Open Access

Role of nitric oxide synthase in the development of bone cancer pain and effect of L‑NMMA

  • Authors:
    • Yan Yang
    • Juan Zhang
    • Yue Liu
    • Yaguo Zheng
    • Jinhua Bo
    • Xiaofang Zhou
    • Junhua Wang
    • Zhengliang Ma
  • View Affiliations

  • Published online on: December 7, 2015     https://doi.org/10.3892/mmr.2015.4647
  • Pages: 1220-1226
  • Copyright: © Yang 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

Spinal nitric oxide is involved in the mechanisms of pain generation and transmission during inflammatory and neuropathic pain. The aim of the present study was to explore the role of spinal nitric oxide in the development of bone cancer pain. 2x105 osteosarcoma cells were implanted into the intramedullary space of right femurs of C3H/HeJ mice to induce a model of ongoing bone cancer. Polymerase chain reaction and immunohistochemical analyses were performed to assess the expression of neuronal nitric oxide synthase (nNOS) and inducible (i)NOS in the spinal cord following inoculation. The results showed that inoculation of osteosarcoma cells induced progressive bone cancer, accompanied with pain‑associated behavior. The levels of nNOS mRNA in the spinal cord of tumor mice began to increase at day 10 and then decreased to the level in sham mice at day 14, while iNOS mRNA markedly increased in the tumor group at days 10 and 14. Immunohistochemical analysis showed that nNOS‑ and iNOS‑positive neurons were mainly located in the superficial dorsal horn and around the central canal of the L3‑L5 spinal cord. Intrathecal injection of 50 µg NOS inhibitor NG‑monomethyl‑l‑arginine (L‑NMMA) attenuated cancer‑evoked pain behaviors at day 14. These findings indicated that an upregulation of nNOS and iNOS in the spinal cord is associated with bone cancer pain and suggests that exogenously administered L‑NMMA may have beneficial effects to alleviate bone cancer pain.

Introduction

Bone cancer pain is a type of severe and chronic pain that affects the quality of life in cancer patients; however, no breakthrough regarding the elucidation of underlying mechanisms and the development of therapeutics for bone cancer pain has been achieved (1). Studies have indicated that nitric oxide (NO) is an important neurotransmitter contributing to the development and maintenance of central and peripheral sensitization in inflammatory (2) and neuropathic pain (3).

NO is synthesized from L-arginine via a family of nitric oxide synthases (NOS), which are key enzymes in NO biosynthesis and comprise neuronal NOS (nNOS), endothelial NOS (eNOS) and inducible NOS (iNOS). The pathophysiological functions of NO are regulated by the expression and activity of these isoforms (4,5). nNOS, expressed in the nervous system, produces NO in neurons and has been shown to contribute to spinal nociceptive processing in several pain models (612). iNOS, which is not constitutively expressed and must be induced to be synthesized, is abundant in a variety of cell types, including glial cells, macrophages, chondrocytes and neutrophils (13). Previous studies demonstrated that iNOS is also involved in the mechanisms of inflammatory, neuropathic pain (5,14) as well as herpetic allodynia (15).

Bone cancer pain is thought to have inflammatory, neuropathic and tumorigenic components. A previous study by our group has shown that NR2B-containing N-methyl-d-aspartate (NMDA) receptor has a critical role in spinal nociceptive processing during bone cancer pain (16). Activation of NMDA receptors allows Ca2+ to enter neurons. Increased intracellular Ca2+ triggers a cascade of events that includes activation of nNOS, leading to the production of NO in the spinal cord (17). Another previous study by our group also demonstrated that upregulation of NR2B and nNOS expression in the spinal dorsal horn contributes to hyperalgesia induced by chronic compression of dorsal root ganglia (18). However, the role of NOS in the development of bone cancer pain has remained elusive.

The present study used a murine model of osteosar-coma-associated bone cancer pain, to explore whether NOS in the spinal L3-L5 segments is responsible for the development of bone cancer pain and to determine the efficacy of NG-monomethyl-l-arginine (L-NMMA) administration in alleviating bone cancer pain.

Materials and methods

Animals

Experiments were approved by the Animal Care and Use Committee of the Medical School of Nanjing University (Nanjing, China) and were in accordance with the guidelines for the use of laboratory animals (19). The numbers of animals and their suffering were minimized in all cases. Male C3H/HeJ mice (n=100; weight, 20–22 g; 4–6 weeks old), purchased from the Model Animal Research Center of Nanjing University (Nanjing, China), were housed in a temperature-controlled (21±1°C) room with a 12-h light/dark cycle with access to food and water ad libitum. Each group used to analyze behavior contained 8 mice, and each group in PCR or immunohisto-chemistry study contained 5 mice. The mice subjected to the behavioral studies were not processed for PCR/immunohistochemistry, and were euthanized after the last behavioral test.

Cell culture and tumor-cell inoculation

The NCTC 2472 osteosarcoma cell line (no. 2087787; American Type Culture Collection, Manassas, VA, USA) was used in the present study. The cells were incubated in NCTC 135 medium (Sigma-Aldrich, St. Louis, USA) containing 10% horse serum (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) in a humidified atmosphere containing 5% CO2 at 37°C.

The mouse model of bone cancer was established according to the method of previous studies by our and another group (16,20). Briefly, 1% pentobarbital sodium (50 mg/kg; Sigma-Aldrich) in normal saline [Baxter Healthcare (Tianjin) Co. Ltd., Tianjin, China] was administered to the mice by intraperitoneal injection and the right femur condyle was perforated with a 30-gauge needle (Jiangsu Zhengkang Medical Equipment Co., Ltd., Changzhou, China). Subsequently, 2×105 NCTC 2472 cells in 20 µl α-minimum essential medium (Thermo Fisher Scientific, Inc.) were injected into the intra-medullary space of the femur. In the sham group, the medium contained no cells. Subsequently, the injection hole was sealed with dental amalgam (AT&M Biomaterials Co., Ltd., Beijing, China), followed by copious irrigation with normal saline and closing of the wound.

In the interaction studies, mice were intrathecally administered 50 µg L-NMMA (Cayman Chemical Company, Ann Arbor, Michigan, USA) dissolved in artificial cerebrospinal fluid (10 µg/µl; 5 µl per mouse; Harvard Apparatus, Holliston, MA, USA) at post-operative day 14, at which pain-associated behavior was observed. Mice in the sham group received vehicle treatment. Intrathecal injections were performed free-hand between the L5 and L6 lumbar space in unanesthetized male mice according to the method of Hylden and Wilcox (21). Pain-associated behavior was assessed prior to, as well as at 2, 12 and 24 h after L-NMMA administration.

The mice assigned for behavioral tests were sacrificed after the last measurement. The mice were deeply anesthetized with pentobarbital (50 mg/kg, intraperitoneally) and then sacrificed by cervical dislocation. The mice assigned for Reverse-transcription quantitative polymerase chain reaction (RT-qPCR) were individually sacrificed at post-operative days 7, 10 or 14, and the spinal cord segments were immediately removed. The mice assigned for immunohistochemical staining were also deeply anesthetized with pentobarbital individually at post-operative days 7, 10 or 14, and then transcardially perfused with saline followed by 4% paraformaldehyde (PFA; Beijing Dingguo Changsheng Biotechnology Co., Ltd., Beijing, China).

Assessment of pain-associated behavior

Withdrawal thresholds as well as latency to mechanical and thermal stimulation of mice were examined prior to surgery as well as at post-operative days 3, 5, 7, 10 and 14. Mice were allowed to acclimatize for at least 30 min prior to each test.

Mechanical allodynia

The paw withdrawal mechanical threshold (PWMT) was assessed according to the method by Chaplan et al (22), which was also used in a previous study by our group (16). Briefly, a set of von Frey filaments (0.16, 0.4, 0.6, 1.0, 1.4 and 2.0g; Stoelting, Wood Dale, IL, USA) was used to vertically push against the plantar surface of the right hind paw. Each mouse was tested five times per stimulus force. The lightest von Frey filament that evoked three or more brisk withdrawal or paw flinching reactions was regarded as the PWMT.

Thermal hyperalgesia

The paw withdrawal thermal latency (PWTL) was measured according to a method of a previous study by our group (23) using a radiant thermal stimulator (BME410A; Institute of Biological Medicine, Academy of Medical Science, Tianjin, China) that focused onto the plantar surface of the hind paw. The PWTL was defined as the latency of the mice to lift or lick their hind paw. Each mouse was tested five times with a 5-min interval. A cut-off time of 20 sec was used to avoid tissue damage. The mean PWTL was obtained from the latter three stimuli.

RT-qPCR

Following the sacrifice of the mice, the L3-L5 lumbar spinal cord segments were immediately harvested, frozen in liquid nitrogen and stored at -80°C. The spinal tissues were homogenized prior to storage by grinding in liquid nitrogen with a small mortar. Total RNA was isolated and purified using TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.) following the manufacturer's instructions. RT was performed using a Moloney murine leukemia virus reverse transcriptase kit (Promega Corp., Madison, WI, USA). The generated cDNA was then used as a template for PCR amplification with Taq DNA polymerase and ROX reference dye provided by the SYBR Premix Ex Taq kit (DRR041A; Takara, Dalian, China) using StepOnePlus RT-PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.). β-actin, nNOS and iNOS were amplified using specific oligonucleotide primers: forward, 5′-AGGAGCAAGGAGGCCATATT-3′ and reverse, 5′-AACACACCAGCATCCTCCTC-3′ for nNOS; forward, 5′-TGATGTGCTGCCTCTGGTCT-3′ and reverse, 5′-ACTTCCTCCAGGATGTTGTA-3′ for iNOS; and forward, 5′-GAGACCTTCAACACCCCAGC-3′ and reverse, 5′-CACAGAGTACTTGCGCTCAG-3′ for β-actin (GenScript, Piscataway, NJ, USA). β-actin was used as internal standard. PCR amplification was performed using the following thermocycling conditions: 94°C for 5 min, followed by 35 cycles of 94°C for 30 sec, 66°C (or 62°C for iNOS and β-actin) for 30 sec and 72°C for 30 sec, and a final elongation at 72°C for 10 min. The relative expression was calculated using the ΔΔCq method (24) and optimized with a standard curve to confirm specificity. Each sample (5 µl) was electrophoresed on 2% agarose gel (Biowest, Nuaillé, France) with ethidium bromide (Biomatik Corporation, Cambridge, ON, Canada) and the intensity of each band was analyzed using a gel imaging analytical system (UVP GDS 8000; UVP, Upland, CA, USA). The sample without reverse transcriptase was used as a negative control and showed no detectable band.

Immunohistochemical staining

To localize and assess the expression of nNOS and iNOS in the spinal cord during bone cancer development, mice in the tumor and sham groups were transcardially perfused with saline, followed by freshly prepared 4% paraformaldehyde at post-operative days 7, 10 and 14 (n=15 per group). The L3-L5 lumbar spinal cord segments were removed, fixed in 4% PFA overnight at 4°C and then embedded in paraffin for sectioning (5 µm; five mice per group and ten sections from each mouse). The sections were dewaxed in xylene (Nanjing Chemical Reagent Co., Ltd., Nanjing, China) for 10 mins and hydrated with a graded ethanol series (100, 95, 90, 85, 80 and 70%; Nanjing Chemical Reagent Co., Ltd.) for 3 mins each, at room temperature. The sections were washed in double distilled water (Nanjing Chemical Reagent Co., Ltd.) for 5 mins and boiled in 0.1 mol/l sodium citrate buffer (pH 6.0; Wuhan Boster Biological Technology, Ltd., Wuhan, China) for 20 min prior to exposure to 3% H2O2 (Shanghai Shenggong Biology Engineering Technology Service, Ltd., Shanghai, China) for 10 min to bleach endogenous peroxidases. Immunostaining was performed with rabbit primary polyclonal antibodies against nNOS (1:10,000; Sigma-Aldrich; SAB4502010) or rabbit primary polyclonal anti-iNOS (1:800; Sigma-Aldrich; SAB4502011) for 30 min at 37°C, followed by washing with PBS 3 times for 10 mins. They were then incubated with a horseradish peroxidase-labeled goat anti-rabbit IgG (heavy and light chain; Beyotime Institute of Biotechnology; A0208) secondary antibody at 4°C overnight. The sections were then washed with PBS 3 times for 10 mins and visualized with diaminobenzidine (Beyotime Institute of Biotechnology, Haimen, China) and observed under a light microscope (Olympus DP11; Olympus, Tokyo, Japan). Optical density of the images was analyzed using Image-Pro Plus 6.0 analysis software (Media Cybernetics, Inc., Rockville, MD, USA). The mean optical density was obtained by averaging the values from five sections.

Statistical analysis

Values are expressed as the mean ± standard deviation. Animals were assigned to various treatment groups in a randomized way. Repeated-measures analysis of variance (ANOVA) was performed to determine overall differences at each time-point for PWMT and PWTL. One-way ANOVA followed by a least-significant differences post-hoc test was used to determine differences in the mRNA levels of nNOS and iNOS among all experimental groups. All analyses were performed using SPSS 13.0 software (SPSS, Inc., Chicago, IL, USA) and P<0.05 was considered to indicate a statistically significant difference.

Results

Pain-associated behavior in tumor-bearing mice

The PWMT and PWTL of mice in the tumor group prior to the operation were not significantly different when compared with those of the mice in the sham group. The right hind limb of the mice in the tumor group displayed a significant decrease in the PWMT at post-operative day 3 (P<0.05), and this was also observed in the mice in the sham group. At post-operative day 5, the PWMT of the two groups reclined to the basal level (Fig. 1A). Subsequently, the PWMT of mice in the tumor group showed a further decline again at day 7, which continuously decreased until the end of the experiment on post-operative day 14 (0.48±0.25 g).

The mice in the tumor group showed a marked decrease in PWTL at post-operative day 3, which recovered to basal levels at days 5 and 7. Until day 7, similar trends were observed in the sham group mice. However, the PWMT of mice in the tumor group decreased again at post-operative day 10 and further declined to 10.5±3.2 sec at day 14, while that in the sham group remained constant (Fig. 1B). Of note, significant decreases in the PWTL at post-operative days 7–10 and in the PWMT at days 10 and 14 were observed in the tumor group when compared with those in the sham group or the time-point prior to surgery, indicating the development of marked bone cancer-associated pain.

nNOS and iNOS mRNA levels are increased in the spinal cords of tumor-bearing mice

The mRNA levels of nNOS were significantly increased in the spinal cord of mice in the tumor group at post-operative day 10 when compared with those in the same mice at day 7 (P<0.05; Fig. 2A) as well as compared with those in the sham group at day 7, while no significant differences were present within the sham group at these time-points. Furthermore, the mRNA levels of iNOS were significantly increased in the spinal cord of mice in the tumor group at post-operative days 10 and 14 when compared with those in the sham group at the same time-points (P<0.05), or compared with those in the tumor group at day 7 (Fig. 2B). However, no significant differences in the mRNA levels of iNOS were observed within the sham group at days 7–10.

Immunocytochemical localization and expression of nNOS and iNOS in the spinal cord of tumor-bearing mice

Next, an immunohistochemical study was performed to assess the localization and expression levels of nNOS and iNOS in the spinal cord during bone cancer development. The results showed that neurons positive for nNOS (Fig. 3A and B) and iNOS (Fig. 3C and D) were mainly located in the superficial dorsal horn (laminae I–II), which is involved in the elaboration of nociceptive stimuli, and around the central canal (lamina X) of the L3–L5 spinal cord (17). A small number of iNOS-positive neurons were observed in the ventral horn and central canal of the spinal cord.

At post-operative day 7, no significant differences in nNOS or iNOS expression were present between the sham and tumor groups (Fig. 3E and F). Compared with those in the sham group, the expression of nNOS and iNOS in the tumor group was significantly increased at post-operative day 10 and further increased at day 14. In the mice of the sham group, the expression of iNOS at day 14 was significantly decreased as compared with that at day 7, while no significant differences in nNOS expression were observed within the sham group at different time-points.

L-NMMA alleviates bone cancer pain in mice

At post-operative day 14, mice in the tumor group showed a marked decline in PWMT and PWTL. Of note, this decrease was attenuated by intrathecal administration of 50 µg L-NMMA (Fig. 4). The PWMT was significantly increased at 2 h (0.90±0.28 vs. 0.40±0.27; P<0.05) and 12 h (0.63±0.25 vs. 0.40±0.27; P<0.05) following administration of L-NMMA (Fig. 4A). Similarly, the PWTL was also markedly increased at 2 h (16.2±0.9 vs. 10.1±2.3; P<0.05) and 12 h (12.7±1.0 vs. 10.1±2.3; P<0.05) following administration of L-NMMA (Fig. 4B).

Discussion

The present study using a murine model of bone cancer pain following a method of a previous study by our group (16) to demonstrate that bone cancer-induced mechanical allodynia and thermal hyperalgesia are accompanied with an upregulation of nNOS and iNOS in the lumbar spinal cord. In addition, the present study found that NOS inhibitor L-NMMA alleviated cancer-induced mechanical and thermal hyperalgesia, suggesting that spinal nNOS and iNOS may participate in the development of bone cancer pain.

While the mRNA levels of nNOS and iNOS in the lumbar spinal cord of mice in the tumor group were increased at day 10 after tumor inoculation, the up-regulation of iNOS was sustained until day 14 following surgery, at which the pain-associated behavior of the mice in the tumor group were prominent and stable. The cells with immunoreactivity for nNOS and iNOS were mainly located in neurons present at the dorsal horn of the spinal cord (laminae I–II), which is an area involved in the elaboration of nociceptive stimuli, and in a few cells in the ventral parts and around the central canal (17).

It has been reported that the mRNA levels of nNOS in the lumbar spinal cord were increased at day one following surgical inflammation (25). Another study by Tang et al (5) observed a low but constitutive expression of iNOS at the mRNA and protein level in the rat spinal cord under non-stimulated conditions. They also reported that constitutively expressed spinal iNOS mediates tissue injury and inflammation-induced hyperalgesia, and that intrathecal administration of a highly selective iNOS inhibitor produced a dose-dependent inhibition of hyperalgesia. The iNOS gene is induced by bacterial lipopolysaccharide or classical pro-inflammatory cytokines, including tumor necrosis factor α. iNOS produces a larger amount of NO over a longer time period compared with the other two constitutive enzymes, nNOS and eNOS (26). Substantial evidence has been provided for the link between the upregulation of spinal iNOS and inflammatory and neuropathic pain, suggesting a potential therapeutic value of iNOS inhibitors for such conditions (15,27). In addition, Pedersen et al (28) reported that an increased expression of iNOS in the dorsal horn was associated with long-term potentiation that contributed to central hyperalgesia. Consistent with a previous study (29), the results of the present study suggested that nNOS contributed to the initial NO release to facilitate nociceptive processing, while iNOS possibly participated in the persistent nocicep-tive responses. This observed difference in the time-course of the expression of nNOS and iNOS suggests a distinct contribution of NOSs to the mechanisms of bone cancer pain development.

L-NMMA, which is a non-selective inhibitor of NOS that can act on all three isoforms of NOS, has been proved to exert anti-nociceptive effects in several models of peripheral inflammation and nerve injury. Intradermal administration (30) or intrathecal injection (31) of L-NMMA has been shown to reduce, or in certain cases, to completely reverse inflammatory hyperalgesia. In addition, intrathecal injection of L-NMMA is able to reduce the mechanical allodynia evoked by nerve injury (32). Furthermore, a randomized crossover trial demonstrated the analgesic effects of L-NMMA in patients with chronic tension-type headache (33). Consistent with these studies, the present study found that the mechanical and thermal hyperalgesia at post-operative day 14 induced by bone cancer was alleviated at 2 and 12 h after intrathecal injection of L-NMMA, while this effect disappeared at 24 h after L-NMMA administration.

In conclusion, the present study demonstrated that spinal NOS may contribute to nociceptive signal processing during central sensitization in the development of bone cancer pain. As a NOS inhibitor, L-NMMA alleviated the cancer-induced mechanical allodynia and thermal hyperalgesia, suggesting its prospective application in bone cancer-associated pain. Further study is required to determine the underlying mechanism of NOS-associated bone cancer pain.

Acknowledgments

The present study was supported by the National Natural Science Foundation of China (nos. 81070892, 81171048, 81171047, 81371207, 81300950 and 81300951), the Natural Science Foundation of Jiangsu Province (no. BK2010105) and a grant from the Department of Health of Jiangsu Province of China (nos. XK201140 and RC2011006).

References

1 

Colvin L and Fallon M: Challenges in cancer pain management-bone pain. Eur J Cancer. 44:1083–1090. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Miyamoto T, Dubin AE, Petrus MJ and Patapoutian A: TRPV1 and TRPA1 mediate peripheral nitric oxide-induced nociception in mice. PLoS One. 4:e75962009. View Article : Google Scholar : PubMed/NCBI

3 

Naik AK, Tandan SK, Kumar D and Dudhgaonkar SP: Nitric oxide and its modulators in chronic constriction injury-induced neuropathic pain in rats. Eur J Pharmacol. 530:59–69. 2006. View Article : Google Scholar

4 

Guan Y, Yaster M, Raja SN and Tao YX: Genetic knockout and pharmacologic inhibition of neuronal nitric oxide synthase attenuate nerve injury-induced mechanical hypersensitivity in mice. Mol Pain. 3:292007. View Article : Google Scholar : PubMed/NCBI

5 

Tang Q, Svensson CI, Fitzsimmons B, Webb M, Yaksh TL and Hua XY: Inhibition of spinal constitutive NOS-2 by 1400W attenuates tissue injury and inflammation-induced hyperalgesia and spinal p38 activation. Eur J Neurosci. 25:2964–2972. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Choi JI, Kim WM, Lee HG, Kim YO and Yoon MH: Role of neuronal nitric oxide synthase in the antiallodynic effects of intrathecal EGCG in a neuropathic pain rat model. Neurosci Lett. 510:53–57. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Lam HH, Hanley DF, Trapp BD, Saito S, Raja S, Dawson TM and Yamaguchi H: Induction of spinal cord neuronal nitric oxide synthase (NOS) after formalin injection in the rat hind paw. Neurosci Lett. 210:201–204. 1996. View Article : Google Scholar : PubMed/NCBI

8 

Chacur M, Matos RJ, Alves AS, Rodrigues AC, Gutierrez V, Cury Y and Britto LR: Participation of neuronal nitric oxide synthase in experimental neuropathic pain induced by sciatic nerve transection. Braz J Med Biol Res. 43:367–376. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Lee JS, Zhang Y and Ro JY: Involvement of neuronal, inducible and endothelial nitric oxide synthases in capsaicin-induced muscle hypersensitivity. Eur J Pain. 13:924–928. 2009. View Article : Google Scholar

10 

Handy RL and Moore PK: Effects of selective inhibitors of neuronal nitric oxide synthase on carrageenan-induced mechanical and thermal hyperalgesia. Neuropharmacology. 37:37–43. 1998. View Article : Google Scholar : PubMed/NCBI

11 

Tanabe M, Nagatani Y, Saitoh K, Takasu K and Ono H: Pharmacological assessments of nitric oxide synthase isoforms and downstream diversity of NO signaling in the maintenance of thermal and mechanical hypersensitivity after peripheral nerve injury in mice. Neuropharmacology. 56:702–708. 2009. View Article : Google Scholar

12 

Chu YC, Guan Y, Skinner J, Raja SN, Johns RA and Tao YX: Effect of genetic knockout or pharmacologic inhibition of neuronal nitric oxide synthase on complete Freund's adjuvant-induced persistent pain. Pain. 119:113–123. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Toda N, Kishioka S, Hatano Y and Toda H: Modulation of opioid actions by nitric oxide signaling. Anesthesiology. 110:166–181. 2009. View Article : Google Scholar

14 

De Alba J, Clayton NM, Collins SD, Colthup P, Chessell I and Knowles RG: GW274150, a novel and highly selective inhibitor of the inducible isoform of nitric oxide synthase (iNOS), shows analgesic effects in rat models of inflammatory and neuropathic pain. Pain. 120:170–181. 2006. View Article : Google Scholar

15 

Sasaki A, Mabuchi T, Serizawa K, Takasaki I, Andoh T, Shiraki K, Ito S and Kuraishi Y: Different roles of nitric oxide synthase-1 and -2 between herpetic and postherpetic allodynia in mice. Neuroscience. 150:459–466. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Gu X, Zhang J, Ma Z, Wang J, Zhou X, Jin Y, Xia X, Gao Q and Mei F: The role of N-methyl-D-aspartate receptor subunit NR2B in spinal cord in cancer pain. Eur J Pain. 14:496–502. 2010. View Article : Google Scholar

17 

Meller ST and Gebhart GF: Nitric oxide (NO) and nociceptive processing in the spinal cord. Pain. 52:127–136. 1993. View Article : Google Scholar : PubMed/NCBI

18 

Ma ZL, Zhang W, Gu XP, Yang WS and Zeng YM: Effects of intrathecal injection of prednisolone acetate on expression of NR2B subunit and nNOS in spinal cord of rats after chronic compression of dorsal root ganglia. Ann Clin Lab Sci. 37:349–355. 2007.PubMed/NCBI

19 

Zimmermann M: Ethical guidelines for investigations of experimental pain in conscious animals. Pain. 16:109–110. 1983. View Article : Google Scholar : PubMed/NCBI

20 

Schwei MJ, Honore P, Rogers SD, Salak-Johnson JL, Finke MP, Ramnaraine ML, Clohisy DR and Mantyh PW: Neurochemical and cellular reorganization of the spinal cord in a murine model of bone cancer pain. J Neurosci. 19:10886–10897. 1999.PubMed/NCBI

21 

Hylden JL and Wilcox GL: Intrathecal morphine in mice: A new technique. Eur J Pharmacol. 67:313–316. 1980. View Article : Google Scholar : PubMed/NCBI

22 

Chaplan SR, Bach FW, Pogrel JW, Chung JM and Yaksh TL: Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods. 53:55–63. 1994. View Article : Google Scholar : PubMed/NCBI

23 

Hargreaves K, Dubner R, Brown F, Flores C and Joris J: A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia. Pain. 32:77–88. 1988. View Article : Google Scholar : PubMed/NCBI

24 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar

25 

Dolan S, Kelly JG, Huan M and Nolan AM: Transient up-regulation of spinal cyclooxygenase-2 and neuronal nitric oxide synthase following surgical inflammation. Anesthesiology. 98:170–180. 2003. View Article : Google Scholar

26 

Conti A, Miscusi M, Cardali S, Germanò A, Suzuki H, Cuzzocrea S and Tomasello F: Nitric oxide in the injured spinal cord: Synthases cross-talk, oxidative stress and inflammation. Brain Res Rev. 54:205–218. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Martucci C, Trovato AE, Costa B, Borsani E, Franchi S, Magnaghi V, Panerai AE, Rodella LF, Valsecchi AE, Sacerdote P and Colleoni M: The purinergic antagonist PPADS reduces pain related behaviours and interleukin-1 beta, interleukin-6, iNOS and nNOS overproduction in central and peripheral nervous system after peripheral neuropathy in mice. Pain. 137:81–95. 2008. View Article : Google Scholar

28 

Pedersen LM, Jacobsen LM, Mollerup S and Gjerstad J: Spinal cord long-term potentiation (LTP) is associated with increased dorsal horn gene expression of IL-1beta, GDNF and iNOS. Eur J Pain. 14:255–260. 2010. View Article : Google Scholar

29 

Guhring H, Tegeder I, Lötsch J, Pahl A, Werner U, Reeh PW, Rehse K, Brune K and Geisslinger G: Role of nitric oxide in zymosan induced paw inflammation and thermal hyperalgesia. Inflamm Res. 50:83–88. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Aley KO, McCarter G and Levine JD: Nitric oxide signaling in pain and nociceptor sensitization in the rat. J Neurosci. 18:7008–7014. 1998.PubMed/NCBI

31 

Malmberg AB and Yaksh TL: Spinal nitric oxide synthesis inhibition blocks NMDA-induced thermal hyperalgesia and produces antinociception in the formalin test in rats. Pain. 54:291–300. 1993. View Article : Google Scholar : PubMed/NCBI

32 

Yonehara N, Kudo C and Kamisaki Y: Involvement of NMDA-nitric oxide pathways in the development of tactile hypersensitivity evoked by the loose-ligation of inferior alveolar nerves in rats. Brain Res. 963:232–243. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Ashina M, Lassen LH, Bendtsen L, Jensen R and Olesen J: Effect of inhibition of nitric oxide synthase on chronic tension-type headache: A randomised crossover trial. Lancet. 353:287–289. 1999. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2016
Volume 13 Issue 2

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
Yang Y, Zhang J, Liu Y, Zheng Y, Bo J, Zhou X, Wang J and Ma Z: Role of nitric oxide synthase in the development of bone cancer pain and effect of L‑NMMA. Mol Med Rep 13: 1220-1226, 2016
APA
Yang, Y., Zhang, J., Liu, Y., Zheng, Y., Bo, J., Zhou, X. ... Ma, Z. (2016). Role of nitric oxide synthase in the development of bone cancer pain and effect of L‑NMMA. Molecular Medicine Reports, 13, 1220-1226. https://doi.org/10.3892/mmr.2015.4647
MLA
Yang, Y., Zhang, J., Liu, Y., Zheng, Y., Bo, J., Zhou, X., Wang, J., Ma, Z."Role of nitric oxide synthase in the development of bone cancer pain and effect of L‑NMMA". Molecular Medicine Reports 13.2 (2016): 1220-1226.
Chicago
Yang, Y., Zhang, J., Liu, Y., Zheng, Y., Bo, J., Zhou, X., Wang, J., Ma, Z."Role of nitric oxide synthase in the development of bone cancer pain and effect of L‑NMMA". Molecular Medicine Reports 13, no. 2 (2016): 1220-1226. https://doi.org/10.3892/mmr.2015.4647