1
|
Hui C, Jin J, Ji M, Wang H, Wang X, Ma J, Wang Y, Si Y, Chen S, Guo T. Neuroprotective properties of the Lilium brownii extracts in the experimental model of Parkinson's disease. Metab Brain Dis 2024; 39:1085-1097. [PMID: 39060803 DOI: 10.1007/s11011-024-01397-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 07/20/2024] [Indexed: 07/28/2024]
Abstract
Lilium brownii (L. brownii) is a plant that can be used for both medicine and food. Its bulbs are commonly used to treat neurological disorders like depression, insomnia, and Parkinson's disease (PD). However, the mechanism by which it treats PD is not yet fully understood. This study aims to investigate the possible mechanism of L. brownii extract in treating PD and to compare the efficacy of ethanol and aqueous extracts of L. brownii. In this study, mice with PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP) were given L. brownii extracts for 30 days, and the effects of both extracts were then evaluated. Our study demonstrated that both extracts of L. brownii effectively improved motor dysfunction in PD mice induced by MPTP. Additionally, they increased the number of neurons in the substantia nigra region of the mice. Moreover, both extracts reduced levels of malondialdehyde (MDA) and ferrous ion (Fe2+), while increasing levels of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) in serum. They also influenced the expression of proteins associated with the p62-Keap1-Nrf2 pathway. Interestingly, while both extracts had similar behavioral effects, the ethanol extract appeared to have a more significant impact on individual proteins in the p62-Keap1-Nrf2 pathway compared to the aqueous extract, possibly due to its higher phenolic acid glyceride content. In conclusion, L. brownii shows promise as an effective and safe treatment for PD.
Collapse
Affiliation(s)
- Chengcheng Hui
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Jinghui Jin
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Mengshan Ji
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Haibo Wang
- Henan Institute for Drug and Medical Device Inspection (Henan Vaccine Issuance Center), NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Chinese Materia Medica and prepared slices), Zhengzhou, 450018, China
| | - Xiaowei Wang
- Henan Institute for Drug and Medical Device Inspection (Henan Vaccine Issuance Center), NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Chinese Materia Medica and prepared slices), Zhengzhou, 450018, China
| | - Jianping Ma
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, China
| | - Ya Wang
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, China
| | - Yanpo Si
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Suiqing Chen
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Tao Guo
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| |
Collapse
|
2
|
Kupnicka P, Listos J, Tarnowski M, Kolasa A, Kapczuk P, Surówka A, Kwiatkowski J, Janawa K, Chlubek D, Baranowska-Bosiacka I. The Effect of Prenatal and Neonatal Fluoride Exposure to Morphine-Induced Neuroinflammation. Int J Mol Sci 2024; 25:826. [PMID: 38255899 PMCID: PMC10815549 DOI: 10.3390/ijms25020826] [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: 12/03/2023] [Revised: 01/01/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Physical dependence is associated with the formation of neuroadaptive changes in the central nervous system (CNS), both at the molecular and cellular levels. Various studies have demonstrated the immunomodulatory and proinflammatory properties of morphine. The resulting neuroinflammation in drug dependence exacerbates substance abuse-related behaviors and increases morphine tolerance. Studies prove that fluoride exposure may also contribute to the development of neuroinflammation and neurodegenerative changes. Morphine addiction is a major social problem. Neuroinflammation increases tolerance to morphine, and neurodegenerative effects caused by fluoride in structures related to the development of dependence may impair the functioning of neuronal pathways, change the concentration of neurotransmitters, and cause memory and learning disorders, which implies this element influences the development of dependence. Therefore, our study aimed to evaluate the inflammatory state of selected brain structures in morphine-dependent rats pre-exposed to fluoride, including changes in cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) expression as well as microglial and astroglial activity via the evaluation of Iba1 and GFAP expression. We provide evidence that both morphine administration and fluoride exposure have an impact on the inflammatory response by altering the expression of COX-1, COX-2, ionized calcium-binding adapter molecule (Iba1), and glial fibrillary acidic protein (GFAP) in brain structures involved in dependence development, such as the prefrontal cortex, striatum, hippocampus, and cerebellum. We observed that the expression of COX-1 and COX-2 in morphine-dependent rats is influenced by prior fluoride exposure, and these changes vary depending on the specific brain region. Additionally, we observed active astrogliosis, as indicated by increased GFAP expression, in all brain structures of morphine-dependent rats, regardless of fluoride exposure. Furthermore, the effect of morphine on Iba1 expression varied across different brain regions, and fluoride pre-exposure may influence microglial activation. However, it remains unclear whether these changes are a result of the direct or indirect actions of morphine and fluoride on the factors analyzed.
Collapse
Affiliation(s)
- Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Joanna Listos
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Maciej Tarnowski
- Department of Physiology in Health Sciences, Pomeranian Medical University, 70-210 Szczecin, Poland
| | - Agnieszka Kolasa
- Department of Histology and Embryology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Patrycja Kapczuk
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Anna Surówka
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland
| | - Jakub Kwiatkowski
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Kamil Janawa
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| |
Collapse
|
3
|
Qiao CM, Quan W, Zhou Y, Niu GY, Hong H, Wu J, Zhao LP, Li T, Cui C, Zhao WJ, Shen YQ. Orally Induced High Serum Level of Trimethylamine N-oxide Worsened Glial Reaction and Neuroinflammation on MPTP-Induced Acute Parkinson's Disease Model Mice. Mol Neurobiol 2023; 60:5137-5154. [PMID: 37266763 DOI: 10.1007/s12035-023-03392-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/18/2023] [Indexed: 06/03/2023]
Abstract
Neuroinflammation mediated by brain glial cells is one of the pathological drivers of Parkinson's disease (PD). Recent studies have shown that higher circulating trimethylamine N-oxide (TMAO, a gut microbiota-derived metabolite) can induce neuroinflammation and are strongly related to a variety of central nervous system diseases and adverse brain events. Herein, we explored the effect of pre-existing higher circulating TMAO on dopamine system and neuroinflammation in acute PD model mice induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydroxypyridine (MPTP). TMAO pretreatment was given by adding 3% (w/v) TMAO to drinking water of mice for 21 days to induce higher circulating TMAO status, then mice were administered with MPTP (20 mg/kg, i.p) for four times in one day to construct an acute PD model mice and treated with TMAO continuously until the end of the experiment. Results demonstrated that TMAO treatment significantly increased serum TMAO levels. Moreover, high serum TMAO significantly increased activation of microglia and astrocytes both in striatum and in substantia nigra. And strikingly, high serum TMAO significantly promoted the metabolism of striatal dopamine (DA) of PD model mice, although it had no significant effect on the number of dopaminergic neurons or the content of DA. Furthermore, immunofluorescence, ELISA, and RT-qPCR results of the hippocampus also showed that high serum TMAO significantly promoted the activation of microglia and astrocytes in the dentate gyrus, increased the levels of TNF-α and IL-1β, and upregulated gene expression of M1 microglia-related markers (including CD16, CD32, and iNOS) and A2 astrocyte-related markers (including S100a10, Ptx3, and Emp1) in mRNA levels. In summary, we found that pre-existing high serum levels of TMAO worsened the PD-related brain pathology by promoting DA metabolism, aggravating neuroinflammation and regulating glial cell polarization.
Collapse
Affiliation(s)
- Chen-Meng Qiao
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Wei Quan
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yu Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Gu-Yu Niu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Hui Hong
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Jian Wu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Li-Ping Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Ting Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Chun Cui
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Wei-Jiang Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yan-Qin Shen
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
4
|
Kaya ZB, Karakoc E, McLean PJ, Saka E, Atilla P. Post-inflammatory administration of N-acetylcysteine reduces inflammation and alters receptor levels in a cellular model of Parkinson's disease. FASEB Bioadv 2023; 5:263-276. [PMID: 37415931 PMCID: PMC10320847 DOI: 10.1096/fba.2022-00145] [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: 12/30/2022] [Revised: 04/10/2023] [Accepted: 04/28/2023] [Indexed: 07/08/2023] Open
Abstract
Parkinson's disease (PD) is a complex, multifactorial neurodegenerative disease with a prevalence of 1% over the age of 55. Neuropathological hallmarks of PD include the loss of dopaminergic neurons in the substantia nigra pars compacta and the accumulation of Lewy bodies that contain a variety of proteins and lipids including alpha-synuclein (α-syn). Although the formation of α-syn occurs intracellularly, it can also be found in the extracellular space where it can be taken up by neighboring cells. Toll-like receptor 2 (TLR2) is an immune system receptor that has been shown to recognize extracellular α-syn and modulate its uptake by other cells. Lymphocyte-activation gene 3 (LAG3), an immune checkpoint receptor, has also been proposed to play a role in extracellular α-syn internalization; however, a recent study has disputed this role. Internalized α-syn can trigger expression and secretion of inflammatory cytokines such as tumor necrosis factor alpha (TNF-α), interleukin (IL)-1β, IL-2, and IL-6 and induce neuroinflammation, apoptosis, and mitophagy that results in cellular death. In this study, we tested if N-acetylcysteine (NAC), an anti-inflammatory and anti-carcinogenic drug, can circumvent the detrimental effects of neuroinflammation and induce an anti-inflammatory response by modulating transcription and expression of TLR2 and LAG3 receptors. Cells overexpressing wild-type α-syn were treated with TNF-α to induce inflammation followed by NAC to inhibit the deleterious effects of TNF-α-induced inflammation and apoptosis. SNCA gene transcription and α-syn protein expression were validated by q-PCR and Western blot (WB), respectively. Cell viability was measured, and apoptosis was evaluated by WB and terminal deoxynucleotidyl transferase nick end labeling methods. Alterations in LAG3 and TLR2 receptor levels were evaluated by immunofluorescent labeling, WB, and q-PCR. TNF-α not only increased inflammation but also increased endogenous and overexpressed α-syn levels. NAC treatment decreased expression of TLR2 and increased transcription of LAG3 receptor and diminished inflammation-mediated toxicity and cell death. Here, we demonstrate that NAC can reduce neuroinflammation that occurs as a result of alpha-synuclein overexpression, via a TLR2-associated pathway, making it a promising candidate for therapeutic intervention. Further studies are needed to elucidate molecular mechanisms and pathways related to neuroinflammation in PD and to develop possible new therapeutic approaches to slow the clinical progression of PD.
Collapse
Affiliation(s)
- Zeynep Bengisu Kaya
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Department of Histology and EmbryologyHacettepe University Faculty of MedicineAnkaraTurkey
| | - Elif Karakoc
- Department of Histology and EmbryologyHacettepe University Faculty of MedicineAnkaraTurkey
| | | | - Esen Saka
- Department of NeurologyHacettepe University Faculty of MedicineAnkaraTurkey
| | - Pergin Atilla
- Department of Histology and EmbryologyHacettepe University Faculty of MedicineAnkaraTurkey
| |
Collapse
|
5
|
Ravenhill SM, Evans AH, Crewther SG. Escalating Bi-Directional Feedback Loops between Proinflammatory Microglia and Mitochondria in Ageing and Post-Diagnosis of Parkinson's Disease. Antioxidants (Basel) 2023; 12:antiox12051117. [PMID: 37237983 DOI: 10.3390/antiox12051117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/12/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Parkinson's disease (PD) is a chronic and progressive age-related neurodegenerative disease affecting up to 3% of the global population over 65 years of age. Currently, the underlying physiological aetiology of PD is unknown. However, the diagnosed disorder shares many common non-motor symptoms associated with ageing-related neurodegenerative disease progression, such as neuroinflammation, microglial activation, neuronal mitochondrial impairment, and chronic autonomic nervous system dysfunction. Clinical PD has been linked to many interrelated biological and molecular processes, such as escalating proinflammatory immune responses, mitochondrial impairment, lower adenosine triphosphate (ATP) availability, increasing release of neurotoxic reactive oxygen species (ROS), impaired blood brain barrier integrity, chronic activation of microglia, and damage to dopaminergic neurons consistently associated with motor and cognitive decline. Prodromal PD has also been associated with orthostatic hypotension and many other age-related impairments, such as sleep disruption, impaired gut microbiome, and constipation. Thus, this review aimed to present evidence linking mitochondrial dysfunction, including elevated oxidative stress, ROS, and impaired cellular energy production, with the overactivation and escalation of a microglial-mediated proinflammatory immune response as naturally occurring and damaging interlinked bidirectional and self-perpetuating cycles that share common pathological processes in ageing and PD. We propose that both chronic inflammation, microglial activation, and neuronal mitochondrial impairment should be considered as concurrently influencing each other along a continuum rather than as separate and isolated linear metabolic events that affect specific aspects of neural processing and brain function.
Collapse
Affiliation(s)
| | - Andrew Howard Evans
- Department of Medicine, The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia
- Epworth Hospital, Richmond 3121, Australia
- Department of Neurology, Royal Melbourne Hospital, Melbourne 3050, Australia
| | | |
Collapse
|
6
|
Xu L, Hao LP, Yu J, Cheng SY, Li F, Ding SM, Zhang R. Curcumin protects against rotenone-induced Parkinson's disease in mice by inhibiting microglial NLRP3 inflammasome activation and alleviating mitochondrial dysfunction. Heliyon 2023; 9:e16195. [PMID: 37234646 PMCID: PMC10208821 DOI: 10.1016/j.heliyon.2023.e16195] [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: 11/17/2022] [Revised: 04/12/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder worldwide. Currently, treatment options can only relieve symptoms but cannot prevent, slow, or halt the neurodegenerative process of PD. Much evidence has suggested that microglia-mediated neuroinflammation is involved in the pathophysiology of PD. As an anti-inflammatory agent, curcumin may exert a neuroprotective effect on PD. However, its mechanism has yet to be demonstrated clearly. Our results indicated that curcumin alleviated rotenone-induced behavioral defects, dopamine neuron loss, and microglial activation. Besides, the NF-κB signaling pathway, the NLRP3 inflammasome, and pro-inflammatory cytokines, including IL-18 and IL-1β, contributed to the microglia-mediated neuroinflammation in PD. Furthermore, Drp1-mediated mitochondrial fission causing mitochondrial dysfunction also had an etiological role in the process. This study suggests that curcumin protects against rotenone-induced PD by inhibiting microglial NLRP3 inflammasome activation and alleviating mitochondrial dysfunction in mice. Thus, curcumin may be a neuroprotective drug with promising prospects in PD.
Collapse
|
7
|
de Siqueira EA, Magalhães EP, de Assis ALC, Sampaio TL, Lima DB, Marinho MM, Martins AMC, de Andrade GM, de Barros Viana GS. 1α,25-Dihydroxyvitamin D3 (VD3) Shows a Neuroprotective Action Against Rotenone Toxicity on PC12 Cells: An In Vitro Model of Parkinson's Disease. Neurochem Res 2023; 48:250-262. [PMID: 36066698 DOI: 10.1007/s11064-022-03735-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 08/10/2022] [Accepted: 08/22/2022] [Indexed: 01/11/2023]
Abstract
Parkinson's disease (PD) is characterized by dopaminergic cell loss in the substantia nigra, and PD brains show neuroinflammation, oxidative stress, and mitochondrial dysfunction. The study evaluated the neuroprotective activity of 1α,25-dihydroxy vitamin D3 (VD3), on the rotenone (ROT)-induced cytotoxicity in PC12 cells. The viability parameters were assessed by the MTT and flow cytometry, on cells treated or not with VD3 and/or ROT. Besides, ROS production, cell death, mitochondrial transmembrane potential, reduced GSH, superoxide accumulation, molecular docking (TH and Keap1-Nrf2), and TH, Nrf2, NF-kB, and VD3 receptor protein contents by western blot were evaluated. VD3 was shown to improve the viability of ROT-exposed cells. Cells exposed to ROT showed increased production of ROS and superoxide, which decreased after VD3. ROT decrease in the mitochondrial transmembrane potential was prevented, after VD3 treatment and, VD3 was shown to interact with tyrosine hydroxylase (TH) and Nrf2. While ROT decreased TH, Nrf2, and NF-kB expressions, these effects were reversed by VD3. In addition, VD3 also increased VD3 receptor protein contents and values went back to those of controls after ROT exposure. VD3 protects PC12 cells against ROT damage, by decreasing oxidative stress and improving mitochondrial function. One target seems to be the TH molecule and possibly an indirect Nrf2 activation could also justify its neuroprotective actions on this PC12 cell model of PD.
Collapse
Affiliation(s)
- Erlânia Alves de Siqueira
- Department of Physiology and Pharmacology, Federal University of Ceará (UFC), Fortaleza, Ceará, Brazil
| | - Emanuel Paula Magalhães
- Department of Clinical and Toxicological Analysis, Federal University of Ceará (UFC), Fortaleza, Ceará, Brazil
| | | | - Tiago Lima Sampaio
- Department of Clinical and Toxicological Analysis, Federal University of Ceará (UFC), Fortaleza, Ceará, Brazil
| | - Danya Bandeira Lima
- Department of Clinical and Toxicological Analysis, Federal University of Ceará (UFC), Fortaleza, Ceará, Brazil
| | - Marcia Machado Marinho
- Department of Clinical and Toxicological Analysis, Federal University of Ceará (UFC), Fortaleza, Ceará, Brazil
| | - Alice Maria Costa Martins
- Department of Clinical and Toxicological Analysis, Federal University of Ceará (UFC), Fortaleza, Ceará, Brazil
| | - Geanne Matos de Andrade
- Department of Physiology and Pharmacology, Federal University of Ceará (UFC), Fortaleza, Ceará, Brazil
| | | |
Collapse
|
8
|
Zhang L, Li Q. Neuroprotective effects of tanshinone IIA in experimental model of Parkinson disease in rats. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
9
|
Leggio L, L'Episcopo F, Magrì A, Ulloa‐Navas MJ, Paternò G, Vivarelli S, Bastos CAP, Tirolo C, Testa N, Caniglia S, Risiglione P, Pappalardo F, Serra A, García‐Tárraga P, Faria N, Powell JJ, Peruzzotti‐Jametti L, Pluchino S, García‐Verdugo JM, Messina A, Marchetti B, Iraci N. Small Extracellular Vesicles Secreted by Nigrostriatal Astrocytes Rescue Cell Death and Preserve Mitochondrial Function in Parkinson's Disease. Adv Healthc Mater 2022; 11:e2201203. [PMID: 35856921 PMCID: PMC11468249 DOI: 10.1002/adhm.202201203] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/05/2022] [Indexed: 01/28/2023]
Abstract
Extracellular vesicles (EVs) are emerging as powerful players in cell-to-cell communication both in healthy and diseased brain. In Parkinson's disease (PD)-characterized by selective dopaminergic neuron death in ventral midbrain (VMB) and degeneration of their terminals in striatum (STR)-astrocytes exert dual harmful/protective functions, with mechanisms not fully elucidated. Here, this study shows that astrocytes from the VMB-, STR-, and VMB/STR-depleted brains release a population of small EVs in a region-specific manner. Interestingly, VMB-astrocytes secreted the highest rate of EVs, which is further exclusively increased in response to CCL3, a chemokine that promotes robust dopaminergic neuroprotection in different PD models. The neuroprotective potential of nigrostriatal astrocyte-EVs is investigated in differentiated versus undifferentiated SH-SY5Y cells exposed to oxidative stress and mitochondrial toxicity. EVs from both VMB- and STR-astrocytes counteract H2 O2 -induced caspase-3 activation specifically in differentiated cells, with EVs from CCL3-treated astrocytes showing a higher protective effect. High resolution respirometry further reveals that nigrostriatal astrocyte-EVs rescue neuronal mitochondrial complex I function impaired by the neurotoxin MPP+ . Notably, only EVs from VMB-astrocyte fully restore ATP production, again specifically in differentiated SH-SY5Y. These results highlight a regional diversity in the nigrostriatal system for the secretion and activities of astrocyte-EVs, with neuroprotective implications for PD.
Collapse
Affiliation(s)
- Loredana Leggio
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
| | | | - Andrea Magrì
- Department of Biological, Geological and Environmental SciencesUniversity of CataniaCatania95125Italy
| | - María José Ulloa‐Navas
- Laboratory of Compared NeurobiologyUniversity of Valencia‐CIBERNEDPaterna46980Spain
- Department of NeuroscienceMayo ClinicJacksonvilleFL32257USA
| | - Greta Paternò
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
| | - Silvia Vivarelli
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
| | | | | | | | | | - Pierpaolo Risiglione
- Department of Biological, Geological and Environmental SciencesUniversity of CataniaCatania95125Italy
| | - Fabrizio Pappalardo
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
| | | | | | - Nuno Faria
- Department of Veterinary MedicineUniversity of CambridgeCambridgeCB3 0ESUK
| | - Jonathan J. Powell
- Department of Veterinary MedicineUniversity of CambridgeCambridgeCB3 0ESUK
| | | | - Stefano Pluchino
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
| | | | - Angela Messina
- Department of Biological, Geological and Environmental SciencesUniversity of CataniaCatania95125Italy
| | - Bianca Marchetti
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
- Oasi Research Institute‐IRCCSTroina94018Italy
| | - Nunzio Iraci
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
| |
Collapse
|
10
|
Transcriptomics analysis of human iPSC-derived dopaminergic neurons reveals a novel model for sporadic Parkinson's disease. Mol Psychiatry 2022; 27:4355-4367. [PMID: 35725899 DOI: 10.1038/s41380-022-01663-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/31/2022] [Accepted: 06/07/2022] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a progressive, neurodegenerative disease affecting over 1% of the population beyond 65 years of age. Although some PD cases are inheritable, the majority of PD cases occur in a sporadic manner. Risk factors comprise next to heredity, age, and gender also exposure to neurotoxins from for instance pesticides and herbicides. As PD is characterized by a loss of dopaminergic neurons in the substantia nigra, it is nearly impossible to access and extract these cells from patients for investigating disease mechanisms. The emergence of induced pluripotent stem (iPSC) technology allows differentiating and growing human dopaminergic neurons, which can be used for in vitro disease modeling. Here, we differentiated human iPSCs into dopaminergic neurons, and subsequently exposed the cells to increasing concentrations of the neurotoxin MPP+. Temporal transcriptomics analysis revealed a strong time- and dose-dependent response with genes over-represented across pathways involved in PD etiology such as "Parkinson's Disease", "Dopaminergic signaling" and "calcium signaling". Moreover, we validated this disease model by showing robust overlap with a meta-analysis of transcriptomics data from substantia nigra from post-mortem PD patients. The overlap included genes linked to e.g. mitochondrial dysfunction, neuron differentiation, apoptosis and inflammation. Our data shows, that MPP+-induced, human iPSC-derived dopaminergic neurons present molecular perturbations as observed in the etiology of PD. Therefore we propose iPSC-derived dopaminergic neurons as a foundation for a novel sporadic PD model to study the pathomolecular mechanisms of PD, but also to screen for novel anti-PD drugs and to develop and test new treatment strategies.
Collapse
|
11
|
Liu CZ, Guo DS, Ma JJ, Dong LR, Chang QQ, Yang HQ, Liang KK, Li XH, Yang DW, Fan YY, Gu Q, Chen SY, Li DS. Correlation of matrix metalloproteinase 3 and matrix metalloproteinase 9 levels with non-motor symptoms in patients with Parkinson’s disease. Front Aging Neurosci 2022; 14:889257. [PMID: 36072482 PMCID: PMC9444063 DOI: 10.3389/fnagi.2022.889257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Matrix metalloproteinases (MMPs) are essential for tissue formation, neuronal network remodeling, and blood–brain barrier integrity. MMPs have been widely studied in acute brain diseases. However, the relationship with Parkinson’s disease (PD) remains unclear. The purpose of this study was to evaluate the serum MMP3 and MMP9 levels of PD patients and analyze their correlation with non-motor symptoms. Methods In this cross-sectional study, we recruited 73 patients with idiopathic PD and 64 healthy volunteers. Serum MMP3 and MMP9 levels were measured by enzyme-linked immunosorbent assay (ELISA). Patients with PD were assessed for non-motor symptoms using the Non-motor Symptoms Scale (NMSS) and Parkinson’s disease sleep scale (PDSS) and Mini Mental State Examination (MMSE). Results Serum MMP3 levels were significantly decreased in PD patients, predominantly those with early-stage PD, compared with controls [12.56 (9.30, 17.44) vs. 15.37 (11.33, 24.41) ng/ml; P = 0.004], and the serum MMP9 levels of PD patients were significantly higher than those of healthy controls [522 (419, 729) vs. 329 (229, 473) ng/ml; P < 0.001]. MMP3 levels were positively correlated with the NMSS total score (r = 0.271, P = 0.020) and the single-item scores for item six, assessing the gastrointestinal tract (r = 0.333, P = 0.004), and there was an inverse correlation between serum MMP3 levels and PDSS score (r = –0.246, P = 0.036); meanwhile, MMP9 levels were positively correlated with the NMSS total score (r = 0.234, P = 0.047), and higher serum MMP9 levels were detected in the cognitive dysfunction subgroup than in the cognitively intact subgroup [658 (504, 877) vs. 502 (397, 608) ng/ml, P = 0.008]. Conclusion The serum MMP3 level of PD patients (especially early-stage patients) was significantly lower than that of the healthy control group, and the MMP9 level was significantly higher than that of the healthy control group. MMP3 and MMP9 levels correlate with sleep disturbance and cognitive function in PD patients, respectively.
Collapse
Affiliation(s)
- Chuan Ze Liu
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Da Shuai Guo
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Jian Jun Ma
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
- *Correspondence: Jian Jun Ma,
| | - Lin Rui Dong
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Qing Qing Chang
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Hong Qi Yang
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Ke Ke Liang
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xiao Huan Li
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Da Wei Yang
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Yong Yan Fan
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Qi Gu
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Si Yuan Chen
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Dong Sheng Li
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| |
Collapse
|
12
|
Yang XX, Yang R, Zhang F. Role of Nrf2 in Parkinson's Disease: Toward New Perspectives. Front Pharmacol 2022; 13:919233. [PMID: 35814229 PMCID: PMC9263373 DOI: 10.3389/fphar.2022.919233] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/12/2022] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is one of the most common and chronic degenerative diseases in the central nervous system. The main pathology of PD formation is the progressive loss of dopaminergic neurons in substantia nigra and the formation of α-synuclein-rich Lewy bodies. The pathogenesis of PD is not caused by any single independent factor. The diversity of these independent factors of PD, such as iron accumulation, oxidative stress, neuroinflammation, mitochondrial dysfunction, age, environment, and heredity, makes the research progress of PD slow. Nrf2 has been well-known to be closely associated with the pathogenesis of PD and could regulate these induced factors development. Nrf2 activation could protect dopaminergic neurons and slow down the progression of PD. This review summarized the role of Nrf2 pathway on the pathogenesis of PD. Regulation of Nrf2 pathway might be one of the promising strategies to prevent and treat PD.
Collapse
Affiliation(s)
- Xin-xing Yang
- Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Rong Yang
- Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Feng Zhang
- Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| |
Collapse
|
13
|
The Anti-Parkinson Potential of Gingko biloba-Supplement Mitigates Cortico-Cerebellar Degeneration and Neuropathobiological Alterations via Inflammatory and Apoptotic Mediators in Mice. Neurochem Res 2022; 47:2211-2229. [DOI: 10.1007/s11064-022-03600-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/28/2022] [Accepted: 04/05/2022] [Indexed: 10/18/2022]
|
14
|
Ayaz M, Anwar F, Saleem U, Shahzadi I, Ahmad B, Mir A, Ismail T. Parkinsonism Attenuation by Antihistamines via Downregulating the Oxidative Stress, Histamine, and Inflammation. ACS OMEGA 2022; 7:14772-14783. [PMID: 35557705 PMCID: PMC9088957 DOI: 10.1021/acsomega.2c00145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/07/2022] [Indexed: 05/17/2023]
Abstract
Parkinson disease (PD) is a neurodegenerative disorder of the motor activity of the brain, regulated by dopaminergic neurons of substantia nigra, resulting in an increased density of histaminergic fibers. This study was aimed to evaluate the effects of H1 antagonist's ebastine and levocetirizine in PD per se and in combination. Animals were divided into 9 groups (n = 10). Group 1 received carboxymethyl cellulose CMC (1 mL/kg). Group II was treated with haloperidol (1 mg/kg) (diseased group). Group III was treated with levodopa/carbidopa (levo 20 mg/kg). Groups IV and V were treated with ebastine at dose levels of 2 and 4 mg/kg, respectively. Groups VI and VII were treated with levocetirizine at dose levels of 0.5 and 1 mg/kg, respectively. Group VIII was treated with ebastine (4 mg/kg) + levo (20 mg/kg) in combination. Group IX was treated with levocetirizine (1 mg/kg) + levo (20 mg/kg). PD was induced with haloperidol (1 mg/kg iv, once daily for 23 days) for a duration of 30 min. Behavioral tests like rotarod, block and triple horizontal bars, actophotometer, and open field were performed. Biochemical markers of oxidative stress, i.e., SOD, CAT, GSH, MDA, dopamine, serotonin, and nor-adrenaline and nitrite, were determined. Histamine, mRNA expression of α-synuclein, and TNF-α level in the serum and brain of mice were analyzed. Endogenous biochemical markers were increased except mRNA expression of α-synuclein, which was reduced. In combination therapy with the standard drug, ebastine (4 mg/kg) significantly improved the cataleptic state and dopamine levels, but no significant difference in the renal and liver functioning tests was observed. This study concluded that ebastine (4 mg/kg) might work in the treatment of PD as it improves the cataleptic state in haloperidol-induced catalepsy.
Collapse
Affiliation(s)
- Maira Ayaz
- Riphah
Institute of Pharmaceutical Sciences, Riphah
International University, Lahore Campus, Lahore 54000, Pakistan
| | - Fareeha Anwar
- Riphah
Institute of Pharmaceutical Sciences, Riphah
International University, Lahore Campus, Lahore 54000, Pakistan
- . Tel: +92-3338883251
| | - Uzma Saleem
- Faculty
of Pharmaceutical Sciences, Government College
University, Faisalabad 38000, Pakistan
| | - Irum Shahzadi
- Department
of Biotechnology, COMSAT University, Abbottabad 22060, Pakistan
| | - Bashir Ahmad
- Riphah
Institute of Pharmaceutical Sciences, Riphah
International University, Lahore Campus, Lahore 54000, Pakistan
| | - Ali Mir
- Riphah
Institute of Pharmaceutical Sciences, Riphah
International University, Lahore Campus, Lahore 54000, Pakistan
| | - Tariq Ismail
- Department
of Pharmacy, COMSAT University, Abbottabad 22060, Pakistan
| |
Collapse
|
15
|
Sun Z, Song L, Li J. Knockdown of small nucleolar RNA host gene 10 (SNHG10) alleviates the injury of human neuroblastoma cells via the miR-1277-5p/insulin substrate receptor 2 axis. Bioengineered 2022; 13:709-720. [PMID: 34967697 PMCID: PMC8805890 DOI: 10.1080/21655979.2021.2012623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/25/2021] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease is a common neurodegenerative disease with a complex physio-pathology. So far, there is no effective medical strategies to prevent the progression of Parkinson's disease. Understanding the mechanisms underlying the progression of Parkinson's disease could provide insights into the formulation of novel preventative or treatment strategies. Small nucleolar RNA host gene 10 (SNHG10) is a lncRNA which has been implicated in the development of many cancers. However, its potential role in Parkinson's disease remains unknown. In this study, we found that SNHG10 was upregulated while miR-1277-5p was downregulated in the Parkinson's disease cell model of 1-Methyl-4-phenyl-pyridine ion (MPP+) induced SH-SY5Y cells. We further revealed that SNHG10 sponged miR-1277-5p to negatively regulate its expression, and miR-1277-5p could bind to the 3'UTR of insulin substrate receptor 2 (IRS2) mRNA to suppress its expression. These data suggest that SNHG10 regulates IRS2 through interacting with miR-1277-5p in the cell model of Parkinson's disease. Through a series of molecular experiments and functional assays, we demonstrated that downregulating SNHG10 in the cell model of Parkinson's disease attenuated the cell injury by reducing the expression of IRS2. Meanwhile, inhibiting miR-1277-5p or overexpressing IRS2 could partially reverse the effect of SNHG10 knockdown. In summary, our data indicate that knockdown of SNHG10 mitigates MPP+ induced damage in SH-SY5Y cells via the miR-1277-5p/IRS2 axis.
Collapse
Affiliation(s)
- Zhaoming Sun
- Department of Neurology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China
| | - Lixiang Song
- Department of Neurology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China
| | - Jiazhen Li
- Department of Neurology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China
| |
Collapse
|
16
|
Ahmad MH, Fatima M, Ali M, Rizvi MA, Mondal AC. Naringenin alleviates paraquat-induced dopaminergic neuronal loss in SH-SY5Y cells and a rat model of Parkinson's disease. Neuropharmacology 2021; 201:108831. [PMID: 34655599 DOI: 10.1016/j.neuropharm.2021.108831] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 09/14/2021] [Accepted: 10/06/2021] [Indexed: 10/20/2022]
Abstract
Parkinson's disease (PD), a common neurodegenerative disease is characterized by the progressive loss of dopaminergic neurons in the substantia nigra. The cause of dopaminergic loss in PD remains unknown for a long time, however, recent reports suggest oxidative stress plays a key role in the pathogenesis of PD. Paraquat (PQ), a widely used herbicide is an oxidative stress inducer that has been implicated as a potential risk factor for the development of PD. Flavonoids are naturally occurring polyphenolic compounds that display a variety of therapeutic properties against oxidative stress. Naringenin (NAR), a natural flavonoid, exhibits neuroprotection against PD-related pathology. However, studies on its neuroprotective role and the underlying mechanisms are scarce, therefore the present study explored the potential neuroprotective role of NAR in PQ-induced parkinsonism in SH-SY5Y cells and rat model. The effect of NAR on PQ-induced cellular toxicity was determined by measuring cell viability, oxidative stress, ATP levels and the same effect was determined by assessing behavioral, biochemical, immunohistochemical, qRT-PCR and Western blot in rat model. NAR treatment in SH-SY5Y cells resulted in increased cell viability, reduced oxidative stress, elevated mitochondrial membrane potential, and higher cellular ATP levels. In rats, NAR treatment resulted in significant neuroprotection against PQ-induced behavioral deficits, oxidative stress, mitochondrial dysfunction, and astrocytosis. NAR treatment significantly modulated PQ-induced mRNA expressions of DRD2, DAT, LRRK2, SNCA, β-catenin, caspase-3, BDNF genes. NAR treatment increased TH protein expression and modulated its immunoreactivity in rat striatum. Also, GFAP decreased in response to NAR treatment. So, in the present study, NAR exhibits neuroprotection against PQ-induced neurotoxicity and neurodegeneration indicating its novel therapeutic potential against PD.
Collapse
Affiliation(s)
- Mir Hilal Ahmad
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India; Genome Biology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Mahino Fatima
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Mansoor Ali
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Moshahid Alam Rizvi
- Genome Biology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Amal Chandra Mondal
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
17
|
Ramos-Martínez IE, Rodríguez MC, Cerbón M, Ramos-Martínez JC, Ramos-Martínez EG. Role of the Cholinergic Anti-Inflammatory Reflex in Central Nervous System Diseases. Int J Mol Sci 2021; 22:ijms222413427. [PMID: 34948222 PMCID: PMC8705572 DOI: 10.3390/ijms222413427] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/30/2021] [Accepted: 12/07/2021] [Indexed: 12/12/2022] Open
Abstract
In several central nervous system diseases, it has been reported that inflammation may be related to the etiologic process, therefore, therapeutic strategies are being implemented to control inflammation. As the nervous system and the immune system maintain close bidirectional communication in physiological and pathological conditions, the modulation of inflammation through the cholinergic anti-inflammatory reflex has been proposed. In this review, we summarized the evidence supporting chemical stimulation with cholinergic agonists and vagus nerve stimulation as therapeutic strategies in the treatment of various central nervous system pathologies, and their effect on inflammation.
Collapse
Affiliation(s)
- Ivan Emmanuel Ramos-Martínez
- Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), Université Paris Est Créteil (UPEC), 94010 Créteil, France;
| | - María Carmen Rodríguez
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, SSA, Morelos 62100, Mexico;
| | - Marco Cerbón
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
- Correspondence: (M.C.); (E.G.R.-M.)
| | - Juan Carlos Ramos-Martínez
- Cardiology Department, Hospital General Regional Lic. Ignacio Garcia Tellez IMSS, Yucatán 97150, Mexico;
| | - Edgar Gustavo Ramos-Martínez
- Escuela de Ciencias, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca 68120, Mexico
- Instituto de Cómputo Aplicado en Ciencias, Oaxaca 68044, Mexico
- Correspondence: (M.C.); (E.G.R.-M.)
| |
Collapse
|
18
|
Giovannini D, Andreola F, Spitalieri P, Krasnowska EK, Colini Baldeschi A, Rossi S, Sangiuolo F, Cozzolino M, Serafino A. Natriuretic peptides are neuroprotective on in vitro models of PD and promote dopaminergic differentiation of hiPSCs-derived neurons via the Wnt/β-catenin signaling. Cell Death Discov 2021; 7:330. [PMID: 34725335 PMCID: PMC8560781 DOI: 10.1038/s41420-021-00723-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/12/2021] [Accepted: 10/15/2021] [Indexed: 12/16/2022] Open
Abstract
Over the last 20 years, the efforts to develop new therapies for Parkinson's disease (PD) have focused not only on the improvement of symptomatic therapy for motor and non-motor symptoms but also on the discovering of the potential causes of PD, in order to develop disease-modifying treatments. The emerging role of dysregulation of the Wnt/β-catenin signaling in the onset and progression of PD, as well as of other neurodegenerative diseases (NDs), renders the targeting of this signaling an attractive therapeutic opportunity for curing this brain disorder. The natriuretic peptides (NPs) atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and C-type natriuretic peptide (CNP), are cardiac and vascular-derived hormones also widely expressed in mammalian CNS, where they seem to participate in numerous brain functions including neural development/differentiation and neuroprotection. We recently demonstrated that ANP affects the Wnt/β-catenin pathway possibly through a Frizzled receptor-mediated mechanism and that it acts as a neuroprotective agent in in vitro models of PD by upregulating this signaling. Here we provide further evidence supporting the therapeutic potential of this class of natriuretic hormones. Specifically, we demonstrate that all the three natriuretic peptides are neuroprotective for SHSY5Y cells and primary cultures of DA neurons from mouse brain, subjected to neurotoxin insult with 6-hydroxydopamine (6-OHDA) for mimicking the neurodegeneration of PD, and these effects are associated with the activation of the Wnt/β-catenin pathway. Moreover, ANP, BNP, CNP are able to improve and accelerate the dopaminergic differentiation and maturation of hiPSCs-derived neural population obtained from two differed healthy donors, concomitantly affecting the canonical Wnt signaling. Our results support the relevance of exogenous ANP, BNP, and CNP as attractive molecules for both neuroprotection and neurorepair in PD, and more in general, in NDs for which aberrant Wnt signaling seems to be the leading pathogenetic mechanism.
Collapse
Affiliation(s)
- Daniela Giovannini
- Institute of Translational Pharmacology-National Research Council of Italy, Rome, Italy
| | - Federica Andreola
- Institute of Translational Pharmacology-National Research Council of Italy, Rome, Italy
| | - Paola Spitalieri
- Department of Biomedicine and Prevention, Genetic Medicine Unit, University of Rome "Tor Vergata", Rome, Italy
| | | | | | - Simona Rossi
- Institute of Translational Pharmacology-National Research Council of Italy, Rome, Italy
| | - Federica Sangiuolo
- Department of Biomedicine and Prevention, Genetic Medicine Unit, University of Rome "Tor Vergata", Rome, Italy
| | - Mauro Cozzolino
- Institute of Translational Pharmacology-National Research Council of Italy, Rome, Italy
| | - Annalucia Serafino
- Institute of Translational Pharmacology-National Research Council of Italy, Rome, Italy.
| |
Collapse
|
19
|
Song Q, Peng S, Zhu X. Baicalein protects against MPP +/MPTP-induced neurotoxicity by ameliorating oxidative stress in SH-SY5Y cells and mouse model of Parkinson's disease. Neurotoxicology 2021; 87:188-194. [PMID: 34666128 DOI: 10.1016/j.neuro.2021.10.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 01/02/2023]
Abstract
Baicalein, a major bioactive flavone constituent isolated from Scutellaria baicalensis Georgi, has neuroprotective properties in several neurological disorders. Many studies suggest that oxidative stress plays a central role in the pathogenesis of Parkinson's disease (PD). Baicalein has also been shown to have antioxidant effects. Therefore, the current study was designed to investigate whether baicalein could protect against MPP+/MPTP-induced neurotoxicity via suppressing oxidative stress in vitro and in vivo. In vitro, our results showed that baicalein increased cell viability in MPP+-treated SH-SY5Y cells. Treatment with baicalein could reversed the increased MDA and ROS levels, and the decreased GSH levels in MPP+-treated SH-SY5Y cells. In MPTP-treated mice, baicalein ameliorated MPTP-induced motor impairment and suppressed the MPTP-induced accumulation of iron and lipid peroxides. Besides, baicalein improved the neurotoxicity induced by MPTP as seen by a significant raise of tyrosine hydroxylase (TH) and simultaneous decrease of monoamine-oxidase-B (MAO-B). The inhibitory effect of baicalein on oxidative stress probably was partially governed by inhibition of ERK activation. In conclusion, our results suggest that baicalein could prevent MPP+/MPTP-induced neurotoxicity via suppressing oxidative stress.
Collapse
Affiliation(s)
- Qingxin Song
- Department of Infection Management, Linyi People's Hospital, Shandong University, Linyi, Shandong 276003, China; Department of Neurosurgery, Linyi People's Hospital, Shandong University, Linyi, Shandong 276003, China
| | - Shanxin Peng
- Department of Infection Management, Linyi People's Hospital, Shandong University, Linyi, Shandong 276003, China; Central Laboratory, Linyi People's Hospital, Shandong University, Linyi, Shandong 276003, China
| | - Xiaosong Zhu
- Department of Infection Management, Linyi People's Hospital, Shandong University, Linyi, Shandong 276003, China; Central Laboratory, Linyi People's Hospital, Shandong University, Linyi, Shandong 276003, China.
| |
Collapse
|
20
|
Zhou S, Yu X, Wang M, Meng Y, Song D, Yang H, Wang D, Bi J, Xu S. Long Non-coding RNAs in Pathogenesis of Neurodegenerative Diseases. Front Cell Dev Biol 2021; 9:719247. [PMID: 34527672 PMCID: PMC8435612 DOI: 10.3389/fcell.2021.719247] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/11/2021] [Indexed: 12/19/2022] Open
Abstract
Emerging evidence addresses the link between the aberrant epigenetic regulation of gene expression and numerous diseases including neurological disorders, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington’s disease (HD). LncRNAs, a class of ncRNAs, have length of 200 nt or more, some of which crucially regulate a variety of biological processes such as epigenetic-mediated chromatin remodeling, mRNA stability, X-chromosome inactivation and imprinting. Aberrant regulation of the lncRNAs contributes to pathogenesis of many diseases, such as the neurological disorders at the transcriptional and post-transcriptional levels. In this review, we highlight the latest research progress on the contributions of some lncRNAs to the pathogenesis of neurodegenerative diseases via varied mechanisms, such as autophagy regulation, Aβ deposition, neuroinflammation, Tau phosphorylation and α-synuclein aggregation. Meanwhile, we also address the potential challenges on the lncRNAs-mediated epigenetic study to further understand the molecular mechanism of the neurodegenerative diseases.
Collapse
Affiliation(s)
- Shiyue Zhou
- Department of Neurology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiao Yu
- Department of Nutrition, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Wang
- Department of Neurology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yujie Meng
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dandan Song
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hui Yang
- Department of Neurology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dewei Wang
- Department of Neurology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jianzhong Bi
- Department of Neurology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shunliang Xu
- Department of Neurology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
21
|
Genkwanin suppresses MPP +-induced cytotoxicity by inhibiting TLR4/MyD88/NLRP3 inflammasome pathway in a cellular model of Parkinson's disease. Neurotoxicology 2021; 87:62-69. [PMID: 34481870 DOI: 10.1016/j.neuro.2021.08.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/09/2021] [Accepted: 08/31/2021] [Indexed: 11/22/2022]
Abstract
Parkinson's disease (PD) is a complicated multifactorial neurodegenerative disorder. Oxidative stress, neuroinflammatory response, and activation of apoptosis have been proposed to be tightly involved in the pathogenesis of PD. Genkwanin is a typical bioactive non-glycosylated flavonoid with anti-inflammatory and anti-oxidant activities. However, the effect of genkwanin on PD remains unclear. Cell viability, lactate dehydrogenase (LDH) release, caspase-3/7 activity, and apoptosis was evaluated by MTT, LDH release assay, caspase-3/7 activity assay, and TUNEL assay, respectively. The secretion of prostaglandin E2 (PGE2), tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6 were measured by respective commercial ELISA kits. The mRNA expression of TNF-α, IL-1β, and IL-6 was detected by qRT-PCR. The protein levels of cycloxygenase-2 (COX-2), toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), and NOD-like receptor (NLR) protein: 3 (NLRP3) were determined by western blot analysis. Genkwanin at concentrations less than 40 μM had no impact on cell viability and LDH release. Genkwanin suppressed MPP+-induced neuroinflammation in SH-SY5Y cells. MPP+ treatment inhibited cell viability, increased LDH release, apoptosis, and ROS generation, and reduced superoxide dismutase (SOD) activity in SH-SY5Y cells, which were abolished by genkwanin treatment. Genkwanin suppressed MPP+-induced activation of TLR4/MyD88/NLRP3 inflammasome pathway in SH-SY5Y cells. TLR4 overexpression weakened the anti-inflammatory and anti-neurotoxicity of genkwanin in SH-SY5Y cells. In conclusion, genkwanin attenuated neuroinflammation and neurotoxicity by inhibiting TLR4/MyD88/NLRP3 inflammasome pathway in MPP+-induced cellular model of PD.
Collapse
|
22
|
Raj K, Gupta GD, Singh S. l-Theanine ameliorates motor deficit, mitochondrial dysfunction, and neurodegeneration against chronic tramadol induced rats model of Parkinson's disease. Drug Chem Toxicol 2021; 45:2097-2108. [PMID: 34210222 DOI: 10.1080/01480545.2021.1907909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Parkinson's disease (PD) is the second most prevalent progressive neurodegenerative disease, characterized by loss of dopaminergic neurons in substantia nigra, with deficiency of dopamine in the striatum. Tramadol is safe analgesic but long-term use confirmed to elevate oxidative stress, neuroinflammation, mitochondrial dysfunction, in brain leads to motor deficits. l-Theanine is an active constituent of green tea which prevents neuronal loss, mitochondrial failure and improves dopamine, gamma-aminobutyric acid (GABA), serotonin levels and in the central nervous system (CNS) via antioxidant, anti-inflammatory, and neuromodulatory properties. In the present study, tramadol was injected intraperitoneally to Wister rats for 28 days at a dose of 50 mg/kg. l-Theanine (25, 50, and 100 mg/kg) was administered orally 3 h before tramadol administration from day 14 to day 28. Behavioral analyses including rotarod, narrow beam walk, open field, and grip strength were used to evaluate motor coordination on a weekly basis. On the day 29, all Wistar rats were sacrificed and striatum homogenates were used for biochemical (lipid peroxidation, nitrite, glutathione, glutathione peroxidase activity, superoxide dismutase, catalase, mitochondrial complex I, IV, and cyclic adenosine monophosphate), neuroinflammatory markers (tumor necrosis factor-α, interleukin-1β, and interleukin-17), and neurotransmitters (dopamine, norepinephrine, serotonin, GABA, and glutamate) analysis. Chronic tramadol treatment caused motor deficits reduced antioxidant enzymes level, increased striatal proinflammatory cytokines release, dysbalanced neurotransmitters, and reduced mitochondrial complex activity I, IV, and cAMP activity. However, l-theanine administration attenuated behavioral, biochemical, neuroinflammatory, neurotransmitters, and mitochondrial activity indicated it as a promising neuroprotective potential against degenerative changes in experimental model of PD.
Collapse
Affiliation(s)
- Khadga Raj
- Department of Pharmacology, Neuropharmacology Division, ISF College of Pharmacy, Moga, India
| | - G D Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, India
| | - Shamsher Singh
- Department of Pharmacology, Neuropharmacology Division, ISF College of Pharmacy, Moga, India
| |
Collapse
|
23
|
Potential Effects of Leukotriene Receptor Antagonist Montelukast in Treatment of Neuroinflammation in Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22115606. [PMID: 34070609 PMCID: PMC8198163 DOI: 10.3390/ijms22115606] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/16/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder where misfolded alpha-synuclein-enriched aggregates called Lewy bodies are central in pathogenesis. No neuroprotective or disease-modifying treatments are currently available. Parkinson’s disease is considered a multifactorial disease and evidence from multiple patient studies and animal models has shown a significant immune component during the course of the disease, highlighting immunomodulation as a potential treatment strategy. The immune changes occur centrally, involving microglia and astrocytes but also peripherally with changes to the innate and adaptive immune system. Here, we review current understanding of different components of the PD immune response with a particular emphasis on the leukotriene pathway. We will also describe evidence of montelukast, a leukotriene receptor antagonist, as a possible anti-inflammatory treatment for PD.
Collapse
|
24
|
Song A, Yang Y, He H, Sun J, Chang Q, Xue Q. Inhibition of Long Non-Coding RNA KCNQ1OT1 Attenuates Neuroinflammation and Neuronal Apoptosis Through Regulating NLRP3 Expression via Sponging miR-30e-3p. J Inflamm Res 2021; 14:1731-1742. [PMID: 33981152 PMCID: PMC8107009 DOI: 10.2147/jir.s291274] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/14/2021] [Indexed: 11/30/2022] Open
Abstract
Background Neuroinflammation and neuronal apoptosis are considered as the critical factors in the pathogenesis of multiple neurological diseases. Recent studies have shown that long non-coding RNA (lncRNA) plays a crucial part in neuroinflammation and neuronal apoptosis. Methods The expression levels of lncRNA KCNQ1OT1, miR-30e-3p and NLRP3 in lipopolysaccharide (LPS)-induced HMC3 cells were analyzed using RT-qPCR. MTT assay, LDH release assay and ELISA were used to assess the effect of KCNQ1OT1 and miR-30e-3p on neuroinflammation and neuronal apoptosis. The targeted regulatory relationships among KCNQ1OT1, miR-30e-3p and NLRP3 were evaluated by bioinformatics analysis, dual-luciferase reporter gene assay, RT-qPCR and Western blot. Results In LPS-induced HMC3 cells, the expression levels of KCNQ1OT1 and NLRP3 were increased, while the expression level of miR-30e-3p was reduced. Knockdown of KCNQ1OT1 alleviated LPS-induced apoptosis and neuroinflammation of HMC3 cells, accompanied by increased cell viability, low LDH release and reduced cell apoptosis rate, and reduced levels of TNF-α, IL-1β and IL-6. Overexpression of miR-30e-3p had a similar effect. Additionally, KCNQ1OT1 could bind with miR-30e-3p and repress its expression in HMC3 cells, and KCNQ1OT1 overexpression counteracted miR-30e-3p’s inhibitory effect on LPS-induced neuronal damage and inflammatory response in HMC3 cells. Furthermore, KCNQ1OT1 could positively regulate the expression of NLRP3 via repressing miR-30e-3p. Conclusion Inhibition of KCNQ1OT1 could reduce neuroinflammation and neuronal apoptosis induced by LPS in HMC3 cells by regulating miR-30e-3p/NLRP3 pathway, suggesting that KCNQ1OT1 and miR-30e-3p could serve as promising therapeutic targets for treating neurological diseases.
Collapse
Affiliation(s)
- Aixia Song
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, 075000, People's Republic of China
| | - Yuying Yang
- Stroke Office, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, 075000, People's Republic of China
| | - Hongmei He
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, 075000, People's Republic of China
| | - Jian Sun
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, 075000, People's Republic of China
| | - Qing Chang
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, 075000, People's Republic of China
| | - Qian Xue
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, 075000, People's Republic of China
| |
Collapse
|
25
|
Li X, Feng Y, Wang XX, Truong D, Wu YC. The Critical Role of SIRT1 in Parkinson's Disease: Mechanism and Therapeutic Considerations. Aging Dis 2020; 11:1608-1622. [PMID: 33269110 PMCID: PMC7673849 DOI: 10.14336/ad.2020.0216] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 02/16/2020] [Indexed: 12/13/2022] Open
Abstract
Silence information regulator 1 (SIRT1), a member of the sirtuin family, targets histones and many non-histone proteins and participates in various physiological functions. The enzymatic activity of SIRT1 is decreased in patients with Parkinson’s disease (PD), which may reduce their ability to resist neuronal damage caused by various neurotoxins. As far as we know, SIRT1 can induce autophagy by regulating autophagy related proteins such as AMP-activated protein kinase, light chain 3, mammalian target of rapamycin, and forkhead transcription factor 1. Furthermore, SIRT1 can regulate mitochondrial function and inhibit oxidative stress mainly by maintaining peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) in a deacetylated state and thus maintaining a constant level of PGC-1α. Other studies have demonstrated that SIRT1 may play a role in the pathophysiology of PD by regulating neuroinflammation. SIRT1 deacetylases nuclear factor-kappa B and thus reduces its transcriptional activity, inhibits inducible nitric oxide synthase expression, and decreases tumor necrosis factor-alpha and interleukin-6 levels. SIRT1 can also upregulate heat shock protein 70 by deacetylating heat shock factor 1 to increase the degradation of α-synuclein oligomers. Few studies have focused on the relationship between SIRT1 single nucleotide polymorphisms and PD risk, so this topic requires further research. Based on the neuroprotective effects of SIRT1 on PD, many in vitro and in vivo experiments have demonstrated that some SIRT1 activators, notably resveratrol, have potential neuroprotective effects against dopaminergic neuronal damage caused by various neurotoxins. Thus, SIRT1 plays a critical role in PD development and might be a potential target for PD therapy.
Collapse
Affiliation(s)
- Xuan Li
- 1Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Ya Feng
- 1Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xi-Xi Wang
- 1Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Daniel Truong
- 2The Truong Neurosciences Institute, Orange Coast Memorial Medical Center, Fountain Valley, CA, USA.,3Department of Neurosciences and Psychiatry, University of California, Riverside, CA, USA
| | - Yun-Cheng Wu
- 1Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
26
|
Aleo SJ, Del Dotto V, Fogazza M, Maresca A, Lodi T, Goffrini P, Ghelli A, Rugolo M, Carelli V, Baruffini E, Zanna C. Drug repositioning as a therapeutic strategy for neurodegenerations associated with OPA1 mutations. Hum Mol Genet 2020; 29:3631-3645. [PMID: 33231680 PMCID: PMC7823107 DOI: 10.1093/hmg/ddaa244] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/19/2020] [Accepted: 10/30/2020] [Indexed: 12/15/2022] Open
Abstract
OPA1 mutations are the major cause of dominant optic atrophy (DOA) and the syndromic form DOA plus, pathologies for which there is no established cure. We used a ‘drug repurposing’ approach to identify FDA-approved molecules able to rescue the mitochondrial dysfunctions induced by OPA1 mutations. We screened two different chemical libraries by using two yeast strains carrying the mgm1I322M and the chim3P646L mutations, identifying 26 drugs able to rescue their oxidative growth phenotype. Six of them, able to reduce the mitochondrial DNA instability in yeast, have been then tested in Opa1 deleted mouse embryonic fibroblasts expressing the human OPA1 isoform 1 bearing the R445H and D603H mutations. Some of these molecules were able to ameliorate the energetic functions and/or the mitochondrial network morphology, depending on the type of OPA1 mutation. The final validation has been performed in patients’ fibroblasts, allowing to select the most effective molecules. Our current results are instrumental to rapidly translating the findings of this drug repurposing approach into clinical trial for DOA and other neurodegenerations caused by OPA1 mutations.
Collapse
Affiliation(s)
- Serena J Aleo
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna 40126, Italy
| | - Valentina Del Dotto
- Unit of Neurology, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna 40139, Italy
| | - Mario Fogazza
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna 40126, Italy
| | - Alessandra Maresca
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna 40139, Italy
| | - Tiziana Lodi
- Department of Chemistry, Life Science and Environmental Sustainability, University of Parma, Parma 43124, Italy
| | - Paola Goffrini
- Department of Chemistry, Life Science and Environmental Sustainability, University of Parma, Parma 43124, Italy
| | - Anna Ghelli
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna 40126, Italy
| | - Michela Rugolo
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna 40126, Italy
| | - Valerio Carelli
- Unit of Neurology, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna 40139, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna 40139, Italy
| | - Enrico Baruffini
- Department of Chemistry, Life Science and Environmental Sustainability, University of Parma, Parma 43124, Italy
| | - Claudia Zanna
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna 40126, Italy
| |
Collapse
|
27
|
Lavisse S, Goutal S, Wimberley C, Tonietto M, Bottlaender M, Gervais P, Kuhnast B, Peyronneau MA, Barret O, Lagarde J, Sarazin M, Hantraye P, Thiriez C, Remy P. Increased microglial activation in patients with Parkinson disease using [ 18F]-DPA714 TSPO PET imaging. Parkinsonism Relat Disord 2020; 82:29-36. [PMID: 33242662 DOI: 10.1016/j.parkreldis.2020.11.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/18/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Increasing evidence suggests that neuroinflammation is active in Parkinson disease (PD) and contributes to neurodegeneration. This process can be studied in vivo with PET and radioligands targeting TSPO, upregulated in activated microglia. Initial PET studies investigating microglial activation in PD with the [11C]-PK11195 have provided inconclusive results. Here we assess the presence and distribution of neuroinflammatory response in PD patients using [18F]-DPA714 and to correlate imaging biomarkers to dopamine transporter imaging and clinical status. METHODS PD patients (n = 24, Hoehn and Yahr I-III) and 28 healthy controls were scanned with [18F]-DPA714 and [11C]-PE2I and analyzed. They were all genotyped for TSPO polymorphism. Regional binding parameters were estimated (reference Logan graphical approach with supervised cluster analysis). Impact of TSPO genotype was analyzed using Wilcoxon signed-rank test. Differences between groups were investigated using a two-way ANOVA and Tukey post hoc tests. RESULTS PD patients showed significantly higher [18F]-DPA714 binding compared to healthy controls bilaterally in the midbrain (p < 0.001), the frontal cortex (p = 0.001), and the putamen contralateral to the more clinically affected hemibody (p = 0.038). Microglial activation in these regions did not correlate with the severity of motor symptoms, disease duration nor putaminal [11C]-PE2I uptake. However, there was a trend toward a correlation between cortical TSPO binding and disease duration (p = 0.015 uncorrected, p = 0.07 after Bonferroni correction). CONCLUSION [18F]-DPA714 binding confirmed that there is a specific topographic pattern of microglial activation in the nigro-striatal pathway and the frontal cortex of PD patients. TRIAL REGISTRATION Trial registration: INFLAPARK, NCT02319382. Registered 18 December 2014- Retrospectively registered, https://clinicaltrials.gov/ct2/show/NCT02319382.
Collapse
Affiliation(s)
- Sonia Lavisse
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France.
| | - Sébastien Goutal
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France.
| | - Catriona Wimberley
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France.
| | - Mattéo Tonietto
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France.
| | - Michel Bottlaender
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France; Université Paris-Saclay, UNIACT, Neurospin, CEA, 91191, Gif-sur-Yvette, France.
| | - Philippe Gervais
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France.
| | - Bertrand Kuhnast
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France.
| | - Marie-Anne Peyronneau
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France.
| | - Olivier Barret
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France.
| | - Julien Lagarde
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France; Department of Neurology of Memory and Language, GHU Paris Psychiatrie & Neurosciences, Sainte-Anne Hospital, Paris, 75014, France; Université de Paris, F-75006, France.
| | - Marie Sarazin
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France; Department of Neurology of Memory and Language, GHU Paris Psychiatrie & Neurosciences, Sainte-Anne Hospital, Paris, 75014, France; Université de Paris, F-75006, France.
| | - Philippe Hantraye
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France.
| | - Claire Thiriez
- Centre Expert Parkinson, Neurologie, CHU Henri Mondor, AP-HP, 51 Avenue du Maréchal de Lattre de Tassigny, Créteil, France.
| | - Philippe Remy
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France; Centre Expert Parkinson, Neurologie, CHU Henri Mondor, AP-HP, 51 Avenue du Maréchal de Lattre de Tassigny, Créteil, France; IMRB, INSERM, Université Paris Est Créteil and NeurATRIS, France.
| |
Collapse
|
28
|
Farombi EO, Awogbindin IO, Owoeye O, Abah VO, Izomoh ER, Ezekiel IO. Kolaviron ameliorates behavioural deficit and injury to striatal dopaminergic terminals via modulation of oxidative burden, DJ-1 depletion and CD45R + cells infiltration in MPTP-model of Parkinson's disease. Metab Brain Dis 2020; 35:933-946. [PMID: 32430695 DOI: 10.1007/s11011-020-00578-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 04/30/2020] [Indexed: 10/24/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. Currently, the precise pathogenic detail of PD is not entirely clear and first line therapeutics fail to attenuate the progress of the disease. In this study, we examined the neuroprotective effect of kolaviron, a natural antioxidant and anti-inflammatory biflavonoid from Garcinia kola seed, on behavioural impairment, neurodegeneration, oxidative stress and neuroinflammation in an acute MPTP-induced PD model. Kolaviron mitigated the frequently interrupted MPTP-associated hyperkinesia, inefficient gait, immobility, inability to pay attention to sizable holes on walking path, habitual clockwise rotations characterized with minimal diversion of movements and impaired balance. Also, kolaviron suppressed MPTP-mediated striatal oxidative stress, depletion as well as degeneration of dopaminergic terminals, reduced DJ-1 secretion and upregulated expression of caspase-3. Kolaviron facilitated cytoprotective antioxidant response and prevented MPTP-mediated neuroinflammation by blocking striatal infiltration of peripheral CD45R positive cells. Additionally, kolaviron reversed MPTP-induced inhibition of acetylcholinesterase activity. Together, our study provides evidence that the neuroprotective capacity of kolaviron to modulate striatal degeneration, behavioural impairment, antioxidant/redox imbalance and neuroinflammation implicated in the pathogenesis of PD may involve upregulation of DJ-1 secretion and inhibition of CD45R cells infiltration. Our data recommend kolaviron as a possible neuroprotective strategy in the management of Parkinson's disease and the associated behavioural complications, albeit the identity of MPTP-associated striatal CD45R infiltrate needs to be further characterized.
Collapse
Affiliation(s)
- Ebenezer O Farombi
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria.
| | - Ifeoluwa O Awogbindin
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria
| | - Olatunde Owoeye
- Department of Anatomy, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Victoria O Abah
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria
| | - Edirin R Izomoh
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria
| | - Ibukunoluwa O Ezekiel
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
29
|
The Inhibition of miR-873 Provides Therapeutic Benefit in a Lipopolysaccharide-Induced Neuroinflammatory Model of Parkinson's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8735249. [PMID: 32724496 PMCID: PMC7381950 DOI: 10.1155/2020/8735249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/05/2020] [Accepted: 06/11/2020] [Indexed: 11/18/2022]
Abstract
Background and Purpose. Alterations in cholesterol homeostasis have been reported in cell and animal models of Parkinson's disease (PD), although there are inconsistent data about the association between serum cholesterol levels and risk of PD. Here, we investigated the effects of miR-873 on lysosomal cholesterol homeostasis and progressive dopaminergic neuron damage in a lipopolysaccharide-(LPS) induced model of PD. Experimental Approach. To evaluate the therapeutic benefit of the miR-873 sponge, rats were injected with a LV-miR-873 sponge or the control vector 3 days before the right-unilateral injection of LPS into the substantia nigra (SN) pars compacta, or 8 and 16 days after LPS injection. Normal SH-SY5Y cells or SH-SY5Y cells overexpressing α-synuclein were used to evaluate the distribution of α-synuclein and cholesterol in lysosomes and to assess the autophagic flux after miR-873 transfection or ABCA1 silencing. The inhibition of miR-873 significantly ameliorated the LPS-induced accumulation of α-synuclein and loss of dopaminergic neurons in the SN at the early stage. miR-873 mediated the inhibition of ABCA1 by LPS. miR-873 transfection or ABCA1 silencing increased the lysosomal cholesterol and α-synuclein levels, and decreased the autophagic flux. The knockdown of ABCA1 or A20, which are the downstream target genes of miR-873, exacerbated the damage to LPS-induced dopaminergic neurons. Conclusion and Implications. The results suggest that the inhibition of miR-873 may play a dual protective role by improving intracellular cholesterol homeostasis and neuroinflammation in PD. The therapeutic effects of the miR-873 sponge in PD may be due to the upregulation of ABCA1 and A20.
Collapse
|
30
|
Xu W, Zhang L, Geng Y, Liu Y, Zhang N. Long noncoding RNA GAS5 promotes microglial inflammatory response in Parkinson's disease by regulating NLRP3 pathway through sponging miR-223-3p. Int Immunopharmacol 2020; 85:106614. [PMID: 32470877 DOI: 10.1016/j.intimp.2020.106614] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/24/2020] [Accepted: 05/14/2020] [Indexed: 01/01/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder. Neuroinflammation induced by microglia plays an important role in the pathogenesis of PD. Long noncoding RNA GAS5 was showed to have significant effects on regulating inflammatory response. Here, we aim to investigate the effects of GAS5 on the inflammatory response of PD, and the underlying mechanism. An in vivo model of PD was established in C57BL/6 mice by rotenone and an in vitro cell model was conducted on microglia by lipopolysaccharide (LPS). Our results indicated that GAS5 was upregulated in tissues in a mice model of PD and microglia activated by LPS. Gain- and loss- of functional experiments demonstrated that GAS5 promoted the inflammation of microglia in vitro. Besides, the knockdown of GAS5 repressed the PD progression in vivo. Mechanistically, GAS5 positively regulated the NLRP3 expression via competitively sponging miR-223-3p. Overall, our finding illuminates that GAS5 accelerates PD progression through targeting miR-223-3p/NLRP3 axis.
Collapse
Affiliation(s)
- Wei Xu
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China.
| | - Ling Zhang
- College of Health Management, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yu Geng
- College of Health Management, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Ye Liu
- Electrocardial Center, the First Affiliated Hospital of Jinzhou Medical University. Jinzhou, Liaoning, China
| | - Ning Zhang
- Department of Hematology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
31
|
He T, Itano MS, Earley LF, Hall NE, Riddick N, Samulski RJ, Li C. The Influence of Murine Genetic Background in Adeno-Associated Virus Transduction of the Mouse Brain. HUM GENE THER CL DEV 2020; 30:169-181. [PMID: 31749390 DOI: 10.1089/humc.2019.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Adeno-associated virus (AAV) vectors have become an important tool for delivering therapeutic genes for a wide range of neurological diseases. AAV serotypes possess differential cellular tropism in the central nervous system. Although several AAV serotypes or mutants have been reported to transduce the brain efficiently, conflicting data occur across studies with the use of various rodent strains from different genetic backgrounds. Herein, we performed a systematic comparison of the brain transduction properties among five AAV serotypes (AAV2, 5, 7, 8, and 9) in two common rodent strains (C57BL/6J and FVB/N), following local intrastriatal injection of AAV vectors encoding enhanced green fluorescent protein (EGFP) driven by the CBh promoter. Important differences were found regarding overall cellular tropism and transduction efficiency, including contralateral transduction among the AAV serotypes and between the mouse strains. We have further found loss of NeuN-immunoreactivity and microglial activation from AAV transduction in the different mouse strains. The important strain-specific differences from our study suggest that the genetic background of the mouse may affect AAV serotype transduction properties in the brain. These data can provide valuable information about how to choose an effective AAV vector for clinical application and interpret the data obtained from preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Ting He
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Michelle S Itano
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina.,Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Lauriel F Earley
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nikita E Hall
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Natallia Riddick
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
32
|
Baicalin Represses C/EBP β via Its Antioxidative Effect in Parkinson's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8951907. [PMID: 32566108 PMCID: PMC7261332 DOI: 10.1155/2020/8951907] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/19/2020] [Accepted: 05/02/2020] [Indexed: 12/19/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by the gradual loss of dopaminergic (DA) neurons in the substantia nigra (SN) and the formation of intracellular Lewy bodies (LB) in the brain, which aggregates α-synuclein (α-Syn) as the main component. The interest of flavonoids as potential neuroprotective agents is increasing due to its high efficiency and low side effects. Baicalin is one of the flavonoid compounds, which is a predominant flavonoid isolated from Scutellaria baicalensis Georgi. However, the key molecular mechanism by which Baicalin can prevent the PD pathogenesis remains unclear. In this study, we used bioinformatic assessment including Gene Ontology (GO) to elucidate the correlation between oxidative stress and PD pathogenesis. RNA-Seq methods were used to examine the global expression profiles of noncoding RNAs and found that C/EBPβ expression was upregulated in PD patients compared with healthy controls. Interestingly, Baicalin could protect DA neurons against reactive oxygen species (ROS) and decreased C/EBPβ and α-synuclein expression in pLVX-Tet3G-α-synuclein SH-SY5Y cells. In a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induced PD mouse model, the results revealed that treatment with Baicalin improved the PD model's behavioral performance and reduced dopaminergic neuron loss in the substantia nigra, associated with the inactivation of proinflammatory cytokines and oxidative stress. Hence, our study supported that Baicalin repressed C/EBPβ via redox homeostasis, which may be an effective potential treatment for PD.
Collapse
|
33
|
Johnson SL, Iannucci J, Seeram NP, Grammas P. Inhibiting thrombin improves motor function and decreases oxidative stress in the LRRK2 transgenic Drosophila melanogaster model of Parkinson's disease. Biochem Biophys Res Commun 2020; 527:532-538. [PMID: 32423817 DOI: 10.1016/j.bbrc.2020.04.068] [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: 04/06/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disease characterized by the presence of tremors, loss of dopaminergic neurons and accumulation of α-synuclein. While there is no single direct cause of PD, genetic mutations, exposure to pesticides, diet and traumatic brain injury have been identified as risk factors. Increasing evidence suggests that oxidative stress and neuroinflammation contribute to the pathogenesis of neuronal injury in neurodegenerative diseases such as PD and Alzheimer's disease (AD). We have previously documented that the multifunctional inflammatory mediator thrombin contributes to oxidative stress and neuroinflammation in AD. Here, for the first time, we explore the role of thrombin in a transgenic PD model, the LRRK2 mutant Drosophila melanogaster. Transgenic flies were treated with the direct thrombin inhibitor dabigatran for 7 days and locomotor activity and indices of oxidative stress evaluated. Our data show that dabigatran treatment significantly (p < 0.05) improved climbing activity, a measurement of locomotor ability, in male but had no effect on locomotor performance in female flies. Dabigatran treatment had no effect on tyrosine hydroxylase levels. Analysis of oxidative stress in male flies showed that dabigatran was able to significantly (p < 0.01) lower reactive oxygen species levels. Furthermore, Western blot analysis showed that the pro-oxidant proteins iNOS and NOX4 are elevated in LRRK2 male flies compared to wildtype and that treatment with dabigatran reduced expression of these proteins. Our results indicate that dabigatran treatment could improve motor function in PD by reducing oxidative stress. These data suggest that targeting thrombin may improve oxidative stress related pathologies in PD.
Collapse
Affiliation(s)
- Shelby L Johnson
- The George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA; Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA.
| | - Jaclyn Iannucci
- The George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA; Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Navindra P Seeram
- The George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA; Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Paula Grammas
- The George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA; Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| |
Collapse
|
34
|
Song G, Xi G, Li Y, Zhao Y, Qi C, Song L, Xiao B, Ma C. Double triggers, nasal induction of a Parkinson's disease mouse model. Neurosci Lett 2020; 724:134869. [PMID: 32114119 DOI: 10.1016/j.neulet.2020.134869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 02/06/2020] [Accepted: 02/24/2020] [Indexed: 12/29/2022]
Abstract
Animal models of Parkinson's disease (PD), a chronic and progressive neurodegenerative disease of the central nervous system (CNS), play a key role in investigating the pathogenesis and developing new therapeutic strategies of PD. However, this goal has been limited by certain weaknesses in the available animal models of PD, e.g., induction by either pro-inflammatory or neurotoxic reagents, or they are too time-/effort-consuming. Here, we report a double triggers, nasal induction of a PD mouse model that mimics the clinical, pathological features and pathogenesis of PD by intranasal (i.n.) administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) combined with lipopolysaccharide (LPS). After administration once every three days for 7 consecutive weeks, these mice displayed enhanced motor dysfunction, loss of dopaminergic neurons, α-synuclein accumulation, as well as activation of microglia and astrocytes in the substantia nigra pars compacta compared with mice that were administered MPTP or LPS alone. This study provides a novel and basic research tool for investigating the pathogenesis and therapeutic intervention of PD.
Collapse
Affiliation(s)
- Guobin Song
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Shanxi Datong University, Datong, 037009, China
| | - Guoping Xi
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Shanxi Datong University, Datong, 037009, China
| | - Yanhua Li
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Shanxi Datong University, Datong, 037009, China
| | - Yijin Zhao
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Shanxi Datong University, Datong, 037009, China
| | - Caixia Qi
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Shanxi Datong University, Datong, 037009, China
| | - Lijuan Song
- Research Center of Neurobiology, The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Baoguo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200025, China
| | - Cungen Ma
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Shanxi Datong University, Datong, 037009, China; Research Center of Neurobiology, The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, 030619, China.
| |
Collapse
|
35
|
Phytoestrogen Agathisflavone Ameliorates Neuroinflammation-Induced by LPS and IL-1β and Protects Neurons in Cocultures of Glia/Neurons. Biomolecules 2020; 10:biom10040562. [PMID: 32272581 PMCID: PMC7225953 DOI: 10.3390/biom10040562] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 02/27/2020] [Accepted: 02/27/2020] [Indexed: 12/27/2022] Open
Abstract
Inflammation and oxidative stress are common aspects of most neurodegenerative diseases in the central nervous system. In this context, microglia and astrocytes are central to mediating the balance between neuroprotective and neurodestructive mechanisms. Flavonoids have potent anti-inflammatory and antioxidant properties. Here, we have examined the anti-inflammatory and neuroprotective potential of the flavonoid agathisflavone (FAB), which is derived from the Brazilian plant Poincianella pyramidalis, in in vitro models of neuroinflammation. Cocultures of neurons/glial cells were exposed to lipopolysaccharide (LPS, 1 µg/mL) or interleukin (IL)-1β (10 ng/mL) for 24 h and treated with FAB (0.1 and 1 µM, 24 h). FAB displayed a significant neuroprotective effect, as measured by nitric oxide (NO) production, Fluoro-Jade B (FJ-B) staining, and immunocytochemistry (ICC) for the neuronal marker β-tubulin and the cell death marker caspase-3, preserving neuronal soma and increasing neurite outgrowth. FAB significantly decreased the LPS-induced microglial proliferation, identified by ICC for Iba-1/bromodeoxyuridine (BrdU) and CD68 (microglia M1 profile marker). In contrast, FAB had no apparent effect on astrocytes, as determined by ICC for glial fibrillary acidic protein (GFAP). Furthermore, FAB protected against the cytodestructive and proinflammatory effects of IL-1β, a key cytokine that is released by activated microglia and astrocytes, and ICC showed that combined treatment of FAB with α and β estrogen receptor antagonists did not affect NF-κB expression. In addition, qPCR analysis demonstrated that FAB decreased the expression of proinflammatory molecules TNF-α, IL-1β, and connexins CCL5 and CCL2, as well as increased the expression of the regulatory molecule IL-10. Together, these findings indicate that FAB has a significant neuroprotective and anti-inflammatory effect in vitro, which may be considered as an adjuvant for the treatment of neurodegenerative diseases.
Collapse
|
36
|
Han D, Zheng W, Wang X, Chen Z. Proteostasis of α-Synuclein and Its Role in the Pathogenesis of Parkinson's Disease. Front Cell Neurosci 2020; 14:45. [PMID: 32210767 PMCID: PMC7075857 DOI: 10.3389/fncel.2020.00045] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 02/18/2020] [Indexed: 12/15/2022] Open
Abstract
Aggregation of α-Synuclein, possibly caused by disturbance of proteostasis, has been identified as a common pathological feature of Parkinson’s disease (PD). However, the initiating events of aggregation have not been fully illustrated, and this knowledge may be critical to understanding the disease mechanisms of PD. Proteostasis is essential in maintaining normal cellular metabolic functions, which regulate the synthesis, folding, trafficking, and degradation of proteins. The toxicity of the aggregating proteins is dramatically influenced by its physical and physiological status. Genetic mutations may also affect the metastable phase transition of proteins. In addition, neuroinflammation, as well as lipid metabolism and its interaction with α-Synuclein, are likely to contribute to the pathogenesis of PD. In this review article, we will highlight recent progress regarding α-Synuclein proteostasis in the context of PD. We will also discuss how the phase transition status of α-Synuclein could correlate with different functional consequences in PD.
Collapse
Affiliation(s)
- Deqiang Han
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Cell Therapy Center, National Clinical Research Center for Geriatric Diseases, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Wei Zheng
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Cell Therapy Center, National Clinical Research Center for Geriatric Diseases, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Xueyao Wang
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Cell Therapy Center, National Clinical Research Center for Geriatric Diseases, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Zhiguo Chen
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Cell Therapy Center, National Clinical Research Center for Geriatric Diseases, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
37
|
Pépin É, Jalinier T, Lemieux GL, Massicotte G, Cyr M. Sphingosine-1-Phosphate Receptors Modulators Decrease Signs of Neuroinflammation and Prevent Parkinson's Disease Symptoms in the 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine Mouse Model. Front Pharmacol 2020; 11:77. [PMID: 32153401 PMCID: PMC7047735 DOI: 10.3389/fphar.2020.00077] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 01/27/2020] [Indexed: 12/20/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a potent bioactive lipid mediator that acts as a natural ligand upon binding to five different receptors that are located in astrocytes, oligodendrocytes, microglial and neuronal cells. Recently, global activation of these receptors by FTY720 (fingolimod) has been suggested to provide neuroprotection in animal model of Parkinson’s disease (PD). Among S1P receptors, the subtype 1 (S1P1R) has been linked to features of neuroprotection and, using the selective agonist SEW2871, the present investigation assessed potential benefits (and mechanisms) of this receptor subtype in an established animal model of PD. We demonstrated that oral treatments with SEW2871 are able to provide protection to the same levels as FTY720 against loss of dopaminergic neurons and motor deficits in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) (30 mg/kg, i.p., 5 days) mouse model of PD. At the molecular level, we observed that the beneficial effects of both S1PR agonists were not associated with alterations in ERK and Akt levels, two markers of molecular adaptations in the striatum neurons. However, these compounds have the capacity to prevent signs of neuroinflammation such as the activation of astrocytes and glial cells, as well as MPTP-induced reduction of BDNF levels in key regions of the brain implicated in motor functions. These findings suggest that selective S1P1R modulation has the ability to provide neuroprotection in response to MPTP neurotoxicity. Targeting S1P1R in PD therapy may represent a prominent candidate for treatment of this neurodegenerative conditions.
Collapse
Affiliation(s)
- Élise Pépin
- Groupe de recherche en signalisation cellulaire, Département de biologie médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Tim Jalinier
- Groupe de recherche en signalisation cellulaire, Département de biologie médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Guillaume L Lemieux
- Groupe de recherche en signalisation cellulaire, Département de biologie médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Guy Massicotte
- Groupe de recherche en signalisation cellulaire, Département de biologie médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Michel Cyr
- Groupe de recherche en signalisation cellulaire, Département de biologie médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| |
Collapse
|
38
|
Lama A, Pirozzi C, Avagliano C, Annunziata C, Mollica MP, Calignano A, Meli R, Mattace Raso G. Nutraceuticals: An integrative approach to starve Parkinson's disease. Brain Behav Immun Health 2020; 2:100037. [PMID: 34589828 PMCID: PMC8474522 DOI: 10.1016/j.bbih.2020.100037] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/08/2020] [Accepted: 01/12/2020] [Indexed: 01/15/2023] Open
Abstract
The therapeutic approach of multifactorial complex diseases is always a challenge; Parkinson's disease (PD) is a heterogeneous neurodegenerative disorder triggered by genetic and environmental factors, contributing to its etiology. Indeed, several pathogenic mechanisms lead to selective dopaminergic neuronal injury, including oxidative stress, mitochondrial dysfunction, alteration of endoplasmic reticulum-to-Golgi protein trafficking, excitotoxicity, and neuroinflammation. Current treatment approaches include mainly dopamine replacement therapy or optimizing dopaminergic transmission; however, these strategies that do not counteract the pathogenic mechanisms underlying PD symptoms and often are less effective over time. Recently, there has been growing interest in the therapeutic use of nutraceuticals, that could represent an integrative approach to the pharmacological standard therapy and specifically affect one or more pathogenic pathways. The intake of nutraceuticals or nutritional modifications are generally safe and can be combined with current common drug therapy in most cases to improve the patient's quality of life and/or mitigate PD symptoms. The current review focuses on several key nutritional compounds and dietary modifications that are effective on several pathogenic pathways involved in PD onset and progression, and further highlights the rationale behind their potential use for the prevention and treatment of PD.
Collapse
Affiliation(s)
- Adriano Lama
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Claudio Pirozzi
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Carmen Avagliano
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Chiara Annunziata
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Maria Pina Mollica
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
- Department of Biology, University of Naples Federico II, Cupa Nuova Cinthia 21-Edificio 7, 80126, Naples, Italy
| | - Antonio Calignano
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Rosaria Meli
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Giuseppina Mattace Raso
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| |
Collapse
|
39
|
Meng F, Guo Z, Hu Y, Mai W, Zhang Z, Zhang B, Ge Q, Lou H, Guo F, Chen J, Duan S, Gao Z. CD73-derived adenosine controls inflammation and neurodegeneration by modulating dopamine signalling. Brain 2020; 142:700-718. [PMID: 30689733 DOI: 10.1093/brain/awy351] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/09/2018] [Accepted: 11/22/2018] [Indexed: 12/21/2022] Open
Abstract
Ectonucleotidase-mediated ATP catabolism provides a powerful mechanism to control the levels of extracellular adenosine. While increased adenosine A2A receptor (A2AR) signaling has been well-documented in both Parkinson's disease models and patients, the source of this enhanced adenosine signalling remains unclear. Here, we show that the ecto-5'-nucleotidase (CD73)-mediated adenosine formation provides an important input to activate A2AR, and upregulated CD73 and A2AR in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinson's disease models coordinatively contribute to the elevated adenosine signalling. Importantly, we demonstrate that CD73-derived adenosine-A2AR signalling modulates microglial immunoresponses and morphological dynamics. CD73 inactivation significantly attenuated lipopolysaccharide-induced pro-inflammatory responses in microglia, but enhanced microglia process extension, movement and morphological transformation in the laser injury and acute MPTP-induced Parkinson's disease models. Limiting CD73-derived adenosine substantially suppressed microglia-mediated neuroinflammation and improved the viability of dopaminergic neurons and motor behaviours in Parkinson's disease models. Moreover, CD73 inactivation suppressed A2AR induction and A2AR-mediated pro-inflammatory responses, whereas replenishment of adenosine analogues restored these effects, suggesting that CD73 produces a self-regulating feed-forward adenosine formation to activate A2AR and promote neuroinflammation. We further provide the first evidence that A2A enhanced inflammation by antagonizing dopamine-mediated anti-inflammation, suggesting that the homeostatic balance between adenosine and dopamine signalling is key to microglia immunoresponses. Our study thus reveals a novel role for CD73-mediated nucleotide metabolism in regulating neuroinflammation and provides the proof-of-principle that targeting nucleotide metabolic pathways to limit adenosine production and neuroinflammation in Parkinson's disease might be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Fan Meng
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhige Guo
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaling Hu
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Weihao Mai
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenjie Zhang
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Zhang
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Qianqian Ge
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Huifang Lou
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Fang Guo
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiangfan Chen
- Molecular Neuropharmacology Laboratory and State Key Laboratory of Optometry, Ophthalmology and Vision Science, School of Optometry and Ophthalmology, Wenzhou, Zhejiang, China
| | - Shumin Duan
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihua Gao
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
40
|
Toscano ECDB, Vieira ÉLM, Portela ACDC, Caliari MV, Brant JAS, Giannetti AV, Suemoto CK, Leite REP, Nitrini R, Rachid MA, Teixeira AL. Microgliosis is associated with visual memory decline in patients with temporal lobe epilepsy and hippocampal sclerosis: A clinicopathologic study. Epilepsy Behav 2020; 102:106643. [PMID: 31805504 DOI: 10.1016/j.yebeh.2019.106643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/27/2019] [Accepted: 10/01/2019] [Indexed: 01/14/2023]
Abstract
Hippocampal sclerosis (HS) is characterized by neuronal loss and gliosis. The intensity and distribution of these histopathological findings over the Cornu Ammonis (CA) subfields are important for the classification of HS and prognostication of patients with temporal lobe epilepsy (TLE). Several studies have associated the neuronal density reduction in the hippocampus with cognitive decline in patients with TLE. The current study aimed at investigating whether the expression of glial proteins in sclerotic hippocampi is associated with presurgical memory performance of patients with TLE. Before amygdalohippocampectomy, patients were submitted to memory tests. Immunohistochemical and morphometric analyses with glial fibrillary acidic protein (GFAP) for astrogliosis and human leucocyte antigen DR (HLA-DR) for microgliosis were performed in paraffin-embedded HS and control hippocampi. Sclerotic hippocampi exhibited increased gliosis in comparison with controls. In patients with TLE, the area and intensity of staining for HLA-DR were associated with worse performance in the memory tests. Glial fibrillary acidic protein was neither associated nor correlated with memory test performance. Our data suggest association between microgliosis, but not astrogliosis, with visual memory decline in patients with TLE.
Collapse
Affiliation(s)
- Eliana Cristina de Brito Toscano
- Departamento de Patologia Geral do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Neuroscience Division, Interdisciplinary Laboratory of Medical Investigation, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Érica Leandro Marciano Vieira
- Neuroscience Division, Interdisciplinary Laboratory of Medical Investigation, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Marcelo Vidigal Caliari
- Departamento de Patologia Geral do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | - Claudia Kimie Suemoto
- Laboratório de Fisiopatologia no Envelhecimento, Universidade de São Paulo, SP, Brazil
| | | | - Ricardo Nitrini
- Laboratório de Fisiopatologia no Envelhecimento, Universidade de São Paulo, SP, Brazil
| | - Milene Alvarenga Rachid
- Departamento de Patologia Geral do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Antônio Lúcio Teixeira
- Neuropsychiatry Program and Immuno-Psychiatry Lab, Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, TX, United States
| |
Collapse
|
41
|
The neuroprotective effect of schisandrol A on 6-OHDA-induced PD mice may be related to PI3K/AKT and IKK/IκBα/NF-κB pathway. Exp Gerontol 2019; 128:110743. [DOI: 10.1016/j.exger.2019.110743] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/17/2019] [Accepted: 09/27/2019] [Indexed: 02/08/2023]
|
42
|
Banqueri M, Méndez M, Gómez-Lázaro E, Arias JL. Early life stress by repeated maternal separation induces long-term neuroinflammatory response in glial cells of male rats. Stress 2019; 22:563-570. [PMID: 31007117 DOI: 10.1080/10253890.2019.1604666] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Childhood maltreatment and neglect lead to a wide range of mental disorders highlighted by hormone and immune alterations in neglected children. This social-health challenge has led to the creation of early stress models such as maternal separation (MS) in rodents. We performed a MS model (4 h per day, 21 days; n = 16 MS and n = 16 control), and then measured three parameters in adult male rat brains, in order to look for long-term effects of early life stress. We used immunocytochemistry to mark glial fibrillary acidic protein (GFAP)-positive cells, which indicates changes in astroglia, and ionized calcium binding adaptor molecule 1 (Iba-1)-positive cells, which inform about reactive microglia. In order to study mRNA levels of some immune mediators, interleukin determination (interleukin-6, IL-6; tumor necrosis factor, TNFα) mRNAs were evaluated by real-time polymerase chain reaction (rt-PCR) in discrete brain regions. Measurements of numbers of GFAP-positive cells, and expression of Iba-1, IL-6 and TNFα mRNAs were performed in prefrontal cortex (PFC): cingulate cortex (CG), prelimbic cortex (PL) and infralimbic cortex (IL), striatal areas (dorsal striatum, STD; and nucleus accumbens, ACC), and dorsal hippocampus (HC: CA1, CA3 and dentate gyrus (DG)). We found that MS produces a dramatic and sustained decrease in the astroglial population in all the areas measured (from -25% in CA1 to -85.7% in ACC), whereas increased numbers of microglia were found, in more restricted regions: STD (72.6%), ACC (31%) and CA3 (33.3%) areas. Regarding mRNA measurements, we found increased IL-6 mRNA expression in HC (104.2%), and after MS.
Collapse
Affiliation(s)
- María Banqueri
- a Laboratory of Neuroscience, Department of Psychology, University of Oviedo , Oviedo , Spain
- b Instituto de Neurociencias del Principado de Asturias (INEUROPA)
| | - Marta Méndez
- a Laboratory of Neuroscience, Department of Psychology, University of Oviedo , Oviedo , Spain
- b Instituto de Neurociencias del Principado de Asturias (INEUROPA)
| | - Eneritz Gómez-Lázaro
- c Department of Basic Psychological Processes and their Development, Basque Country University , San Sebastian , Spain
| | - Jorge L Arias
- a Laboratory of Neuroscience, Department of Psychology, University of Oviedo , Oviedo , Spain
- b Instituto de Neurociencias del Principado de Asturias (INEUROPA)
| |
Collapse
|
43
|
Yazar HO, Yazar T, Cihan M. A preliminary data: Evaluation of serum Galectin-3 levels in patients with Idiopathic Parkinson's Disease. J Clin Neurosci 2019; 70:164-168. [PMID: 31471077 DOI: 10.1016/j.jocn.2019.08.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/05/2019] [Indexed: 12/11/2022]
Abstract
AIM In our study, we aimed to collect data for the hypothesis that Galectin-3 might be used as a new prognostic and therapeutic biomarker in Idiopathic Parkinson's Disease (IPD). METHOD In this prospective and cross-sectional study, the Unified Parkinson's Disease Rating Scale (UPDRS) and Modified Hoehn and Yahr (H&Y) scales were applied to each patient diagnosed as IPD according to the UK Brain Bank diagnostic criteria. The control group consisted of healthy individuals with the same age, gender, and body mass index characteristics as the patients meeting the exclusion criteria. RESULTS A total of 111 cases were included in the study, 48 were IPD, and 63 were healthy controls. There were no statistically significant differences between the IPD and control groups in terms of demographic, anthropometric, and blood parameters (p > 0.05). Serum galectin-3 levels were significantly higher in IPD than the control group (p < 0.001). Serum galectin-3 levels, UPDRS scores, and duration of disease were significantly higher in patients with IPD in parallel with the progression of the disease (p < 0.001; 0.001; 0.009). No significant relationship was detected between the stage of the disease and other parameters (p < 0.05). CONCLUSION Our study supports the hypothesis that serum galectin-3 level might be associated with IPD. Our data suggest that serum galectin-3 levels might be an accessible biomarker for the detection and prevention of chronic, progressive diseases such as IPH.
Collapse
Affiliation(s)
- Hülya Olgun Yazar
- Ordu University Training and Research Hospital, Clinic of Neurology, Turkey.
| | - Tamer Yazar
- Ordu State Hospital, Clinic of Neurology, Turkey
| | - Murat Cihan
- Ordu University Training and Research Hospital, Clinical Biochemist, Turkey
| |
Collapse
|
44
|
Tao D, Wang Y, Bao XQ, Yang BB, Gao F, Wang L, Zhang D, Li L. Discovery of coumarin Mannich base derivatives as multifunctional agents against monoamine oxidase B and neuroinflammation for the treatment of Parkinson's disease. Eur J Med Chem 2019; 173:203-212. [DOI: 10.1016/j.ejmech.2019.04.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/08/2019] [Accepted: 04/08/2019] [Indexed: 11/28/2022]
|
45
|
Watson L, Keatinge M, Gegg M, Bai Q, Sandulescu MC, Vardi A, Futerman AH, Schapira AH, Burton EA, Bandmann O. Ablation of the pro-inflammatory master regulator miR-155 does not mitigate neuroinflammation or neurodegeneration in a vertebrate model of Gaucher's disease. Neurobiol Dis 2019; 127:563-569. [DOI: 10.1016/j.nbd.2019.04.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/03/2019] [Accepted: 04/10/2019] [Indexed: 01/22/2023] Open
|
46
|
Modulation of the neurotransmitter systems through the anti-inflammatory and antidepressant-like effects of squalene from Aurantiochytrium sp. PLoS One 2019; 14:e0218923. [PMID: 31251788 PMCID: PMC6599144 DOI: 10.1371/journal.pone.0218923] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 06/13/2019] [Indexed: 11/19/2022] Open
Abstract
Although algae have been the focal point of biofuel research, studies on their biological activities have been limited. In recent years, however, the importance of algae as sources of functional ingredients has been recognized due to their health beneficial effects. In this study, we evaluated the antidepressant-like activities of ethanol extract of Aurantiochytrium sp. (EEA) in the forced swimming test (FST)-induced depression in ICR mice. Imipramine, a commercially available tricyclic antidepressant drug, was used as positive control. Animals were administered EEA orally for 14 consecutive days and were subjected to the locomotor activity testing. Additionally, changes in gene expression in mice brain were assessed by real-time PCR and microarray assays to understand the molecular mechanisms underlying the effect of EEA. We found that the immobility time in FST was significantly reduced in the EEA-treated mice compared to that of in the control mice. Microarray and real-time PCR results revealed that EEA treatment induced changes in several genes in mice brain associated with pro-inflammation and dopaminergic, cholinergic, glutamatergic, and serotonergic synapses. It has previously been reported that several cytokines, such as IL-6 and TNF-α, which mediate neuroinflammation, are also responsible for indirectly altering brain neurotransmitter levels in neuropsychiatric disorders. Therefore, the regulation of the expression of pro-inflammatory genes in EEA-administered mice brain is considered to contribute to the enhancement of neurotransmitter systems-related gene expression in our study. Moreover, our in vitro study suggested that squalene, a component produced by Aurantiochytrium, was one of the active substances in EEA. In conclusion, our study provides the first evidence that Aurantiochytrium sp. can reduce neuroinflammation that may contribute to the modulation of the neurotransmitter systems, which could underlie its antistress and antidepressant effects.
Collapse
|
47
|
Antonazzo M, Botta M, Bengoetxea H, Ruiz-Ortega JÁ, Morera-Herreras T. Therapeutic potential of cannabinoids as neuroprotective agents for damaged cells conducing to movement disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 146:229-257. [PMID: 31349929 DOI: 10.1016/bs.irn.2019.06.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The basal ganglia (BG), an organized network of nuclei that integrates cortical information, play a crucial role in controlling motor function. In fact, movement disorders such as Parkinson's disease (PD) and Huntington's disease (HD) are caused by the degeneration of specific structures within the BG. There is substantial evidence supporting the idea that cannabinoids may constitute novel promising compounds for the treatment of movement disorders as neuroprotective and anti-inflammatory agents. This potential therapeutic role of cannabinoids is based, among other qualities, on their capacity to reduce oxidative injury and excitotoxicity, control calcium influx and limit the toxicity of reactive microglia. The mechanisms involved in these effects are related to CB1 and CB2 receptor activation, although some of the effects are CB receptor independent. Thus, taking into account the aforementioned properties, compounds that act on the endocannabinoid system could be useful as a basis for developing disease-modifying therapies for PD and HD.
Collapse
Affiliation(s)
- Mario Antonazzo
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain; Neurodegenerative Diseases Group, BioCruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - María Botta
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Harkaitz Bengoetxea
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - José Ángel Ruiz-Ortega
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain; Neurodegenerative Diseases Group, BioCruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain; Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Teresa Morera-Herreras
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain; Neurodegenerative Diseases Group, BioCruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain.
| |
Collapse
|
48
|
Rebenkova MS, Gombozhapova AE, Rogovskaya YV, Ryabov VV, Kzhyshkowska YG, Kim BE, Prohorova YA. [Dynamics of brain CD68+ and stabilin-1+ macrophage infiltration in patients with myocardial infarction]. ACTA ACUST UNITED AC 2019; 59:44-50. [PMID: 31131759 DOI: 10.18087/cardio.2584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 11/18/2022]
Abstract
Te aim of the study was to evaluate the temporal dynamics of brain CD68+ and stabilin-1+ macrophage infltration in patients with fatal myocardial infarction (MI) type 1. MATERIALS AND METHODS Te study included 31 patients with fatal MI type I. Te control group comprised 10 patients of 18-40 age group who died from injuries incompatible with life. Patients with MI were divided into two groups. Group 1 comprised patients who died during the frst 72 hours of MI, group 2 comprised patients who died on days 4‒28. Macrophage infltration in the brain was assessed by immunohistochemical analysis. We used CD68 as a marker for the cells of the macrophage lineage and stabilin-1 as an M2-like macrophage biomarker. RESULTS In group 1 the number of brain CD68+ macrophages was signifcantly higher than in the control group. In group 2 the intensity of brain CD68+ cells infltration was lower than in group 1 and higher than in the control group. Tere was a small amount of stabilin-1+ macrophages in the brain of healthy people, as well as of patients who died from MI. Tere were no signifcant differences in the number of stabilin-1+ cells between group 1 and group 2. Correlation analysis revealed the presence of positive correlation between the number of CD68 + macrophages in the infarct, peri-infarct, and non-infarct areas of the myocardium and the number of CD68+ macrophages in the brain in patients with MI. Tere were not correlations between the number of CD68 + and stabilin-1+ cells and the presence of diabetes mellitus, history of stroke, history of MI, and pre-infarction angina. CONCLUSION Te number of brain CD68+ macrophages signifcantly increased during the frst three days of MI. Te number of brain stabilin-1+ macrophages did not increase and did not differ from the control values. We observed a positive correlation between the number of CD68+ macrophages in the brain and myocardium.
Collapse
Affiliation(s)
- M S Rebenkova
- Tomsk National Research Medical Center of the Russian Academy of Science; National Research Tomsk State University
| | - A E Gombozhapova
- Tomsk National Research Medical Center of the Russian Academy of Science; National Research Tomsk State University
| | - Yu V Rogovskaya
- Tomsk National Research Medical Center of the Russian Academy of Science; National Research Tomsk State University
| | - V V Ryabov
- Tomsk National Research Medical Center of the Russian Academy of Science; National Research Tomsk State University; Siberian State Medical University
| | | | - B E Kim
- National Research Tomsk State University
| | | |
Collapse
|
49
|
Ilie IM, Caflisch A. Simulation Studies of Amyloidogenic Polypeptides and Their Aggregates. Chem Rev 2019; 119:6956-6993. [DOI: 10.1021/acs.chemrev.8b00731] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Ioana M. Ilie
- Department of Biochemistry, University of Zürich, Zürich CH-8057, Switzerland
| | - Amedeo Caflisch
- Department of Biochemistry, University of Zürich, Zürich CH-8057, Switzerland
| |
Collapse
|
50
|
Farombi EO, Awogbindin IO, Farombi TH, Oladele JO, Izomoh ER, Aladelokun OB, Ezekiel IO, Adebambo OI, Abah VO. Neuroprotective role of kolaviron in striatal redo-inflammation associated with rotenone model of Parkinson's disease. Neurotoxicology 2019; 73:132-141. [PMID: 30930291 DOI: 10.1016/j.neuro.2019.03.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 03/20/2019] [Accepted: 03/20/2019] [Indexed: 02/06/2023]
Abstract
Parkinson's disease is the most prevalent movement disorder. Currently, therapies are palliative with associated irreversible behavioural incompetence. Here, we investigated the ability of kolaviron (KV), an anti-inflammatory biflavonoid isolated form Garcinia kola seeds, to rescue striatal neuronal damage and redo-inflammation in rats exposed to rotenone (ROT). Aged rats exposed to 11 days of rotenone intoxication were treated with KV either concurrently or for 18 days. The 18-day regimen included 7 days of pre-treatment prior 11-day concurrent ROT-KV treatment. Rotenone-exposed rats lost weight appreciably and travelled less distance with reduced speed, decline efficiency to maintain a straight path, enhanced freezing, increased immobile episodes and poor hole recognition. The motor incompetence was attributed to enhanced striatal neurodegeneration, increased alpha synuclein formation and reduced tyrosine hydroxylase expression. ROT intoxication significantly increased reactive species production, which co-existed with induction of striatal antioxidant system and damage to biomolecules. ROT additionally upregulated COX-2 expression, enhanced myeloperoxidase activity and increased concentration of striatal inteleukine-6 (IL-6), IL-1β and tumour necrosis factor (TNF-α). Treatment with kolaviron reversed the rotenone-associated locomotor impairment and exploratory deficits, motor/neuromuscular incompetence, striatal neurodegeneration, neurobiochemical imbalance, altered antioxidant defence system and neuroinflammation. KV-treated rats showed improved capacity to maintain efficient gait with minimal rigidity and enhanced coordination. Taken together, kolaviron exhibited neuroprotective properties, which may be beneficial for the prevention and management of Parkinson's disease, via antioxidant, anti-inflammatory and anti-apoptotic mechanisms.
Collapse
Affiliation(s)
- Ebenezer O Farombi
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| | - Ifeoluwa O Awogbindin
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Temitope H Farombi
- Department of Medicine, College of Medicine, University of Ibadan, University College Hospital, Ibadan, Nigeria; Chief Tony Anenih Geriatric Center, University College Hospital, Ibadan, Nigeria
| | - Johnson O Oladele
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Edirin R Izomoh
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Oladimeji B Aladelokun
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Ibukunoluwa O Ezekiel
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Oluwabunmi I Adebambo
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Victoria O Abah
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|