全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

17 -Ethynyl-androst-5-ene-3 ,7 ,17 -triol (HE3286) Is Neuroprotective and Reduces Motor Impairment and Neuroinflammation in a Murine MPTP Model of Parkinson’s Disease

DOI: 10.1155/2012/969418

Full-Text   Cite this paper   Add to My Lib

Abstract:

17α-Ethynyl-androst-5-ene-3β,7β,17β-triol (HE3286) is a synthetic androstenetriol in Phase II clinical development for the treatment of inflammatory diseases. HE3286 was evaluated for blood-brain barrier (BBB) permeability in mice, and efficacy in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) murine model of Parkinson’s disease (PD). We found that HE3286 freely penetrated the BBB. HE3286 treatment significantly improved motor function compared to vehicle in the rotarod test (mean 58.2?sec versus 90.9?sec, ), and reduced inflammatory mediator gene expression in the brain (inducible nitric oxide synthase, 20%, ; tumor necrosis factor α, 40%, , and interleukin-1β, 33%, ) measured by reverse-transcriptase polymerase chain reaction. Brain tissue histopathology and immunohistochemistry showed that HE3286 treatment increased the numbers of tyrosine hydroxylase-positive cells by 17% compared to vehicle ( ), and decreased the numbers of damaged neurons by 38% relative to vehicle ( ). L-3,4-dihydroxyphenylalanine (L-DOPA) efficacy was not enhanced by concurrent administration of HE3286. HE3286 administration prior to MPTP did not enhance efficacy. Our data suggest a potential role for HE3286 in PD treatment, and provides incentive for further investigation. 1. Introduction PD is a neurodegenerative disorder characterized by a progressive degeneration of dopaminergic (DAergic) neurons in the substantia nigra pars compacta (SNpc) and decreased levels of dopamine in the putamen of the dorsolateral striatum. The loss of dopamine in the striatum manifests clinically as motor disabilities that include bradykinesia, resting tremor, and muscular rigidity. Diagnosis is based on motor symptoms, which become evident only after the loss of more than 50% of the SNpc DAergic neurons and 60–80% of striatal dopamine [1]. Prolonged treatment of PD with L-DOPA usually results in a dyskinesia that can be more disabling than the disease itself; therefore, there is a great need for alternative therapeutic modalities. The acute MPTP mouse model of nigrostriatal degeneration recapitulates the DAergic neuron loss seen in PD and currently represents the most commonly used toxin-induced mouse model of PD [2]. MPTP’s mechanism of toxicity is complex, and exerted through its toxic metabolite, methyl-4-phenylpyridinium (MPP+) ion, which is taken up selectively by DAergic neurons through the dopamine transporter. Inside the cell, MPP+ is a mitochondrial toxin, which induces neuronal death through several mechanisms that include oxidative stress [3], excitotoxicity [4], and

References

[1]  H. Bernheimer, W. Birkmayer, O. Hornykiewicz, K. Jellinger, and F. Seitelberger, “Brain dopamine and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations,” Journal of the Neurological Sciences, vol. 20, pp. 415–455, 1973.
[2]  V. Jackson-Lewis and S. Przedborski, “Protocol for the MPTP mouse model of Parkinson's disease,” Nature Protocols, vol. 2, no. 1, pp. 141–151, 2007.
[3]  T. Obata, Y. Yamanaka, H. Kinemuchi, and L. Oreland, “Release of dopamine by perfusion with 1-methyl-4-phenylpyridinium ion (MPP+) into the striatum is associated with hydroxyl free radical generation,” Brain Research, vol. 906, no. 1-2, pp. 170–175, 2001.
[4]  G. E. Meredith, S. Totterdell, M. Beales, and C. K. Meshul, “Impaired glutamate homeostasis and programmed cell death in a chronic MPTP mouse model of Parkinson's disease,” Experimental Neurology, vol. 219, no. 1, pp. 334–340, 2009.
[5]  I. Kurkowska-Jastrzebska, E. Ba?kowiec-Iskra, A. Ciesielska et al., “Decreased inflammation and augmented expression of trophic factors correlate with MOG-induced neuroprotection of the injured nigrostriatal system in the murine MPTP model of Parkinson's disease,” International Immunopharmacology, vol. 9, no. 6, pp. 781–791, 2009.
[6]  M. S. Perkinton, J. K. Ip, G. L. Wood, A. J. Crossthwaite, and R. J. Williams, “Phosphatidylinositol 3-kinase is a central mediator of NMDA receptor signalling to MAP kinase (Erkl/2), Akt/PKB and CREB in striatal neurones,” Journal of Neurochemistry, vol. 80, no. 2, pp. 239–254, 2002.
[7]  L. Yang, L. Mao, Q. Tang, S. Samdani, Z. Liu, and J. Q. Wang, “A novel Ca2+-independent signaling pathway to extracellular signal-regulated protein kinase by coactivation of NMDA receptors and metabotropic glutamate receptor 5 in neurons,” Journal of Neuroscience, vol. 24, no. 48, pp. 10846–10857, 2004.
[8]  G. Y. Chen and G. Nu?ez, “Sterile inflammation: sensing and reacting to damage,” Nature Reviews Immunology, vol. 10, no. 12, pp. 826–837, 2010.
[9]  S. F. Yan, R. Ramasamy, and A. M. Schmidt, “The RAGE axis a fundamental mechanism signaling danger to the vulnerable vasculature,” Circulation Research, vol. 106, no. 5, pp. 842–853, 2010.
[10]  D. Pisetsky, “Cell death in the pathogenesis of immune-mediated diseases: the role of HMGB1 and DAMP-PAMP complexes,” Swiss Medical Weekly, vol. 141, article w13256, 2011.
[11]  H. M. Gao, H. Zhou, F. Zhang, B. C. Wilson, W. Kam, and J. S. Hong, “HMGB1 acts on microglia Mac1 to mediate chronic neuroinflammation that drives progressive neurodegeneration,” Journal of Neuroscience, vol. 31, no. 3, pp. 1081–1092, 2011.
[12]  X. Hu, D. Zhang, H. Pang et al., “Macrophage antigen complex-1 mediates reactive microgliosis and progressive dopaminergic neurodegeneration in the MPTP model of Parkinson's disease,” Journal of Immunology, vol. 181, pp. 7194–7204, 2008.
[13]  C. N. Ahlem, M. R. Kennedy, T. M. Page et al., “Studies of the pharmacology of 17α-ethynyl-androst-5-ene-3β,7β,17β-triol, a synthetic anti-inflammatory androstene,” International Journal of Clinical and Experimental Medicine, vol. 4, no. 2, pp. 119–135, 2011.
[14]  D. L. Auci, C. N. Ahlem, M. R. Kennedy, T. M. Page, C. L. Reading, and J. M. Frincke, “A potential role for 5-androstene-3β,7β,17β-triol in obesity and metabolic syndrome,” Obesity, vol. 19, no. 4, pp. 806–811, 2011.
[15]  C. N. Ahlem, D. L. Auci, F. Nicoletti et al., “Pharmacology and immune modulating properties of 5-androstene-3β,7β,17β-triol, a DHEA metabolite in the human metabolome,” Journal of Steroid Biochemistry and Molecular Biology, vol. 126, no. 3–5, pp. 87–94, 2011.
[16]  D. L. Auci, L. Kaler, S. Subramanian et al., “A new orally bioavailable synthetic androstene inhibits collagen-induced arthritis in the mouse: androstene hormones as regulators of regulatory T cells,” Annals of the New York Academy of Sciences, vol. 1110, pp. 630–640, 2007.
[17]  M. M. Kosiewicz, D. L. Auci, P. Fagone et al., “HE3286, an orally bioavailable synthetic analogue of an active DHEA metabolite suppresses spontaneous autoimmune diabetes in the non-obese diabetic (NOD) mouse,” European Journal of Pharmacology, vol. 658, no. 2-3, pp. 257–262, 2011.
[18]  C. Ahlem, D. Auci, K. Mangano et al., “HE3286: a novel synthetic steroid as an oral treatment for autoimmune disease,” Annals of the New York Academy of Sciences, vol. 1173, pp. 781–790, 2009.
[19]  C. L. Reading, J. M. Frincke, and S. K. White, “Molecular targets for 17α-ethynyl-5-androstene-3β,7β,17β-triol, an anti-inflammatory agent derived from the human metabolome,” PLoS One, vol. 7, article e32147, 2012.
[20]  M. Lu, D. Patsouris, P. Li et al., “A new antidiabetic compound attenuates inflammation and insulin resistance in Zucker diabetic fatty rats,” American Journal of Physiology, vol. 298, no. 5, pp. E1036–E1048, 2010.
[21]  S. Subramaniam and K. Unsicker, “ERK and cell death: ERK1/2 in neuronal death,” FEBS Journal, vol. 277, no. 1, pp. 22–29, 2010.
[22]  B. J. Turunen, H. Ge, J. Oyetunji et al., “Paclitaxel succinate analogs: anionic and amide introduction as a strategy to impart blood-brain barrier permeability,” Bioorganic and Medicinal Chemistry Letters, vol. 18, no. 22, pp. 5971–5974, 2008.
[23]  X. Liu, M. Tu, R. S. Kelly, C. Chen, and B. J. Smith, “Development of a computational approach to predict blood-brain barrier permeability,” Drug Metabolism and Disposition, vol. 32, no. 1, pp. 132–139, 2004.
[24]  D. D. Dischino, M. J. Welch, M. R. Kilbourn, and M. E. Raichle, “Relationship between lipophilicity and brain extraction of C-11-labeled radiopharmaceuticals,” Journal of Nuclear Medicine, vol. 24, no. 11, pp. 1030–1038, 1983.
[25]  S. Fasano, E. Bezard, A. D'Antoni et al., “Inhibition of Ras-guanine nucleotide-releasing factor 1 (Ras-GRF1) signaling in the striatum reverts motor symptoms associated with L-dopa-induced dyskinesia,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, pp. 21824–21829, 2010.
[26]  V. Jackson-Lewis, M. Jakowec, R. E. Burke, and S. Przedborski, “Time course and morphology of dopaminergic neuronal death caused by the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine,” Neurodegeneration, vol. 4, no. 3, pp. 257–269, 1995.
[27]  W. G. Tatton, C. E. Greenwood, N. A. Seniuk, and P. T. Salo, “Interactions between MPTP-induced and age-related neuronal death in a murine model of Parkinson's disease,” Canadian Journal of Neurological Sciences, vol. 19, no. 1, pp. 124–133, 1992.
[28]  E. Brouillet and M. F. Beal, “NMDA antagonists partially protect against MPTP induced neurotoxicity in mice,” NeuroReport, vol. 4, no. 4, pp. 387–390, 1993.
[29]  J. B. Schulz, R. T. Matthews, M. M. K. Muqit, S. E. Browne, and M. F. Beal, “Inhibition of neuronal nitric oxide synthase by 7-nitroindazole protects against MPTP-induced neurotoxicity in mice,” Journal of Neurochemistry, vol. 64, no. 2, pp. 936–939, 1995.
[30]  M. Maroso, S. Balosso, T. Ravizza et al., “Toll-like receptor 4 and high-mobility group box-1 are involved in ictogenesis and can be targeted to reduce seizures,” Nature Medicine, vol. 16, no. 4, pp. 413–419, 2010.
[31]  C. Agulhon, M. Y. Sun, T. Murphy, T. Myers, K. Lauderdale, and T. A. Fiacco, “Calcium signaling and gliotransmission in normal versus reactive astrocytes,” Frontiers in Pharmacology, vol. 3, article 139, 2012.
[32]  D. Conrad, A. Wang, R. Pieters et al., “HE3286, an oral synthetic steroid, treats lung inflammation in mice without immune suppression,” Journal of Inflammation, vol. 7, article 52, 2010.
[33]  P. C. Orban, P. F. Chapman, and R. Brambilla, “Is the Ras-MAPK signalling pathway necessary for long-term memory formation?” Trends in Neurosciences, vol. 22, no. 1, pp. 38–44, 1999.
[34]  S. Cagnol and J. C. Chambard, “ERK and cell death: mechanisms of ERK-induced cell death—apoptosis, autophagy and senescence,” FEBS Journal, vol. 277, no. 1, pp. 2–21, 2010.
[35]  L. Xiao, C. Hu, C. Feng, and Y. Chen, “Switching of N-Methyl-D-aspartate (NMDA) receptor-favorite intracellular signal pathways from ERK1/2 protein to p38 mitogen-activated protein kinase leads to developmental changes in NMDA neurotoxicity,” Journal of Biological Chemistry, vol. 286, no. 23, pp. 20175–20193, 2011.
[36]  S. W. Kim, C. M. Lim, J. B. Kim et al., “Extracellular HMGB1 released by NMDA treatment confers neuronal apoptosis via RAGE-p38 MAPK/ERK signaling pathway,” Neurotoxicity Research, vol. 20, pp. 159–169, 2011.
[37]  A. Vezzani, M. Maroso, S. Balosso, M. A. Sanchez, and T. Bartfai, “IL-1 receptor/Toll-like receptor signaling in infection, inflammation, stress and neurodegeneration couples hyperexcitability and seizures,” Brain, Behavior, and Immunity, vol. 25, pp. 1281–1289, 2011.
[38]  E. D. Crown, Y. S. Gwak, Z. Ye, K. M. Johnson, and C. E. Hulsebosch, “Activation of p38 MAP kinase is involved in central neuropathic pain following spinal cord injury,” Experimental Neurology, vol. 213, no. 2, pp. 257–267, 2008.
[39]  Z. Liu, C. Lv, W. Zhao, Y. Song, D. Pei, and T. Xu, “NR2B-containing NMDA receptors expression and their relationship to apoptosis in hippocampus of Alzheimer's disease-like rats,” Neurochemical Research, vol. 37, no. 7, pp. 1420–1427, 2012.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133