft, B. (2000). p120 cate- substrate properties of CNS white matter. Neuron 1, 85-96. nin regulates the actin via Rho family GTPases. J. Chen, M.S., Huber, A.B., van der Haar, M.E., Frank, M., Schnell, L., Cell Biol. 150, 567-580. Spillmann, A.A., Christ, F., and Schwab, M.E. (2000). Nogo-A is a Peer, D., Zhu, P., Carman, C.V., Lieberman, J., and Shimaoka, M. myelin-associated neurite outgrowth inhibitor and an antigen for (2007). Selective gene silencing in activated leukocytes by target- monoclonal antibody IN-1. Nature 403, 434-439. ing siRNAs to the lymphocyte function-associated anti- Choi, Y.S., Lee, J.Y., Suh, J.S., Kwon, Y.M., Lee, S.J., Chung, J.K., gen-1. Proc. Natl. Acad. Sci. USA 104, 4095-4100. Lee, D.S., Yang, V.C., Chung, C.P., and Park, Y.J. (2010). The Pham-Dinh, D., Allinquant, B., Ruberg, M., Della Gaspera, B., Nuss- systemic delivery of siRNAs by a cell penetrating peptide, low baum, J.L., and Dautigny, A. (1994). Characterization and ex- molecular weight protamine. Biomaterials 31, 1429-1443. pression of the cDNA coding for the human mye- Ellezam, B., Dubreuil, C., Winton, M., Loy, L., Dergham, P., Selles- lin/oligodendrocyte glycoprotein. J. Neurochem. 63, 2353-2356. Navarro, I., and McKerracher, L. (2002). Inactivation of intracellu- Schachner, M. (1994). Neural recognition molecules in disease and lar Rho to stimulate axon growth and regeneration. Prog. Brain regeneration. Curr. Opin. Neurobiol. 4, 726-734. Res. 137, 371-380. Spencer, T., and Filbin, M.T. (2004). A role for cAMP in regeneration Fischer, D., Heiduschka, P., and Thanos, S. (2001). Lens-injury- of the adult mammalian CNS. J. Anat. 204, 49-55. stimulated axonal regeneration throughout the optic pathway of Wang K.C., Koprivica V., Kim J.A., Sivasankaran R., Guo Y., Neve adult rats. Exp. Neurol. 172, 257-272. R.L., and He Z. (2002a). Oligodendrocyte-myelin glycoprotein is a Fischer, D., Petkova, V., Thanos, S., and Benowitz, L.I. (2004). Nogo receptor that inhibits neurite outgrowth. Nature 417, Switching mature retinal ganglion cells to a robust growth state in 941-944. vivo: gene expression and synergy with RhoA inactivation. J. Wang, K.C., Kim, J.A., Sivasankaran, R., Segal, R., and He, Z. Neurosci. 24, 8726-8740. (2002b). P75 interacts with the Nogo receptor as a co-receptor Frade, J.M., Bovolenta P., and Rodriguez-Tebar, A. (1999). Neuro- for Nogo, MAG and OMgp. Nature 420, 74-78. trophins and other growth factors in the generation of retinal neu- Wen, W.H., Liu, J.Y., Qin, W.J., Zhao, J., Wang, T., Jia L.T., Meng, rons. Microsc. Res. Tech. 45, 243-251. Y.L., Gao H., Xue, C.F., Jin, B.Q., et al. (2007). Targeted inhibi- GrandPre, T., Nakamura, F., Vartanian, T., and Strittmatter, S.M. tion of HBV gene expression by single-chain antibody mediated (2000). Identification of the Nogo inhibitor of axon regeneration as small interfering RNA delivery. Hepatology 46, 84-94. a Reticulon protein. Nature 403, 439-444. Winzeler, A., and Wang, J.T. (2013). Purification and culture of retin- GrandPre, T., Li, S., and Strittmatter, S.M. (2002). Nogo-66 receptor al ganglion cells from rodents. Cold Spring Harb. Protoc. 2013, antagonist peptide promotes axonal regeneration. Nature 417, 643-652. 547-551. Yin, Y., Cui, Q., Li Y., Irwin, N., Fischer, D., Harvey, A.R., and Beno- Huo, Y., Yuan, R.D., Ye, J., Yin, X.L., and Zou, H. (2011). Expres- witz L.I. (2003). Macrophage-derived factors stimulate optic nerve sion of Nogo-A and Nogo receptor in neonatal rats visual system regeneration. J. Neurosci. 23, 2284-2293. during development. Zhonghua Yan Ke Za Zhi 47, 54-58. Yin, Y., Henzl, M.T., Lorber, B., Nakazawa, T., Thomas, T.T., Jiang, Huo, Y., Yin, X.L., Ji, S.X., Zou, H., Lang, M., Zheng, Z., Cai, X.F., F., Langer, R., and Benowitz, L.I. (2006). Oncomodulin is a ma- Liu, W., Chen, C.L., Zhou, Y.G., et al. (2013). Inhibition of retinal crophage-derived signal for axon regeneration in retinal ganglion ganglion cell axonal outgrowth through the Amino-Nogo-A signal- cells. Nat. Neurosci. 9, 843-852. ing pathway. Neurochem. Res. 38, 1365-1374. You, J., Kamihira, M., and Iijima, S. (1999). Enhancement of trans- Inoh, Y., Furuno, T., Hirashima, N., Kitamoto, D., and Nakanishi, M. fection efficiency by protamine in DDAB lipid vesicle-mediated (2013). Synergistic effect of a biosurfactant and protamine on gene transfer. J. Biochem. 125, 1160-1167. gene transfection efficiency. Eur. J. Pharm. Sci. 49, 1-9.

http://molcells.org Mol. Cells 619