Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

An Engineered Endomorphin-2 Gene for Morphine Withdrawal Syndrome

  • Fei-xiang Wu,

    Affiliations Department of Anesthesiology & Intensive Care, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China, Department of Anesthesiology and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, United States of America

  • Yan He,

    Affiliation Department of Anesthesiology, Dongfang Hospital, Fujian, 354200, China

  • Hui-ting Di,

    Affiliation Department of Anesthesiology & Intensive Care, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China

  • Yu-ming Sun,

    Affiliation Department of Anesthesiology & Intensive Care, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China

  • Rui-rui Pan,

    Affiliation Department of Anesthesiology & Intensive Care, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China

  • Wei-feng Yu ,

    ywf808@sohu.com (WFW); liur@uphs.upenn.edu (RYL)

    Affiliation Department of Anesthesiology & Intensive Care, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China

  • Renyu Liu

    ywf808@sohu.com (WFW); liur@uphs.upenn.edu (RYL)

    Affiliation Department of Anesthesiology and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, United States of America

Abstract

An optimal therapeutics to manage opioid withdrawal syndrome is desired for opioid addiction treatment. Down-regulation of endogenous endomorphin-2 (EM2) level in the central nervous system after continuous morphine exposure was observed, which suggested that increase of EM2 could be an alternative novel method for opioid dependence. As a short peptide, the short half-life of EM2 limits its clinical usage through conventional administration. In the present study, we engineered an EM2 gene using a signal peptide of mouse growth factor for an out-secretory expression of EM2 and an adenovirus as a vector, which ultimately sustained the release of EM-2. After administration of the adenovirus in central nervous system, a sustained increase of EM2 level in the cerebral spinal fluid (CSF) was observed along with a reduction of morphine withdrawal syndrome. These findings suggest that the engineered EM2 gene delivered to the central nervous system could be a novel therapeutics for withdrawal syndrome in opioid dependent subjects.

Introduction

Opioid dependence affects more than 1 million people in North America alone each year [1, 2]. Opioid dependence associated with various infectious diseases, serious medical complications, overdose mortality, tolerance, loss of social functioning, financial instability, and serious crime [36]. Sudden discontinuation or decreased opioid intake leads to withdrawal symptoms including nausea, diarrhea, severe itching, tachycardia, and depression [7, 8]. Current managements of opioid dependence are using full mu opioid receptor (MOR) agonist, methadone, or partial MOR agonist, buprenorphine [911]. However, both methadone and buprenorphine are addictive and are ineffective in 15% to 25% patients [12], and a long-term monitored treatment is required due to potential relapse [13].

Endomorphin-2 (EM2) is an endogenous agonist for MOR with high affinity and high selectivity, which has an exciting and promising therapeutic potential for pain or opioid dependence with less deleterious side effects of opiates [1416]. Spinal EM2 was decreased by the activation of MOR but enhanced by the blockade of spinal MOR, which suggested that EM2 is involved in the pathophysiologic state of dependence after chronic exposure to opioids and could be used as an alternative drug for opioid dependence [17]. However, the usefulness of synthetic EM2 was limited due to its short half-life [18]. The alternative approach is to increase the half-life of EM2 by structural modifications of the EM2 protein [1921] or using a genetic engineering approach to sustain the release of EM2 [22]. In the present study, we hypothesized that the sustained release of endogenous EM2 by genetic engineering could be used to manage opioid withdrawal syndrome.

Materials and Methods

Materials

The recombinant adenovirus (Deletion of the E1/E3 regions of genome), Ad-EM2 (an adenovirus with EM2) and Ad-EGFP (an adenovirus with enhanced green fluorescence protein) were produced, purified, and stored as previously described [22, 23]. The titers of the virus were adjusted to 1×1011 PFU/ml (PFU = plaque-forming units). The target gene of Ad-EM2 was a fusion gene complex consisting of a gene for human EM2 and a gene for a signal peptide of mouse nerve growth factor (Fig 1). The fusion gene expression cassette was engineered under the control of a cytomegalovirus (CMV) promoter. The signal peptide of mouse growth factor was introduced as an out-secretory peptide for EM2.

thumbnail
Fig 1. The fusion gene complex for endogenous endomorphin-2 (EM2) consisted of a gene for human EM2 and a gene for a signal peptide of mouse nerve growth factor (NGF).

The fusion gene expression cassette was engineered under the control of a cytomegalovirus (CMV) promoter. The white area represented the cleavage site for Furin.

https://doi.org/10.1371/journal.pone.0149877.g001

Animals

All animal experiments were carried out under the guidelines issued by the Administrative Committee of Experimental Animal Care and Use of Shanghai, and conformed to the National Institute of Health guidelines on the ethical use of animals. The institutional review committee of Eastern Hepatobiliary Surgery Hospital approved the experimental protocol. Male Sprague-Dawley rats (Weight: 200-250g, Age: 10–12 weeks from Shanghai Experimental Animal Center of Chinese Academy of Sciences) were housed in a pathogen free condition with a 12:12 hour light /dark cycle. The room and cage conditions were monitored twice a day. Monitoring for health problems were performed three times a day. All rats were healthy and no death was found during the experiments. The rats were euthanized by CO2 after the whole experiments were finished.

Animal model and protocol

Rats were divided into four groups randomly (n = 10/group): an Ad-EM2 group (Ad-EM2 with titers of 5×109 PFU); an Ad-EGFP group (Ad-EGFP with titers of 5×109 PFU); a NS group (Normal Saline injection); and a sham group. The morphine dependent model was constructed by subcutaneous administration of daily escalating doses of morphine three times/day for 6 consecutive days (5, 10, 20, 40, 50, 60 mg/kg) [24]. The same volumes of normal saline (NS) were received subcutaneously in the sham group. In the Ad-EM2 group and Ad-EGFP group, dependent rats received intrathecal 50μl of the various virus solutions after the last dosage of morphine was administered. In the NS group and sham group, the same volumes of NS were injected intrathecally.

Lumbar subarachnoid catheterization

Lumbar subarachnoid catheterization was carried out 3 days before starting the morphine administration. Rats were anesthetized with intraperitoneal injection of 40 mg/kg of sodium pentobarbital (Chemical Reagent Company, Shanghai, China). The vital signs of rats were monitored by the MouseOx® Plus pulse oximeter (Starr Life Sciences Corporation, USA). A sterile PE10 polyethylene catheter (Becton Dickinson, Sparks, MD, USA) was used for intrathecal placement between lumbar vertebrae 5(L5) and lumbar vertebrae 6 (L5) [25]. The external part of the indwelling catheter was protected under the skin to the cervical region according to the Milligan’s method [26]. A lidocaine test was performed to determine the position of the catheter. The catheter was utilized to deliver the virus and to collect cerebrospinal fluid (CSF) samples. On the 6th day of establishment of the morphine dependent model, the Ad-EM2, Ad-EGFP or NS were delivered through the catheters after the last dosage of morphine. Before injection of the virus and on each of the 1st, 3rd, 5th, 7th, and 10th day after injection of the virus, the CSF samples were collected.

The scores of the withdrawal syndrome

On the 1st, 3rd, 5th, 7th, and 10th day after intrathecal delivery of the adenovirus carried gene, morphine withdrawal syndrome was induced by intraperitoneal injection of naloxone (2mg/kg). The symptoms of morphine withdrawal syndrome in each animal were observed for 30 min after naloxone injection. The scores of withdrawal symptoms were determined according to Maldonado's modified method as described in our previous study (Table 1) [27, 28]. The body weight was measured after scoring.

thumbnail
Table 1. Scores of the symptoms of morphine withdrawal syndrome.

https://doi.org/10.1371/journal.pone.0149877.t001

ELISA assay

The EM2 concentration in CSF was measured using a sandwich ELISA kit (Uscn Life Science Inc, Wuhan, China) according to the manufacturer’s protocol. 96-well micro-titers were used for incubation and measurement. The optical density of each sample was read at 450 nm by a microplate reader (Bio-Rad Laboratories, Shanghai, China). Endomorphin-2 concentration was determined according to the standard curve as described previously [22].

Green fluorescence assay

On the 10th day after intrathecal injection, rats were anesthetized with intraperitoneal injection of 40 mg/kg of sodium pentobarbital and perfused through the ascending aorta with saline, followed by 4% paraformaldehyde in 0.16 M phosphate buffer (pH 7.2–7.4) containing 1.5% picric acid. After perfusion, the tissues of L5 spinal cord were collected and fixed in the same fixation solution for 3 h and then in 15% sucrose overnight. Transverse spinal sections (30μm) were obtained on a cryostat and processed for EGFP expression estimation with a green fluorescence assay. These sections were examined under an Olympus fluorescence microscope (Olympus, Japan).

Statistical analysis

All data were presented as mean ± SEM. Statistical analysis was carried out using two-way ANOVA followed by post-hoc Tukey's multiple comparison using GraphPad Prism software (Version 5. GraphPad Software Inc, CA, USA). P<0.05 was considered statistically significant.

Results

The persistent increase of EM2 in CSF

There were no significant changes of the EM2 in CSF on the 1st day as indicated in Fig 2. On the 3rd, 5th, 7th, and 10th day, EM2 in CSF increased significantly in the Ad-EM2 groups compared to the Ad-EGFP group and NS group (aP<0.0001). The up-regulation of EM2 in CSF after intrathecal administration of Ad-EM2 indicated an out-secretory expression of EM2. After intrathecal injection, the EGFP expression was detected in the meningeal pia mater cells in the Ad-EGFP group (Fig 3), suggesting that the adenovirus was transfected into the same location after intrathecal injection and the expression of EM2 occurred in an out-secretory manner.

thumbnail
Fig 2. Endomorphin-2 (EM2) expression in CSF after injection.

On the 3rd, 5th, 7th, and 10th day after intrathecal injection, the EM2 in cerebral spinal fluid (CSF) increased significantly in the Ad-EM2 groups compared to that in the Ad-EGFP group and NS group (aP<0.0001). Endomorphin-2 was indicated as EM2; adenovirus with EM2 was indicated as Ad-EM2; adenovirus with enhanced green fluorescence protein was indicated as Ad-EGFP; normal saline group with subcutaneous morphine injection and intrathecal NS administration was indicated as NS; Sham group with subcutaneous NS injection and intrathecal NS administration was indicated as Sham.

https://doi.org/10.1371/journal.pone.0149877.g002

thumbnail
Fig 3. Enhanced green fluorescence protein (EGFP) expression in the meningeal pia mater cells.

EGFP expression was located in the meningeal pia mater cells in the Ad-EGFP group after intrathecal injection (see red arrow in the panel A), suggesting that adenovirus was transfected into the same location. This was not observed in the normal saline group as indicated in panel B.

https://doi.org/10.1371/journal.pone.0149877.g003

The alleviation of the morphine withdrawal syndrome

Compared to the sham group, the scores of the symptoms of morphine withdrawal were significantly higher in the NS and Ad-EGFP group after cessation of morphine (aP<0.0001), and the scores were higher in the Ad-EM2 on the 1st, 3rd day (aP<0.0001). On the 1st day after intrathecal injection, there were no significant differences for the scores among three groups of dependent rats. On the 3rd, 5th, 7th, and 10th day, withdrawal scores decreased significantly in the Ad-EM2 group compared to the Ad-EGFP and NS groups (bP<0.0001) as shown in Fig 4, suggesting that the increase of EM2 in the CSF associated with alleviation of the morphine withdrawal syndrome.

thumbnail
Fig 4. Total score of morphine withdrawal symptoms.

On the 3rd, 5th, 7th, and 10th day after intrathecal injection, withdrawal scores were decreased markedly in the Ad-EM2 group compared to that in the Ad-EGFPand NS groups (bP<0.0001). Endomorphin-2 was indicated as EM2; adenovirus with EM2 was indicated as Ad-EM2; adenovirus with enhanced green fluorescence protein was indicated as Ad-EGFP; normal saline group with subcutaneous morphine injection and intrathecal NS administration was indicated as NS; Sham group with subcutaneous NS injection and intrathecal NS administration was indicated as Sham.

https://doi.org/10.1371/journal.pone.0149877.g004

The change of body weights

Loss of body weight was found at 3 days after cessation of morphine in the morphine-dependent rats (Fig 5). Compared to the sham group, there was a significant weight loss in the other three groups receiving morphine injection on the 3rd, 5th, 7th, and 10th day (aP<0.0001). In the Ad-EM2 group, the body weight significantly increased compared to the Ad-EGFP and NS groups on the 5th, 7th, and 10th day after intrathecal injection (bP<0.0001), suggesting that EM2 expressed by Ad-EM2 associated with the reduction of the loss of body weight in the morphine dependent rats.

thumbnail
Fig 5. Body weight changes among groups.

In the Ad-EM2 group, on the 5th, 7th, and 10th day after intrathecal administration, the body weight increased significantly compared to the Ad-EGFP and the NS groups (bP>0.0001), suggesting that EM2 expressed by Ad-EM2 reduced the loss of body weight in morphine dependent rats. Endomorphine-2 was indicated as EM2; adenovirus with EM2 was indicated as Ad-EM2; adenovirus with enhanced green fluorescence protein was indicated as Ad-EGFP; normal saline group with subcutaneous morphine injection and intrathecal NS administration was indicated as NS; Sham group with subcutaneous NS injection and intrathecal NS administration was indicated as Sham.

https://doi.org/10.1371/journal.pone.0149877.g005

Discussion

In the present study, EM2 gene was introduced into the central nervous system intrathecally via an adenovirus resulting in sustained increase of EM2 in CSF. This led to a significant reduction of the morphine withdrawal syndrome in the morphine dependent rat model.

The sustained EM2 production

In 1997, EM2 was discovered as an endogenous ligand for MOR [29]. The potent analgesic effect of EM2 has encouraged many studies for potential clinical usage. However, there are two major obstacles for its clinical usage. 1) EM2, with four short amino acid residues, is easily degraded by various enzymes [30] 2) Unknown original gene sequences of EM2 limit the possibility of engineering through transgene methods. In the present study, we bypassed these obstacles by engineering a complex of gene for EM2 and a type I signal peptide of mouse growth factor. A cleavage site of the enzyme, Furin, was introduced for out-secretory expression. Furin cleaves off the signal peptide from the fusion protein and releases EM2 from the membrane of cells [31, 32]. In a neuropathic pain rat model, a single intrathecal injection of the adenovirus carrying Ad-EM2 gene induced 35 days of anti-nociception effect without showing any tolerance or addiction [22]. Thus, this novel Ad-EM2 would be a useful tool for the prolonged release of EM2 for therapeutic purposes.

Gene therapy of opioid dependence

In opioid dependence, the tone of the endogenous opioid system is too low to restore the deficiency caused by long-term external opioids exposure [33]. After cessation of external opioids, the addict enters an acute withdrawal syndrome due to the shortage of endogenous opioid. Compensation of opioids, such as enkephalins [34], β-endorphin [35] and endomorphin [36], have shown potential applications in treatment of dependence. Using trans-gene technique, dependence related protein such β-endorphin and Cocaine Hydrolase, have been transducted into central nervous system and shown the attenuation of dependence, which results suggest that transduction of substantial mounts of an engineered human protein in or near the cells that are affected in dependence could directly alter the pathophysiologic state [37, 28]. This study demonstrated that EM2 was increased in the CSF 3 days after intrathecal injection of Ad-EM2 and that the acute withdrawal syndrome of morphine was reduced with the increase of EM2. The body mass of the rats also increased after administration of Ad-EM2. These findings supported the hypothesis that EM2 expressed by Ad-EM2 has therapeutic effects for acute opioid withdrawal syndrome.

The adenoviral vector

The replication deficient adenovirus is a useful vector to deliver genes for neurological disorders [38]. As a tool to deliver EM2, the adenovirus had several advantages. 1) Unlike retroviral vector targeting proliferation cells, the adenoviral vector transfected into both proliferation and non-proliferating cells. It was beneficial for the application of most non-proliferating cells in the nervous system [39]. 2) The protein in CSF delivered by the adenovirus quickly affected neighboring neurons and directly targeted receptors such as mu opioid receptor without neural toxicity. This minimized the dosage and reduced the systemic side effects [40, 41]. 3) The adenovirus did not integrate into the chromosome of the hosts, which could decrease the likelihood of insertion mutation [42, 43]. However, one of the disadvantages of the adenovirus was that it may induce an immune response of the host, which would eliminate the virus gradually [44, 45]. This elimination of the adenovirus resulted in the gradual decrease of EM2 in the CSF [22]. However, this gradual decrease of EM2 could be an advantage of gene therapy, similar to the decreasing dose regimen for methadone [46]. In previous clinical trials, the adenovirus has been used to deliver into the normal brain parenchyma to manage tumors or brain diseases with a safe and successful profile [47, 48]. Thus, such approach could be potentially used to deliver EM2 to manage opioid dependence and withdrawal syndrome.

The route of administration

For the treatment of acute withdrawal syndrome using adenoviral vectors, the vectors were usually delivered intracerebroventricularly [28] or directly into the brain tissue, in the nucleus accumbens and ventromedial striatum [37]. These methods require complicated procedures and induce additional tissue trauma and potential infections. In the present study, the adenovirus was delivered intrathecally, which provides a safer and more convenient way of administration compared to an intracerebroventricular or brain tissue injection. The adenovirus was detected in the location of the meningeal pia mater cells surrounding the spinal cord, which was consistent in the study of Finegold AA, et al [49]. This alternative route of administration of adenovirus could be a safe and convenient way to manage withdrawal syndrome.

Limitation of the study

In clinical practice, the levels and timing of EM2 expression are required to be controlled precisely, thus needing an inducible gene expression system such as an RU486 regulating system. Other models such as conditioned place preference model are also required for observation of long-term effect on chronic models. Therefore, further studies are warranted.

Conclusions

In summary, an engineered EM2 gene delivered to the central nervous system via intrathecal administration reduced withdraw syndrome in an opioid dependent rat model, indicating that this novel engineered EM2 gene could be an alternative therapeutics for management of withdrawal syndrome in opioid dependent subjects.

Acknowledgments

The authors appreciate the support in vector construction from Xuewu Xu, M., PhD, at the Department of Anesthesiology, Hospital 306, Beijing, China, the academic advice and review from Professor William Armstead at the Department of Anesthesiology and Critical Care at the University of Pennsylvania, and the technical support from Ms. Jingyuan Ma and Megumi Sugita at the University of Pennsylvania.

Author Contributions

Conceived and designed the experiments: FXW YMS WFY RYL. Performed the experiments: FXW YH HTD RRP. Analyzed the data: FXW HY YMS. Contributed reagents/materials/analysis tools: FXW HY YMS HTD RRP WFY RYL. Wrote the paper: FXW.

References

  1. 1. Gartry CC, Oviedo-Joekes E, Laliberte N, Schechter MT. NAOMI: The trials and tribulations of implementing a heroin assisted treatment study in North America. Harm Reduct J 2009; 6: 2. pmid:19159475
  2. 2. Oviedo-Joekes E, Brissette S, Marsh DC, Lauzon P, Guh D, Anis A et al. Diacetylmorphine versus methadone for the treatment of opioid addiction. N Engl J Med 2009; 361: 777–86. pmid:19692689
  3. 3. March JC, Oviedo-Joekes E, Romero M. Drugs and social exclusion in ten European cities. Eur Addict Res 2006; 12: 33–41. pmid:16352901
  4. 4. Miller CL, Kerr T, Frankish JC, Spittal PM, Li K, Schechter MT et al. Binge drug use independently predicts HIV seroconversion among injection drug users: implications for public health strategies. Subst Use Misuse 2006; 41: 199–210. pmid:16393742
  5. 5. Wood E, Li K, Palepu A, Marsh DC, Schechter MT, Hogg RS et al. Sociodemographic disparities in access to addiction treatment among a cohort of Vancouver injection drug users. Subst Use Misuse 2005; 40: 1153–67. pmid:16040375
  6. 6. Manzoni P, Brochu S, Fischer B, Rehm R. Determinants of property crime among illicit opiate users outside of treatment across Canada. Deviant Behav 2006; 27: 351–76.
  7. 7. Stevens RC, Rapaport S, Maroldo-Connelly L, Patterson JB, Bertz R.. Lack of methadone dose alterations or withdrawal symptoms during therapy with lopinavir/ritonavir. J Acquir Immune Defic Syndr 2003; 33: 650–1. pmid:12902812
  8. 8. Karila L, Berlin I, Benyamina A, Reynaud M. Psychotic symptoms following buprenorphine withdrawal. Am J Psychiatry 2008; 165: 400–1. pmid:18316438
  9. 9. Vigezzi P, Guglielmino L, Marzorati P, Silenzio R, De Chiara M, Corrado F et al. Multimodal drug addiction treatment: a field comparison of methadone and buprenorphine among heroin- and cocaine-dependent patients. J Subst Abuse Treat 2006; 31: 3–7. pmid:16814005
  10. 10. Farrell M, Ward J, Mattick R, Hall W, Stimson GV, des Jarlais D et al. Methadone maintenance treatment in opiate dependence: a review. Bmj 1994; 309: 997–1001. pmid:7950725
  11. 11. Mattick RP, Breen C, Kimber J, Davoli M. Methadone maintenance therapy versus no opioid replacement therapy for opioid dependence. Cochrane Database Syst Rev 2009: CD002209. pmid:19588333
  12. 12. Johnson RE, Chutuape MA, Strain EC, Walsh SL, Stitzer ML, Bigelow GE. A comparison of levomethadyl acetate, buprenorphine, and methadone for opioid dependence. N Engl J Med 2000; 343: 1290–7. pmid:11058673
  13. 13. Termorshuizen F, Krol A, Prins M, Geskus R, van den Brink W, van Ameijden EJ. Prediction of relapse to frequent heroin use and the role of methadone prescription: An analysis of the Amsterdam Cohort Study among drug users. Drug Alcohol Depend 2005; 79: 231–40. pmid:16002032
  14. 14. Tseng LF, Narita M, Suganuma C, Mizoguchi H, Ohsawa M, Nagase H et al. Differential antinociceptive effects of endomorphin-1 and endomorphin-2 in the mouse. Journal of Pharmacology and Experimental Therapeutics 2000; 292: 576–83. pmid:10640294
  15. 15. Przewlocka B, Mika J, Labuz D, Toth G, Przewlocki R. Spinal analgesic action of endomorphins in acute, inflammatory and neuropathic pain in rats. Eur J Pharmacol 1999; 367: 189–96. pmid:10078992
  16. 16. Bian D, Nichols ML, Ossipov MH, Lai J, Porreca F. Characterization of the Antiallodynic Efficacy of Morphine in a Model of Neuropathic Pain in Rats. Neuroreport 1995; 6: 1981–4. pmid:8580422
  17. 17. Gupta DS, von Gizycki H, Gintzler AR. Sex-/ovarian steroid-dependent release of endomorphin 2 from spinal cord. Journal of Pharmacology and Experimental Therapeutics 2007; 321: 635–41. pmid:17308039
  18. 18. Sugimoto-Watanabe A, Kubota K, Fujibayashi K, Saito K. Antinociceptive effect and enzymatic degradation of endomorphin-1 in newborn rat spinal cord. Jpn J Pharmacol 1999; 81: 264–70. pmid:10622214
  19. 19. Hau VS, Huber JD, Campos CR, Lipkowski AW, Misicka A, Davis TP. Effect of guanidino modification and proline substitution on the in vitro stability and blood-brain barrier permeability of endomorphin II. J Pharm Sci 2002; 91: 2140–9. pmid:12226841
  20. 20. Liu HM, Zhang BZ, Liu XF, Wang C, Ni J, Wang R. Endomorphin-1 analogs with enhanced metabolic stability and systemic analgesic activity: Design, synthesis, and pharmacological characterization. Bioorg Med Chem 2007; 15: 1694–702. pmid:17188879
  21. 21. Honda T, Shirasu N, Isozaki K, Kawano M, Shigehiro D, Chuman Y et al. Differential receptor binding characteristics of consecutive phenylalanines in mu-opioid specific peptide ligand endomorphin-2. Bioorg Med Chem 2007; 15: 3883–8. pmid:17395470
  22. 22. Wu F, Xu X, Miao X, Chen J, Sun Y, Yu W. Effect of recombinant adenovirus coding for endomorphin-2 on neuropathic pain in rats. Int J Clin Exp Patho 2012; 5: 914–23.
  23. 23. Wu F, Liu Y, Lv X, Miao X, Sun Y, Yu W. Small interference RNA targeting TLR4 gene effectively attenuates pulmonary inflammation in a rat model. J Biomed Biotechnol 2012; 2012: 406435. pmid:22496605
  24. 24. Xiao H, Zhai DX, Yan BB, Wang JH, Xu WS, Wang GY et al. A role for the parafascicular thalamic nucleus in the development of morphine dependence and withdrawal. Brain Res 2009; 1271: 74–82. pmid:19332040
  25. 25. Storkson RV, Kjorsvik A, Tjolsen A, Hole K. Lumbar catheterization of the spinal subarachnoid space in the rat. J Neurosci Methods 1996; 65: 167–72. pmid:8740594
  26. 26. Milligan ED, Hinde JL, Mehmert KK, Maier SF, Watkins LR. A method for increasing the viability of the external portion of lumbar catheters placed in the spinal subarachnoid space of rats. J Neurosci Methods 1999; 90: 81–6. pmid:10517276
  27. 27. Maldonado R, Negus S, Koob GF. Precipitation of morphine withdrawal syndrome in rats by administration of mu-, delta- and kappa-selective opioid antagonists. Neuropharmacology 1992; 31: 1231–41. pmid:1335131
  28. 28. He Y, Wu FX, Miao XR, Xu XW, Sun YM, Chen CY et al. Suppression of acute morphine withdrawal syndrome by adenovirus-mediated beta-endorphin in rats. Brain Res 2011; 1422: 13–9. pmid:21983207
  29. 29. Zadina JE, Hackler L, Ge LJ, Kastin AJ. A potent and selective endogenous agonist for the mu-opiate receptor. Nature 1997; 386: 499–502. pmid:9087409
  30. 30. Peter A, Toth G, Tomboly C, Laus G, Tourwe D. Liquid chromatographic study of the enzymatic degradation of endomorphins, with identification by electrospray ionization mass spectrometry. J Chromatogr A 1999; 846: 39–48. pmid:10420597
  31. 31. Dalbey RE, Lively MO, Bron S, van Dijl JM. The chemistry and enzymology of the type I signal peptidases. Protein Sci 1997; 6: 1129–38. pmid:9194173
  32. 32. Knappskog S, Ravneberg H, Gjerdrum C, Trosse C, Stern B, Pryme IF. The level of synthesis and secretion of Gaussia princeps luciferase in transfected CHO cells is heavily dependent on the choice of signal peptide. J Biotechnol 2007; 128: 705–15. pmid:17316861
  33. 33. Mayer P, Hollt V. Pharmacogenetics of opioid receptors and addiction. Pharmacogenetics and genomics 2006; 16: 1–7. pmid:16344716
  34. 34. Fukunaga Y, Inoue N, Miyamoto M, Kishioka S, Yamamoto H. Effects of peptidase inhibitors, [D-Ala2, Met5]-enkephalinamide and antiserum to methionine-enkephalin microinjected into the caudal periaqueductal gray on morphine withdrawal in rats. Jpn J Pharmacol 1998; 78: 455–61. pmid:9920202
  35. 35. Capasso A, Loizzo A. Beta-endorphin fragments (DT gamma E and DE gamma E) reduce opiate withdrawal in guinea pig ileum. Progress in neuro-psychopharmacology & biological psychiatry 2002; 26: 313–9.
  36. 36. Okada Y, Tsuda Y, Salvadori S, Lazarus LH. Developmental potential for endomorphin opioidmimetic drugs. International journal of medicinal chemistry 2012; 2012: 715123. pmid:25954530
  37. 37. Gao Y, Brimijoin S. Viral transduction of cocaine hydrolase in brain reward centers. Cell Mol Neurobiol 2006; 26: 357–63. pmid:16708286
  38. 38. Mata M, Glorioso JC, Fink DJ. Gene therapy: novel treatments for polyneuropathy and chronic pain. Curr Neurol Neurosci Rep 2004; 4: 1–2. pmid:14683620
  39. 39. George JA. Gene therapy progress and prospects: adenoviral vectors. Gene Ther 2003; 10: 1135–41. pmid:12833122
  40. 40. Wilson SP, Yeomans DC. Genetic therapy for pain management. Curr Rev Pain 2000; 4: 445–50. pmid:11060590
  41. 41. Pohl M, Braz J. Gene therapy of pain: emerging strategies and future directions. Eur J Pharmacol 2001; 429: 39–48. pmid:11698025
  42. 42. Volpers C, Kochanek S. Adenoviral vectors for gene transfer and therapy. J Gene Med 2004; 6 Suppl 1: S164–71. pmid:14978759
  43. 43. Brunetti-Pierri N, Ng P. Progress and prospects: gene therapy for genetic diseases with helper-dependent adenoviral vectors. Gene Ther 2008; 15: 553–60. pmid:18288209
  44. 44. Ishii K, Isono M, Inoue R, Hori S. Attempted gene therapy for intractable pain: dexamethasone-mediated exogenous control of beta-endorphin secretion in genetically modified cells and intrathecal transplantation. Exp Neurol 2000; 166: 90–8. pmid:11031086
  45. 45. Cao H, Koehler DR, Hu J. Adenoviral vectors for gene replacement therapy. Viral Immunol 2004; 17: 327–33. pmid:15357899
  46. 46. Robles E, Gilmore-Thomas KK, Miller FB, McMillan DE. Sensitivity to acute methadone dose changes in maintenance patients. J Subst Abuse Treat 2002; 23: 409–13. pmid:12495803
  47. 47. Muhammad AK, Xiong W, Puntel M, Farrokhi C, Kroeger KM, Salem A et al. Safety profile of gutless adenovirus vectors delivered into the normal brain parenchyma: implications for a glioma phase 1 clinical trial. Hum Gene Ther Methods 2012; 23: 271–84. pmid:22950971
  48. 48. Puntel M, A KM G, Farrokhi C, Vanderveen N, Paran C, Appelhans A et al. Safety profile, efficacy, and biodistribution of a bicistronic high-capacity adenovirus vector encoding a combined immunostimulation and cytotoxic gene therapy as a prelude to a phase I clinical trial for glioblastoma. Toxicol Appl Pharmacol 2013; 268: 318–30. pmid:23403069
  49. 49. Finegold AA, Mannes AJ, Iadarola MJ. A paracrine paradigm for in vivo gene therapy in the central nervous system: treatment of chronic pain. Hum Gene Ther 1999; 10: 1251–7. pmid:10340556