1
|
Larochelle J, Howell JA, Yang C, Liu L, Gunraj RE, Stansbury SM, de Oliveira ACP, Baksh S, Candelario-Jalil E. Pharmacological inhibition of receptor-interacting protein kinase 2 (RIPK2) elicits neuroprotective effects following experimental ischemic stroke. Exp Neurol 2024; 377:114812. [PMID: 38729551 DOI: 10.1016/j.expneurol.2024.114812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/19/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
Ischemic stroke induces a debilitating neurological insult, where inflammatory processes contribute greatly to the expansion and growth of the injury. Receptor-interacting protein kinase 2 (RIPK2) is most well-known for its role as the obligate kinase for NOD1/2 pattern recognition receptor signaling and is implicated in the pathology of various inflammatory conditions. Compared to a sham-operated control, ischemic stroke resulted in a dramatic increase in the active, phosphorylated form of RIPK2, indicating that RIPK2 may be implicated in the response to stroke injury. Here, we assessed the effects of pharmacological inhibition of RIPK2 to improve post-stroke outcomes in mice subjected to experimental ischemic stroke. We found that treatment at the onset of reperfusion with a RIPK2 inhibitor, which inhibits the phosphorylation and activation of RIPK2, resulted in marked improvements in post-stroke behavioral outcomes compared to the vehicle-administered group assessed 24 h after stroke. RIPK2 inhibitor-treated mice exhibited dramatic reductions in infarct volume, concurrent with reduced damage to the blood-brain barrier, as evidenced by reduced levels of active matrix metalloproteinase-9 (MMP-9) and leakage of blood-borne albumin in the ipsilateral cortex. To explore the protective mechanism of RIPK2 inhibition, we next pretreated mice with RIPK2 inhibitor or vehicle and examined transcriptomic alterations occurring in the ischemic brain 6 h after stroke. We observed a dramatic reduction in neuroinflammatory markers in the ipsilateral cortex of the inhibitor-treated group while also attaining a comprehensive view of the vast transcriptomic alterations occurring in the brain with inhibitor treatment through bulk RNA-sequencing of the injured cortex. Overall, we provide significant novel evidence that RIPK2 may represent a viable target for post-stroke pharmacotherapy and potentially other neuroinflammatory conditions.
Collapse
Affiliation(s)
- Jonathan Larochelle
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - John Aaron Howell
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Lei Liu
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Rachel E Gunraj
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Sofia M Stansbury
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | | | - Shairaz Baksh
- BioImmuno Designs, Inc., Edmonton, Alberta, Canada; Bio-Stream Diagnostics, Inc., Edmonton, Alberta, Canada
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
2
|
Abu-Elfotuh K, Hamdan AME, Mohamed SA, Bakr RO, Ahmed AH, Atwa AM, Hamdan AM, Alanzai AG, Alnahhas RK, Gowifel AMH, Salem MA. The potential anti-Alzheimer's activity of Oxalis corniculata Linn. Methanolic extract in experimental rats: Role of APOE4/LRP1, TLR4/NF-κβ/NLRP3, Wnt 3/β-catenin/GSK-3β, autophagy and apoptotic cues. JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117731. [PMID: 38218505 DOI: 10.1016/j.jep.2024.117731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/23/2023] [Accepted: 01/06/2024] [Indexed: 01/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Oxalis corniculata (O. corniculata) is a member of Oxalidaceae family, widely distributed in Asia, Europe, America, and Africa, used extensively as food and its traditional folkloric uses include management of epilepsy, gastric disorders, and neurodegenerative diseases, together with its use in enhancing health. Numerous pharmacological benefits of O. corniculata are linked to its anti-inflammatory and antioxidant abilities. One of the most prevalent neurodegenerative disorders is Alzheimer's disease (AD) in which neuroinflammation and oxidative stress are its main pathogenic processes. AIM OF THE STUDY Our research aimed to study the neuroprotective effect of the methanolic extract of Oxalis corniculata Linn. (O. corniculata ME), compared to selenium (Se) against AlCl3-induced AD. MATERIALS AND METHODS Forty male albino rats were allocated into four groups (Gps). Gp I a control group, the rest of the animals received AlCl3 (Gp II-Gp IV). Rats in Gp III and IV were treated with Se and O. corniculata ME, respectively. RESULTS The chemical profile of O. corniculata ME was studied using ultraperformance liquid chromatography-electrospray ionization-quadrupole time-of-flight mass spectrometry, allowing the tentative identification of sixty-six compounds, including organic acids, phenolics and others, cinnamic acid and its derivatives, fatty acids, and flavonoids. AlCl3 showed deterioration in short-term memory and brain histological pictures. Our findings showed that O. corniculata ME and selenium helped to combat oxidative stress produced by accumulation of AlCl3 in the brain and in prophylaxis against AD. Thus, Selenium (Se) and O. corniculata ME restored antioxidant defense, via enhancing Nrf2/HO-1 hub, hampered neuroinflammation, via TLR4/NF-κβ/NLRP3, along with dampening apoptosis, Aβ generation, tau hyperphosphorylation, BACE1, ApoE4 and LRP1 levels. Treatments also promoted autophagy and modulated Wnt 3/β-catenin/GSK3β cue. CONCLUSIONS It was noted that O. corniculata ME showed a notable ameliorative effect compared to Se on Nrf2/HO-1, TLR4/NF-κβ/NLRP3, APOE4/LRP1, Wnt 3/β-catenin/GSK-3β and PERK axes.
Collapse
Affiliation(s)
- Karema Abu-Elfotuh
- Clinical Pharmacy Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt; Al-Ayen Iraqi University, Thi-Qar, 64001, Iraq.
| | - Ahmed M E Hamdan
- Pharmacy Practice Department, Faculty of Pharmacy, University of Tabuk, Tabuk 74191, Saudi Arabia.
| | - Shaza A Mohamed
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt.
| | - Riham O Bakr
- Department of Pharmacognosy, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA) University, Giza 11787, Egypt.
| | - Amal H Ahmed
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt.
| | - Ahmed M Atwa
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo-Suez Road, Cairo 11829, Egypt.
| | - Amira M Hamdan
- Oceanography Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt.
| | | | | | - Ayah M H Gowifel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 11571, Egypt.
| | - Maha A Salem
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 11571, Egypt.
| |
Collapse
|
3
|
Buchanan AM, Mena S, Choukari I, Vasa A, Crawford JN, Fadel J, Maxwell N, Reagan L, Cruikshank A, Best J, Nijhout HF, Reed M, Hashemi P. Serotonin as a biomarker of toxin-induced Parkinsonism. Mol Med 2024; 30:33. [PMID: 38429661 PMCID: PMC10908133 DOI: 10.1186/s10020-023-00773-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 12/18/2023] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Loss of dopaminergic neurons underlies the motor symptoms of Parkinson's disease (PD). However stereotypical PD symptoms only manifest after approximately 80% of dopamine neurons have died making dopamine-related motor phenotypes unreliable markers of the earlier stages of the disease. There are other non-motor symptoms, such as depression, that may present decades before motor symptoms. METHODS Because serotonin is implicated in depression, here we use niche, fast electrochemistry paired with mathematical modelling and machine learning to, for the first time, robustly evaluate serotonin neurochemistry in vivo in real time in a toxicological model of Parkinsonism, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). RESULTS Mice treated with acute MPTP had lower concentrations of in vivo, evoked and ambient serotonin in the hippocampus, consistent with the clinical comorbidity of depression with PD. These mice did not chemically respond to SSRI, as strongly as control animals did, following the clinical literature showing that antidepressant success during PD is highly variable. Following L-DOPA administration, using a novel machine learning analysis tool, we observed a dynamic shift from evoked serotonin release in the hippocampus to dopamine release. We hypothesize that this finding shows, in real time, that serotonergic neurons uptake L-DOPA and produce dopamine at the expense of serotonin, supporting the significant clinical correlation between L-DOPA and depression. Finally, we found that this post L-DOPA dopamine release was less regulated, staying in the synapse for longer. This finding is perhaps due to lack of autoreceptor control and may provide a ground from which to study L-DOPA induced dyskinesia. CONCLUSIONS These results validate key prior hypotheses about the roles of serotonin during PD and open an avenue to study to potentially improve therapeutics for levodopa-induced dyskinesia and depression.
Collapse
Affiliation(s)
- Anna Marie Buchanan
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, USA
- Department of Pharmacology, Physiology, & Neuroscience, University of South Carolina SOM, Columbia, SC, 29209, USA
| | - Sergio Mena
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Iman Choukari
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Aditya Vasa
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, USA
| | - Jesseca N Crawford
- Department of Pharmacology, Physiology, & Neuroscience, University of South Carolina SOM, Columbia, SC, 29209, USA
| | - Jim Fadel
- Department of Pharmacology, Physiology, & Neuroscience, University of South Carolina SOM, Columbia, SC, 29209, USA
| | - Nick Maxwell
- Department of Pharmacology, Physiology, & Neuroscience, University of South Carolina SOM, Columbia, SC, 29209, USA
| | - Lawrence Reagan
- Department of Pharmacology, Physiology, & Neuroscience, University of South Carolina SOM, Columbia, SC, 29209, USA
- Columbia VA Health Care System, Columbia, SC, 29208, USA
| | | | - Janet Best
- Department of Mathematics, The Ohio State University, Columbus, OH, USA
| | | | - Michael Reed
- Department of Mathematics, Duke University, Durham, NC, USA
| | - Parastoo Hashemi
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, USA.
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
4
|
Larochelle J, Tishko RJ, Yang C, Ge Y, Phan LT, Gunraj RE, Stansbury SM, Liu L, Mohamadzadeh M, Khoshbouei H, Candelario-Jalil E. Receptor-interacting protein kinase 2 (RIPK2) profoundly contributes to post-stroke neuroinflammation and behavioral deficits with microglia as unique perpetrators. J Neuroinflammation 2023; 20:221. [PMID: 37777791 PMCID: PMC10543871 DOI: 10.1186/s12974-023-02907-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023] Open
Abstract
BACKGROUND Receptor-interacting protein kinase 2 (RIPK2) is a serine/threonine kinase whose activity propagates inflammatory signaling through its association with pattern recognition receptors (PRRs) and subsequent TAK1, NF-κB, and MAPK pathway activation. After stroke, dead and dying cells release a host of damage-associated molecular patterns (DAMPs) that activate PRRs and initiate a robust inflammatory response. We hypothesize that RIPK2 plays a damaging role in the progression of stroke injury by enhancing the neuroinflammatory response to stroke and that global genetic deletion or microglia-specific conditional deletion of Ripk2 will be protective following ischemic stroke. METHODS Adult (3-6 months) male mice were subjected to 45 min of transient middle cerebral artery occlusion (tMCAO) followed by 24 h, 48 h, or 28 days of reperfusion. Aged male and female mice (18-24 months) were subjected to permanent ischemic stroke and sacrificed 48 h later. Infarct volumes were calculated using TTC staining (24-48 h) or Cresyl violet staining (28d). Sensorimotor tests (weight grip, vertical grid, and open field) were performed at indicated timepoints. Blood-brain barrier (BBB) damage, tight junction proteins, matrix metalloproteinase-9 (MMP-9), and neuroinflammatory markers were assessed via immunoblotting, ELISA, immunohistochemistry, and RT-qPCR. Differential gene expression profiles were generated through bulk RNA sequencing and nanoString®. RESULTS Global genetic deletion of Ripk2 resulted in decreased infarct sizes and reduced neuroinflammatory markers 24 h after stroke compared to wild-type controls. Ripk2 global deletion also improved both acute and long-term behavioral outcomes with powerful effects on reducing infarct volume and mortality at 28d post-stroke. Conditional deletion of microglial Ripk2 (mKO) partially recapitulated our results in global Ripk2 deficient mice, showing reductive effects on infarct volume and improved behavioral outcomes within 48 h of injury. Finally, bulk transcriptomic profiling and nanoString data demonstrated that Ripk2 deficiency in microglia decreases genes associated with MAPK and NF-κB signaling, dampening the neuroinflammatory response after stroke injury by reducing immune cell activation and peripheral immune cell invasion. CONCLUSIONS These results reveal a hitherto unknown role for RIPK2 in the pathogenesis of ischemic stroke injury, with microglia playing a distinct role. This study identifies RIPK2 as a potent propagator of neuroinflammatory signaling, highlighting its potential as a therapeutic target for post-stroke intervention.
Collapse
Affiliation(s)
- Jonathan Larochelle
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Ryland J Tishko
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Yong Ge
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health, San Antonio, TX, USA
| | - Leah T Phan
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Rachel E Gunraj
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Sofia M Stansbury
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Lei Liu
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Mansour Mohamadzadeh
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health, San Antonio, TX, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA.
| |
Collapse
|
5
|
Schmit KJ, Garcia P, Sciortino A, Aho VTE, Pardo Rodriguez B, Thomas MH, Gérardy JJ, Bastero Acha I, Halder R, Cialini C, Heurtaux T, Ostahi I, Busi SB, Grandmougin L, Lowndes T, Singh Y, Martens EC, Mittelbronn M, Buttini M, Wilmes P. Fiber deprivation and microbiome-borne curli shift gut bacterial populations and accelerate disease in a mouse model of Parkinson's disease. Cell Rep 2023; 42:113071. [PMID: 37676767 PMCID: PMC10548091 DOI: 10.1016/j.celrep.2023.113071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 07/01/2023] [Accepted: 08/16/2023] [Indexed: 09/09/2023] Open
Abstract
Parkinson's disease (PD) is a neurological disorder characterized by motor dysfunction, dopaminergic neuron loss, and alpha-synuclein (αSyn) inclusions. Many PD risk factors are known, but those affecting disease progression are not. Lifestyle and microbial dysbiosis are candidates in this context. Diet-driven gut dysbiosis and reduced barrier function may increase exposure of enteric neurons to toxins. Here, we study whether fiber deprivation and exposure to bacterial curli, a protein cross-seeding with αSyn, individually or together, exacerbate disease in the enteric and central nervous systems of a transgenic PD mouse model. We analyze the gut microbiome, motor behavior, and gastrointestinal and brain pathologies. We find that diet and bacterial curli alter the microbiome and exacerbate motor performance, as well as intestinal and brain pathologies, but to different extents. Our results shed important insights on how diet and microbiome-borne insults modulate PD progression via the gut-brain axis and have implications for lifestyle management of PD.
Collapse
Affiliation(s)
- Kristopher J Schmit
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Institute for Medical Genetics and Applied Genomics, Hospital University Tubingen, 72076 Tubingen, Germany; Luxembourg Center of Neuropathology, 3555 Dudelange, Luxembourg.
| | - Pierre Garcia
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Luxembourg Center of Neuropathology, 3555 Dudelange, Luxembourg
| | - Alessia Sciortino
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Luxembourg Center of Neuropathology, 3555 Dudelange, Luxembourg
| | - Velma T E Aho
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Beatriz Pardo Rodriguez
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Luxembourg Center of Neuropathology, 3555 Dudelange, Luxembourg
| | - Mélanie H Thomas
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Luxembourg Center of Neuropathology, 3555 Dudelange, Luxembourg
| | - Jean-Jacques Gérardy
- Luxembourg Center of Neuropathology, 3555 Dudelange, Luxembourg; National Center of Pathology, Laboratoire National de Santé, 3555 Dudelange, Luxembourg
| | - Irati Bastero Acha
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Luxembourg Center of Neuropathology, 3555 Dudelange, Luxembourg
| | - Rashi Halder
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Camille Cialini
- Luxembourg Center of Neuropathology, 3555 Dudelange, Luxembourg; Department of Cancer Research, Luxembourg Institute of Health, 1526 Luxembourg, Luxembourg
| | - Tony Heurtaux
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Luxembourg Center of Neuropathology, 3555 Dudelange, Luxembourg; Department of Life Sciences and Medicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Irina Ostahi
- National Center of Pathology, Laboratoire National de Santé, 3555 Dudelange, Luxembourg
| | - Susheel B Busi
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Léa Grandmougin
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Tuesday Lowndes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Yogesh Singh
- Institute for Medical Genetics and Applied Genomics, Hospital University Tubingen, 72076 Tubingen, Germany
| | - Eric C Martens
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Michel Mittelbronn
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Luxembourg Center of Neuropathology, 3555 Dudelange, Luxembourg; National Center of Pathology, Laboratoire National de Santé, 3555 Dudelange, Luxembourg; Department of Cancer Research, Luxembourg Institute of Health, 1526 Luxembourg, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
| | - Manuel Buttini
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Luxembourg Center of Neuropathology, 3555 Dudelange, Luxembourg
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
6
|
Li M, Zhang J, Jiang L, Wang W, Feng X, Liu M, Yang D. Neuroprotective effects of morroniside from Cornus officinalis sieb. Et zucc against Parkinson's disease via inhibiting oxidative stress and ferroptosis. BMC Complement Med Ther 2023; 23:218. [PMID: 37393274 PMCID: PMC10314491 DOI: 10.1186/s12906-023-03967-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 04/19/2023] [Indexed: 07/03/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenera-tive disorder after Alzheimer disease accompanied by the death of dopaminergic neurons and brain nigrostriatal mitochondrial damage in the elderly population. The features of the disease include tremor, rigidity, postural instability, and motor retardation. The pathogenesis of Parkinson's disease is complex, and abnormal lipid metabolism resulting in ferroptosis due to the excessive accumulation of free radicals from oxidative stress in the substantia nigra of the brain was thought to be one of the factors causing the disease. Morroniside has been reported to have significant neuroprotective effects, although it has not been studied in PD. Therefore, this study focused on determining the neuroprotective effects of morroniside (25, 50, and 100 mg/kg) on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP, 30 mg/kg)-induced mice models of PD and explored 1-methyl-4-phenylpyridinium MPP+-induced ferroptosis in PC12 cells. Morroniside restored impaired motor function in the PD mice models while reducing neuronal injury. The activation of nuclear factor erythroid 2-related factor 2/antioxidant response elements (Nrf2/ARE) by morroniside promoted antioxidation, the content of reducing agent glutathione (GSH) increased, and the level of the lipid metabolite malondialdehyde (MDA) decreased. Notably, morroniside inhibited ferroptosis in substantia nigra of the brain and PC12 cells, reduced iron levels, and upregulated the expression of the iron-regulated proteins glutathione peroxidase 4 (GPX4), solute carrier family 7 member 11 (SLC7A11), ferritin heavy chain 1 (FTH-1), and ferroportin (FPN). More importantly, morroniside repaired the mitochondrial damage, restored the mitochondrial respiratory chain, and inhibited the production of reactive oxygen species (ROS). These data indicated that morroniside could activate the Nrf2/ARE signaling pathway to increase the antioxidant capacity, thereby inhibiting abnormal lipid metabolism and protecting dopaminergic neurons from ferroptosis in PD.
Collapse
Affiliation(s)
- Mao Li
- The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Junli Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lianyan Jiang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wujun Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Xianrong Feng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Meijun Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Dongdong Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
7
|
Simons E, Fleming SM. Role of rodent models in advancing precision medicine for Parkinson's disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:3-16. [PMID: 36803818 DOI: 10.1016/b978-0-323-85555-6.00002-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
With a current lack of disease-modifying treatments, an initiative toward implementing a precision medicine approach for treating Parkinson's disease (PD) has emerged. However, challenges remain in how to define and apply precision medicine in PD. To accomplish the goal of optimally targeted and timed treatment for each patient, preclinical research in a diverse population of rodent models will continue to be an essential part of the translational path to identify novel biomarkers for patient diagnosis and subgrouping, understand PD disease mechanisms, identify new therapeutic targets, and screen therapeutics prior to clinical testing. This review highlights the most common rodent models of PD and discusses how these models can contribute to defining and implementing precision medicine for the treatment of PD.
Collapse
Affiliation(s)
- Emily Simons
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Sheila M Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States.
| |
Collapse
|
8
|
Santoro M, Fadda P, Klephan KJ, Hull C, Teismann P, Platt B, Riedel G. Neurochemical, histological, and behavioral profiling of the acute, sub-acute, and chronic MPTP mouse model of Parkinson's disease. J Neurochem 2023; 164:121-142. [PMID: 36184945 PMCID: PMC10098710 DOI: 10.1111/jnc.15699] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/25/2022] [Accepted: 09/30/2022] [Indexed: 02/04/2023]
Abstract
Parkinson's disease (PD) is a heterogeneous multi-systemic disorder unique to humans characterized by motor and non-motor symptoms. Preclinical experimental models of PD present limitations and inconsistent neurochemical, histological, and behavioral readouts. The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD is the most common in vivo screening platform for novel drug therapies; nonetheless, behavioral endpoints yielded amongst laboratories are often discordant and inconclusive. In this study, we characterized neurochemically, histologically, and behaviorally three different MPTP mouse models of PD to identify translational traits reminiscent of PD symptomatology. MPTP was intraperitoneally (i.p.) administered in three different regimens: (i) acute-four injections of 20 mg/kg of MPTP every 2 h; (ii) sub-acute-one daily injection of 30 mg/kg of MPTP for 5 consecutive days; and (iii) chronic-one daily injection of 4 mg/kg of MPTP for 28 consecutive days. A series of behavioral tests were conducted to assess motor and non-motor behavioral changes including anxiety, endurance, gait, motor deficits, cognitive impairment, circadian rhythm and food consumption. Impairments in balance and gait were confirmed in the chronic and acute models, respectively, with the latter showing significant correlation with lesion size. The sub-acute model, by contrast, presented with generalized hyperactivity. Both, motor and non-motor changes were identified in the acute and sub-acute regime where habituation to a novel environment was significantly reduced. Moreover, we report increased water and food intake across all three models. Overall, the acute model displayed the most severe lesion size, while across the three models striatal dopamine content (DA) did not correlate with the behavioral performance. The present study demonstrates that detection of behavioral changes following MPTP exposure is challenging and does not correlate with the dopaminergic lesion extent.
Collapse
Affiliation(s)
- Matteo Santoro
- Institute of Medical SciencesUniversity of AberdeenAberdeenUK
- Present address:
Department of Neurosurgery, School of MedicineStanford UniversityPalo AltoCaliforniaUSA
| | - Paola Fadda
- Department of NeuroscienceUniversity of CagliariCagliariItaly
| | - Katie J. Klephan
- Newcastle UniversitySchool of Biomedical, Nutritional, and Sport SciencesNewcastle upon TyneUK
- Present address:
AccuRXLondonLondonUK
| | - Claire Hull
- Institute of Medical SciencesUniversity of AberdeenAberdeenUK
| | - Peter Teismann
- Institute of Medical SciencesUniversity of AberdeenAberdeenUK
| | - Bettina Platt
- Institute of Medical SciencesUniversity of AberdeenAberdeenUK
| | - Gernot Riedel
- Institute of Medical SciencesUniversity of AberdeenAberdeenUK
| |
Collapse
|
9
|
Aniszewska A, Bergström J, Ingelsson M, Ekmark-Lewén S. Modeling Parkinson's disease-related symptoms in alpha-synuclein overexpressing mice. Brain Behav 2022; 12:e2628. [PMID: 35652155 PMCID: PMC9304846 DOI: 10.1002/brb3.2628] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/08/2022] [Accepted: 04/15/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Intracellular deposition of alpha-synuclein (α-syn) as Lewy bodies and Lewy neurites is a central event in the pathogenesis of Parkinson's disease (PD) and other α-synucleinopathies. Transgenic mouse models overexpressing human α-syn, are useful research tools in preclinical studies of pathogenetic mechanisms. Such mice develop α-syn inclusions as well as neurodegeneration with a topographical distribution that varies depending on the choice of promoter and which form of α-syn that is overexpressed. Moreover, they display motor symptoms and cognitive disturbances that to some extent resemble the human conditions. PURPOSE One of the main motives for assessing behavior in these mouse models is to evaluate the potential of new treatment strategies, including their impact on motor and cognitive symptoms. However, due to a high within-group variability with respect to such features, the behavioral studies need to be applied with caution. In this review, we discuss how to make appropriate choices in the experimental design and which tests that are most suitable for the evaluation of PD-related symptoms in such studies. METHODS We have evaluated published results on two selected transgenic mouse models overexpressing wild type (L61) and mutated (A30P) α-syn in the context of their validity and utility for different types of behavioral studies. CONCLUSIONS By applying appropriate behavioral tests, α-syn transgenic mouse models provide an appropriate experimental platform for studies of symptoms related to PD and other α-synucleinopathies.
Collapse
Affiliation(s)
- Agata Aniszewska
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Joakim Bergström
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, Sweden.,Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada.,Department of Medicine and Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Ontario, Canada
| | - Sara Ekmark-Lewén
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| |
Collapse
|
10
|
Targeted BRD4 protein degradation by dBET1 ameliorates acute ischemic brain injury and improves functional outcomes associated with reduced neuroinflammation and oxidative stress and preservation of blood-brain barrier integrity. J Neuroinflammation 2022; 19:168. [PMID: 35761277 PMCID: PMC9237998 DOI: 10.1186/s12974-022-02533-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/16/2022] [Indexed: 11/22/2022] Open
Abstract
Bromodomain-containing protein 4 (BRD4), a member of the bromodomain and extra-terminal domain (BET) protein family, plays a crucial role in regulating inflammation and oxidative stress that are tightly related to stroke development and progression. Consequently, BRD4 blockade has attracted increasing interest for associated neurological diseases, including stroke. dBET1 is a novel and effective BRD4 degrader through the proteolysis-targeting chimera (PROTAC) strategy. We hypothesized that dBET1 protects against brain damage and neurological deficits in a transient focal ischemic stroke mouse model by reducing inflammation and oxidative stress and preserving the blood–brain barrier (BBB) integrity. Post-ischemic dBET1 treatment starting 4 h after stroke onset significantly ameliorated severe neurological deficits and reduced infarct volume 48 h after stroke. dBET1 markedly reduced inflammation and oxidative stress after stroke, indicated by multiple pro-inflammatory cytokines and chemokines including IL-1β, IL-6, TNF-α, CCL2, CXCL1 and CXCL10, and oxidative damage markers 4-hydroxynonenal (4-HNE) and gp91phox and antioxidative proteins SOD2 and GPx1. Meanwhile, stroke-induced BBB disruption, increased MMP-9 levels, neutrophil infiltration, and increased ICAM-1 were significantly attenuated by dBET1 treatment. Post-ischemic dBET1 administration also attenuated ischemia-induced reactive gliosis in microglia and astrocytes. Overall, these findings demonstrate that BRD4 degradation by dBET1 improves acute stroke outcomes, which is associated with reduced neuroinflammation and oxidative stress and preservation of BBB integrity. This study identifies a novel role of BET proteins in the mechanisms resulting in ischemic brain damage, which can be leveraged to develop novel therapies.
Collapse
|
11
|
Troshev D, Voronkov D, Pavlova A, Abaimov D, Latanov A, Fedorova T, Berezhnoy D. Time Course of Neurobehavioral Disruptions and Regional Brain Metabolism Changes in the Rotenone Mice Model of Parkinson’s Disease. Biomedicines 2022; 10:biomedicines10020466. [PMID: 35203675 PMCID: PMC8962442 DOI: 10.3390/biomedicines10020466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 12/10/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by slow progression with a long prodromal stage and the gradual evolution of both neuropsychological symptoms and subtle motor changes, preceding motor dysfunction. Thus, in order for animal models of PD to be valid, they should reproduce these characteristics of the disease. One of such models, in which neuropathology is induced by chronic injections of low doses of mitochondrial toxin rotenone, is well established in rats. However, data on this model adapted to mice remain controversial. We have designed the study to describe the timecourse of motor and non-motor symptoms during chronic subcutaneous administration of rotenone (4 mg/kg daily for 35 days) in C57BL/6 mice. We characterize the underlying neuropathological processes (dopaminergic neuron degeneration, regional brain metabolism, monoamine neurotransmitter and lipid peroxidation changes) at different timepoints: 1 day, 2 weeks and 5 weeks of daily rotenone exposure. Based on the behavioral data, we can describe three stages of pathology: cognitive changes from week 2 of rotenone exposure, subtle motor changes in week 3–4 and motor dysfunction starting roughly from week 4. Neuropathological changes in this model include a general decrease in COX activity in different areas of the brain (acute effect of rotenone) and a more specific decrease in midbrain (chronic effect), followed by significant neurodegeneration in SNpc but not VTA by the 5th week of rotenone exposure. However, we were unable to find changes in the level of monoamine neurotransmitters neither in the striatum nor in the cortex, nor in the level of lipid peroxidation in the brainstem. Thus, the gradual progression of pathology in this model is linked with metabolic changes, rather than with oxidative stress or tonic neurotransmitter release levels. Overall, this study supports the idea that a low-dose rotenone mouse model can also reproduce different stages of PD as well as rats.
Collapse
Affiliation(s)
- Dmitry Troshev
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Vavilov Street, 26, 119334 Moscow, Russia;
| | - Dmitry Voronkov
- Research Center of Neurology, Laboratory of Clinical and Experimental Neurochemistry, Volokolamskoeshosse, 80, 125367 Moscow, Russia; (D.V.); (D.A.); (T.F.)
| | - Anastasia Pavlova
- Biological Faculty, Moscow State University, Leninskie Gory, 1s12, 119234 Moscow, Russia; (A.P.); (A.L.)
| | - Denis Abaimov
- Research Center of Neurology, Laboratory of Clinical and Experimental Neurochemistry, Volokolamskoeshosse, 80, 125367 Moscow, Russia; (D.V.); (D.A.); (T.F.)
| | - Alexander Latanov
- Biological Faculty, Moscow State University, Leninskie Gory, 1s12, 119234 Moscow, Russia; (A.P.); (A.L.)
| | - Tatiana Fedorova
- Research Center of Neurology, Laboratory of Clinical and Experimental Neurochemistry, Volokolamskoeshosse, 80, 125367 Moscow, Russia; (D.V.); (D.A.); (T.F.)
| | - Daniil Berezhnoy
- Research Center of Neurology, Laboratory of Clinical and Experimental Neurochemistry, Volokolamskoeshosse, 80, 125367 Moscow, Russia; (D.V.); (D.A.); (T.F.)
- Biological Faculty, Moscow State University, Leninskie Gory, 1s12, 119234 Moscow, Russia; (A.P.); (A.L.)
- Correspondence:
| |
Collapse
|
12
|
Sangarapillai K, Norman BM, Almeida QJ. Boxing vs Sensory Exercise for Parkinson's Disease: A Double-Blinded Randomized Controlled Trial. Neurorehabil Neural Repair 2021; 35:769-777. [PMID: 34121511 PMCID: PMC8414806 DOI: 10.1177/15459683211023197] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background. Exercise is increasingly becoming recognized as an important adjunct to medications in the clinical management of Parkinson's disease (PD). Boxing and sensory exercise have shown immediate benefits, but whether they continue beyond program completion is unknown. This study aimed to investigate the effects of boxing and sensory training on motor symptoms of PD, and whether these benefits remain upon completion of the intervention. Methods. In this 20-week double-blinded randomized controlled trial, 40 participants with idiopathic PD were randomized into 2 treatment groups, (n = 20) boxing or (n = 20) sensory exercise. Participants completed 10 weeks of intervention. Motor symptoms were assessed at (week 0, 10, and 20) using the Unified Parkinson's Disease Rating Scale (UPDRS-III). Data were analyzed using SPSS, and repeated-measures ANOVA was conducted. Results. A significant interaction effect between groups and time were observed F(1, 39) = 4.566, P = .036, where the sensory group improved in comparison to the boxing group. Post hoc analysis revealed that in comparison to boxing, the effects of exercise did not wear off at washout (week 20) P < .006. Conclusion. Future rehabilitation research should incorporate similar measures to explore whether effects of exercise wear off post intervention.
Collapse
Affiliation(s)
- Kishoree Sangarapillai
- Movement Disorders Research and
Rehabilitation Centre, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Benjamin M. Norman
- Movement Disorders Research and
Rehabilitation Centre, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Quincy J. Almeida
- Movement Disorders Research and
Rehabilitation Centre, Wilfrid Laurier University, Waterloo, ON, Canada
| |
Collapse
|
13
|
Koppula S, Alluri R, Kopalli SR. Coriandrum sativum attenuates microglia mediated neuroinflammation and MPTP-induced behavioral and oxidative changes in Parkinson's disease mouse model. EXCLI JOURNAL 2021; 20:835-850. [PMID: 34177406 PMCID: PMC8222636 DOI: 10.17179/excli2021-3668] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022]
Abstract
Coriandrum sativum Linn. (family: Umbelliferae; C. sativum), is a potential herb widely used as a spice and traditional medicine. In the present work, the effects of C. sativum fruit extract (CSE), against lipopolysaccharide (LPS)-stimulated BV-2 microglia-mediated neuroinflammation in vitro and 1-methyl-4 phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced Parkinson's disease (PD) animal model in vivo was investigated. LPS-stimulated increase in nitric oxide (NO), inducible NO synthase, cyclooxygenase-2, interleukin-6 and tumor necrosis factor-alpha were significantly (p < 0.05 ~ p < 0.001) inhibited by CSE (25, 50 and 100 μg/mL) in BV-2 microglial cells. Further, CSE inhibited the LPS-induced nuclear factor of kappa-beta activation and IκB-α phosphorylation in BV-2 microglia. In vivo studies, CSE (100, 200 and 300 mg/kg) ameliorated the MPTP (25 mg/kg, i.p.)-induced changes in locomotor, cognitive and behavior functions evaluated by rotarod, passive avoidance and open field test significantly (p < 0.05 ~ p < 0.001). The MPTP-induced changes in brain oxidative enzyme levels such as superoxide dismutase, catalase, and lipid peroxidation were significantly (p < 0.01 and p < 0.001 at 200 and 300 mg/kg, respectively) restored with CSE treatment. High-performance thin-layer chromatography fingerprinting analysis of CSE exhibited several distinctive peaks with quercetin and kaempferol-3O-glucoside as identifiable compounds. In conclusion, our study indicated that CSE attenuated neuroinflammatory processes in LPS-stimulated microglia in vitro and restored the MPTP-induced behavioral deficits and brain oxidative enzyme status in vivo proving its therapeutic potential in the treatment of neuroinflammatory and oxidative stress-mediated neurodegeneration seen in PD.
Collapse
Affiliation(s)
- Sushruta Koppula
- College of Biomedical and Health Science, Konkuk University, Chungju-Si, Chungcheongbuk Do, 380-701, Republic of Korea
| | - Ramesh Alluri
- Department of Pharmacy, Vishnu Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| |
Collapse
|
14
|
Liu W, Lao W, Zhang R, Zhu H. Altered expression of voltage gated sodium channel Nav1.1 is involved in motor ability in MPTP-treated mice. Brain Res Bull 2021; 170:187-198. [PMID: 33610724 DOI: 10.1016/j.brainresbull.2021.02.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 01/12/2021] [Accepted: 02/16/2021] [Indexed: 11/25/2022]
Abstract
Parkinson's disease (PD) is a motor disabling disorder owing to the progressive degeneration of dopaminergic neurons in the substantia nigra (SN). The mechanisms causing motor deficits remain debated. High synchronized oscillations in the basal ganglia (BG) were proposed to be associated with motor symptoms in PD patients and animal models of PD. Voltage-gated sodium channels play a vital role in the initiation and propagation of action potentials. Here, we investigated the expression patterns of a VGSC subtype Nav1.1 in the BG of a PD animal model induced by MPTP intraperitoneal injection. The results showed that Nav1.1 was significantly reduced in tyrosine hydroxylase (TH) positive dopaminergic neurons of the SN. Moreover, Nav1.1 expression was significantly increased in calcium binding protein parvalbumin (PV) positive neurons of the globus pallidus (GP) in MPTP-treated mice compared to the rarely undetectable expression of Nav1.1 in the control GP. Furthermore, the administration of phenytoin, a VGSCs blocker, can effectively improve motor disabilities and reduce the synchronous oscillations in the BG of MPTP-treated mice. These findings suggested that the alterations of Nav1.1 expression may be associated with the high synchronous oscillations in the BG of PD animals.
Collapse
Affiliation(s)
- Weitang Liu
- School of Life Science, Shanghai University, Shanghai, China
| | - Wenwen Lao
- School of Life Science, Shanghai University, Shanghai, China
| | - Renxing Zhang
- School of Life Science, Shanghai University, Shanghai, China
| | - Hongyan Zhu
- School of Life Science, Shanghai University, Shanghai, China.
| |
Collapse
|
15
|
Motor Coordination Disorders Evaluated through the Grid Test and Changes in the Nigral Nrf2 mRNA Expression in Rats with Pedunculopontine Lesion. Behav Sci (Basel) 2020; 10:bs10100156. [PMID: 33066049 PMCID: PMC7600924 DOI: 10.3390/bs10100156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/22/2020] [Accepted: 09/30/2020] [Indexed: 11/16/2022] Open
Abstract
Neurotoxic lesion of the pedunculopontine nucleus (PPN) is known to cause subtle motor dysfunctions. However, motor coordination during advance on a discontinuous and elevated surface has not been studied. It is also not known whether there are changes in the mRNA expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) in nigral tissue. Methods: The effects of the unilateral neurotoxic lesion of the PPN in motor coordination evaluated through grid test and Nrf2 mRNA expression in nigral tissue were evaluated. Two experimental designs (ED) were organized: ED#1 behavioral study (7 and 30 days after PPN lesion) and ED#2 molecular biology study (24 h, 48 h and 7 days) after PPN lesion. Results: ED#1—The number of faults made with left limbs, were significant higher in the lesioned groups (p < 0.01) both 7 and 30 days post-lesion. The number of failures made by the right limbs, was also significantly higher (p < 0.05) vs. control groups. ED#2—Nrf2 mRNA expression showed an increase 24 h after PPN injury (p < 0.01), followed by a peak of expression 48 h post injury (p < 0.001). Conclusions: Disorders of motor coordination associated with PPN injury are bilateral. The increased Nrf2 mRNA expression could represent an adaptive response to oxidative stress in the nigral tissue following pontine injury.
Collapse
|
16
|
Siddu A, David LS, Lauinger N, Chen X, Saint-Pierre M, Alpaugh M, Durcan T, Cicchetti F. Beneficial effects of cysteamine in Thy1-α-Syn mice and induced pluripotent stem cells with a SNCA gene triplication. Neurobiol Dis 2020; 145:105042. [PMID: 32798729 DOI: 10.1016/j.nbd.2020.105042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 06/29/2020] [Accepted: 08/08/2020] [Indexed: 12/15/2022] Open
Abstract
A number of publications have reported that cysteamine has significant therapeutic effects on several aspects of Parkinson's disease (PD)-related pathology but none of these studies have evaluated its impact on pathological forms of α-Synuclein (α-Syn), one of the main hallmarks of PD. We therefore tested the efficacy of cysteamine on the Thy1-α-Syn mouse model which over-expresses full-length human wild-type α-Syn. Two-month (early stage disease) and 6-month old (late stage disease) mice and littermate controls were treated daily with cysteamine (20 mg/kg, i.p.) to assess the protective and restorative properties of this compound. After 6 weeks of treatment, animals were tested using a battery of motor tests. Cysteamine-treated transgenic mice displayed significant improvements in motor performance as compared to saline-treated transgenic littermates. Post-mortem readouts revealed a reduction in fibrillation, phosphorylation and total levels of overexpresed human α-Syn. To determine if such outcomes extended to human cells, the benefits of cysteamine were additionally tested using 6-hydroxydopamine (6-OHDA) treated neurons differentiated from induced pluripotent stem cells (iPSCs) derived from a PD patient harbouring a triplication of the SNCA gene. SNCA neurons treated with cysteamine exhibited significantly more intact/healthy neurites than cells treated with 6-OHDA alone. Additionally, SNCA neurons treated with cysteamine in the absence of 6-OHDA showed a trend towards lower total α-Syn levels. Overall, our in vivo and in vitro findings suggest that cysteamine can act as a disease-modifying molecule by enhancing -the survival of dopaminergic neurons and reducing pathological forms of α-Syn.
Collapse
Affiliation(s)
- Alberto Siddu
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705 Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, 1050, avenue de la Médecine, Québec, QC G1V 0A6, Canada
| | - Linda Suzanne David
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705 Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, 1050, avenue de la Médecine, Québec, QC G1V 0A6, Canada
| | - Nadine Lauinger
- Department of Neurology and Neurosurgery, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Xiuqing Chen
- Department of Neurology and Neurosurgery, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; Montreal Neurological Institute and Hospital, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Martine Saint-Pierre
- Département de Psychiatrie & Neurosciences, Université Laval, 1050, avenue de la Médecine, Québec, QC G1V 0A6, Canada
| | - Melanie Alpaugh
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705 Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, 1050, avenue de la Médecine, Québec, QC G1V 0A6, Canada
| | - Thomas Durcan
- Department of Neurology and Neurosurgery, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; Montreal Neurological Institute and Hospital, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Francesca Cicchetti
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705 Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, 1050, avenue de la Médecine, Québec, QC G1V 0A6, Canada.
| |
Collapse
|
17
|
Gezer AO, Kochmanski J, VanOeveren SE, Cole-Strauss A, Kemp CJ, Patterson JR, Miller KM, Kuhn NC, Herman DE, McIntire A, Lipton JW, Luk KC, Fleming SM, Sortwell CE, Bernstein AI. Developmental exposure to the organochlorine pesticide dieldrin causes male-specific exacerbation of α-synuclein-preformed fibril-induced toxicity and motor deficits. Neurobiol Dis 2020; 141:104947. [PMID: 32422283 PMCID: PMC7343230 DOI: 10.1016/j.nbd.2020.104947] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/22/2020] [Accepted: 05/13/2020] [Indexed: 12/21/2022] Open
Abstract
Human and animal studies have shown that exposure to the organochlorine pesticide dieldrin is associated with increased risk of Parkinson's disease (PD). Previous work showed that developmental dieldrin exposure increased neuronal susceptibility to MPTP toxicity in male C57BL/6 mice, possibly via changes in dopamine (DA) packaging and turnover. However, the relevance of the MPTP model to PD pathophysiology has been questioned. We therefore studied dieldrin-induced neurotoxicity in the α-synuclein (α-syn)-preformed fibril (PFF) model, which better reflects the α-syn pathology and toxicity observed in PD pathogenesis. Specifically, we used a "two-hit" model to determine whether developmental dieldrin exposure increases susceptibility to α-syn PFF-induced synucleinopathy. Dams were fed either dieldrin (0.3 mg/kg, every 3-4 days) or vehicle corn oil starting 1 month prior to breeding and continuing through weaning of pups at postnatal day 22. At 12 weeks of age, male and female offspring received intrastriatal α-syn PFF or control saline injections. Consistent with the male-specific increased susceptibility to MPTP, our results demonstrate that developmental dieldrin exposure exacerbates PFF-induced toxicity in male mice only. Specifically, in male offspring, dieldrin exacerbated PFF-induced motor deficits on the challenging beam and increased DA turnover in the striatum 6 months after PFF injection. However, male offspring showed neither exacerbation of phosphorylated α-syn aggregation (pSyn) in the substantia nigra (SN) at 1 or 2 months post-PFF injection, nor exacerbation of PFF-induced TH and NeuN loss in the SN 6 months post-PFF injection. Collectively, these data indicate that developmental dieldrin exposure produces a male-specific exacerbation of synucleinopathy-induced behavioral and biochemical deficits. This sex-specific result is consistent with both previous work in the MPTP model, our previously reported sex-specific effects of this exposure paradigm on the male and female epigenome, and the higher prevalence and more severe course of PD in males. The novel two-hit environmental toxicant/PFF exposure paradigm established in this project can be used to explore the mechanisms by which other PD-related exposures alter neuronal vulnerability to synucleinopathy in sporadic PD.
Collapse
Affiliation(s)
- Aysegul O Gezer
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America; Cell and Molecular Biology Graduate Program, College of Natural Sciences, Michigan State University, East Lansing, MI, United States of America; College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States of America
| | - Joseph Kochmanski
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Sarah E VanOeveren
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Allyson Cole-Strauss
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Christopher J Kemp
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Joseph R Patterson
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Kathryn M Miller
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Nathan C Kuhn
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Danielle E Herman
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, United States of America
| | - Alyssa McIntire
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, United States of America
| | - Jack W Lipton
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America; Mercy Health St. Mary's, Grand Rapids, MI, United States of America
| | - Kelvin C Luk
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Sheila M Fleming
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, United States of America
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America; Mercy Health St. Mary's, Grand Rapids, MI, United States of America
| | - Alison I Bernstein
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America; Mercy Health St. Mary's, Grand Rapids, MI, United States of America.
| |
Collapse
|
18
|
Jiang S, Berger S, Hu Y, Bartsch D, Tian Y. Alterations of the Motor and Olfactory Functions Related to Parkinson's Disease in Transgenic Mice With a VMAT2-Deficiency in Dopaminergic Neurons. Front Neurosci 2020; 14:356. [PMID: 32410942 PMCID: PMC7198702 DOI: 10.3389/fnins.2020.00356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/24/2020] [Indexed: 12/26/2022] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases, with approximately six million people affected worldwide. Vesicular monoamine transporter 2 (VMAT2) dysfunction has recently become a hot topic in the pathophysiology of PD, and the advent of transgenic mice has also accelerated the development of behavioral studies in animal models. However, there are only a few systematic behavioral tests that embrace abundant motor and non-motor performance in a unique mutant mouse model which correspond to the varied symptoms observed in human PD. The aim of this study is to evaluate the responsibility of the unique reduction of dopamine in the varied motor and non-motor symptoms of PD via a transgenic mice model. We analyzed neurotransmitter concentrations in the brain tissue of 18-month-old mutant mice, with selective inactivation of one allele of Vmat2 in dopaminergic neurons (VMAT2DATcre-HET) to confirm the selective reduction of dopamine, and then examined behavioral functions. Neurochemical tests showed lower dopamine concentrations in specific brain regions of VMAT2DATcre-HET mice, especially the ventral tegmental area/substantia nigra and striatum, together with relatively unchanging concentrations of norepinephrine and serotonin, demonstrating the dopaminergic specificity of this mouse model. Behavioral tasks showed impairments in several motor functions and major defects in olfactory abilities in the VMAT2DATcre-HET mice. However, no significant changes were found in the majority of non-motor tests, such as emotional performance and sleep patterns. We concluded from this study that the selective inactivation of one allele of the Vmat2 gene in dopaminergic neurons was related to dopamine reduction, resulting in phenotypes resembling some of the major deficits in PD, especially those of motor symptoms and olfactory functions.
Collapse
Affiliation(s)
- Song Jiang
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Molecular Biology, Central Institute of Mental Health, Heidelberg University Faculty of Medicine in Mannheim, Mannheim, Germany
| | - Stefan Berger
- Department of Molecular Biology, Central Institute of Mental Health, Heidelberg University Faculty of Medicine in Mannheim, Mannheim, Germany
| | - Yajuan Hu
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Anhui Medical University, Hefei, China
| | - Dusan Bartsch
- Department of Molecular Biology, Central Institute of Mental Health, Heidelberg University Faculty of Medicine in Mannheim, Mannheim, Germany
| | - Yanghua Tian
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Anhui Medical University, Hefei, China.,Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| |
Collapse
|
19
|
Cromolyn sodium delays disease onset and is neuroprotective in the SOD1 G93A Mouse Model of amyotrophic lateral sclerosis. Sci Rep 2019; 9:17728. [PMID: 31776380 PMCID: PMC6881366 DOI: 10.1038/s41598-019-53982-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 11/05/2019] [Indexed: 12/12/2022] Open
Abstract
Accumulating evidence suggests that neuroinflammatory processes are implicated in the initiation and progression of amyotrophic lateral sclerosis (ALS). Previous reports have demonstrated an increase in microgliosis and astrogliosis in the lumbar spinal cord of SOD1G93A transgenic mice before the onset of symptoms, a neuroinflammatory response which correlated with disease progression. Importantly, early stage homeostatic microglia enhanced motor neuron survival, while pro-inflammatory microglia were toxic to motor neurons in the SOD1G93A mice. Recent studies from our group have demonstrated that cromolyn sodium, an FDA approved compound, exerts neuroprotective effects in mouse models of Alzheimer's disease by altering microglial cell activation. Here, we tested the neuroprotective and anti-inflammatory effects of cromolyn sodium in the SOD1G93A mouse model of ALS. Our results indicate that cromolyn sodium treatment significantly delayed the onset of neurological symptoms, and improved deficits in PaGE performance in both male and female mice, however, there was only an effect on survival in female mice. Furthermore, there was a significant increase in motor neuron survival in the lumbar spinal cord as well as a significant decrease in the denervation of the neuromuscular junction of the tibialis anterior muscle in cromolyn treated transgenic SOD1G93A mice. Lastly, cromolyn treatment decreased the expression of pro-inflammatory cytokines/chemokines in the lumbar spinal cord and plasma and decreased mast cell degranulation in the tibialis anterior muscle of transgenic SOD1G93A mice. Together, these findings suggest that cromolyn sodium provides neuroprotection in the SOD1G93A mice by decreasing the inflammatory response.
Collapse
|
20
|
Ablation of RIP3 protects from dopaminergic neurodegeneration in experimental Parkinson's disease. Cell Death Dis 2019; 10:840. [PMID: 31690718 PMCID: PMC6831575 DOI: 10.1038/s41419-019-2078-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/18/2019] [Indexed: 12/14/2022]
Abstract
Parkinson’s disease (PD) is driven by dopaminergic neurodegeneration in the substantia nigra pars compacta (SN) and striatum. Although apoptosis is considered the main neurodegenerative mechanism, other cell death pathways may be involved. In this regard, necroptosis is a regulated form of cell death dependent on receptor interacting protein 3 (RIP3), a protein also implicated in apoptosis and inflammation independently of its pro-necroptotic activity. Here, we explored the role of RIP3 genetic deletion in in vivo and in vitro PD models. Firstly, wild-type (Wt) and RIP3 knockout (RIP3ko) mice were injected intraperitoneally with MPTP (40 mg/kg, i.p.), and sacrificed after either 6 or 30 days. RIP3ko protected from dopaminergic neurodegeneration in the SN of MPTP-injected mice, but this effect was independent of necroptosis. In keeping with this, necrostatin-1s (10 mg/kg/day, i.p.) did not afford full neuroprotection. Moreover, MPTP led to DNA fragmentation, caspase-3 activation, lipid peroxidation and BAX expression in Wt mice, in the absence of caspase-8 cleavage, suggesting intrinsic apoptosis. This was mimicked in primary cortical neuronal cultures exposed to the active MPTP metabolite. RIP3 deficiency in cultured cells and in mouse brain abrogated all phenotypes. Curiously, astrogliosis was increased in the striatum of MPTP-injected Wt mice and further exacerbated in RIP3ko mice. This was accompanied by absence of microgliosis and reposition of glial cell line-derived neurotrophic factor (GDNF) levels in the striata of MPTP-injected RIP3ko mice when compared to MPTP-injected Wt mice, which in turn showed a massive GDNF decrease. RIP3ko primary mixed glial cultures also presented decreased expression of inflammation-related genes upon inflammatory stimulation. These findings hint at possible undescribed non-necroptotic roles for RIP3 in inflammation and MPTP-driven cell death, which can contribute to PD progression.
Collapse
|
21
|
Palasz E, Niewiadomski W, Gasiorowska A, Wysocka A, Stepniewska A, Niewiadomska G. Exercise-Induced Neuroprotection and Recovery of Motor Function in Animal Models of Parkinson's Disease. Front Neurol 2019; 10:1143. [PMID: 31736859 PMCID: PMC6838750 DOI: 10.3389/fneur.2019.01143] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/11/2019] [Indexed: 12/19/2022] Open
Abstract
Parkinson's disease (PD) is manifested by progressive motor, autonomic, and cognitive disturbances. Dopamine (DA) synthesizing neurons in the substantia nigra (SN) degenerate, causing a decline in DA level in the striatum that leads to the characteristic movement disorders. A disease-modifying therapy to arrest PD progression remains unattainable with current pharmacotherapies, most of which cause severe side effects and lose their efficacy with time. For this reason, there is a need to seek new therapies supporting the pharmacological treatment of PD. Motor therapy is recommended for pharmacologically treated PD patients as it alleviates the symptoms. Molecular mechanisms behind the beneficial effects of motor therapy are unknown, nor is it known whether such therapy may be neuroprotective in PD patients. Due to obvious limitations, human studies are unlikely to answer these questions; therefore, the use of animal models of PD seems indispensable. Motor therapy in animal models of PD characterized by the loss of dopaminergic neurons has neuroprotective and neuroregenerative effects, and the completeness of neuronal protection may depend on (i) degree of neuronal loss, (ii) duration and intensity of exercise, and (iii) time elapsed between insult and commencing of training. As the physical activity is neuroprotective for dopaminergic neurons, the question arises what is the mechanism of this protective action. A current hypothesis assumes a central role of neurotrophic factors in the neuroprotection of dopaminergic neurons, even though it is still not clear whether increased DA level in the nigrostriatal axis results from neurogenesis of dopaminergic neurons in the SN, recovery of the phenotype of dopaminergic neurons, increased sprouting of the residual dopaminergic axons in the striatum, or generation of local striatal neurons from inhibitory interneurons. In the present review, we discuss studies describing the influence of physical exercise on the PD-like changes manifested in animal models of the disease and focus our interest on the current state of knowledge on the mechanism of neuroprotection induced by physical activity as a supportive therapy in PD.
Collapse
Affiliation(s)
- Ewelina Palasz
- Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw, Poland
| | - Wiktor Niewiadomski
- Department of Applied Physiology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Gasiorowska
- Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw, Poland.,Department of Applied Physiology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Adrianna Wysocka
- Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw, Poland
| | - Anna Stepniewska
- Department of Applied Physiology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Grazyna Niewiadomska
- Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw, Poland
| |
Collapse
|
22
|
Trichloroethylene and its metabolite TaClo lead to degeneration of substantia nigra dopaminergic neurones: Effects in wild type and human A30P mutant α-synuclein mice. Neurosci Lett 2019; 711:134437. [PMID: 31422098 PMCID: PMC6892271 DOI: 10.1016/j.neulet.2019.134437] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 11/24/2022]
Abstract
Parkinson's disease (PD) is characterised pathologically by degeneration of the dopaminergic (DA) neurones of the substantia nigra pars compacta (SNpc) and the presence of α-synuclein containing Lewy body inclusions. Trichloroethylene (TCE) has been suggested as a potential environmental chemical that may contribute to the development of PD, via conversion to the neurotoxin, 1-Trichloromethyl-1,2,3,4-tetrahydro-β-carboline (TaClo). We investigated the effect of an 8 week exposure to TCE or TaClo on wild type and, as an experimental model of PD, A30P mutant α-synuclein overexpressing mice using a combination of behaviour and pathology. TCE or TaClo exposure caused significant DA neuronal loss within the SNpc in both wild type and transgenic mice. Cell numbers were lower in A30P animals than wild type, however, no additive effect of TCE or TaClo exposure and A30P overexpression was found. TCE or TaClo did not appear to lead to acceleration of motor or cognitive deficits in either wild type or A30P mutant mice, potentially because of the modest reductions of DA neuronal number in the SNpc. Our results do however suggest that TCE exposure could be a possible factor in development of PD like changes following exposure.
Collapse
|
23
|
Samal J, Rebelo AL, Pandit A. A window into the brain: Tools to assess pre-clinical efficacy of biomaterials-based therapies on central nervous system disorders. Adv Drug Deliv Rev 2019; 148:68-145. [PMID: 30710594 DOI: 10.1016/j.addr.2019.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/04/2019] [Accepted: 01/28/2019] [Indexed: 12/13/2022]
Abstract
Therapeutic conveyance into the brain is a cardinal requirement for treatment of diverse central nervous system (CNS) disorders and associated pathophysiology. Effectual shielding of the brain by the blood-brain barrier (BBB) sieves out major proportion of therapeutics with the exception of small lipophilic molecules. Various nano-delivery systems (NDS) provide an effective solution around this obstacle owing to their small size and targeting properties. To date, these systems have been used for several pre-clinical disease models including glioma, neurodegenerative diseases and psychotic disorders. An efficacy screen for these systems involves a test battery designed to probe into the multiple facets of therapeutic delivery. Despite their wide application in redressing various disease targets, the efficacy evaluation strategies for all can be broadly grouped into four modalities, namely: histological, bio-imaging, molecular and behavioural. This review presents a comprehensive insight into all of these modalities along with their strengths and weaknesses as well as perspectives on an ideal design for a panel of tests to screen brain nano-delivery systems.
Collapse
Affiliation(s)
- Juhi Samal
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Ana Lucia Rebelo
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
24
|
Kline EM, Butkovich LM, Bradner JM, Chang J, Gelbard H, Goodfellow V, Caudle WM, Tansey MG. The second generation mixed lineage kinase-3 (MLK3) inhibitor CLFB-1134 protects against neurotoxin-induced nigral dopaminergic neuron loss. Exp Neurol 2019; 318:157-164. [PMID: 31077715 PMCID: PMC6592621 DOI: 10.1016/j.expneurol.2019.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 05/01/2019] [Accepted: 05/07/2019] [Indexed: 11/29/2022]
Abstract
Dopaminergic neurons express mixed lineage kinases which regulate the expression of cell death genes. In Parkinson's disease, cell death via apoptosis is prevalent, and previous work testing mixed lineage kinase inhibitors in animal models suggested the inhibitors had some neuroprotective potential. CLFB-1134 is a new, brain-penetrant inhibitor specific for MLK3, tested here in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of dopaminergic depletion and nigral neuron death in mice. After ensuring that treatment with CLFB-1134 did not alter conversion of MPTP to MPP+, we demonstrated CLFB-1134's inhibition of MLK3 and neuroprotective efficacy. Specifically we evaluated the integrity of the nigrostriatal dopamine system following MPTP by assessing protein expression, high performance liquid chromatography, and immunohistology with stereology. We found that CLFB-1134 achieves protection of striatal dopaminergic terminals and nigral cell bodies when dosed simultaneously or following MPTP treatment. By preventing phosphorylation of JNK and other downstream targets of MLK3, CLFB-1134 protects against the neurotoxin MPTP. Inhibition of MLK3 may be a valid target for future work investigating treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Elizabeth M Kline
- Emory University, 615 Michael St, Atlanta, GA 30322, United States of America.
| | - Laura M Butkovich
- Emory University, 615 Michael St, Atlanta, GA 30322, United States of America.
| | - Joshua M Bradner
- Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, United States of America.
| | - Jianjun Chang
- Emory University, 615 Michael St, Atlanta, GA 30322, United States of America.
| | - Harris Gelbard
- University of Rochester Medical Center, Box 645, 601 Elmwood Avenue, Rochester, NY 14642, United States of America.
| | - Val Goodfellow
- Califia Bio Inc., San Diego, CA, United States of America.
| | - W Michael Caudle
- Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, United States of America.
| | - Malú G Tansey
- Emory University, 615 Michael St, Atlanta, GA 30322, United States of America.
| |
Collapse
|
25
|
Wertman V, Gromova A, La Spada AR, Cortes CJ. Low-Cost Gait Analysis for Behavioral Phenotyping of Mouse Models of Neuromuscular Disease. J Vis Exp 2019. [PMID: 31380846 DOI: 10.3791/59878] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Measurement of animal locomotion is a common behavioral tool used to describe the phenotype of a given disease, injury, or drug model. The low-cost method of gait analysis demonstrated here is a simple but effective measure of gait abnormalities in murine models. Footprints are analyzed by painting a mouse's feet with non-toxic washable paint and allowing the subject to walk through a tunnel on a sheet of paper. The design of the testing tunnel takes advantage of natural mouse behavior and their affinity for small dark places. The stride length, stride width, and toe spread of each mouse is easily measured using a ruler and a pencil. This is a well-established and reliable method, and it generates several metrics that are analogous to digital systems. This approach is sensitive enough to detect changes in stride early in phenotype presentation, and due to its non-invasive approach, it allows for testing of groups across life-span or phenotypic presentation.
Collapse
Affiliation(s)
- Virginia Wertman
- Department of Neurology, Duke University School of Medicine; Duke Center for Neurodegeneration & Neurotherapeutics, Duke University School of Medicine
| | - Anastasia Gromova
- Department of Neurology, Duke University School of Medicine; Duke Center for Neurodegeneration & Neurotherapeutics, Duke University School of Medicine; Biomedical Sciences Graduate Program, University of California San Diego
| | - Albert R La Spada
- Department of Neurology, Duke University School of Medicine; Duke Center for Neurodegeneration & Neurotherapeutics, Duke University School of Medicine; Department of Neurobiology, Duke University School of Medicine; Department of Cell Biology, Duke University School of Medicine
| | - Constanza J Cortes
- Department of Neurology, Duke University School of Medicine; Duke Center for Neurodegeneration & Neurotherapeutics, Duke University School of Medicine;
| |
Collapse
|
26
|
Valek L, Auburger G, Tegeder I. Sensory neuropathy and nociception in rodent models of Parkinson's disease. Dis Model Mech 2019; 12:12/6/dmm039396. [PMID: 31248900 PMCID: PMC6602317 DOI: 10.1242/dmm.039396] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) often manifests with prodromal pain and sensory losses whose etiologies are not well understood. Multiple genetic and toxicity-based rodent models of PD partly recapitulate the histopathology and motor function deficits. Although far less studied, there is some evidence that rodents, similar to humans, develop sensory manifestations of the disease, which may precede motor disturbances and help to elucidate the underlying mechanisms of PD-associated pain at the molecular and neuron circuit levels. The present Review summarizes nociception and other sensory functions in frequently used rodent PD models within the context of the complex phenotypes. In terms of mechanisms, it appears that the acute loss of dopaminergic neurons in systemic toxicity models (MPTP, rotenone) primarily causes nociceptive hyperexcitability, presumably owing to a loss of inhibitory control, whereas genetic models primarily result in a progressive loss of heat perception, reflecting sensory fiber neuropathies. At the molecular level, neither α-synuclein deposits alone nor failure of mitophagy alone appear to be strong enough to result in axonal or synaptic pathology of nociceptive neurons that manifest at the behavioral level, and peripheral sensory loss may mask central ‘pain’ in behavioral tests. Hence, allostatic combinations or additional challenges and novel behavioral assessments are needed to better evaluate PD-associated sensory neuropathies and pain in rodents. Summary: Rodent models of Parkinson's disease partially develop prodromal somatosensory and olfactory dysfunctions reminiscent of sensory neuropathies in patients and reveal mechanistic insight, but data are incomplete and fragmented.
Collapse
Affiliation(s)
- Lucie Valek
- Institute of Clinical Pharmacology, Goethe-University Hospital, 60590 Frankfurt, Germany
| | - Georg Auburger
- Experimental Neurology, Goethe-University Hospital, 60590 Frankfurt, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University Hospital, 60590 Frankfurt, Germany
| |
Collapse
|
27
|
Luo Z, Ahlers-Dannen KE, Spicer MM, Yang J, Alberico S, Stevens HE, Narayanan NS, Fisher RA. Age-dependent nigral dopaminergic neurodegeneration and α-synuclein accumulation in RGS6-deficient mice. JCI Insight 2019; 5:126769. [PMID: 31120439 DOI: 10.1172/jci.insight.126769] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Parkinson's is primarily a non-familial, age-related disorder caused by α-synuclein accumulation and the progressive loss of dopamine neurons in the substantia nigra pars compacta (SNc). G protein-coupled receptor (GPCR)-cAMP signaling has been linked to a reduction in human Parkinson's incidence and α-synuclein expression. Neuronal cAMP levels are controlled by GPCRs coupled to Gs or Gi/o, which increase or decrease cAMP, respectively. Regulator of G protein signaling 6 (RGS6) powerfully inhibits Gi/o signaling. Therefore, we hypothesized that RGS6 suppresses D2 autoreceptor- Gi/o signaling in SNc dopamine neurons promoting neuronal survival and reducing α-synuclein expression. Here we provide novel evidence that RGS6 critically suppresses late-age-onset SNc dopamine neuron loss and α-synuclein accumulation. RGS6 is restrictively expressed in human SNc dopamine neurons and, despite their loss in Parkinson's, all surviving neurons express RGS6. RGS6-/- mice exhibit hyperactive D2 autoreceptors with reduced cAMP signaling in SNc dopamine neurons. Importantly, RGS6-/- mice recapitulate key sporadic Parkinson's hallmarks, including: SNc dopamine neuron loss, reduced nigrostriatal dopamine, motor deficits, and α-synuclein accumulation. To our knowledge, Rgs6 is the only gene whose loss phenocopies these features of human Parkinson's. Therefore, RGS6 is a key regulator of D2R-Gi/o signaling in SNc dopamine neurons, protecting against Parkinson's neurodegeneration and α-synuclein accumulation.
Collapse
Affiliation(s)
- Zili Luo
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Katelin E Ahlers-Dannen
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Mackenzie M Spicer
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Interdisciplinary Graduate Program of Molecular Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Jianqi Yang
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | | | - Hanna E Stevens
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Nandakumar S Narayanan
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Rory A Fisher
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
28
|
Agnihotri SK, Sun L, Yee BK, Shen R, Akundi RS, Zhi L, Duncan MJ, Cass WA, Büeler H. PINK1 deficiency is associated with increased deficits of adult hippocampal neurogenesis and lowers the threshold for stress-induced depression in mice. Behav Brain Res 2019; 363:161-172. [PMID: 30735759 DOI: 10.1016/j.bbr.2019.02.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 12/29/2022]
Abstract
Parkinson's disease (PD) is characterized by motor impairments and several non-motor features, including frequent depression and anxiety. Stress-induced deficits of adult hippocampal neurogenesis (AHN) have been linked with abnormal affective behavior in animals. It has been speculated that AHN defects may contribute to affective symptoms in PD, but this hypothesis remains insufficiently tested in animal models. Mice that lack the PD-linked kinase PINK1 show impaired differentiation of adult-born neurons in the hippocampus. Here, we examined the relationship between AHN deficits and affective behavior in PINK1-/- mice under basal (no stress) conditions and after exposure to chronic stress. PINK1 loss and corticosterone negatively and jointly affected AHN, leading to lower numbers of neural stem cells and newborn neurons in the dentate gyrus of corticosterone-treated PINK1-/- mice. Despite increased basal AHN deficits, PINK1-deficient mice showed normal affective behavior. However, lack of PINK1 sensitized mice to corticosterone-induced behavioral despair in the tail suspension test at a dose where wildtype mice were unaffected. Moreover, after two weeks of chronic restraint stress male PINK1-/- mice displayed increased immobility in the forced swim test, and protein expression of the glucocorticoid receptor in the hippocampus was reduced. Thus, while impaired AHN as such is insufficient to cause affective dysfunction in this PD model, PINK1 deficiency may lower the threshold for chronic stress-induced depression in PD. Finally, PINK1-deficient mice displayed reduced basal voluntary wheel running but normal rotarod performance, a finding whose mechanisms remain to be determined.
Collapse
Affiliation(s)
- Sandeep K Agnihotri
- School of Life Science and Technology, Harbin Institute of Technology, 150080 Harbin, China
| | - Liuke Sun
- School of Life Science and Technology, Harbin Institute of Technology, 150080 Harbin, China
| | - Benjamin K Yee
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China
| | - Ruifang Shen
- School of Life Science and Technology, Harbin Institute of Technology, 150080 Harbin, China
| | - Ravi S Akundi
- Department of Neuroscience, University of Kentucky, Lexington KY 40536, USA
| | - Lianteng Zhi
- Department of Neuroscience, University of Kentucky, Lexington KY 40536, USA
| | - Marilyn J Duncan
- Department of Neuroscience, University of Kentucky, Lexington KY 40536, USA
| | - Wayne A Cass
- Department of Neuroscience, University of Kentucky, Lexington KY 40536, USA
| | - Hansruedi Büeler
- School of Life Science and Technology, Harbin Institute of Technology, 150080 Harbin, China.
| |
Collapse
|
29
|
Bhat PV, Anand T, Mohan Manu T, Khanum F. Restorative effect of l-Dopa treatment against Ochratoxin A induced neurotoxicity. Neurochem Int 2018; 118:252-263. [DOI: 10.1016/j.neuint.2018.04.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 02/07/2018] [Accepted: 04/04/2018] [Indexed: 11/30/2022]
|
30
|
Ekimova IV, Plaksina DV, Pastukhov YF, Lapshina KV, Lazarev VF, Mikhaylova ER, Polonik SG, Pani B, Margulis BA, Guzhova IV, Nudler E. New HSF1 inducer as a therapeutic agent in a rodent model of Parkinson's disease. Exp Neurol 2018; 306:199-208. [PMID: 29704482 DOI: 10.1016/j.expneurol.2018.04.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/17/2018] [Accepted: 04/24/2018] [Indexed: 01/03/2023]
Abstract
Molecular chaperone HSP70 (HSPA1A) has therapeutic potential in conformational neurological diseases. Here we evaluate the neuroprotective function of the chaperone in a rat model of Parkinson's disease (PD). We show that the knock-down of HSP70 (HSPA1A) in dopaminergic neurons of the Substantia nigra causes an almost 2-fold increase in neuronal death and multiple motor disturbances in animals. Conversely, pharmacological activation of HSF1 transcription factor and enhanced expression of inducible HSP70 with the echinochrome derivative, U-133, reverses the process of neurodegeneration, as evidenced by а increase in the number of tyrosine hydroxylase-containing neurons, and prevents the motor disturbances that are typical of the clinical stage of the disease. The neuroprotective effect caused by the elevation of HSP70 in nigral neurons is due to the ability of the chaperone to prevent α-synuclein aggregation and microglia activation. Our findings support the therapeutic relevance of HSP70 induction for the prevention and/or deceleration of PD-like neurodegeneration.
Collapse
Affiliation(s)
- Irina V Ekimova
- Laboratory of Comparative Thermophysiology, I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, pr. Maurice Thorez, 44, St. Petersburg 194223, Russia.
| | - Daria V Plaksina
- Laboratory of Comparative Thermophysiology, I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, pr. Maurice Thorez, 44, St. Petersburg 194223, Russia
| | - Yuri F Pastukhov
- Laboratory of Comparative Thermophysiology, I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, pr. Maurice Thorez, 44, St. Petersburg 194223, Russia
| | - Ksenia V Lapshina
- Laboratory of Comparative Thermophysiology, I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, pr. Maurice Thorez, 44, St. Petersburg 194223, Russia
| | - Vladimir F Lazarev
- Cell Protection Mechanisms Laboratory, Institute of Cytology Russian of Academy of Sciences, Tikhoretsky pr., 4, St. Petersburg 194064, Russia
| | - Elena R Mikhaylova
- Cell Protection Mechanisms Laboratory, Institute of Cytology Russian of Academy of Sciences, Tikhoretsky pr., 4, St. Petersburg 194064, Russia
| | - Sergey G Polonik
- G.B.Elyakov Pacific Institute of Bioorganic Chemistry of Far East Branch of Russian Academy of Sciences, pr. 100 let Vladivostoku 159, Vladivostok 690022, Russia
| | - Bibhusita Pani
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine NY, NY 10016, USA
| | - Boris A Margulis
- Cell Protection Mechanisms Laboratory, Institute of Cytology Russian of Academy of Sciences, Tikhoretsky pr., 4, St. Petersburg 194064, Russia
| | - Irina V Guzhova
- Cell Protection Mechanisms Laboratory, Institute of Cytology Russian of Academy of Sciences, Tikhoretsky pr., 4, St. Petersburg 194064, Russia.
| | - Evgeny Nudler
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine NY, NY 10016, USA; Howard Hughes Medical Institute, New York University School of Medicine NY, NY 10016, USA.
| |
Collapse
|
31
|
Safandeev VV, Kolacheva AA, Ivanov DE, Ugryumov MV. Detection of the latent functional insufficiency of dopaminergic neurons in the nigrostriatal system in a chronic model of Parkinson’s disease. NEUROCHEM J+ 2017. [DOI: 10.1134/s1819712417040079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
32
|
Beier EE, Neal M, Alam G, Edler M, Wu LJ, Richardson JR. Alternative microglial activation is associated with cessation of progressive dopamine neuron loss in mice systemically administered lipopolysaccharide. Neurobiol Dis 2017; 108:115-127. [PMID: 28823928 DOI: 10.1016/j.nbd.2017.08.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/17/2017] [Accepted: 08/16/2017] [Indexed: 12/12/2022] Open
Abstract
Inflammation arising from central and/or peripheral sources contributes to the pathogenesis of multiple neurodegenerative diseases including Parkinson's disease (PD). Emerging data suggest that differential activation of glia could lead to the pathogenesis and progression of PD. Here, we sought to determine the relationship between lipopolysaccharide (LPS) treatment, loss of dopaminergic neurons and differential activation of glia. Using a model of repeated injections with LPS (1mg/kg, i.p. for 4days), we found that LPS induced a 34% loss of dopamine neurons in the substantia nigra 19days after initiation of treatment, but no further cell loss was observed at 36days. LPS induced a strong pro-inflammatory response with increased mRNA expression of pro-inflammatory markers, including tumor necrosis factor-α (4.8-fold), inducible nitric oxide synthase (2.0-fold), interleukin-1 beta (8.9-fold), interleukin-6 (10.7-fold), and robust glial activation were observed at 1day after final dose of LPS. These pro-inflammatory genes were then reduced at 19days after treatment, when there was a rise in the anti-inflammatory genes Ym1 (1.8-fold) and arginase-1 (2.6-fold). Additionally, 36days after the last LPS injection there was a significant increase in interleukin-10 (2.1-fold) expression. The qPCR data results were supported by protein data, including cytokine measurements, western blotting, and immunofluorescence in brain microglia. Taken together, these data demonstrate that progressive neurodegeneration in the substantia nigra following LPS is likely arrested by microglia shifting to an anti-inflammatory phenotype. Thus, strategies to promote resolution of neuroinflammation may be a promising avenue to slow the progressive loss of dopamine neurons in PD.
Collapse
Affiliation(s)
- Eric E Beier
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, United States
| | - Matthew Neal
- Department of Pharmaceutical Sciences, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Gelerah Alam
- Department of Pharmaceutical Sciences, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Melissa Edler
- Department of Pharmaceutical Sciences, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Jason R Richardson
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, United States; Department of Pharmaceutical Sciences, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, OH, United States.
| |
Collapse
|
33
|
Viveros-Paredes JM, González-Castañeda RE, Gertsch J, Chaparro-Huerta V, López-Roa RI, Vázquez-Valls E, Beas-Zarate C, Camins-Espuny A, Flores-Soto ME. Neuroprotective Effects of β-Caryophyllene against Dopaminergic Neuron Injury in a Murine Model of Parkinson's Disease Induced by MPTP. Pharmaceuticals (Basel) 2017; 10:E60. [PMID: 28684694 PMCID: PMC5620604 DOI: 10.3390/ph10030060] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders and is characterized by the loss of dopaminergic neurons in the substantia nigra (SN). Although the causes of PD are not understood, evidence suggests that its pathogenesis is associated with oxidative stress and inflammation. Recent studies have suggested a protective role of the cannabinoid signalling system in PD. β-caryophyllene (BCP) is a natural bicyclic sesquiterpene that is an agonist of the cannabinoid type 2 receptor (CB2R). Previous studies have suggested that BCP exerts prophylactic and/or curative effects against inflammatory bowel disease through its antioxidative and/or anti-inflammatory action. The present study describes the neuroprotective effects of BCP in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced murine model of PD, and we report the results of our investigation of its neuroprotective mechanism in neurons and glial cells. In the murine model, BCP pretreatment ameliorated motor dysfunction, protected against dopaminergic neuronal losses in the SN and striatum, and alleviated MPTP-induced glia activation. Additionally, BCP inhibited the levels of inflammatory cytokines in the nigrostriatal system. The observed neuroprotection and inhibited glia activation were reversed upon treatment with the CB2R selective antagonist AM630, confirming the involvement of the CB2R. These results indicate that BCP acts via multiple neuroprotective mechanisms in our murine model and suggest that BCP may be viewed as a potential treatment and/or preventative agent for PD.
Collapse
Affiliation(s)
- Juan M Viveros-Paredes
- Departamento de Farmacobiología CUCEI, Universidad de Guadalajara, 44430 Guadalajara, Mexico.
| | - Rocio E González-Castañeda
- Laboratorio de Microscopía de Alta Resolución, Departamento de Neurociencias, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, 44340 Guadalajara, Mexico.
| | - Juerg Gertsch
- Institute of Biochemistry and Molecular Medicine, NCCR Trans Cure, University of Bern, CH-3012 Bern, Switzerland.
| | - Veronica Chaparro-Huerta
- Laboratorio de Neurobiología Celular y Molecular, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, 44421 Guadalajara, Mexico.
| | - Rocio I López-Roa
- Departamento de Farmacobiología CUCEI, Universidad de Guadalajara, 44430 Guadalajara, Mexico.
| | - Eduardo Vázquez-Valls
- Laboratorio de Inmunodeficiencias y Retrovirus Humanos, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, 44421 Guadalajara, Mexico.
| | - Carlos Beas-Zarate
- Laboratorio de Regeneración y Desarrollo Neural, Instituto de Neurobiología, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, 44340 Guadalajara, Mexico.
| | - Antoni Camins-Espuny
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia i Ciencias de l'Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain.
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain.
| | - Mario E Flores-Soto
- Departamento de Farmacobiología CUCEI, Universidad de Guadalajara, 44430 Guadalajara, Mexico.
- Laboratorio de Microscopía de Alta Resolución, Departamento de Neurociencias, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, 44340 Guadalajara, Mexico.
| |
Collapse
|
34
|
Stephenson DT, Childs MA, Li Q, Carvajal-Gonzalez S, Opsahl A, Tengowski M, Meglasson MD, Merchant K, Emborg ME. Differential Loss of Presynaptic Dopaminergic Markers in Parkinsonian Monkeys. Cell Transplant 2017; 16:229-44. [PMID: 17503735 DOI: 10.3727/000000007783464704] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Assessment of dopamine nerve terminal function and integrity is a strategy employed to monitor deficits in Parkinson's disease (PD) patients and in preclinical models of PD. Dopamine replacement therapies effectively replenish the diminished supply of endogenous dopamine and provide symptomatic benefit to patients. Tyrosine hydroxylase (TH), dopamine transporter (DAT), vesicular monoamine transporter 2 (VMAT2), and amino acid decarboxylase (AADC) are widely used markers of dopaminergic neurons and terminals. The present studies were initiated to: (a) assess alterations in all four markers in the MPTP primate model of dopaminergic degeneration and (b) to determine whether L-DOPA treatment may itself modulate the expression of these markers. MPTP treatment induced a significant decline of dopaminergic immunoreactive fiber and terminal density in the basal ganglia. The amount of reduction varied between markers. The rank order of presynaptic marker loss, from most to least profound reduction, was TH > VMAT2 > DAT > AADC. Semiquantitative image analysis of relative dopaminergic presynaptic fiber and terminal density illustrated region-specific reduction of all four markers. Double immunofluorescence colocalization of two presynaptic markers on the same tissue section confirmed there was a more dramatic loss of TH than of VMAT2 or of DAT following MPTP treatment. L-DOPA treatment was associated with a significantly higher level of AADC and VMAT2 immunoreactivity in the caudate nucleus compared to placebo. These results illustrate that neurotoxic injury of the dopamine system in primates leads to altered and differential expression of presynaptic dopaminergic markers in the basal ganglia and that expression of such markers may be modulated by L-DOPA therapy. These findings have implications for the use of biomarkers of disease progression as well as for the assessment of neurorestorative strategies, such as cell replacement, for the treatment of PD.
Collapse
|
35
|
Chronic mild stress augments MPTP induced neurotoxicity in a murine model of Parkinson's disease. Physiol Behav 2017; 173:132-143. [DOI: 10.1016/j.physbeh.2017.01.046] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/05/2017] [Accepted: 01/23/2017] [Indexed: 12/30/2022]
|
36
|
Alam G, Edler M, Burchfield S, Richardson JR. Single low doses of MPTP decrease tyrosine hydroxylase expression in the absence of overt neuron loss. Neurotoxicology 2017; 60:99-106. [PMID: 28377118 DOI: 10.1016/j.neuro.2017.03.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 03/26/2017] [Accepted: 03/27/2017] [Indexed: 10/19/2022]
Abstract
Parkinson's disease (PD) is the second most common age-related neurodegenerative disease. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a prototypical neurotoxicant used in mice to mimic primary features of PD pathology including striatal dopamine depletion and dopamine neuron loss in the substantia nigra pars compacta (SNc). In the literature, there are several experimental paradigms involving multiple doses of MPTP that are used to elicit dopamine neuron loss. However, a recent study reported that a single low dose caused significant loss of dopamine neurons. Here, we determined the effect of a single intraperitoneal injection of one of three doses of MPTP (0.1, 2 and 20mg/kg) on dopamine neurons, labeled by tyrosine hydroxylase (TH+), and total neuron number (Nissl+) in the SNc using unbiased stereological counting. Data reveal a significant loss of neurons in the SNc (TH+ and Nissl+) only in the group treated with 20mg/kg MPTP. Groups treated with lower dose of MPTP (0.1 and 2mg/kg) only showed significant loss of TH+ neurons rather than TH+ and Nissl+ neurons. Striatal dopamine levels were decreased in the groups treated with 2 and 20mg/kg MPTP and striatal terminal markers including, TH and the dopamine transporter (DAT), were only decreased in the groups treated with 20mg/kg MPTP. These data demonstrate that lower doses of MPTP likely result in loss of TH expression rather than actual dopamine neuron loss in the SN. This finding reinforces the need to measure both total neuron number along with TH+ cells in determining dopamine neuron loss.
Collapse
Affiliation(s)
- Gelareh Alam
- Department of Pharmaceutical Sciences, College of Pharmacy, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, OH 44272, United States
| | - Melissa Edler
- Department of Pharmaceutical Sciences, College of Pharmacy, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, OH 44272, United States
| | - Shelbie Burchfield
- Department of Pharmaceutical Sciences, College of Pharmacy, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, OH 44272, United States
| | - Jason R Richardson
- Department of Pharmaceutical Sciences, College of Pharmacy, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, OH 44272, United States.
| |
Collapse
|
37
|
Girard-Joyal O, Ismail N. Effect of LPS treatment on tyrosine hydroxylase expression and Parkinson-like behaviors. Horm Behav 2017; 89:1-12. [PMID: 28025041 DOI: 10.1016/j.yhbeh.2016.12.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 12/13/2016] [Accepted: 12/20/2016] [Indexed: 01/18/2023]
Abstract
Puberty is a critical period of development during which the brain undergoes reorganizing and remodeling. Exposure to stress during this period is thought to interfere with normal brain development and increase susceptibility to mental illnesses. In female mice, pubertal exposure to lipopolysaccharide (LPS), a bacterial endotoxin, has been shown to alter sexual, anxiety-like, and depression-like behaviors and cognition in an enduring manner. However, the mechanisms underlying these effects remain unknown. The present study examined age and sex difference in tyrosine hydroxylase (TH) expression and dopamine-dependent and Parkinson-like behaviors following LPS treatment. The results show that LPS treatment during adulthood causes an enduring increase in TH expression in many of the brain regions examined. In contrast, there is no change in TH expression following LPS treatment during puberty. However, pubertal LPS treatment induces enduring behavioral deficits in tests of Parkinson-like behaviors, more so in male than in female mice. These results suggest that the low levels of TH following exposure to pubertal immune challenge may predispose mice to Parkinson-like behavior. These findings add to our understanding of stress and immune responses during puberty and their impact on mental health later in life.
Collapse
Affiliation(s)
| | - Nafissa Ismail
- School of Psychology, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
38
|
Synaptic vesicle glycoprotein 2C (SV2C) modulates dopamine release and is disrupted in Parkinson disease. Proc Natl Acad Sci U S A 2017; 114:E2253-E2262. [PMID: 28246328 PMCID: PMC5358362 DOI: 10.1073/pnas.1616892114] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Here we describe a role for the synaptic vesicle glycoprotein 2C (SV2C) in dopamine neurotransmission and Parkinson disease (PD). SV2C is expressed on the vesicles of dopamine-producing neurons, and genetic deletion of SV2C causes a reduction in synaptic release of dopamine. The reduced dopamine release is associated with a decrease in motor activity. SV2C is suspected of mediating the neuroprotective effects of nicotine, and we show an ablated neurochemical response to nicotine in SV2C-knockout mice. Last, we demonstrate that SV2C expression is specifically disrupted in mice that express mutated α-synuclein and in humans with PD. Together, these data establish SV2C as an important mediator of dopamine homeostasis and a potential contributor to PD pathogenesis. Members of the synaptic vesicle glycoprotein 2 (SV2) family of proteins are involved in synaptic function throughout the brain. The ubiquitously expressed SV2A has been widely implicated in epilepsy, although SV2C with its restricted basal ganglia distribution is poorly characterized. SV2C is emerging as a potentially relevant protein in Parkinson disease (PD), because it is a genetic modifier of sensitivity to l-DOPA and of nicotine neuroprotection in PD. Here we identify SV2C as a mediator of dopamine homeostasis and report that disrupted expression of SV2C within the basal ganglia is a pathological feature of PD. Genetic deletion of SV2C leads to reduced dopamine release in the dorsal striatum as measured by fast-scan cyclic voltammetry, reduced striatal dopamine content, disrupted α-synuclein expression, deficits in motor function, and alterations in neurochemical effects of nicotine. Furthermore, SV2C expression is dramatically altered in postmortem brain tissue from PD cases but not in Alzheimer disease, progressive supranuclear palsy, or multiple system atrophy. This disruption was paralleled in mice overexpressing mutated α-synuclein. These data establish SV2C as a mediator of dopamine neuron function and suggest that SV2C disruption is a unique feature of PD that likely contributes to dopaminergic dysfunction.
Collapse
|
39
|
Niewiadomski W, Palasz E, Skupinska M, Zylinski M, Steczkowska M, Gasiorowska A, Niewiadomska G, Riedel G. TracMouse: A computer aided movement analysis script for the mouse inverted horizontal grid test. Sci Rep 2016; 6:39331. [PMID: 27982134 PMCID: PMC5159816 DOI: 10.1038/srep39331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 11/22/2016] [Indexed: 11/19/2022] Open
Abstract
In rodents, detection and quantification of motor impairments is difficult. The traction test (inverted grid with mice clinging to the underside) currently has no objective rating system. We here developed and validated the semi-automatic MATLAB script TracMouse for unbiased detection of video-recorded movement patterns. High precision videos were analyzed by: (i) principal identification of anatomical paw details frame-by-frame by an experimentally blinded rater; (ii) automatic retrieval of proxies by TracMouse for individual paws. The basic states of Hold and Step were discriminated as duration and frequency, and these principle parameters were converted into static and dynamic endpoints and their discriminating power assessed in a dopaminergic lesion model. Relative to hind paws, forepaws performed ~4 times more steps, they were ~20% longer, and Hold duration was ~5 times shorter in normal C57Bl/6 mice. Thus, forepaw steps were classified as exploratory, hind paw movement as locomotive. Multiple novel features pertaining to paw sequence, step lengths and exploratory touches were accessible through TracMouse and revealed subtle Parkinsonian phenotypes. Novel proxies using TracMouse revealed previously unidentified features of movement and may aid the understanding of (i) brain circuits related to motor planning and execution, and (ii) phenotype detection in experimental models of movement disorders.
Collapse
Affiliation(s)
- W. Niewiadomski
- Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
- Warsaw Medical University, Warsaw, Poland
| | | | | | - M. Zylinski
- Warsaw University of Technology, Warsaw, Poland
| | | | - A. Gasiorowska
- Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
- Nencki Institute, Warsaw, Poland
| | | | - G. Riedel
- Institute of Medical Sciences, University of Aberdeen, UK
| |
Collapse
|
40
|
Glajch KE, Ferraiuolo L, Mueller KA, Stopford MJ, Prabhkar V, Gravanis A, Shaw PJ, Sadri-Vakili G. MicroNeurotrophins Improve Survival in Motor Neuron-Astrocyte Co-Cultures but Do Not Improve Disease Phenotypes in a Mutant SOD1 Mouse Model of Amyotrophic Lateral Sclerosis. PLoS One 2016; 11:e0164103. [PMID: 27716798 PMCID: PMC5055348 DOI: 10.1371/journal.pone.0164103] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/20/2016] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease caused by loss of motor neurons. ALS patients experience rapid deterioration in muscle function with an average lifespan of 3–5 years after diagnosis. Currently, the most effective therapeutic only extends lifespan by a few months, thus highlighting the need for new and improved therapies. Neurotrophic factors (NTFs) are important for neuronal development, maintenance, and survival. NTF treatment has previously shown efficacy in pre-clinical ALS models. However, clinical trials using NTFs produced no major improvements in ALS patients, due in part to the limited blood brain barrier (BBB) penetration. In this study we assessed the potential neuroprotective effects of a novel class of compounds known as MicroNeurotrophins (MNTs). MNTs are derivatives of Dehydroepiandrosterone (DHEA), an endogenous neurosteroid that can cross the BBB and bind to tyrosine kinase receptors mimicking the pro-survival effects of NTFs. Here we sought to determine whether MNTs were neuroprotective in two different models of ALS. Our results demonstrate that BNN27 (10 μM) attenuated loss of motor neurons co-cultured with astrocytes derived from human ALS patients with SOD1 mutations via the reduction of oxidative stress. Additionally, in the G93A SOD1 mouse, BNN27 (10 mg/kg) treatment attenuated motor behavioral impairment in the paw grip endurance and rotarod tasks at postnatal day 95 in female but not male mice. In contrast, BNN27 (10 mg/kg and 50 mg/kg) treatment did not alter any other behavioral outcome or neuropathological marker in male or female mice. Lastly, BNN27 was not detected in post-mortem brain or spinal cord tissue of treated mice due to the rapid metabolism of BNN27 by mouse hepatocytes relative to human hepatocytes. Together, these findings demonstrate that BNN27 treatment failed to yield significant neuroprotective effects in the G93A SOD1 model likely due to its rapid rate of metabolism in mice.
Collapse
Affiliation(s)
- Kelly E. Glajch
- NeuroEpigenetics Laboratory, MassGeneral Institute for Neurodegenerative Disease (MIND), Massachusetts General Hospital, Boston, MA, 02129–4404, United States of America
| | - Laura Ferraiuolo
- Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, United Kingdom
| | - Kaly A. Mueller
- NeuroEpigenetics Laboratory, MassGeneral Institute for Neurodegenerative Disease (MIND), Massachusetts General Hospital, Boston, MA, 02129–4404, United States of America
| | - Matthew J. Stopford
- Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, United Kingdom
| | - Varsha Prabhkar
- NeuroEpigenetics Laboratory, MassGeneral Institute for Neurodegenerative Disease (MIND), Massachusetts General Hospital, Boston, MA, 02129–4404, United States of America
| | - Achille Gravanis
- Department of Pharmacology, School of Medicine, University of Crete, Institute of Molecular Biology & Biotechnology-FORTH, Heraklion 71003, Greece
| | - Pamela J. Shaw
- Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, United Kingdom
| | - Ghazaleh Sadri-Vakili
- NeuroEpigenetics Laboratory, MassGeneral Institute for Neurodegenerative Disease (MIND), Massachusetts General Hospital, Boston, MA, 02129–4404, United States of America
- * E-mail:
| |
Collapse
|
41
|
Li D, Tong L, Kawano H, Liu N, Yan HJ, Zhao L, Li HP. Regulation and role of ERK phosphorylation in glial cells following a nigrostriatal pathway injury. Brain Res 2016; 1648:90-100. [PMID: 27402431 DOI: 10.1016/j.brainres.2016.07.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 06/30/2016] [Accepted: 07/07/2016] [Indexed: 02/05/2023]
Abstract
This study was undertaken to examine the function of extracellular signal-regulated kinase (ERK) signaling pathway on the proliferation and activation of microglia/macrophage and astrocytes after brain injury in mice. The result of Western blot showed that p-ERK was immediately activated after injury (<4h), but the duration was short (<4 days). According to immunofluorescence double staining, it was found that at 4 and 8h after injury, p-ERK was expressed in microglia/macrophages, and that more cells were co-expressed by p-ERK and IBA-1 (microglia/macrophage marker) at 8h; at days 1 and 4, p-ERK was expressed in astrocytes, and more cells were co-expressed by p-ERK and GFAP (astrocyte marker) at day 4. After injury, the mice were injected with U0126 (MAPK/ERK signaling pathway inhibitor) via the femoral vein. Compared with those injected with DMSO, the cell number co-expressed by p-ERK and IBA-1 or GFAP significantly decreased (P<0.05). The increase of microglia/macrophage and astrocyte caused by injury was remitted, and the positive cell number significantly decreased (P<0.05). Western blot showed that the expression quantity of IBA-1 and GFAP significantly decreased (P<0.05). Furthermore, the ERK signaling pathway was involved in the proliferation and activation of the two glial cells types and improved long-term neurobehavioral function after brain injury. Therefore, the exploration of the formation mechanism of glial scar after injury and further research on the therapeutic method of neural regeneration are essential.
Collapse
Affiliation(s)
- Dan Li
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Lei Tong
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Hitoshi Kawano
- Department of Health and Dietetics, Faculty of Health and Medical Science. Teikyo Heisei University, Tokyo 170-8445, Japan
| | - Nan Liu
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Hong-Jing Yan
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Liang Zhao
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Hong-Peng Li
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
42
|
The misfolded pro-inflammatory protein S100A9 disrupts memory via neurochemical remodelling instigating an Alzheimer's disease-like cognitive deficit. Behav Brain Res 2016; 306:106-16. [PMID: 26965570 DOI: 10.1016/j.bbr.2016.03.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 02/15/2016] [Accepted: 03/05/2016] [Indexed: 12/14/2022]
Abstract
Memory deficits may develop from a variety of neuropathologies including Alzheimer's disease dementia. During neurodegenerative conditions there are contributory factors such as neuroinflammation and amyloidogenesis involved in memory impairment. In the present study, dual properties of S100A9 protein as a pro-inflammatory and amyloidogenic agent were explored in the passive avoidance memory task along with neurochemical assays in the prefrontal cortex and hippocampus of aged mice. S100A9 oligomers and fibrils were generated in vitro and verified by AFM, Thioflavin T and A11 antibody binding. Native S100A9 as well as S100A9 oligomers and fibrils or their combination were administered intranasally over 14 days followed by behavioral and neurochemical analysis. Both oligomers and fibrils evoked amnestic activity which correlated with disrupted prefrontal cortical and hippocampal dopaminergic neurochemistry. The oligomer-fibril combination produced similar but weaker neurochemistry to the fibrils administered alone but without passive avoidance amnesia. Native S100A9 did not modify memory task performance even though it generated a general and consistent decrease in monoamine levels (DA, 5-HT and NA) and increased metabolic marker ratios of DA and 5-HT turnover (DOPAC/DA, HVA/DA and 5-HIAA) in the prefrontal cortex. These results provide insight into a novel pathogenetic mechanism underlying amnesia in a fear-aggravated memory task based on amyloidogenesis of a pro-inflammatory factor leading to disrupted brain neurochemistry in the aged brain. The data further suggests that amyloid species of S100A9 create deleterious effects principally on the dopaminergic system and this novel finding might be potentially exploited during dementia management through a neuroprotective strategy.
Collapse
|
43
|
Sridharan S, Mohankumar K, Jeepipalli SPK, Sankaramourthy D, Ronsard L, Subramanian K, Thamilarasan M, Raja K, Chandra VK, Sadras SR. Neuroprotective effect of Valeriana wallichii rhizome extract against the neurotoxin MPTP in C57BL/6 mice. Neurotoxicology 2015; 51:172-83. [PMID: 26522450 DOI: 10.1016/j.neuro.2015.10.012] [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: 11/17/2014] [Revised: 10/20/2015] [Accepted: 10/26/2015] [Indexed: 01/08/2023]
Abstract
Oxidative stress and inflammation are some of the contributing factors for dopaminergic neurodegeneration in Parkinson's disease (PD). Though Valeriana wallichii D.C. is known for its nervine activities its effect against PD is yet to be studied. This is the first report on the antioxidant and anti-inflammatory effect of V. wallichii rhizome extract (VWE) in MPTP induced PD mice. GC-MS analysis of VWE indicated the presence of phytoconstituents like isovaleric acid and acacetin. PD induced mice were treated orally with three different doses (50, 100 and 200mg/kg body weight (BW)) of VWE for 14 days and their behavioural changes were studied on days 0, 8, 13 and 21. The levels of striatal dopamine, mid brain tyrosine hydroxylase positive (TH(+)) cell count, TH protein expression, reactive oxygen species (ROS), lipid peroxidation (LPO), antioxidants and inflammatory cytokines were analysed. Mid brain glial fibrillary acidic protein (GFAP) expression was assessed by immunohistochemistry and western blotting. Also mid brain histopathological analysis was performed. VWE treatment significantly recuperated the altered behavioural test scores, striatal dopamine levels, mid brain TH(+) cell count and TH protein levels, increased GFAP expression and the histopathological changes observed in PD mice. Similarly, diminished levels of antioxidants, elevated levels of ROS, LPO and inflammatory cytokines were also significantly ameliorated following VWE treatment. The effective dose of VWE was found to be 200mg/kg BW. Conclusively, V. wallichii rhizome extract has the potential to mitigate oxidative stress and inflammatory damage in PD.
Collapse
Affiliation(s)
- Subhashree Sridharan
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Kumaravel Mohankumar
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Syam Praveen Kumar Jeepipalli
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Divya Sankaramourthy
- Department of Pharmacology, Mother Theresa Post Graduate and Research Institute of Health Sciences, Puducherry 605006, India
| | - Larance Ronsard
- Virology Laboratory-II, National Institute of Immunology, New Delhi, Delhi 110067, India
| | - Kavimani Subramanian
- Department of Pharmacology, Mother Theresa Post Graduate and Research Institute of Health Sciences, Puducherry 605006, India
| | - Manivasagam Thamilarasan
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar 608002, India
| | - Kumar Raja
- Department of Pathology, Rajiv Gandhi Institute of Veterinary Education and Research, Puducherry 605009, India
| | - Varshney Khub Chandra
- Department of Pathology, Rajiv Gandhi Institute of Veterinary Education and Research, Puducherry 605009, India
| | - Sudha Rani Sadras
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry 605014, India.
| |
Collapse
|
44
|
Lv E, Deng J, Yu Y, Wang Y, Gong X, Jia J, Wang X. Nrf2-ARE signals mediated the anti-oxidative action of electroacupuncture in an MPTP mouse model of Parkinson's disease. Free Radic Res 2015; 49:1296-307. [PMID: 26118717 DOI: 10.3109/10715762.2015.1067696] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Oxidative stress and neuroinflammation are early events associated with dopaminergic neuronal degeneration in Parkinson's disease (PD). Previous studies indicated that electroacupuncture (EA) stimulation is effective in protecting dopaminergic neurons from degeneration in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. In this study, we further characterized the effect of EA on MPTP-induced oxidative responses in the mouse dopamine system. We found that subacute administration of MPTP enhanced lipid and protein oxidation and reduced expression of endogenous antioxidant enzymes (such as superoxide dismutase and catalase) in the striatum. MPTP also reduced expression of an antioxidant transcription factor, nuclear factor-E2-related factor-2 (Nrf2), and Nrf2-regulated antioxidant enzymes (nicotinamide adenine dinucleotide phosphate quinone oxidoreductase-1 and heme oxygenase-1) in the striatum and/or midbrain. Using human placental alkaline phosphatase (hPAP) as a reporter of Nrf2-regulated gene expression in hPAP transgenic mice, we found that MPTP suppressed hPAP expression in the striatum and midbrain. Application of EA at an effective frequency (100 Hz) was sufficient to reverse these changes induced by MPTP. In addition, EA reduced microglia activation and astrogliosis in the striatum and midbrain, increased tyrosine hydroxylase levels in the striatum, and improved vertical movement in MPTP mice. These results provide further evidence supporting that EA produces a series of anti-oxidative effects that effectively counteract with the oxidative stress in the nigrostriatal dopamine system induced by MPTP in a mouse model of PD.
Collapse
Affiliation(s)
- E Lv
- a Departments of Neurobiology and Physiology , Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Capital Medical University; Beijing Institute for Brain Disorders , Beijing , 100069 , China
| | - J Deng
- a Departments of Neurobiology and Physiology , Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Capital Medical University; Beijing Institute for Brain Disorders , Beijing , 100069 , China
| | - Y Yu
- a Departments of Neurobiology and Physiology , Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Capital Medical University; Beijing Institute for Brain Disorders , Beijing , 100069 , China
| | - Y Wang
- a Departments of Neurobiology and Physiology , Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Capital Medical University; Beijing Institute for Brain Disorders , Beijing , 100069 , China
| | - X Gong
- a Departments of Neurobiology and Physiology , Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Capital Medical University; Beijing Institute for Brain Disorders , Beijing , 100069 , China
| | - J Jia
- a Departments of Neurobiology and Physiology , Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Capital Medical University; Beijing Institute for Brain Disorders , Beijing , 100069 , China
| | - X Wang
- a Departments of Neurobiology and Physiology , Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Capital Medical University; Beijing Institute for Brain Disorders , Beijing , 100069 , China
| |
Collapse
|
45
|
Genskow KR, Bradner JM, Hossain MM, Richardson JR, Caudle WM. Selective damage to dopaminergic transporters following exposure to the brominated flame retardant, HBCDD. Neurotoxicol Teratol 2015; 52:162-9. [PMID: 26073293 DOI: 10.1016/j.ntt.2015.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 06/05/2015] [Accepted: 06/09/2015] [Indexed: 12/01/2022]
Abstract
Over the last several decades, the use of halogenated organic compounds has become the cause of environmental and human health concerns. Of particular notoriety has been the establishment of the neurotoxicity of polychlorinated biphenyls (PCBs) and polybrominated diphenyl ethers (PBDEs). The subsequent banning of PBDEs has led to greatly increased use of 1,2,5,6,9,10-hexabromocyclododecane (HBCDD, also known as HBCD) as a flame retardant in consumer products. The physiochemical similarities between HBCDD and PBDEs suggest that HBCDD may also be neurotoxic to the dopamine system, which is specifically damaged in Parkinson disease (PD). The purpose of this study was to assess the neurotoxicity of HBCDD on the nigrostriatal dopamine system using an in vitro and in vivo approach. We demonstrate that exposure to HBCDD (0-25 μM) for 24 h causes significant cell death in the SK-N-SH catecholaminergic cell line, as well as reductions in the growth and viability of TH+ primary cultured neurons at lower concentrations (0-10 μM) after 72 h of treatment. Assessment of the in vivo neurotoxicity of HBCDD (25 mg/kg for 30 days) resulted in significant reductions in the expression of the striatal dopamine transporter and vesicular monoamine transporter 2, both of which are integral in mediating dopamine homeostasis and neurotransmission in the dopamine circuit. However, no changes were seen in the expression of tyrosine hydroxylase in the dopamine terminal, or striatal levels of dopamine. To date, these are the first data to demonstrate that exposure to HBCDD disrupts the nigrostriatal dopamine system. Given these results and the ubiquitous nature of HBCDD in the environment, its possible role as an environmental risk factor for PD should be further investigated.
Collapse
Affiliation(s)
- Kelly R Genskow
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322-3090, USA
| | - Joshua M Bradner
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322-3090, USA; Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA 30322-3090, USA
| | - Muhammad M Hossain
- Department of Environmental and Occupational Medicine, Robert Wood Johnson Medical School Piscataway, NJ 08854, USA
| | - Jason R Richardson
- Department of Environmental and Occupational Medicine, Robert Wood Johnson Medical School Piscataway, NJ 08854, USA
| | - W Michael Caudle
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322-3090, USA; Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA 30322-3090, USA.
| |
Collapse
|
46
|
Kim BW, Koppula S, Kumar H, Park JY, Kim IW, More SV, Kim IS, Han SD, Kim SK, Yoon SH, Choi DK. α-Asarone attenuates microglia-mediated neuroinflammation by inhibiting NF kappa B activation and mitigates MPTP-induced behavioral deficits in a mouse model of Parkinson's disease. Neuropharmacology 2015; 97:46-57. [PMID: 25983275 DOI: 10.1016/j.neuropharm.2015.04.037] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 04/28/2015] [Accepted: 04/29/2015] [Indexed: 12/21/2022]
Abstract
The selective loss of dopaminergic neurons in Parkinson's disease (PD) is associated with microglial activation. Therefore, the importance of early therapeutic intervention to inhibit microglial activation would be an effective strategy to alleviate the progression of PD. α-Asarone, an active compound found in Araceae and Annonaceae plant species has been used to improve various disease conditions including central nervous system disorders. In the present study the in vitro and in vivo therapeutic effects of α-asarone isolated from the rhizome of Acorus gramineus Solander was evaluated on microglia-mediated neuroinflammation and neuroprotection. Lipopolysaccharide (LPS)-stimulated BV-2 microglial cells were used to evaluate in vitro effects. 1-methyl-4 phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced mouse model of PD was developed to study the neuroprotective effects of α-asarone in vivo. The results indicated that α-asarone significantly attenuated the LPS-stimulated increase in neuroinflammatory responses and suppressed pro-inflammatory cytokine production in BV-2 cells. Mechanistic study revealed that α-asarone inhibited the LPS-stimulated activation via regulation of nuclear factor kappa-B by blocking degradation of inhibitor kappa B-alpha signaling in BV-2 microglial cells. In in vivo studies, MPTP intoxication to mice resulted in brain microglial activation and significant behavioral deficits. Prophylactic treatment with α-asarone suppressed microglial activation and attenuated PD-like behavioral impairments as assessed by the Y-maze and pole tests. Taken together, these data demonstrate that α-asarone is a promising neuroprotective agent that should be further evaluated and developed for future prevention and treatment of microglia-mediated neuroinflammatory conditions including PD.
Collapse
Affiliation(s)
- Byung-Wook Kim
- Department of Biotechnology, Konkuk University, 380-701, South Korea
| | - Sushruta Koppula
- Department of Biotechnology, Konkuk University, 380-701, South Korea
| | - Hemant Kumar
- Department of Biotechnology, Konkuk University, 380-701, South Korea
| | - Ju-Young Park
- Department of Molecular Science and Technology, Ajou University, Suwon, 443-749, South Korea
| | - Il-Woung Kim
- Department of Biomedical Chemistry, Konkuk University, 380-701, South Korea
| | - Sandeep V More
- Department of Biotechnology, Konkuk University, 380-701, South Korea
| | - In-Su Kim
- Department of Biotechnology, Konkuk University, 380-701, South Korea
| | - Sang-Don Han
- Department of Neurology, School of Medicine, Konkuk University, 380-704, South Korea
| | - Si-Kwan Kim
- Department of Biomedical Chemistry, Konkuk University, 380-701, South Korea
| | - Sung-Hwa Yoon
- Department of Molecular Science and Technology, Ajou University, Suwon, 443-749, South Korea
| | - Dong-Kug Choi
- Department of Biotechnology, Konkuk University, 380-701, South Korea.
| |
Collapse
|
47
|
Tousson E, Salama AF, Ibrahim W, Sakr S, Masoud A, Akela MA, El-Rahman MAA. Epigenetic Study of Parkinson's Disease in Experimental Animal Model. PHARMACOLOGIA 2015; 6:52-62. [DOI: 10.5567/pharmacologia.2015.52.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
48
|
Gruden MA, Davydova TV, Narkevich VB, Fomina VG, Wang C, Kudrin VS, Morozova-Roche LA, Sewell RD. Noradrenergic and serotonergic neurochemistry arising from intranasal inoculation with α-synuclein aggregates which incite parkinsonian-like symptoms. Behav Brain Res 2015; 279:191-201. [DOI: 10.1016/j.bbr.2014.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 10/28/2014] [Accepted: 11/01/2014] [Indexed: 12/13/2022]
|
49
|
|
50
|
Effect of Resveratrol as Caloric Restriction Mimetic and Environmental Enrichment on Neurobehavioural Responses in Young Healthy Mice. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/545170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Caloric restriction and environmental enrichment have been separately reported to possess health benefits such as improvement in motor and cognitive functions. Resveratrol, a natural polyphenolic compound, has been reported to be caloric restriction mimetic. This study therefore aims to investigate the potential benefit of the combination of resveratrol as CR and EE on learning and memory, motor coordination, and motor endurance in young healthy mice. Fifty mice of both sexes were randomly divided into five groups of 10 animals each: group I animals received carboxymethylcellulose (CMC) orally per kg/day (control), group II animals were maintained on every other day feeding, group III animals received resveratrol 50 mg/kg, suspended in 10 g/L of (CMC) orally per kg/day, group IV animals received CMC and were kept in an enriched environment, and group V animals received resveratrol 50 mg/kg and were kept in EE. The treatment lasted for four weeks. On days 26, 27, and 28 of the study period, the animals were subjected to neurobehavioural evaluation. The results obtained showed that there was no significant change (P>0.05) in neurobehavioural responses in all the groups when compared to the control which indicates that 50 mg/kg of resveratrol administration and EE have no significant effects on neurobehavioural responses in young healthy mice over a period of four weeks.
Collapse
|