1
|
Kang X, Wang W, Zuo Y, Wang Y, Zhang L, Liu L. Dopamine receptor agonist pramipexole exerts neuroprotection on global cerebral ischemia/reperfusion injury by inhibiting ferroptosis. J Stroke Cerebrovasc Dis 2024:108101. [PMID: 39490461 DOI: 10.1016/j.jstrokecerebrovasdis.2024.108101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024] Open
Abstract
OBJECTIVE To explore the mechanism of dopamine receptor agonist pramipexole in exerting neuroprotection on global cerebral ischemia/reperfusion injury (GCI/R). MATERIAL AND METHOD Male Sprague-Dawley rats were randomly divided into four groups (n = 36 in each group), and the Pulsinelli's four-vessel occlusion method was used to establish the rat model of GCI/R injury. Pramipexole administration group was intraperitoneally injected with pramipexole 0.5 mg/kg once a day for 14 days. Pramipexole combined with levodopa administration group was intraperitoneally injected with pramipexole 0.5 mg/kg and levodopa 50 mg/kg once a day for 14 days. The mNSS scores and Y maze test were used to evaluate neurological behaviors. Nissl staining and transmission electron microscopy were used to respectively observe hippocampal neurons and mitochondrial ultrastructure. Molecular biological tests including tissue iron concentration, GSH, MDA were used to detect the degree of ferroptosis. Western blotting was used to detect the expression levels of Nrf2, GPX4, X-CT and p53 proteins at 3 days, 7 days and 14 days after GCI/R injury. RESULTS Pramipexole alone or combined with levodopa for 14 days improved neurological behaviors, improved the morphology of neurons, increased the number of surviving neurons in the hippocampal CA1 region of GCI/R rats, which showed similar neuroprotective effects. Pramipexole alone or combined with levodopa for 14 days restored mitochondrial ultrastructure, decreased malondialdehyde concentration and increased glutathione concentration in the brain of GCI/R rats, which also induced the relative expressions of Nrf2, GPX4 and X-CT proteins and reduced p53 protein. CONCLUSION Pramipexole alone or combined with levodopa exert neuroprotection by inhibiting ferroptosis after GCI/R injury via Nrf2/GPX4/SLC7A11 pathway, and long-term intervention could be applied as an effective therapeutic strategy for neuroprotection against GCI/R injury.
Collapse
Affiliation(s)
- Xiaoyu Kang
- School of Rehabilitation, Capital Medical University, Beijing, China; Beijing Bo'ai hospital, China Rehabilitation Research Center, Beijing, China.
| | - Wenzhu Wang
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China; Institute of Rehabilitation Medicine of China, Chinese Institute of Rehabilitation Science, Beijing, China.
| | - Yao Zuo
- Beijing Bo'ai hospital, China Rehabilitation Research Center, Beijing, China; Shandong University Cheeloo College of Medicine, Jinan, Shandong, China.
| | - Yunlei Wang
- School of Rehabilitation, Capital Medical University, Beijing, China; Beijing Bo'ai hospital, China Rehabilitation Research Center, Beijing, China.
| | - Linyao Zhang
- School of Rehabilitation, Capital Medical University, Beijing, China; Beijing Bo'ai hospital, China Rehabilitation Research Center, Beijing, China.
| | - Lixu Liu
- School of Rehabilitation, Capital Medical University, Beijing, China; Beijing Bo'ai hospital, China Rehabilitation Research Center, Beijing, China; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China.
| |
Collapse
|
2
|
Prajapati R, Seong SH, Paudel P, Park SE, Jung HA, Choi JS. In Vitro and In Silico Characterization of Kurarinone as a Dopamine D 1A Receptor Antagonist and D 2L and D 4 Receptor Agonist. ACS OMEGA 2021; 6:33443-33453. [PMID: 34926894 PMCID: PMC8674921 DOI: 10.1021/acsomega.1c04109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/24/2021] [Indexed: 06/14/2023]
Abstract
Alterations in the expression and/or activity of brain G-protein-coupled receptors (GPCRs) such as dopamine D1R, D2LR, D3R, and D4R, vasopressin V1AR, and serotonin 5-HT1AR are noted in various neurodegenerative diseases (NDDs). Since studies have indicated that flavonoids can target brain GPCRs and provide neuroprotection via inhibition of monoamine oxidases (hMAOs), our study explored the functional role of kurarinone, an abundant lavandulated flavonoid in Sophora flavescens, on dopamine receptor subtypes, V1AR, 5-HT1AR, and hMAOs. Radioligand binding assays revealed considerable binding of kurarinone on D1R, D2LR, and D4R. Functional GPCR assays unfolded the compound's antagonist behavior on D1R (IC50 42.1 ± 0.35 μM) and agonist effect on D2LR and D4R (EC50 22.4 ± 3.46 and 71.3 ± 4.94 μM, respectively). Kurarinone was found to inhibit hMAO isoenzymes in a modest and nonspecific manner. Molecular docking displayed low binding energies during the intermolecular interactions of kurarinone with the key residues of the deep orthosteric binding pocket and the extracellular loops of D1R, D2LR, and D4R, validating substantial binding affinities to these prime targets. With appreciable D2LR and D4R agonism and D1R antagonism, kurarinone might be a potential compound that can alleviate clinical symptoms of Parkinson's disease and other NDDs.
Collapse
Affiliation(s)
- Ritu Prajapati
- Department
of Food and Life Science, Pukyong National
University, Busan 48513, Republic of Korea
| | - Su Hui Seong
- Department
of Food and Life Science, Pukyong National
University, Busan 48513, Republic of Korea
- Natural
Products Research Division, Honam National
Institute of Biological Resource, Mokpo 58762, Republic
of Korea
| | - Pradeep Paudel
- Department
of Food and Life Science, Pukyong National
University, Busan 48513, Republic of Korea
- National
Center for Natural Products Research, The
University of Mississippi, Oxford, Mississippi 38677, United States
| | - Se Eun Park
- Department
of Food and Life Science, Pukyong National
University, Busan 48513, Republic of Korea
- Department
of Biomedical Science, Asan Medical Institute
of Convergence Science and Technology, Seoul 05505, Republic
of Korea
| | - Hyun Ah Jung
- Department
of Food Science and Human Nutrition, Jeonbuk
National University, Jeonju 54896, Republic of Korea
| | - Jae Sue Choi
- Department
of Food and Life Science, Pukyong National
University, Busan 48513, Republic of Korea
| |
Collapse
|
3
|
Liu P, Qin D, Lv H, Fan W, Tao Z, Xu Y. Neuroprotective effects of dopamine D2 receptor agonist on neuroinflammatory injury in olfactory bulb neurons in vitro and in vivo in a mouse model of allergic rhinitis. Neurotoxicology 2021; 87:174-181. [PMID: 34624383 DOI: 10.1016/j.neuro.2021.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 08/22/2021] [Accepted: 10/01/2021] [Indexed: 11/17/2022]
Abstract
Available evidence indicates that dopamine D2 receptor modulates the neurotoxic effects induced by glutamate. However, neurotoxicity mediated by AMPA-subtype glutamate receptor has rarely been studied in the olfactory bulb. This study mainly explores the neuroprotective effects of dopamine D2 receptor agonist on AMPA receptor-mediated neurotoxicity in the olfactory bulb in a mouse model of allergic rhinitis (AR) with olfactory dysfunction (OD). In our study, we found that AR with OD was closely associated with increased surface expression of the AMPA receptor GluR1, reduced surface expression of GluR2, and apoptosis damage in the olfactory bulb in vivo. Quinpirole (a dopamine D2 receptor agonist) improved olfactory function in mice, ameliorated apoptosis injury in the olfactory bulb but not in the olfactory mucosa, and inhibited the internalization of GluR2-containing AMPA receptor in vitro and in vivo. In addition, phosphorylation plays a crucial role in the regulation of AMPA receptor trafficking. Our results showed that quinpirole reduced the phosphorylation of GluR1 S845 and GluR2 S880 in olfactory bulb neurons in vitro, but it had no obvious effect on GluR1 S831. Therefore, dopamine D2 receptor agonist may inhibit the phosphorylation of GluR1 S845 and GluR2 S880, thereby reducing AMPA receptor-mediated neurotoxicity and alleviating neurotoxic injury to the olfactory bulb caused by AR.
Collapse
Affiliation(s)
- Peiqiang Liu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Danxue Qin
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hao Lv
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenjun Fan
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zezhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yu Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
4
|
Shin EJ, Jeong JH, Hwang Y, Sharma N, Dang DK, Nguyen BT, Nah SY, Jang CG, Bing G, Nabeshima T, Kim HC. Methamphetamine-induced dopaminergic neurotoxicity as a model of Parkinson's disease. Arch Pharm Res 2021; 44:668-688. [PMID: 34286473 DOI: 10.1007/s12272-021-01341-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/06/2021] [Indexed: 12/01/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with a high prevalence, approximately 1 % in the elderly population. Numerous studies have demonstrated that methamphetamine (MA) intoxication caused the neurological deficits and nigrostriatal damage seen in Parkinsonian conditions, and subsequent rodent studies have found that neurotoxic binge administration of MA reproduced PD-like features, in terms of its symptomatology and pathology. Several anti-Parkinsonian medications have been shown to attenuate the motor impairments and dopaminergic damage induced by MA. In addition, it has been recognized that mitochondrial dysfunction, oxidative stress, pro-apoptosis, proteasomal/autophagic impairment, and neuroinflammation play important roles in inducing MA neurotoxicity. Importantly, MA neurotoxicity has been shown to share a common mechanism of dopaminergic toxicity with that of PD pathogenesis. This review describes the major findings on the neuropathological features and underlying neurotoxic mechanisms induced by MA and compares them with Parkinsonian pathogenesis. Taken together, it is suggested that neurotoxic binge-type administration of MA in rodents is a valid animal model for PD that may provide knowledge on the neuropathogenesis of PD.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, 06974, Seoul, Republic of Korea
| | - Yeonggwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.,Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, 06974, Seoul, Republic of Korea
| | - Duy-Khanh Dang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.,Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, 900000, Can Tho City, Vietnam
| | - Bao-Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory, Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, 05029, Seoul, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, 16419, Suwon, Republic of Korea
| | - Guoying Bing
- Department of Neuroscience, College of Medicine, University of Kentucky, KY, 40536, Lexington, USA
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Graduate School of Health Science, Fujita Health University, 470-1192, Toyoake, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea. .,Neuropsychopharmacology & Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.
| |
Collapse
|
5
|
G-Protein-Coupled Receptors and Ischemic Stroke: a Focus on Molecular Function and Therapeutic Potential. Mol Neurobiol 2021; 58:4588-4614. [PMID: 34120294 DOI: 10.1007/s12035-021-02435-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/18/2021] [Indexed: 01/22/2023]
Abstract
In ischemic stroke, there is only one approved drug, tissue plasminogen activator, to be used in clinical conditions for thrombolysis. New neuroprotective therapies for ischemic stroke are desperately needed. Several targets and pathways have been shown to confer neuroprotective effects in ischemic stroke. G-protein-coupled receptors (GPCRs) are one of the most frequently targeted receptors for developing novel therapeutics for central nervous system disorders. GPCRs are a large family of cell surface receptors that response to a wide variety of extracellular stimuli. GPCRs are involved in a wide range of physiological and pathological processes. More than 90% of the identified non-sensory GPCRs are expressed in the brain, where they play important roles in regulating mood, pain, vision, immune responses, cognition, and synaptic transmission. There is also good evidence that GPCRs are implicated in the pathogenesis of stroke. This review narrates the pathophysiological role and possible targeted therapy of GPCRs in ischemic stroke.
Collapse
|
6
|
Sharma N, Shin EJ, Pham DT, Sharma G, Dang DK, Duong CX, Kang SW, Nah SY, Jang CG, Lei XG, Nabeshima T, Bing G, Jeong JH, Kim HC. GPx-1-encoded adenoviral vector attenuates dopaminergic impairments induced by methamphetamine in GPx-1 knockout mice through modulation of NF-κB transcription factor. Food Chem Toxicol 2021; 154:112313. [PMID: 34082047 DOI: 10.1016/j.fct.2021.112313] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 01/10/2023]
Abstract
We suggested that selenium-dependent glutathione peroxidase (GPx) plays a protective role against methamphetamine (MA)-induced dopaminergic toxicity. We focused on GPx-1, a major selenium-dependent enzyme and constructed a GPx-1 gene-encoded adenoviral vector (Ad-GPx-1) to delineate the role of GPx-1 in MA-induced dopaminergic neurotoxicity. Exposure to Ad-GPx-1 significantly induced GPx activity and GPx-1 protein levels in GPx-1-knockout (GPx-1-KO) mice. MA-induced dopaminergic impairments [i.e., hyperthermia; increased nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) DNA-binding activity; and decreased dopamine levels, TH activity, and behavioral activity] were more pronounced in GPx-1-KO mice than in WT mice. In contrast, exposure to Ad-GPx-1 significantly attenuated MA-induced dopaminergic loss in GPx-1-KO mice. The protective effect exerted by Ad-GPx-1 was comparable to that exerted by pyrrolidine dithiocarbamate (PDTC), an NF-κB inhibitor against MA insult. Consistently, GPx-1 overexpression significantly attenuated MA dopaminergic toxicity in mice. PDTC did not significantly impact the protective effect of GPx-1 overexpression, suggesting that interaction between NF-κB and GPx-1 is critical for dopaminergic protection. Thus, NF-κB is a potential therapeutic target for GPx-1-mediated dopaminergic protective activity. This study for the first time demonstrated that Ad-GPx-1 rescued dopaminergic toxicity in vivo following MA insult. Furthermore, GPx-1-associated therapeutic interventions may be important against dopaminergic toxicity.
Collapse
Affiliation(s)
- Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, South Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, South Korea
| | - Duc Toan Pham
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, South Korea
| | - Garima Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, South Korea
| | - Duy-Khanh Dang
- Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, Can Tho City, 900000, Viet Nam
| | - Chu Xuan Duong
- Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, Can Tho City, 900000, Viet Nam
| | - Sang Won Kang
- Department of Life Science, College of Natural Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, 05029, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, Republic of Korea
| | - Xin Gen Lei
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Toyoake, 470-1192, Japan
| | - Guoying Bing
- Anatomy and Neurobiology, University of Kentucky Medical Center, Medical Center MN208 800 Rose Strees, Lexington, KY, 40536, USA
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea.
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, South Korea.
| |
Collapse
|
7
|
Gao H, Wang D, Wang YL, Mao JP, Jiang S, Yang XL. Pramipexole attenuates 6-OHDA-induced Parkinson's disease by mediating the Nurr1/NF-κB pathway. Mol Biol Rep 2021; 48:3079-3087. [PMID: 33891271 DOI: 10.1007/s11033-021-06343-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 04/07/2021] [Indexed: 11/30/2022]
Abstract
Neuroinflammation is the key factor associated with the progression of Parkinson's disease (PD). Pramipexole (PPX) has anti-inflammatory and antioxidant properties. This study explored the effects of PPX on PD and its related mechanisms. A PD rat model was established using 6-hydroxydopamine (6-OHDA). Thirty rats were divided into the following three groups: control, PD, and PD + PPX. The rats in the PD and PD + PPX groups were first administered 6-OHDA and then respectively treated with saline and PPX. Afterward, rotational behavior tests were performed to evaluate the efficiency of PPX. The level of tyrosine hydroxylase (TH) was measured using immunohistochemical staining. Subsequently, real-time quantitative PCR (RT-qPCR) and western blot were used to determine the expression of α-synuclein (α-syn), nuclear receptor subfamily 4 group A member 2 (Nurr1), and nuclear factor kappa B (NF-κB). PPX improved the motor behavior of PD rats caused by 6-OHDA. The number of TH-positive neurons in the PD group was significantly lower than that in the control group (P < 0.05), while PPX could rescue 6-OHDA-induced TH loss. RT-qPCR and western blot showed that Nurr1 expression was significantly downregulated in the PD group compared to that of the control group (P < 0.05), while after PPX treatment, its expression was significantly upregulated (P < 0.05). For α-syn and NF-κB, 6-OHDA significantly upregulated their expressions (P < 0.05), whereas PPX reversed them. PPX improved the motor behavior of PD through mediating the inflammatory response and regulating the Nurr1/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Hua Gao
- Department of Neurology, Second Hospital Affiliated of Xinjiang Medical University, No. 38, South Lake Road, Ürümqi, 830063, Xinjiang, China.,Department of Neurology, Fifth Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830011, Xinjiang, China
| | - Dan Wang
- Department of Neurology, Second Hospital Affiliated of Xinjiang Medical University, No. 38, South Lake Road, Ürümqi, 830063, Xinjiang, China
| | - Yu-Ling Wang
- Cadre Health Center, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, China
| | - Jie-Ping Mao
- Department of Neurology, Urumqi Friendship Hospital, Ürümqi, 830049, Xinjiang, China
| | - Sen Jiang
- Department of Neurology, Second Hospital Affiliated of Xinjiang Medical University, No. 38, South Lake Road, Ürümqi, 830063, Xinjiang, China
| | - Xin-Ling Yang
- Department of Neurology, Second Hospital Affiliated of Xinjiang Medical University, No. 38, South Lake Road, Ürümqi, 830063, Xinjiang, China.
| |
Collapse
|
8
|
Sridhar V, Tiwari A, Wairkar S, Gupta GL, Gaud R. Pramipexole thermosensitive nasal gel for experimental parkinsonism in rats. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
9
|
Mallah K, Couch C, Borucki DM, Toutonji A, Alshareef M, Tomlinson S. Anti-inflammatory and Neuroprotective Agents in Clinical Trials for CNS Disease and Injury: Where Do We Go From Here? Front Immunol 2020; 11:2021. [PMID: 33013859 PMCID: PMC7513624 DOI: 10.3389/fimmu.2020.02021] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Neurological disorders are major contributors to death and disability worldwide. The pathology of injuries and disease processes includes a cascade of events that often involve molecular and cellular components of the immune system and their interaction with cells and structures within the central nervous system. Because of this, there has been great interest in developing neuroprotective therapeutic approaches that target neuroinflammatory pathways. Several neuroprotective anti-inflammatory agents have been investigated in clinical trials for a variety of neurological diseases and injuries, but to date the results from the great majority of these trials has been disappointing. There nevertheless remains great interest in the development of neuroprotective strategies in this arena. With this in mind, the complement system is being increasingly discussed as an attractive therapeutic target for treating brain injury and neurodegenerative conditions, due to emerging data supporting a pivotal role for complement in promoting multiple downstream activities that promote neuroinflammation and degeneration. As we move forward in testing additional neuroprotective and immune-modulating agents, we believe it will be useful to review past trials and discuss potential factors that may have contributed to failure, which will assist with future agent selection and trial design, including for complement inhibitors. In this context, we also discuss inhibition of the complement system as a potential neuroprotective strategy for neuropathologies of the central nervous system.
Collapse
Affiliation(s)
- Khalil Mallah
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Christine Couch
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Health Sciences and Research, College of Health Professions, Medical University of South Carolina, Charleston, SC, United States
| | - Davis M. Borucki
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, United States
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, United States
| | - Amer Toutonji
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, United States
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, United States
| | - Mohammed Alshareef
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurological Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Ralph Johnson VA Medical Center, Charleston, SC, United States
| |
Collapse
|
10
|
Salamon A, Zádori D, Szpisjak L, Klivényi P, Vécsei L. Neuroprotection in Parkinson's disease: facts and hopes. J Neural Transm (Vienna) 2019; 127:821-829. [PMID: 31828513 PMCID: PMC7242234 DOI: 10.1007/s00702-019-02115-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/03/2019] [Indexed: 12/15/2022]
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease worldwide. Behind the symptoms there is a complex pathological mechanism which leads to a dopaminergic cell loss in the substantia nigra pars compacta. Despite the strong efforts, curative treatment has not been found yet. To prevent a further cell death, numerous molecules were tested in terms of neuroprotection in preclinical (in vitro, in vivo) and in clinical studies as well. The aim of this review article is to summarize our knowledge about the extensively tested neuroprotective agents (Search period: 1991–2019). We detail the underlying pathological mechanism and summarize the most important results of the completed animal and clinical trials. Although many positive results have been reported in the literature, there is still no evidence that any of them should be used in clinical practice (Cochrane analysis was performed). Therefore, further studies are needed to better understand the pathomechanism of PD and to find the optimal neuroprotective agent(s).
Collapse
Affiliation(s)
- András Salamon
- Department of Neurology, Faculty of Medicine, Interdisciplinary Excellence Centre, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6., Szeged, 6725, Hungary
| | - Dénes Zádori
- Department of Neurology, Faculty of Medicine, Interdisciplinary Excellence Centre, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6., Szeged, 6725, Hungary
| | - László Szpisjak
- Department of Neurology, Faculty of Medicine, Interdisciplinary Excellence Centre, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6., Szeged, 6725, Hungary
| | - Péter Klivényi
- Department of Neurology, Faculty of Medicine, Interdisciplinary Excellence Centre, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6., Szeged, 6725, Hungary
| | - László Vécsei
- Department of Neurology, Faculty of Medicine, Interdisciplinary Excellence Centre, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6., Szeged, 6725, Hungary. .,MTA-SZTE Neuroscience Research Group, Szeged, Hungary.
| |
Collapse
|
11
|
Sabrini S, Russell B, Wang G, Lin J, Kirk I, Curley L. Methamphetamine induces neuronal death: Evidence from rodent studies. Neurotoxicology 2019; 77:20-28. [PMID: 31812708 DOI: 10.1016/j.neuro.2019.12.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 10/23/2019] [Accepted: 12/04/2019] [Indexed: 12/19/2022]
Abstract
Animal studies have consistently observed neuronal death following methamphetamine (MA) administration, however, these have not been systematically reviewed. This systematic review aims to present the evidence for MA-induced neuronal death in animals (rodents) and identify the regions affected. Locating the brain regions in which neuronal death occurs in animal studies will provide valuable insight into the linkage between MA consumption and the structural alterations observed in the human brain. The data were collected from three databases: Scopus, Ovid, and the Web of Science. Thirty-seven studies met the inclusion criteria and were divided into two sub-groups, i.e. acute and repeated administration. Twenty-six (of 27) acute and ten (of 11) repeated administration studies observed neuronal death. A meta-analysis was not possible due to different variables between studies, i.e. species, treatment regimens, withdrawal periods, methods of quantification, and regions studied. Acute MA treatment induced neuronal death in the frontal cortex, striatum, and substantia nigra, but not in the hippocampus, whereas repeated MA administration led to neuronal loss in the hippocampus, frontal cortex, and striatum. In addition, when animals self-administered the drug, neuronal death was observed at much lower doses than the doses administered by experimenters. There is some overlap in the regions where neuronal death occurred in animals and the identified regions from human studies. For instance, gray matter deficits have been observed in the prefrontal cortex and hippocampus of MA users. The findings presented in this review implicate that not only does MA induce neuronal death in animals, but it also damages the same regions affected in human users. Despite the inter-species differences, animal studies have contributed significantly to addiction research, and are still of great assistance for future research with a more relevant model of compulsive drug use in humans.
Collapse
Affiliation(s)
- Sabrini Sabrini
- School of Pharmacy, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142 New Zealand.
| | - Bruce Russell
- School of Pharmacy, University of Otago, New Zealand.
| | - Grace Wang
- Department of Psychology, Faculty of Health and Environmental Sciences, Auckland University of Technology, New Zealand.
| | - Joanne Lin
- School of Pharmacy, Faculty of Medical and Health Sciences, The University of Auckland, New Zealand.
| | - Ian Kirk
- School of Psychology, Faculty of Science, The University of Auckland, New Zealand.
| | - Louise Curley
- School of Pharmacy, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142 New Zealand.
| |
Collapse
|
12
|
Andrabi SS, Ali M, Tabassum H, Parveen S, Parvez S. Pramipexole prevents ischemic cell death via mitochondrial pathways in ischemic stroke. Dis Model Mech 2019; 12:dmm.033860. [PMID: 31235613 PMCID: PMC6737958 DOI: 10.1242/dmm.033860] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 06/06/2019] [Indexed: 12/21/2022] Open
Abstract
A dopamine D2 receptor agonist, pramipexole, has been found to elicit neuroprotection in patients with Parkinson's disease and restless leg syndrome. Recent evidence has shown that pramipexole mediates its neuroprotection through mitochondria. Considering this, we examined the possible mitochondrial role of pramipexole in promoting neuroprotection following an ischemic stroke of rat. Male Wistar rats underwent transient middle cerebral artery occlusion (tMCAO) and then received pramipexole (0.25 mg and 1 mg/kg body weight) at 1, 6, 12 and 18 h post-occlusion. A panel of neurological tests and 2,3,5-triphenyl tetrazolium chloride (TTC) staining were performed at 24 h after the surgery. Flow cytometry was used to detect the mitochondrial membrane potential, and mitochondrial levels of reactive oxygen species (ROS) and Ca2+, respectively. Mitochondrial oxidative phosphorylation was analyzed by oxygraph (oxygen electrode). Western blotting was used to analyze the expression of various proteins such as Bax, Bcl-2 and cytochrome c Pramipexole promoted the neurological recovery as shown by the panel of neurobehavioral tests and TTC staining. Post-stroke treatment with pramipexole reduced levels of mitochondrial ROS and Ca2+ after ischemia. Pramipexole elevated the mitochondrial membrane potential and mitochondrial oxidative phosphorylation. Western blotting showed that pramipexole inhibited the transfer of cytochrome c from mitochondria to cytosol, and hence inhibited the mitochondrial permeability transition pore. Thus, our results have demonstrated that post-stroke administration of pramipexole induces the neurological recovery through mitochondrial pathways in ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Syed Suhail Andrabi
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Mubashshir Ali
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Heena Tabassum
- Division of Basic Medical Sciences, Indian Council of Medical Research, Ministry of Health and Family Welfare, Government of India, V. Ramalingaswamy Bhawan, New Delhi 110 029, India
| | - Sabiha Parveen
- Department of Communication Sciences and Disorders, Oklahoma State University, Stillwater, OK 74078, USA
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
13
|
Effectiveness of Fragment C Domain of Tetanus Toxin and Pramipexole in an Animal Model of Parkinson’s Disease. Neurotox Res 2019; 35:699-710. [DOI: 10.1007/s12640-018-9990-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 12/05/2018] [Accepted: 12/11/2018] [Indexed: 12/13/2022]
|
14
|
Wang Y, Yu X, Zhang P, Ma Y, Wang L, Xu H, Sui D. Neuroprotective effects of pramipexole transdermal patch in the MPTP-induced mouse model of Parkinson's disease. J Pharmacol Sci 2018; 138:31-37. [DOI: 10.1016/j.jphs.2018.08.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 07/30/2018] [Accepted: 08/07/2018] [Indexed: 01/12/2023] Open
|
15
|
Aripiprazole exerts a neuroprotective effect in mouse focal cerebral ischemia. Exp Ther Med 2017; 15:745-750. [PMID: 29399080 PMCID: PMC5772374 DOI: 10.3892/etm.2017.5443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/29/2017] [Indexed: 01/24/2023] Open
Abstract
Previous studies have demonstrated that aripiprazole (APZ), a third-generation atypical antipsychotic drug, exhibits anti-depressant and neuroprotective effects by promoting dopaminergic neuronal cell recovery in stroke. To investigate the neuroprotective effects of APZ, behavioral and histopathological experiments were performed in the current study a mouse model of middle cerebral artery occlusion (MCAO)-induced ischemia following administration of APZ. The subacute phase of ischemic assaults was divided into 3 periods, each with a duration of 5 days, according to the start of APZ (3 mg/kg) administration (1–5, 5–9 or 10–14 days following MCAO). The beneficial effects of APZ on motor behavior demonstrated in the cylinder, rotarod and wire suspension tests were greatest when APZ was administered 1–5 days following MCAO, with clear improvements in motor function compared with vehicle-treated mice. Histopathological analysis revealed that prominent atrophic changes occurred in the striatum of MCAO mice and that these changes were reduced following APZ treatment. APZ also attenuated dopaminergic neuronal injury in the striatum. Cell death and microglial activation were decreased and the expression of Ca2+/calmodulin-dependent protein kinase II δ was enhanced following APZ treatment. These results indicate that the atypical antipsychotic drug, APZ, exhibits a neuroprotective effect in dopaminergic neuronal cells that may improve behavioral function following ischemic stroke.
Collapse
|
16
|
Sadeghi H, Parishani M, Akbartabar Touri M, Ghavamzadeh M, Jafari Barmak M, Zarezade V, Delaviz H, Sadeghi H. Pramipexole reduces inflammation in the experimental animal models of inflammation. Immunopharmacol Immunotoxicol 2017; 39:80-86. [PMID: 28162057 DOI: 10.1080/08923973.2017.1284230] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Pramipexole is a dopamine (DA) agonist (D2 subfamily receptors) that widely use in the treatment of Parkinson's diseases. Some epidemiological and genetic studies propose a role of inflammation in the pathophysiology of Parkinson's disease. To our knowledge, there is no study regarding the anti-inflammatory activity of pramipexol. Therefore, the aim of the study was to investigate anti-inflammatory effect of pramipexol. Anti-inflammatory effects of pramipexole were studied in three well-characterized animal models of inflammation, including carrageenan- or formalin-induced paw inflammation in rats, and 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced ear edema in mice. The animals received pramipexol (0.25, 0.5 and 1 mg/kg, I.P.) 30 min before subplantar injection of carrageenan or formalin. Pramipexol (0.5 and 1 mg/kg) was also injected 30 min before topical application of TPA on the ear mice. Serum malondialdehyde (MDA) levels were evaluated in the carrageenan test. Finally, pathological examination of the inflamed tissues was carried out. Pramipexole significantly inhibited paw inflammation 1, 2, 3 and 4 h after carrageenan challenge compared with the control group (p < .001). Pramipexol also showed considerable anti-inflammatory activity against formalin-evoked paw edema over a period of 24 h (p < .001). TPA-induced ear edema was markedly decreased by pramipexol (p < .001). The pathological evaluation of the paws and ears revealed that pramipexole reduced tissue injury, neutrophil infiltration, and subcutaneous edema. Pramipexole did not alter the increased serum levels of MDA due to carrageenan injection. These data clearly indicate that pramipexol possesses significant anti-inflammatory activity. It seems that its antioxidants do not play an important role in these effects.
Collapse
Affiliation(s)
- Heibatollah Sadeghi
- a Cellular and Molecular Research Center, Yasuj University of Medical Sciences , Yasuj , Iran.,b Medicinal Plants Research Center, Yasuj University of Medical Sciences , Yasuj , Iran
| | - Mohammad Parishani
- c Student Research Committee , Yasuj University of Medical Sciences , Yasuj , Iran
| | - Mehdi Akbartabar Touri
- d Social Determinants of Health Research Center, Yasuj University of Medical Sciences , Yasuj , Iran
| | - Mehdi Ghavamzadeh
- c Student Research Committee , Yasuj University of Medical Sciences , Yasuj , Iran
| | - Mehrzad Jafari Barmak
- a Cellular and Molecular Research Center, Yasuj University of Medical Sciences , Yasuj , Iran
| | - Vahid Zarezade
- e Department of Biochemistry, School of Medicine, Behbahan Faculty of Medical Sciences , Behbahan , Iran
| | - Hamdollah Delaviz
- a Cellular and Molecular Research Center, Yasuj University of Medical Sciences , Yasuj , Iran
| | - Hossein Sadeghi
- a Cellular and Molecular Research Center, Yasuj University of Medical Sciences , Yasuj , Iran.,b Medicinal Plants Research Center, Yasuj University of Medical Sciences , Yasuj , Iran.,f Department of Pharmacology, Faculty of Medicine , Yasuj University of Medical Sciences , Yasuj , Iran
| |
Collapse
|
17
|
Osier ND, Dixon CE. Catecholaminergic based therapies for functional recovery after TBI. Brain Res 2015; 1640:15-35. [PMID: 26711850 DOI: 10.1016/j.brainres.2015.12.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/11/2015] [Accepted: 12/14/2015] [Indexed: 11/15/2022]
Abstract
Among the many pathophysiologic consequences of traumatic brain injury are changes in catecholamines, including dopamine, epinephrine, and norepinephrine. In the context of TBI, dopamine is the one most extensively studied, though some research exploring epinephrine and norepinephrine have also been published. The purpose of this review is to summarize the evidence surrounding use of drugs that target the catecholaminergic system on pathophysiological and functional outcomes of TBI using published evidence from pre-clinical and clinical brain injury studies. Evidence of the effects of specific drugs that target catecholamines as agonists or antagonists will be discussed. Taken together, available evidence suggests that therapies targeting the catecholaminergic system may attenuate functional deficits after TBI. Notably, it is fairly common for TBI patients to be treated with catecholamine agonists for either physiological symptoms of TBI (e.g. altered cerebral perfusion pressures) or a co-occuring condition (e.g. shock), or cognitive symptoms (e.g. attentional and arousal deficits). Previous clinical trials are limited by methodological limitations, failure to replicate findings, challenges translating therapies to clinical practice, the complexity or lack of specificity of catecholamine receptors, as well as potentially counfounding effects of personal and genetic factors. Overall, there is a need for additional research evidence, along with a need for systematic dissemination of important study details and results as outlined in the common data elements published by the National Institute of Neurological Diseases and Stroke. Ultimately, a better understanding of catecholamines in the context of TBI may lead to therapeutic advancements. This article is part of a Special Issue entitled SI:Brain injury and recovery.
Collapse
Affiliation(s)
- Nicole D Osier
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; School of Nursing, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - C Edward Dixon
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15260, USA; V.A. Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA.
| |
Collapse
|
18
|
Huck JHJ, Freyer D, Böttcher C, Mladinov M, Muselmann-Genschow C, Thielke M, Gladow N, Bloomquist D, Mergenthaler P, Priller J. De novo expression of dopamine D2 receptors on microglia after stroke. J Cereb Blood Flow Metab 2015; 35:1804-11. [PMID: 26104289 PMCID: PMC4635235 DOI: 10.1038/jcbfm.2015.128] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 04/01/2015] [Accepted: 04/27/2015] [Indexed: 11/09/2022]
Abstract
Dopamine is the predominant catecholamine in the brain and functions as a neurotransmitter. Dopamine is also a potent immune modulator. In this study, we have characterized the expression of dopamine receptors on murine microglia. We found that cultured primary microglia express dopamine D1, D2, D3, D4, and D5 receptors. We specifically focused on the D2 receptor (D2R), a major target of antipsychotic drugs. Whereas D2Rs were strongly expressed on striatal neurons in vivo, we did not detect any D2R expression on resident microglia in the healthy brains of wild-type mice or transgenic mice expressing the green fluorescent protein (GFP) under the control of the Drd2 promoter. However, cerebral ischemia induced the expression of D2R on Iba1-immunoreactive inflammatory cells in the infarct core and penumbra. Notably, D2R expression was confined to CD45(hi) cells, and GFP BM chimeras revealed that D2R was expressed on activated resident microglia as well as on peripherally derived macrophages in the ischemic brain. Importantly, the D2/3R agonist, pramipexole, enhanced the secretion of nitrite by cultured microglia in response to proinflammatory stimuli. Thus, dopamine may serve as a modulator of microglia function during neuroinflammation.
Collapse
Affiliation(s)
- Jojanneke H J Huck
- Laboratory of Molecular Psychiatry, Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Dorette Freyer
- Departments of Experimental Neurology, Neurology and Center for Stroke Research, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Chotima Böttcher
- Laboratory of Molecular Psychiatry, Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Mihovil Mladinov
- Laboratory of Molecular Psychiatry, Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Muselmann-Genschow
- Departments of Experimental Neurology, Neurology and Center for Stroke Research, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Mareike Thielke
- Departments of Experimental Neurology, Neurology and Center for Stroke Research, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Nadine Gladow
- Laboratory of Molecular Psychiatry, Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Dana Bloomquist
- Laboratory of Molecular Psychiatry, Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Philipp Mergenthaler
- Departments of Experimental Neurology, Neurology and Center for Stroke Research, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Cluster of Excellence 'NeuroCure', Berlin, Germany.,Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Josef Priller
- Laboratory of Molecular Psychiatry, Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Cluster of Excellence 'NeuroCure', Berlin, Germany.,BIH and DZNE, Berlin, Germany
| |
Collapse
|
19
|
Mohammad Ahmadi Soleimani S, Ekhtiari H, Cadet JL. Drug-induced neurotoxicity in addiction medicine: From prevention to harm reduction. PROGRESS IN BRAIN RESEARCH 2015; 223:19-41. [PMID: 26806769 DOI: 10.1016/bs.pbr.2015.07.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neurotoxicity is considered as a major cause of neurodegenerative disorders. Most drugs of abuse have nonnegligible neurotoxic effects many of which are primarily mediated by several dopaminergic and glutamatergic neurotransmitter systems. Although many researchers have investigated the medical and cognitive consequences of drug abuse, the neurotoxicity induced by these drugs still requires comprehensive attention. The science of neurotoxicity promises to improve preventive and therapeutic strategies for brain disorders such as Alzheimer disease and Parkinson's disease. However, its clinical applications for addiction medicine remain to be defined adequately. This chapter reviews the most commonly discussed mechanisms underlying neurotoxicity induced by common drugs of abuse including amphetamines, cocaine, opiates, and alcohol. In addition, the known factors that trigger and/or predispose to drug-induced neurotoxicity are discussed. These factors include drug-related, individual-related, and environmental insults. Moreover, we introduce some of the potential pharmacological antineurotoxic interventions deduced from experimental animal studies. These interventions involve various targets such as dopaminergic system, mitochondria, cell death signaling, and NMDA receptors, among others. We conclude the chapter with a discussion of addicted patients who might benefit from such interventions.
Collapse
Affiliation(s)
- S Mohammad Ahmadi Soleimani
- Neurocognitive Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran; Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hamed Ekhtiari
- Neurocognitive Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran; Translational Neuroscience Program, Institute for Cognitive Science Studies (ICSS), Tehran, Iran; Research Center for Molecular and Cellular Imaging (RCMCI), Tehran University of Medical Sciences, Tehran, Iran
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
20
|
Winter B, Brunecker P, Fiebach JB, Jungehulsing GJ, Kronenberg G, Endres M. Striatal Infarction Elicits Secondary Extrafocal MRI Changes in Ipsilateral Substantia Nigra. PLoS One 2015; 10:e0136483. [PMID: 26325192 PMCID: PMC4556671 DOI: 10.1371/journal.pone.0136483] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 12/24/2014] [Indexed: 02/05/2023] Open
Abstract
Focal ischemia may induce pathological alterations in brain areas distant from the primary lesion. In animal models, exofocal neuron death in the ipsilateral midbrain has been described after occlusion of the middle cerebral artery (MCA). Using sequential magnetic resonance imaging (T2- and diffusion-weighted) at 3 Tesla, we investigated acute ischemic stroke patients on days 1, 2, 6, 8, and 10 after stroke onset. Sixteen consecutive patients who had suffered a stroke involving the caudate nucleus and/or putamen of either hemisphere were recruited into the study. Four additional patients with strokes sparing the caudate nucleus and putamen but encompassing at least one-third of the MCA territory served as controls. Ischemic lesions involving striatal structures resulted in hyperintense lesions in ipsilateral midbrain that emerged between days 6 and 10 after stroke and were not present on the initial scans. In contrast, none of the control stroke patients developed secondary midbrain lesions. Hyperintense lesions in the pyramidal tract or the brain stem caused by degeneration of the corticospinal tract could be clearly distinguished from these secondary midbrain gray matter lesions and were detectable from day 2 after ischemia. Co-registration of high-resolution images with a digitized anatomic atlas revealed localization of secondary lesions primarily in the substantia nigra pars compacta. Apparent diffusion coefficient (ADC) values in the secondary lesions showed a delayed sharp decline through day 10. Normalization of ADC values was observed at late measurements. Taken together, our study demonstrates that striatal infarction elicits delayed degenerative changes in ipsilateral substantia nigra pars compacta.
Collapse
Affiliation(s)
- Benjamin Winter
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Peter Brunecker
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
| | - Jochen B. Fiebach
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
| | - Gerhard Jan Jungehulsing
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
- Department of Neurology, Jüdisches Krankenhaus Berlin, Heinz-Galinski-Strasse 1, 13347, Berlin, Germany
| | - Golo Kronenberg
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Psychiatry, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Max-Delbrück Center and Charité Medical Faculty, Experimental and Clinical Research Center, Lindenbergerweg 80, 13125, Berlin, Germany
| | - Matthias Endres
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Excellence Cluster Neurocure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ludwig-Erhard-Allee, 53175, Bonn, Germany
- German Centre for Cardiovascular Research (DZHK), Oudenarder Straße 16, 13347, Berlin, Germany
- * E-mail:
| |
Collapse
|
21
|
Shibagaki K, Okamoto K, Katsuta O, Nakamura M. Beneficial protective effect of pramipexole on light-induced retinal damage in mice. Exp Eye Res 2015. [PMID: 26213307 DOI: 10.1016/j.exer.2015.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We investigated the effects of pramipexole, a potent dopamine receptor D2/D3 agonist, on light-induced retinal damage in mice, H2O2-induced retinal pigment epithelium ARPE-19 cell injury in humans, and hydroxyl radical scavenging activity in a cell-free system. Pramipexole (0.1 and 1 mg/kg body weight) was orally administered to mice 1 h before light exposure (5000 lux, 2 h). Electrophysiological and morphologic studies were performed to evaluate the effects of the pramipexole on light-induced retinal damage in mice. Pramipexole significantly prevented the reduction of the a- and b-wave electroretinogram (ERG) amplitudes caused by light exposure in a dose-dependent manner. In parallel, damage to the inner and outer segments (IS/OS) of the photoreceptors, loss of photoreceptor nuclei, and the number of Tdt-mediated dUTP nick-end labeling (TUNEL)-positive cells in the outer nuclear layer (ONL) caused by light exposure were notably ameliorated by pramipexole. Additionally, pramipexole suppressed H2O2-induced ARPE-19 cell death in vitro in a concentration-dependent manner. The effect of pramipexole was significant at concentrations of 10(-6) M or higher. Pramipexole also significantly prevented H2O2-induced activation of caspases-3/7 and the intracellular accumulation of reactive oxygen species (ROS) in a concentration-dependent manner ranging from 10(-5) to 10(-3) M. Furthermore, pramipexole increased the scavenging activity toward a hydroxyl radical generated from H2O2 in a Fenton reaction. Our results suggest that pramipexole protects against light-induced retinal damage as an antioxidant and that it may be a novel and effective therapy for retinal degenerative disorders, such as dry age-related macular degeneration.
Collapse
Affiliation(s)
- Keiichi Shibagaki
- Research and Development Division, Santen Pharmaceutical Co., Ltd., 4-20, Ofuka-cho, Kita-ku, Osaka 530-8552, Japan; Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma-shi, Nara 630-0192, Japan.
| | - Kazuyoshi Okamoto
- Corporate Development Division, Santen Pharmaceutical Co., Ltd., 4-20, Ofuka-cho, Kita-ku, Osaka 530-8552, Japan
| | - Osamu Katsuta
- Research and Development Division, Santen Pharmaceutical Co., Ltd., 4-20, Ofuka-cho, Kita-ku, Osaka 530-8552, Japan
| | - Masatsugu Nakamura
- Research and Development Division, Santen Pharmaceutical Co., Ltd., 4-20, Ofuka-cho, Kita-ku, Osaka 530-8552, Japan
| |
Collapse
|
22
|
Van der Schyf CJ. Rational drug discovery design approaches for treating Parkinson’s disease. Expert Opin Drug Discov 2015; 10:713-41. [DOI: 10.1517/17460441.2015.1041495] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
23
|
Hellem TL, Lundberg KJ, Renshaw PF. A review of treatment options for co-occurring methamphetamine use disorders and depression. J Addict Nurs 2015; 26:14-23; quiz E1. [PMID: 25761159 PMCID: PMC5510330 DOI: 10.1097/jan.0000000000000058] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Co-occurring methamphetamine use and depression interferes with treatment outcomes. Female methamphetamine users are known to have higher rates of depression than male methamphetamine users, although this is also true for the general population. There are limited treatment options for the management of depression among methamphetamine users. In this integrative review, we summarize data on treatment strategies for co-occurring depression and methamphetamine use disorders. English-language articles were identified from PsychINFO, CINAHL, PubMed, and Medline as well as from reference lists of key articles. Search terms included "methamphetamine," "depression," and "treatment." Research articles describing psychological (n = 3), pharmacological (n = 6), nutritional supplement (n = 1), and psychological combined with pharmacological (n = 3) approaches for the treatment of methamphetamine use or withdrawal and/or depression are included in this review. Psychological and combination of psychological with pharmacological approaches have not been shown to be effective in treating these co-occurring conditions. Antidepressants have been determined to be ineffective and/or to introduce side effects. Gender differences with response to treatment were examined in only one of the published studies. There is a large gap in knowledge regarding treatment of co-occurring methamphetamine use disorders and depression. Considering that female methamphetamine users experience higher rates of depression than men, a focus on gender-specific treatment approaches is warranted.
Collapse
Affiliation(s)
- Tracy L Hellem
- Tracy L. Hellem, PhD, RN, College of Nursing and The Brain Institute, University of Utah, Salt Lake City. Kelly J. Lundberg, PhD, Department of Psychiatry, University of Utah, Salt Lake City. Perry F. Renshaw, MD, PhD, MBA, The Brain Institute and Department of Psychiatry, University of Utah, and VISN 19 MIRECC, Salt Lake City
| | | | | |
Collapse
|
24
|
Farias CCD, Bonifácio KL, Matsumoto AK, Higachi L, Casagrande R, Moreira EG, Barbosa DS. Comparison of the antioxidant potential of antiparkinsonian drugs in different in vitro models. BRAZ J PHARM SCI 2014. [DOI: 10.1590/s1984-82502014000400017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Parkinson's disease (PD) is characterized by progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta. Furthermore, oxidative stress plays a role in PD, causing or contributing to the neurodegenerative process. Currently PD has only symptomatic treatment and still nothing can be done to stop the degenerative process of the disease. This study aimed to comparatively evaluate the antioxidant capacity of pramipexole, selegeline and amantadine in different in vitrostudies and to offer possible explanations on the molecular antioxidant mechanisms of these drugs. In vitro, the antioxidant capacity of the drugs was assessed by the ability of antiparkinsonian drugs to decrease or scavenge ROS in the neutrophil respiratory burst, ability of antiparkinsonian drugs to donate hydrogen and stabilize the free radical 2,2-diphenyl-1-picryl-hydrazyl (DPPH•), to scavenge 2,2'-azino-di-(3-ethylbenzthiazoline-6-sulphonic acid (ABTS+) and evaluation of the ferric reducing antioxidant power (FRAP). This study demonstrated that both pramipexole and selegiline, but not amantadine, have antioxidant effects in vitro by scavenging superoxide anion on the respiratory burst, donating electron in the ABTS+ assay and presenting ferric reduction antioxidant power. This chemical structure-related antioxidant capacity suggests a possible neuroprotective mechanism of these drugs beyond their already recognized mechanism of action.
Collapse
Affiliation(s)
| | | | | | | | - Rúbia Casagrande
- State University of Londrina, Brazil; State University of Londrina, Brazil
| | | | | |
Collapse
|
25
|
Silindir M, Ozer AY. The benefits of pramipexole selection in the treatment of Parkinson's disease. Neurol Sci 2014; 35:1505-11. [PMID: 25038745 DOI: 10.1007/s10072-014-1891-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 07/14/2014] [Indexed: 12/21/2022]
Abstract
Levodopa administration as a gold standard in Parkinson's disease (PD) treatment is very valuable, however, long-term administration may cause some motor complications such as abnormal unintended movements and shortening response to each dose (wearing off phenomenon). Dopamine agonists were developed to reduce duration of immobile off periods and dependence to levodopa for improving motor impairments (Clarke et al., Cochrane Libr 1:1-23, 2000). Pramipexole is one of these nonergot dopamine agonists with high relative in vitro specificity and full intrinsic activity at D2 subfamily of dopamine receptors, with a higher binding affinity to D3 than to D4 or D2 receptor subtypes (Piercey, Clin Neuropharmacol 21:141-151, 1998). It can be advantageously administered as monotherapy or adjunctive therapy to levodopa to decrease side effects and increase effectiveness in both early and advanced PD treatment.
Collapse
Affiliation(s)
- Mine Silindir
- Department of Radiopharmacy, Faculty of Pharmacy, Hacettepe University, 06100, Ankara, Turkey,
| | | |
Collapse
|
26
|
Selective neuronal loss in ischemic stroke and cerebrovascular disease. J Cereb Blood Flow Metab 2014; 34:2-18. [PMID: 24192635 PMCID: PMC3887360 DOI: 10.1038/jcbfm.2013.188] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/15/2013] [Accepted: 10/17/2013] [Indexed: 01/23/2023]
Abstract
As a sequel of brain ischemia, selective neuronal loss (SNL)-as opposed to pannecrosis (i.e. infarction)-is attracting growing interest, particularly because it is now detectable in vivo. In acute stroke, SNL may affect the salvaged penumbra and hamper functional recovery following reperfusion. Rodent occlusion models can generate SNL predominantly in the striatum or cortex, showing that it can affect behavior for weeks despite normal magnetic resonance imaging. In humans, SNL in the salvaged penumbra has been documented in vivo mainly using positron emission tomography and (11)C-flumazenil, a neuronal tracer validated against immunohistochemistry in rodent stroke models. Cortical SNL has also been documented using this approach in chronic carotid disease in association with misery perfusion and behavioral deficits, suggesting that it can result from chronic or unstable hemodynamic compromise. Given these consequences, SNL may constitute a novel therapeutic target. Selective neuronal loss may also develop at sites remote from infarcts, representing secondary 'exofocal' phenomena akin to degeneration, potentially related to poststroke behavioral or mood impairments again amenable to therapy. Further work should aim to better characterize the time course, behavioral consequences-including the impact on neurological recovery and contribution to vascular cognitive impairment-association with possible causal processes such as microglial activation, and preventability of SNL.
Collapse
|
27
|
Asanuma M, Miyazaki I, Diaz-Corrales FJ, Shimizu M, Tanaka KI, Ogawa N. Pramipexole has ameliorating effects on levodopa-induced abnormal dopamine turnover in parkinsonian striatum and quenching effects on dopamine-semiquinone generatedin vitro. Neurol Res 2013; 27:533-9. [PMID: 15978181 DOI: 10.1179/016164105x22093] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES AND METHODS To clarify the effects of a non-ergot dopamine agonist pramipexole on levodopa-induced abnormal dopamine metabolism in the parkinsonian model, we examined striatal changes in dopamine and its metabolites after repeated administration of pramipexole and/or levodopa using 6-hydroxydopamine-lesioned hemi-parkinsonian mice. Moreover, the effects of pramipexole on dopamine-semiquinones were also accessed using an in vitro dopamine-semiquinone generating system to elucidate its neuroprotective property against dopamine quinone-induced neurotoxicity that appears as dopamine neuron-specific oxidative stress. RESULTS Combined administration of pramipexole (0.5 or 1 mg/kg/day, 7 days) selectively suppressed the levodopa-induced (50 mg/kg/day) increase of striatal dopamine turnover in the parkinsonian side, but not in the non-lesioned side. In addition to the antioxidant properties previously reported, it was clarified that pramipexole scavenged dopamine-semiquinones generated in a dose-dependent manner either in simultaneous incubation or post-incubation. DISCUSSION The neurotoxicity of dopamine quinones that appear as dopaminergic neuron-specific oxidative stress has recently been known to play a role in the pathogenesis of Parkinson's disease and neurotoxin-induced parkinsonism. Therefore, the present results revealed that pramipexole possesses neuroprotective effects against abnormal dopamine metabolism in excessively levodopa-administered parkinsonian brains and against cytotoxic dopamine quinones generated from excess dopamine, preventing consequently dopaminergic neuronal damage induced by excess dopamine or levodopa.
Collapse
Affiliation(s)
- Masato Asanuma
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | | | | | | | | | | |
Collapse
|
28
|
Corcia P, Gordon PH. Amyotrophic lateral sclerosis and the clinical potential of dexpramipexole. Ther Clin Risk Manag 2012; 8:359-66. [PMID: 22956874 PMCID: PMC3431958 DOI: 10.2147/tcrm.s21981] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder that leads to progressive weakness from loss of motor neurons and death on average in less than 3 years after symptom onset. No clear causes have been found and just one medication, riluzole, extends survival. Researchers have identified some of the cellular processes that occur after disease onset, including mitochondrial dysfunction, protein aggregation, oxidative stress, excitotoxicity, inflammation, and apoptosis. Mitochondrial disease may be a primary event in neurodegeneration or occur secondary to other cellular processes, and may itself contribute to oxidative stress, excitotoxicity, and apoptosis. Clinical trials currently aim to slow disease progression by testing drugs that impact one or more of these pathways. While every agent tested in the 18 years after the approval of riluzole has been ineffective, basic and clinical research methods in ALS have become dramatically more sophisticated. Dexpramipexole (RPPX), the R(+) enantiomer of pramiprexole, which is approved for symptomatic treatment of Parkinson disease, carries perhaps the currently largest body of pre-and early clinical data that support testing in ALS. The neuroprotective properties of RPPX in various models of neurodegeneration, including the ALS murine model, may be produced through protective actions on mitochondria. Early phase trials in human ALS suggest that the drug can be taken safely by patients in doses that provide neuroprotection in preclinical models. A Phase III trial to test the efficacy of RPPX in ALS is underway.
Collapse
Affiliation(s)
- Philippe Corcia
- Centre SLA, CHRU de Tours, Tours, France; UMR INSERM U930, Université François Rabelais de Tours (PC), Tours, France
| | | |
Collapse
|
29
|
Wang Q, Shin EJ, Nguyen XKT, Li Q, Bach JH, Bing G, Kim WK, Kim HC, Hong JS. Endogenous dynorphin protects against neurotoxin-elicited nigrostriatal dopaminergic neuron damage and motor deficits in mice. J Neuroinflammation 2012; 9:124. [PMID: 22695044 PMCID: PMC3409049 DOI: 10.1186/1742-2094-9-124] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 06/13/2012] [Indexed: 11/25/2022] Open
Abstract
Background The striato-nigral projecting pathway contains the highest concentrations of dynorphin in the brain. The functional role of this opioid peptide in the regulation of mesencephalic dopaminergic (DAergic) neurons is not clear. We reported previously that exogenous dynorphin exerts potent neuroprotective effects against inflammation-induced dopaminergic neurodegeneration in vitro. The present study was performed to investigate whether endogenous dynorphin has neuroprotective roles in vivo. Methods 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and methamphetamine (MA), two commonly used neurotoxins in rodent models of Parkinson’s disease, were administered to wild-type (Dyn+/+) and prodynorphin-deficient mice (Dyn−/−). We examined dopaminergic neurotoxicity by using an automated video tracking system, HPLC, immunocytochemistry, and reverse transcription and polymerase chain reaction (RT-PCR). Results Treatment with MPTP resulted in behavioral impairments in both strains. However, these impairments were more pronounced in Dyn-l- than in Dyn+/+. Dyn−/− showed more severe MPTP-induced dopaminergic neuronal loss in the substantia nigra and striatum than Dyn+/+. Similarly, the levels of dopamine and its metabolites in the striatum were depleted to a greater extent in Dyn−/− than in Dyn+/+. Additional mechanistic studies revealed that MPTP treatment caused a higher degree of microglial activation and M1 phenotype differentiation in Dyn−/− than in Dyn+/+. Consistent with these observations, prodynorphin deficiency also exacerbated neurotoxic effects induced by MA, although this effect was less pronounced than that of MPTP. Conclusions The in vivo results presented here extend our previous in vitro findings and further indicate that endogenous dynorphin plays a critical role in protecting dopaminergic neurons through its anti-inflammatory effects.
Collapse
Affiliation(s)
- Qingshan Wang
- Neuropsychopharmacology and Toxicology Program, College of PharmacyKangwon National University, Chunchon 200-701, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Zhang YF, Wang XY, Guo F, Burns K, Guo QY, Wang XM. Simultaneously changes in striatum dopaminergic and glutamatergic parameters following hypoxic-ischemic neuronal injury in newborn piglets. Eur J Paediatr Neurol 2012; 16:271-8. [PMID: 21723167 DOI: 10.1016/j.ejpn.2011.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 04/28/2011] [Accepted: 05/27/2011] [Indexed: 12/21/2022]
Abstract
Basal ganglia injury (BGI) is a type of perinatal hypoxic-ischemic (H-I) brain injury. Both malfunctions of glutamatergic and dopaminergic pathways in striatum were suggested to contribute to BGI. In current study, we investigated the imaging profile of glutamate (Glx) levels by proton magnetic resonance spectroscopy ((1)H-MRS), and the expression of dopamine D2 receptors (D2R) and dopamine transporter (DAT) by immunohistochemical staining in a newborn piglet model of H-I brain injury. We found that the number of striatal D2R positive neurons decreased following H-I brain injury, and the decrease in positive neuron number was consistent with the degree of striatum. Following H-I brain insult, the number of striatal DAT positive neurons and glutamate level were simultaneously increased initially, followed by a gradual decline toward control level. There was a positive correlation between the changes in striatal DAT positive neurons and glutamate level following H-I brain insults in newborn piglets. Our findings suggest that following H-I brain insult, striatal D2R positive neurons decreased due to neuron death; straital DAT initially increased to compensate for dopamine uptake; and glutamatergic and dopaminergic systems in striatum may act in an interdependent way in the striatum of newborn piglets.
Collapse
Affiliation(s)
- Yan-Fen Zhang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | | | | | | | | | | |
Collapse
|
31
|
van Bregt DR, Thomas TC, Hinzman JM, Cao T, Liu M, Bing G, Gerhardt GA, Pauly JR, Lifshitz J. Substantia nigra vulnerability after a single moderate diffuse brain injury in the rat. Exp Neurol 2012; 234:8-19. [PMID: 22178300 PMCID: PMC3294202 DOI: 10.1016/j.expneurol.2011.12.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 11/28/2011] [Accepted: 12/01/2011] [Indexed: 11/25/2022]
Abstract
Dementia and parkinsonism are late-onset symptoms associated with repetitive head injury, as documented in multiple contact-sport athletes. Clinical symptomatology is the likely phenotype of chronic degeneration and circuit disruption in the substantia nigra (SN). To investigate the initiating neuropathology, we hypothesize that a single diffuse brain injury is sufficient to initiate SN neuropathology including neuronal loss, vascular disruption and microglial activation, contributing to neurodegeneration and altered dopamine regulation. Adult, male Sprague-Dawley rats were subjected to sham or moderate midline fluid percussion brain injury. Stereological estimates indicated a significant 44% loss of the estimated total neuron number in the SN at 28-days post-injury, without atrophy of neuronal nuclear volumes, including 25% loss of tyrosine hydroxylase positive neurons by 28-days post-injury. Multi-focal vascular compromise occurred 1-2 days post-injury, with ensuing microglial activation (significant 40% increase at 4-days). Neurodegeneration (silver-stain technique) encompassed on average 21% of the SN by 7-days post-injury and increased to 29% by 28-days compared to sham (1%). Whole tissue SN, but not striatum, dopamine metabolism was altered at 28-days post-injury, without appreciable gene or protein changes in dopamine synthesis or regulation elements. Together, single moderate diffuse brain injury resulted in SN neurovascular pathology potentially associated with neuroinflammation or dopamine dysregulation. Compensatory mechanisms may preserve dopamine signaling acutely, but subsequent SN damage with aging or additional injury may expose clinical symptomatology of motor ataxias and dementia.
Collapse
Affiliation(s)
- Daniel R. van Bregt
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Biology and Biochemistry, University of Bath, Bath, UK
| | - Theresa Currier Thomas
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Jason M. Hinzman
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
- Morris K. Udall Parkinson's Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, USA
- Center for Microelectrode Technology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Tuoxin Cao
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Mei Liu
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Guoying Bing
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Greg A. Gerhardt
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
- Morris K. Udall Parkinson's Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, USA
- Center for Microelectrode Technology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - James R. Pauly
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Pharmaceutical Sciences University of Kentucky College of Pharmacy, Lexington, KY, USA
| | - Jonathan Lifshitz
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Physical Medicine & Rehabilitation, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
32
|
Delis F, Benveniste H, Xenos M, Grandy D, Wang GJ, Volkow ND, Thanos PK. Loss of dopamine D2 receptors induces atrophy in the temporal and parietal cortices and the caudal thalamus of ethanol-consuming mice. Alcohol Clin Exp Res 2011; 36:815-25. [PMID: 22017419 DOI: 10.1111/j.1530-0277.2011.01667.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND The need of an animal model of alcoholism becomes apparent when we consider the genetic diversity of the human populations, an example being dopamine D2 receptor (DRD2) expression levels. Research suggests that low DRD2 availability is associated with alcohol abuse, while higher DRD2 levels may be protective against alcoholism. This study aims to establish whether (i) the ethanol-consuming mouse is a suitable model of alcohol-induced brain atrophy and (ii) DRD2 protect the brain against alcohol toxicity. METHODS Adult Drd2+/+ and Drd2-/- mice drank either water or 20% ethanol solution for 6 months. At the end of the treatment period, the mice underwent magnetic resonance (MR) imaging under anesthesia. MR images were registered to a common space, and regions of interest were manually segmented. RESULTS We found that chronic ethanol intake induced a decrease in the volume of the temporal and parietal cortices as well as the caudal thalamus in Drd2-/- mice. CONCLUSIONS The result suggests that (i) normal DRD2 expression has a protective role against alcohol-induced brain atrophy and (ii) in the absence of Drd2 expression, prolonged ethanol intake reproduces a distinct feature of human brain pathology in alcoholism, the atrophy of the temporal and parietal cortices.
Collapse
Affiliation(s)
- Foteini Delis
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disease that results in increasing disability and that is uniformly fatal. Since its approval in the 1990s, riluzole remains the sole treatment for ALS offering modest survival benefit. While significant advances have been made in the symptomatic management of the disease, more effective drug therapy targeting disease progression is sorely needed. AREAS COVERED Advances in the understanding of pathogenic mechanisms involved in disease development and progression have provided multiple avenues for developing effective treatment strategies. This review highlights recent discoveries relating to these diverse mechanisms and their implications for the development of drug therapy. Previous human clinical trials that have targeted these pathways are mentioned and ongoing drug trials are discussed. EXPERT OPINION The search for effective drug therapy faces important challenges in the areas of basic science and animal research, translation of these results into human clinical trials, inherent bias in human studies and issues related to delays in clinical diagnosis. How these issues may be addressed and why ALS research constitutes fertile grounds for drug development not only for this devastating disease, but also for other more prevalent neurodegenerative diseases, is discussed in this review.
Collapse
Affiliation(s)
- Ali Aamer Habib
- The Neurological Institute of Columbia University, Eleanor and Lou Gehrig MDA/ALS Center, NY 10032, USA.
| | | |
Collapse
|
34
|
Elhak SG, Ghanem AA, Abdelghaffar H, Eldakroury S, Eltantawy D, Eldosouky S, Salama M. The role of pramipexole in a severe Parkinson's disease model in mice. Ther Adv Neurol Disord 2011; 3:333-7. [PMID: 21179594 DOI: 10.1177/1756285610389655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Pramipexole is one of a new generation of dopamine agonists. Recently there have been questions regarding its neuroprotective effects. These effects have been tested against various insults, which have yielded conflicting results. METHODS In this study, we introduced a combination of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/paraquat to induce a severe Parkinson's disease model. The mice, after receiving the combination of toxins, were evaluated using mortality rates and immunohistochemistry for degenerating tyrosine hydroxylase-positive neurons. RESULTS AND CONCLUSIONS Pramipexole was tested for its capacity to offer protection against neurotoxic the effects of MPTP/paraquat in this model; however, the results showed no improvement with pramipexole therapy.
Collapse
|
35
|
Youdim MBH. Why do we need multifunctional neuroprotective and neurorestorative drugs for Parkinson's and Alzheimer's disorders? Rambam Maimonides Med J 2010; 1:e0011. [PMID: 23908783 PMCID: PMC3678780 DOI: 10.5041/rmmj.10011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Parkinson's disease (PD) and Alzheimer's disease (AD) are severe neurodegenerative disorders, with no drugs that are currently approved to prevent the neuronal cell loss characteristic in brains of patients suffering from PD and AD, and all drug treatments are symptomatic and monomodal in their action. Due to the complex pathophysiology, including a cascade of neurotoxic molecular events that result in neuronal death and predisposition to depression and eventual dementia, and etiology of these disorders, an innovative approach towards neuroprotection or neurorestoration (neurorescue) is the development and use of multifunctional pharmaceuticals which can act at different brain regions and neurons. Such drugs target an array of pathological pathways, each of which is believed to contribute to the cascades that ultimately lead to neuronal cell death. In this short review, we discuss examples of novel multifunctional ligands that may have potential as neuroprotective-neurorestorative therapeutics in PD and AD, some of which are under development. The compounds discussed originate from synthetic chemistry as well as from natural sources.
Collapse
|
36
|
Youdim MBH. Why do we need multifunctional neuroprotective and neurorestorative drugs for Parkinson's and Alzheimer's diseases as disease modifying agents. Exp Neurobiol 2010; 19:1-14. [PMID: 22110336 PMCID: PMC3214798 DOI: 10.5607/en.2010.19.1.1] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 06/20/2010] [Indexed: 01/01/2023] Open
Abstract
Parkinson's disease (PD) and Alzheimer's Disease (AD) are severe neurodegenerative disorders, with no drugs that are currently approved to prevent the neuronal cell loss characteristic in brains of patients suffering from PD and AD and all drug treatment are synptomactic. Due to the complex pathophysiology, including a cascade of neurotoxic molecular events that results in neuronal death and predisposition to depression and eventual dementia and etiology of these disorders, an innovative approach towards neuroprotection or neurorestoration (neurorescue) may be the development and use of multifunctional pharmaceuticals. Such drugs target an array of pathological pathways, each of which is believed to contribute to the cascades that ultimately lead to neuronal cell death. In this short review, we discuss examples of novel multifunctional ligands that may have potential as neuroprotective-neurorestorative therapeutics in PD and AD. The compounds discussed originate from synthetic chemistry as well as from natural sources.
Collapse
Affiliation(s)
- Moussa B H Youdim
- Eve Topf and US National Parkinson Foundation Centers of Excellence for Neurodegenerative Diseases, Technion-Faculty of Medicine, Efron St., Haifa 31096, Israel
| |
Collapse
|
37
|
Thrash B, Thiruchelvan K, Ahuja M, Suppiramaniam V, Dhanasekaran M. Methamphetamine-induced neurotoxicity: the road to Parkinson's disease. Pharmacol Rep 2010; 61:966-77. [PMID: 20081231 DOI: 10.1016/s1734-1140(09)70158-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 10/29/2009] [Indexed: 01/22/2023]
Abstract
Studies have implicated methamphetamine exposure as a contributor to the development of Parkinson's disease. There is a significant degree of striatal dopamine depletion produced by methamphetamine, which makes the toxin useful in the creation of an animal model of Parkinson's disease. Parkinson's disease is a progressive neurodegenerative disorder associated with selective degeneration of nigrostriatal dopaminergic neurons. The immediate need is to understand the substances that increase the risk for this debilitating disorder as well as these substances'neurodegenerative mechanisms. Currently, various approaches are being taken to develop a novel and cost-effective anti-Parkinson's drug with minimal adverse effects and the added benefit of a neuroprotective effect to facilitate and improve the care of patients with Parkinson's disease. Amethamphetamine-treated animal model for Parkinson's disease can help to further the understanding of the neurodegenerative processes that target the nigrostriatal system. Studies on widely used drugs of abuse, which are also dopaminergic toxicants, may aid in understanding the etiology, pathophysiology and progression of the disease process and increase awareness of the risks involved in such drug abuse. In addition, this review evaluates the possible neuroprotective mechanisms of certain drugs against methamphetamine-induced toxicity.
Collapse
Affiliation(s)
- Bessy Thrash
- Department of Pharmacological Sciences, Harrison School of Pharmacy, Auburn University, 4306 Walker building, Auburn, AL 36849, USA
| | | | | | | | | |
Collapse
|
38
|
Neuroprotective effect of the antiparkinsonian drug pramipexole against nigrostriatal dopaminergic degeneration in rotenone-treated mice. Neurochem Int 2009; 55:760-7. [DOI: 10.1016/j.neuint.2009.07.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Revised: 07/23/2009] [Accepted: 07/23/2009] [Indexed: 12/21/2022]
|
39
|
Abstract
BACKGROUND The death of dopaminergic neurons in Parkinson's disease (PD) appears to have various causes, including oxidative stress, excitotoxicity, mitochondrial dysfunction (and associated apoptosis), ubiquitin/proteasomal dysfunction, and inflammation, any of which could in principle be the therapeutic target of a neuroprotective drug. The biology of dopaminergic neurons offers further potential targets, involving neurotrophic factors, dopamine-neuron genes, and even neurogenesis. OBJECTIVE To outline each hypothetical neuroprotective mechanism, the evidence suggesting its relevance to PD, and the research on pharmacologic intervention. METHODS A PubMed search was conducted to identify relevant preclinical and clinical literature published between 1989 and 2009. Additional articles were identified by reviewing the reference lists of papers selected in the original search. To circumscribe the survey and facilitate consideration of the conditions required for a neuroprotective effect, emphasis was placed on a single drug class, dopamine agonists, and in particular pramipexole. REVIEW OF THE FIELD: In a variety of in vitro and in vivo PD models, pramipexole exhibited preclinical evidence of neuroprotective actions of all hypothesized types, and in human neuroimaging studies it slowed the rate of loss of markers of dopaminergic function, consistent with drug-conferred neuroprotection in PD itself. Interpretation of the preclinical data was hampered by differences among models and by uncertainties concerning each model's mimicry of PD. Overall, the identified neuroprotection almost always required pretreatment (i.e., before insult) and high drug concentration. Interpretation of the clinical data was hampered by absence of placebo control and of a direct measure of neuroprotection. CONCLUSIONS Although the evidence is promising, neuroprotection in PD remains an elusive goal. In whatever form it emerges, neuroprotective therapy would be a strong argument against deferring PD treatment until symptoms are a significant life impediment, and thus would add urgency to early PD identification.
Collapse
|
40
|
Castri P, Busceti C, Battaglia G, Girardi F, Cavallari M, Orzi F, Fornai F. Protection by Apomorphine in Two Independent Models of Acute Inhibition of Oxidative Metabolism in Rodents. Clin Exp Hypertens 2009; 28:387-94. [PMID: 16833051 DOI: 10.1080/10641960600549827] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Apomorphine was administered by continuous infusion in the mouse following acute inhibition of oxidative metabolism induced by systemic administration of MPTP, and in the gerbil following transient occlusion of the carotid arteries. The dosage employed was comparable to the one used in the treatment of severe on-off fluctuations in Parkinson's disease. The results show that apomorphine significantly diminishes the striatal lesion caused by MPTP and the size of the infarct associated with the transient global ischemia. These data suggest that apomorphine is neuroprotective, probably by means of an antioxidant effect, at doses that are clinically used. The finding may be relevant to brain ischemia as well to chronic neurodegeneration.
Collapse
Affiliation(s)
- Paola Castri
- Department of Neurological Sciences, University of Roma, "La Sapienza", Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
41
|
Li Q, Wong JH, Lu G, Antonio GE, Yeung DK, Ng TB, Forster LE, Yew DT. Gene expression of synaptosomal-associated protein 25 (SNAP-25) in the prefrontal cortex of the spontaneously hypertensive rat (SHR). Biochim Biophys Acta Mol Basis Dis 2009; 1792:766-76. [PMID: 19482079 DOI: 10.1016/j.bbadis.2009.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Revised: 05/20/2009] [Accepted: 05/21/2009] [Indexed: 12/20/2022]
Abstract
Dopamine is believed to play an important role in the etiology of attention-deficit/hyperactivity disorder (ADHD). In our previous study, we showed that gene expression of dopamine D4 receptor decreased in the spontaneously hypertensive rat (SHR) in the prefrontal cortex (PFC). In the present study, we explored the potential causes of dysfunction in the dopamine system in ADHD. It is the first time that neuronal activities in both juvenile SHR and WKY rats have been measured by functional MRI (fMRI). Our results showed that in PFC the Blood Oxygenation Level Dependent (BOLD) signal response in SHR was much higher than WKY under stressful situations. We tested the effects of acute and repeated administration of amphetamine on behavioral changes in SHR combined with the expression of the neuronal activity marker, c-fos, in the PFC. Meanwhile dopamine-related gene expression was measured in the PFC after repeated administration of amphetamine. We found that potential neuronal damage occurred through deficit of D2-like receptor protective functions in the PFC of the SHR. We also measured the expression of synaptosomal-associated protein 25 (SNAP-25) in SHR in PFC. The results showed decreased expression of SNAP-25 mRNA in the PFC of SHR; this defect disappeared after repeated injection of D-AMP.
Collapse
Affiliation(s)
- Qi Li
- The Department of Psychiatry, Faculty of Medicine, The University of Hong Kong, 21 Sasson Road, Pokfulam, Hong Kong, China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Why should we use multifunctional neuroprotective and neurorestorative drugs for Parkinson's disease? Parkinsonism Relat Disord 2009; 13 Suppl 3:S281-91. [PMID: 18267251 DOI: 10.1016/s1353-8020(08)70017-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Parkinson's disease (PD) is a severe neurodegenerative disorder, with no available drugs able to prevent the neuronal cell loss characteristic in brains of patients suffering from PD. Due to the complex cascade of molecular events involved in the etiology of PD, an innovative approach towards neuroprotection or neurorescue may entail the use of multifunctional pharmaceuticals that target an array of pathological pathways, each of which is believed to contribute to events that ultimately lead to neuronal cell death. Here we discuss examples of novel multifunctional ligands that may have potential as neuroprotective and neurorestorative therapeutics in PD. The compounds discussed originate from synthetic chemistry as well as from natural sources where various moieties, identified in research to possess neuroprotective and neurorestorative properties, have been introduced into the structures of several monomodal drugs, some of which are used in the clinic.
Collapse
|
43
|
Hall ED, Traystman RJ. Role of animal studies in the design of clinical trials. FRONTIERS OF NEUROLOGY AND NEUROSCIENCE 2009; 25:10-33. [PMID: 19478492 DOI: 10.1159/000209470] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
44
|
Gribkoff VK, Bozik ME. KNS-760704 [(6R)-4,5,6,7-tetrahydro-N6-propyl-2, 6-benzothiazole-diamine dihydrochloride monohydrate] for the treatment of amyotrophic lateral sclerosis. CNS Neurosci Ther 2008; 14:215-26. [PMID: 18801114 DOI: 10.1111/j.1755-5949.2008.00048.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Developing effective treatments for chronic neurodegenerative disorders such as amyotrophic lateral sclerosis (ALS) has proven extremely difficult. ALS is universally fatal, characterized by progressive weakness due to the degeneration of upper and lower motor neurons, and leads eventually to respiratory failure which is the usual cause of death. Only a single treatment has been approved, the modestly effective nonspecific neuroprotectant Rilutek (riluzole; 2-amino-6-(trifluoromethoxy)benzothiazole). KNS-760704 [(6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine dihydrochloride, RPPX], a synthetic amino-benzothiazole with demonstrated activity in maintaining mitochondrial function, is being developed as a treatment for ALS. It has proven to be effective in multiple in vitro and in vivo assays of neuroprotection, including the G93A-SOD1 mutant mouse model; however, its specific mechanism of action remains unknown. The potential of KNS-760604 as a treatment for ALS was first suggested by studies showing that its optical enantiomer, Mirapex[(6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine; pramipexole dihydrochloride; PPX], a high-affinity agonist at dopamine D2, D3, and D4 receptors, exhibits important neuroprotective properties independent of its dopamine receptor agonism. In cell-based assays, both RPPX and PPX reduce the production of reactive oxygen species (ROS), attenuate the activation of apoptotic pathways, and increase cell survival in response to a variety of neurotoxins. However, PPX has limited utility as a clinical neuroprotective agent because the drug concentrations required for neuroprotection would likely produce unacceptable dopaminergic side effects. RPPX, on the other hand, while possessing the same neuroprotective potential as PPX, is a much lower-affinity dopamine receptor agonist and may therefore be more useful in the treatment of ALS. This review will examine the data supporting the hypothesis that the RPPX may have therapeutic potential for the treatment of neurodegenerative disorders including ALS. In addition, we will briefly review recent preclinical data in support of RPPX, and discuss the current status of its clinical development.
Collapse
|
45
|
Scheller D, Ullmer C, Berkels R, Gwarek M, Lübbert H. The in vitro receptor profile of rotigotine: a new agent for the treatment of Parkinson’s disease. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2008; 379:73-86. [DOI: 10.1007/s00210-008-0341-4] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Accepted: 07/16/2008] [Indexed: 11/24/2022]
|
46
|
Iravani MM, Sadeghian M, Leung CCM, Tel BC, Rose S, Schapira AH, Jenner P. Continuous subcutaneous infusion of pramipexole protects against lipopolysaccharide-induced dopaminergic cell death without affecting the inflammatory response. Exp Neurol 2008; 212:522-31. [PMID: 18571649 DOI: 10.1016/j.expneurol.2008.04.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Revised: 04/02/2008] [Accepted: 04/30/2008] [Indexed: 12/27/2022]
Abstract
The D2/D3 dopamine receptor agonist pramipexole, protects against toxin-induced dopaminergic neuronal destruction but its mechanism of action is unknown. Inflammation following glial cell activation contributes to cell death in Parkinson's disease and we now report on the effects of acute or chronic administration of pramipexole on lipopolysaccharide (LPS) induced inflammation and nigral dopaminergic cell death in the rat. At 48 h and 30 days following supranigral administration of LPS, approximately 70% of tyrosine hydroxylase (TH) immunoreactive (-ir) cells in substantia nigra had degenerated with a corresponding loss of TH-ir terminals in the striatum. In rats acutely treated with pramipexole (2x1 mg/kg; s.c.) 48 h following LPS application, there was no difference in the number of TH-ir cells or terminals compared to LPS-treated rats receiving vehicle. However, the continuous subcutaneous infusion of pramipexole for 7 days prior to LPS and 21 days subsequently, produced a marked preservation of both TH-ir cells and terminals. At 48 h or 30 days, LPS induced an up-regulation of ubiquitin-ir within the nigral TH-ir neurones, which was reduced by pramipexole treatment. Thirty days following supranigral LPS administration (9 days after the end of infusion), (+)-amphetamine (5 mg/kg, i.p.) caused robust ipsiversive rotation. In rats treated with LPS but receiving continuous subcutaneous administration of pramipexole, (+)-amphetamine-induced rotation was markedly reduced. LPS-induced increase in the levels of inflammatory markers, were not affected by either acute administration or continuous infusion of pramipexole. Continuous infusion of pramipexole protected dopaminergic neurones against inflammation induced degeneration but without modification of the inflammatory response.
Collapse
Affiliation(s)
- Mahmoud M Iravani
- Neurodegenerative Disease Research Centre, School of Health and Biomedical Sciences, King's College, London SE1 1UL, UK
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Pramipexole is a non-ergot dopamine agonist shown to be efficacious in the treatment of Parkinson's disease (PD). This review addresses the literature concerning pramipexole's efficacy in treating motor and non-motor symptoms in PD, its impact on the development of dyskinesias and response fluctuations, the issue of neuroprotection, and the risk for developing adverse events such as increased somnolence, attacks of sudden onset of sleep, cardiac valvulopathy and impulse control disturbances.
Collapse
Affiliation(s)
- Radu Constantinescu
- Department of Neurology, Sahlgrenska University Hospital 413 45 Göteborg, Sweden.
| |
Collapse
|
48
|
Izumi Y, Yamamoto N, Kume T, Katsuki H, Sawada H, Akaike A. Regulation of intracellular dopamine levels by dopaminergic drugs: Involvement of vesicular monoamine transporter. Eur J Pharmacol 2008; 582:52-61. [DOI: 10.1016/j.ejphar.2007.12.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Revised: 11/28/2007] [Accepted: 12/16/2007] [Indexed: 11/30/2022]
|
49
|
Ferrante M, Blackwell KT, Migliore M, Ascoli GA. Computational models of neuronal biophysics and the characterization of potential neuropharmacological targets. Curr Med Chem 2008; 15:2456-71. [PMID: 18855673 PMCID: PMC3560392 DOI: 10.2174/092986708785909094] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The identification and characterization of potential pharmacological targets in neurology and psychiatry is a fundamental problem at the intersection between medicinal chemistry and the neurosciences. Exciting new techniques in proteomics and genomics have fostered rapid progress, opening numerous questions as to the functional consequences of ligand binding at the systems level. Psycho- and neuro-active drugs typically work in nerve cells by affecting one or more aspects of electrophysiological activity. Thus, an integrated understanding of neuropharmacological agents requires bridging the gap between their molecular mechanisms and the biophysical determinants of neuronal function. Computational neuroscience and bioinformatics can play a major role in this functional connection. Robust quantitative models exist describing all major active membrane properties under endogenous and exogenous chemical control. These include voltage-dependent ionic channels (sodium, potassium, calcium, etc.), synaptic receptor channels (e.g. glutamatergic, GABAergic, cholinergic), and G protein coupled signaling pathways (protein kinases, phosphatases, and other enzymatic cascades). This brief review of neuromolecular medicine from the computational perspective provides compelling examples of how simulations can elucidate, explain, and predict the effect of chemical agonists, antagonists, and modulators in the nervous system.
Collapse
Affiliation(s)
| | - Kim T. Blackwell
- Krasnow Institute for Advanced Study, George Mason University
- Department of Molecular Neuroscience, George Mason University, Fairfax, Virginia
| | - Michele Migliore
- Institute of Biophysics, National Research Council, Palermo, Italy
| | - Giorgio A. Ascoli
- Krasnow Institute for Advanced Study, George Mason University
- Department of Molecular Neuroscience, George Mason University, Fairfax, Virginia
| |
Collapse
|
50
|
Caffeine activates the PI3K/Akt pathway and prevents apoptotic cell death in a Parkinson's disease model of SH-SY5Y cells. Neurosci Lett 2007; 432:146-50. [PMID: 18201823 DOI: 10.1016/j.neulet.2007.12.034] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Revised: 11/22/2007] [Accepted: 12/10/2007] [Indexed: 11/24/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. Recent epidemiological studies suggest that caffeine, one of the major components of coffee, has a protective effect against developing PD. However, the detailed mechanisms of how caffeine suppresses neuronal death have not been fully elucidated. We investigated the cytoprotective mechanisms of caffeine using human dopaminergic neuroblastoma SH-SY5Y cells as a PD model. Caffeine prevented the apoptotic cell death induced by serum/retinoic acid (RA) deprivation, MPP+, rotenone, and 6-OHDA in SH-SY5Y cells in a dose dependent manner. Caffeine lowered caspase-3 activity induced by serum/RA deprivation and 6-OHDA administration, and also decreased the number of apoptotic condensed and/or fragmented nuclei. Akt was phosphorylated 60 min after caffeine administration in a dose dependent manner; PI3K inhibitors, wortmannin and LY294002 canceled this cytoprotective effect of caffeine. On the other hand, MAPKs such as Erk1/2, p38, or JNK were not activated by caffeine. These results suggest that caffeine has a cytoprotective effect due to the activation of the PI3K/Akt pathways in SH-SY5Y cells.
Collapse
|