1
|
Abolarin PO, Amin A, Nafiu AB, Ogundele OM, Owoyele BV. Optimization of Parkinson's disease therapy with plant extracts and nutrition's evolving roles. IBRO Neurosci Rep 2024; 17:1-12. [PMID: 38872839 PMCID: PMC11167367 DOI: 10.1016/j.ibneur.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 05/01/2024] [Accepted: 05/24/2024] [Indexed: 06/15/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by death of dopaminergic neurons in the substantia nigra pars compacta (SNpc). Death of dopaminergic cells in the SNpc leads to manifestations of motor dysfunction and non-motor symptoms of PD. The progression of PD symptoms severely affects the quality of life of patients and poses socio-economic problems to families and society at large. The clinical and neuropathological characteristics of PD are triggered by multiple factors such as oxidative stress, neuroinflammation, mitochondrial dysfunction, and protein aggregation. Notwithstanding the advancements in pharmacological therapy in PD management, there is burgeoning interest in alternative and complementary approaches, essentially nutrition and plant extracts strategies. This review gives widespread analysis of the role of nutrition and plant extracts in the management of PD. Studies that investigated the effects of various dietary compounds and plant extract on PD symptoms and progression were reviewed from existing literatures. Nutraceuticals, including vitamins and phytochemicals such as Mucuna pruriens have shown potential neuroprotective functions in preclinical and clinical studies. Indeed, these strategies ameliorate mitochondrial dysfunction, oxidative stress, and neuroinflammation, all which are implicated in the pathogenesis of PD. The neuroprotective mechanisms of nutrition and plant extracts in PD, with emphasis on their capacity to target multiple pathways implicated in PD are discussed. Additionally, challenges and limitations related with translating preclinical findings into clinical practice including standardization of dosing regimens, bioavailability, and inter-individual variability are discussed. Largely, this review elucidates on the role of nutrition and plant extracts as adjunctive therapy in PD management.
Collapse
Affiliation(s)
- Patrick Oluwole Abolarin
- Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Abdulbasit Amin
- Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | | | - Olalekan Michael Ogundele
- Department of Comparative Biomedical Sciences, Louisiana State University, School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Bamidele Victor Owoyele
- Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| |
Collapse
|
2
|
Li H, Dan QQ, Chen YJ, Chen L, Zhang HT, Mu DZ, Wang TH. Cellular Localization and Distribution of TGF-β1, GDNF and PDGF-BB in the Adult Primate Central Nervous System. Neurochem Res 2023; 48:2406-2423. [PMID: 36976393 DOI: 10.1007/s11064-023-03909-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/29/2023]
Abstract
The available data on the localization of transforming growth factor beta1 (TGF-β1), glial cell line-derived neurotrophic factor (GDNF), and platelet-derived growth factor-BB (PDGF-BB) in the adult primate and human central nervous system (CNS) are limited and lack comprehensive and systematic information. This study aimed to investigate the cellular localization and distribution of TGF-β1, GDNF, and PDGF-BB in the CNS of adult rhesus macaque (Macaca mulatta). Seven adult rhesus macaques were included in the study. The protein levels of TGF-β1, PDGF-BB, and GDNF in the cerebral cortex, cerebellum, hippocampus, and spinal cord were analyzed by western blotting. The expression and location of TGF-β1, PDGF-BB, and GDNF in the brain and spinal cord was examined by immunohistochemistry and immunofluorescence staining, respectively. The mRNA expression of TGF-β1, PDGF-BB, and GDNF was detected by in situ hybridization. The molecular weight of TGF-β1, PDGF-BB, and GDNF in the homogenate of spinal cord was 25 KDa, 30 KDa, and 34 KDa, respectively. Immunolabeling revealed GDNF was ubiquitously distributed in the cerebral cortex, hippocampal formation, basal nuclei, thalamus, hypothalamus, brainstem, cerebellum, and spinal cord. TGF-β1 was least distributed and found only in the medulla oblongata and spinal cord, and PDGF-BB expression was also limited and present only in the brainstem and spinal cord. Besides, TGF-β1, PDGF-BB, and GDNF were localized in the astrocytes and microglia of spinal cord and hippocampus, and their expression was mainly found in the cytoplasm and primary dendrites. The mRNA of TGF-β1, PDGF-BB, and GDNF was localized to neuronal subpopulations in the spinal cord and cerebellum. These findings suggest that TGF-β1, GDNF and PDGF-BB may be associated with neuronal survival, neural regeneration and functional recovery in the CNS of adult rhesus macaques, providing the potential insights into the development or refinement of therapies based on these factors.
Collapse
Affiliation(s)
- Hui Li
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Department of Intensive Care Unit of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Qi-Qin Dan
- Institute of Neurological Disease, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
| | - Yan-Jun Chen
- Institute of Neurological Disease, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
| | - Li Chen
- Institute of Neurological Disease, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
| | - Hong-Tian Zhang
- Institute of Neurological Disease, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China
| | - De-Zhi Mu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Ting-Hua Wang
- Institute of Neurological Disease, Sichuan University West China Hospital, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
3
|
de Almeida EJR, Ibrahim HJ, Chitolina Schetinger MR, de Andrade CM, Cardoso AM. Modulation of Inflammatory Mediators and Microglial Activation Through Physical Exercise in Alzheimer's and Parkinson's Diseases. Neurochem Res 2022; 47:3221-3240. [PMID: 35962936 DOI: 10.1007/s11064-022-03713-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 12/19/2022]
Abstract
Neuroinflammation is an inflammatory process in the central nervous system (CNS), in addition to being one of the main features of Alzheimer's disease (AD) and Parkinson's disease (PD). Microglia are known for their immune functions and have multiple reactive phenotypes related to the types of stages involving neurodegenerative diseases. Depending on the state of activation of microglia in the CNS, it can be neuroprotective or neurotoxic. In this context, AD is a neurodegenerative and neuroinflammatory disease characterized by the deposition of beta-amyloid plaques, formation of fibrillar tangles of tau protein, and loss of neurons due to neurotoxic activation of microglia. However, PD is characterized by the loss of dopaminergic neurons in the substantia nigra and accumulation of alpha-synuclein in the cortical regions, spinal cord, and brain stem, which occurs by microglial activation, contributing to the neuroinflammatory process. In this aspect, the activation of microglia in both pathologies triggers high levels of inflammatory markers, such as interleukins, and causes the neuroinflammatory process of the diseases. Thus, physical exercise is pointed out as neuroprotective, as it can act to strengthen neurogenesis and reduce the inflammatory process. Therefore, the present review addresses the neuroprotective effect of microglia after different types of physical exercise protocols and evaluates the activity and effects of inflammatory and anti-inflammatory parameters and mechanisms of AD and PD. This review will discuss the anti-inflammatory effects of physical exercise through microglia activation with neuroprotective activity and the role of pro-and anti-inflammatory cytokines in AD and PD.
Collapse
Affiliation(s)
| | | | | | - Cinthia Melazzo de Andrade
- Department of Small Animal Clinic, Center of Rural Sciences, Federal University of Santa Maria-RS, Room 121, Veterinary Hospital Building, Avenue Roraima No. 1000, Santa Maria, RS, 97105-900, Brazil
| | - Andréia Machado Cardoso
- Graduate Program in Physical Education From Federal University of Santa Maria, Santa Maria, RS, Brazil. .,Graduate Program in Biomedical Sciences From Federal University of Fronteira Sul, Campus Chapecó, Rodovia SC 484 - Km 02, Fronteira Sul, SC, 89815-899, Brazil.
| |
Collapse
|
4
|
The promise of the TGF-β superfamily as a therapeutic target for Parkinson's disease. Neurobiol Dis 2022; 171:105805. [PMID: 35764291 DOI: 10.1016/j.nbd.2022.105805] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/20/2022] Open
Abstract
A large body of evidence underscore the regulatory role of TGF-β superfamily in the central nervous system. Components of the TGF-β superfamily modulate key events during embryonic brain development and adult brain tissue injury repair. With respect to Parkinson's disease (PD), TGF-ß signaling pathways are implicated in the differentiation, maintenance and synaptic function of the dopaminergic neurons, as well as in processes related to the activation state of astrocytes and microglia. In vitro and in vivo studies using toxin models, have interrogated on the dopaminotrophic and protective role of the TGF-β superfamily members. The evolution of genetic and animal models of PD that more closely recapitulate the disease condition has made possible the dissection of intracellular pathways in response to TGF-ß treatment. Although the first clinical trials using GDNF did not meet their primary endpoints, substantial work has been carried out to reappraise the TGF-β superfamily's clinical benefit.
Collapse
|
5
|
Yoon S, Parnell E, Penzes P. TGF-β-Induced Phosphorylation of Usp9X Stabilizes Ankyrin-G and Regulates Dendritic Spine Development and Maintenance. Cell Rep 2021; 31:107685. [PMID: 32460012 PMCID: PMC7324065 DOI: 10.1016/j.celrep.2020.107685] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/02/2020] [Accepted: 05/04/2020] [Indexed: 12/30/2022] Open
Abstract
Signaling by the cytokine transforming growth factor β (TGF-β) has been implicated in a multitude of biological functions; however, TGF-β signaling, particularly in the CNS, remains largely unexplored. ANK3 variants (encoding ankyrin-G) are associated with bipolar disorder, intellectual disability, and autism spectrum disorder, while mutations in USP9X, which encodes a deubiquitinase, are associated with X-linked intellectual disability and autism in humans. Here, we show that TGF-β signaling promotes Usp9X phosphorylation, which enhances its interaction with ankyrin-G and stabilizes ankyrin-G in spines, leading to spine enlargement. Using in situ proximity ligation combined with structured illumination superresolution microscopy, we characterize the postsynaptic spatial organization of phosphorylation-dependent regulation of Usp9X/ankyrin-G interactions in dendrites and its quantitative relationship with spine morphology and number. These data reveal a cytokine-mediated mechanism regulating protein stability in spines and suggest a role for deubiquitination and TGF-β signaling in neurodevelopmental disorder pathogenesis and treatment. Yoon et al. show that phosphorylation of a deubiquitinating enzyme by a cytokine enhances the stabilization of synaptic scaffolding protein during dendritic spine development, and its alterations result in deficient synaptic structural maintenance, with relevance for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Sehyoun Yoon
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Euan Parnell
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Northwestern University, Center for Autism and Neurodevelopment, Chicago, IL 60611, USA.
| |
Collapse
|
6
|
Jaumotte JD, Saarma M, Zigmond MJ. Protection of dopamine neurons by CDNF and neurturin variant N4 against MPP+ in dissociated cultures from rat mesencephalon. PLoS One 2021; 16:e0245663. [PMID: 33534843 PMCID: PMC7857574 DOI: 10.1371/journal.pone.0245663] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 01/05/2021] [Indexed: 01/27/2023] Open
Abstract
Parkinson's disease is associated with the loss of dopamine (DA) neurons in ventral mesencephalon. We have previously reported that no single neurotrophic factor we tested protected DA neurons from the dopaminergic toxin 1-methyl-4-phenylpyridinium (MPP+) in dissociated cultures isolated from the P0 rat substantia nigra, but that a combination of five neurotrophic factors was protective. We now report that cerebral DA neurotrophic factor (CDNF) and a variant of neurturin (NRTN), N4, were also not protective when provided alone but were protective when added together. In cultures isolated from the substantia nigra, MPP+ (10 μM) decreased tyrosine hydroxylase-positive cells to 41.7 ± 5.4% of vehicle control. Although treatment of cultures with 100 ng/ml of either CDNF or N4 individually before and after toxin exposure did not significantly increase survival in MPP+-treated cultures, when the two trophic factors were added together at 100 ng/ml each, survival of cells was increased 28.2 ± 6.1% above the effect of MPP+ alone. In cultures isolated from the ventral tegmental area, another DA rich area, a higher dose of MPP+ (1 mM) was required to produce an EC50 in TH-positive cells but, as in the substantia nigra, only the combination of CDNF and N4 (100 ng/ml each) was successful at increasing the survival of these cells compared to MPP+ alone (by 22.5 ± 3.5%). These data support previous findings that CDNF and N4 may be of therapeutic value for treatment of PD, but suggest that they may need to be administered together.
Collapse
Affiliation(s)
- Juliann D. Jaumotte
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
- Pittsburgh Institute of Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Michael J. Zigmond
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
- Pittsburgh Institute of Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|
7
|
Excess amounts of 3-iodo-l-tyrosine induce Parkinson-like features in experimental approaches of Parkinsonism. Neurotoxicology 2018; 67:178-189. [DOI: 10.1016/j.neuro.2018.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/08/2018] [Accepted: 06/04/2018] [Indexed: 10/14/2022]
|
8
|
Let's make microglia great again in neurodegenerative disorders. J Neural Transm (Vienna) 2017; 125:751-770. [PMID: 29027011 DOI: 10.1007/s00702-017-1792-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022]
Abstract
All of the common neurodegenerative disorders-Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and prion diseases-are characterized by accumulation of misfolded proteins that trigger activation of microglia; brain-resident mononuclear phagocytes. This chronic form of neuroinflammation is earmarked by increased release of myriad cytokines and chemokines in patient brains and biofluids. Microglial phagocytosis is compromised early in the disease process, obfuscating clearance of abnormal proteins. This review identifies immune pathologies shared by the major neurodegenerative disorders. The overarching concept is that aberrant innate immune pathways can be targeted for return to homeostasis in hopes of coaxing microglia into clearing neurotoxic misfolded proteins.
Collapse
|
9
|
Guo X, Liu T, Zhao D, Wang X, Liu D, He Y, Shan C, Kong Y, Hu W, Tao B, Sun L, Zhao H, Li S, Liu J. FGF18 protects against 6-hydroxydopamine-induced nigrostriatal damage in a rat model of Parkinson’s disease. Neuroscience 2017; 356:229-241. [DOI: 10.1016/j.neuroscience.2017.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 04/23/2017] [Accepted: 05/03/2017] [Indexed: 02/07/2023]
|
10
|
Evidence for an Additive Neurorestorative Effect of Simultaneously Administered CDNF and GDNF in Hemiparkinsonian Rats: Implications for Different Mechanism of Action. eNeuro 2017; 4:eN-NWR-0117-16. [PMID: 28303260 PMCID: PMC5346176 DOI: 10.1523/eneuro.0117-16.2017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 02/03/2017] [Accepted: 02/08/2017] [Indexed: 12/25/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder associated with a progressive loss of dopaminergic (DAergic) neurons of the substantia nigra (SN) and the accumulation of intracellular inclusions containing α-synuclein. Current therapies do not stop the progression of the disease, and the efficacy of these treatments wanes over time. Neurotrophic factors (NTFs) are naturally occurring proteins promoting the survival and differentiation of neurons and the maintenance of neuronal contacts. CDNF (cerebral dopamine NTF) and GDNF (glial cell line-derived NTF) are able to protect DAergic neurons against toxin-induced degeneration in experimental models of PD. Here, we report an additive neurorestorative effect of coadministration of CDNF and GDNF in the unilateral 6-hydroxydopamine (6-OHDA) lesion model of PD in rats. NTFs were given into the striatum four weeks after unilateral intrastriatal injection of 6-OHDA (20 µg). Amphetamine-induced (2.5 mg/kg, i.p.) rotational behavior was measured every two weeks. Number of tyrosine hydroxylase (TH)-positive cells from SN pars compacta (SNpc) and density of TH-positive fibers in the striatum were analyzed at 12 weeks after lesion. CDNF and GDNF alone restored the DAergic function, and one specific dose combination had an additive effect: CDNF (2.5µg) and GDNF (1µg) coadministration led to a stronger trophic effect relative to either of the single treatments alone. The additive effect may indicate different mechanism of action for the NTFs. Indeed, both NTFs activated the survival promoting PI3 kinase (PI3K)-Akt signaling pathway, but only CDNF decreased the expression level of tested endoplasmatic reticulum (ER) stress markers ATF6, glucose-regulated protein 78 (GRP78), and phosphorylation of eukaryotic initiation factor 2α subunit (eIF2α).
Collapse
|
11
|
Haas SJP, Zhou X, Machado V, Wree A, Krieglstein K, Spittau B. Expression of Tgfβ1 and Inflammatory Markers in the 6-hydroxydopamine Mouse Model of Parkinson's Disease. Front Mol Neurosci 2016; 9:7. [PMID: 26869879 PMCID: PMC4737885 DOI: 10.3389/fnmol.2016.00007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 01/14/2016] [Indexed: 11/13/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that is characterized by loss of midbrain dopaminergic (mDA) neurons in the substantia nigra (SN). Microglia-mediated neuroinflammation has been described as a common hallmark of PD and is believed to further trigger the progression of neurodegenerative events. Injections of 6-hydroxydopamine (6-OHDA) are widely used to induce degeneration of mDA neurons in rodents as an attempt to mimic PD and to study neurodegeneration, neuroinflammation as well as potential therapeutic approaches. In the present study, we addressed microglia and astroglia reactivity in the SN and the caudatoputamen (CPu) after 6-OHDA injections into the medial forebrain bundle (MFB), and further analyzed the temporal and spatial expression patterns of pro-inflammatory and anti-inflammatory markers in this mouse model of PD. We provide evidence that activated microglia as well as neurons in the lesioned SN and CPu express Transforming growth factor β1 (Tgfβ1), which overlaps with the downregulation of pro-inflammatory markers Tnfα, and iNos, and upregulation of anti-inflammatory markers Ym1 and Arg1. Taken together, the data presented in this study suggest an important role for Tgfβ1 as a lesion-associated factor that might be involved in regulating microglia activation states in the 6-OHDA mouse model of PD in order to prevent degeneration of uninjured neurons by microglia-mediated release of neurotoxic factors such as Tnfα and nitric oxide (NO).
Collapse
Affiliation(s)
| | - Xiaolai Zhou
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-UniversityFreiburg, Germany; Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell UniversityIthaca, NY, USA
| | - Venissa Machado
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-UniversityFreiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-UniversityFreiburg, Germany; Faculty of Biology, Albert-Ludwigs-UniversityFreiburg, Germany
| | - Andreas Wree
- Institute of Anatomy, Rostock University Medical Center Rostock, Germany
| | - Kerstin Krieglstein
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, Germany
| | - Björn Spittau
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, Germany
| |
Collapse
|
12
|
Liu Z, Chen HQ, Huang Y, Qiu YH, Peng YP. Transforming growth factor-β1 acts via TβR-I on microglia to protect against MPP(+)-induced dopaminergic neuronal loss. Brain Behav Immun 2016; 51:131-143. [PMID: 26254549 DOI: 10.1016/j.bbi.2015.08.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 07/07/2015] [Accepted: 08/04/2015] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation is associated with pathogenesis of Parkinson's disease (PD), a neurodegenerative disorder characterized by a progressive loss of dopaminergic (DAergic) neurons within the substantia nigra. Transforming growth factor (TGF)-β1 exerts anti-inflammatory and neuroprotective properties. However, it is unclear if microglia are required for TGF-β1 neuroprotection in PD. Here we used both shRNA and pharmacologic inhibition to determine the role of microglial TGF-β receptor (TβR)-I and its downstream signaling pathways in 1-methyl-4-phenylpyridinium (MPP(+))-induced DAergic neuronal toxicity. As expected, MPP(+) reduced the number of tyrosine hydroxylase (TH)-immunoreactive cells in ventral mesencephalic cell cultures. We found that MPP(+) activated microglia as determined by an upregulation in expression of CD11b and inducible nitric oxide synthase (iNOS), an increase in expression and secretion of tumor necrosis factor (TNF)-α and interleukin (IL)-1β, and a decrease in expression and secretion of the neurotrophic factor, insulin-like growth factor (IGF)-1. Pretreatment with TGF-β1 significantly inhibited all these changes caused by MPP(+). Expression of microglial TβR-I was upregulated by TGF-β1. Silencing of the TβR-I gene in microglia abolished both the neuroprotective and anti-inflammatory properties of TGF-β1. TGF-β1 increased microglial p38 MAPK and Akt phosphorylation, both of which were blocked by the p38 inhibitor SB203580 and the PI3K inhibitor LY294002, respectively. Pretreatment of microglia with either SB203580 or LY294002 impaired the ability of TGF-β1 to inhibit MPP(+)-induced DAergic neuronal loss and microglial activation. These findings establish that TGF-β1 activates TβR-I and its downstream p38 MAPK and PI3K-Akt signaling pathways in microglia to protect against DAergic neuronal loss that characterizes in PD.
Collapse
Affiliation(s)
- Zhan Liu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China
| | - Hui-Qiao Chen
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China
| | - Yan Huang
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China.
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China.
| |
Collapse
|
13
|
Gouarné C, Giraudon-Paoli M, Seimandi M, Biscarrat C, Tardif G, Pruss RM, Bordet T. Olesoxime protects embryonic cortical neurons from camptothecin intoxication by a mechanism distinct from BDNF. Br J Pharmacol 2015; 168:1975-88. [PMID: 23278424 DOI: 10.1111/bph.12094] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 11/28/2012] [Accepted: 12/10/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Olesoxime is a small cholesterol-oxime promoting rat embryonic motor neurons survival in the absence of trophic factors. Because olesoxime can substitute for neurotrophic factors in many situations, and to gain further understanding of its mechanism of action, we wondered if it could prevent neuronal death induced by camptothecin (CPT) and compared its effects with those of brain-derived neurotrophic factor (BDNF). EXPERIMENTAL APPROACH E17 rat embryonic cortical neurons were treated with olesoxime, BDNF or vehicle and intoxicated with CPT. Caspase-dependent and caspase-independent death pathways along with pro-survival pathways activation were explored. KEY RESULTS As previously reported for BDNF, olesoxime dose-dependently delayed CPT-induced cell death. Both compounds acted downstream of p53 activation preventing cytochrome c release and caspases activation. When caspase activation was blocked, both olesoxime and BDNF provided additional neuroprotective effect, potentially through the prevention of apoptosis-inducing factor release from mitochondria. While BDNF activates both the PI3K/Akt and the ERK pathway, olesoxime induced only a late activation of the ERK pathways, which did not seem to play a major role in its neuroprotection against CPT. Rather, our results favour preserved mitochondrial membrane integrity by olesoxime. CONCLUSIONS AND IMPLICATIONS Albeit different, olesoxime and BDNF mechanisms for neuroprotection converge to preserve mitochondrial function. These findings emphasize the importance of targeting the mitochondria in the process of neurodegeneration. Importantly olesoxime, by mimicking neurotrophin pro-survival activities without impacting PI3K/Akt and ERK signalling, may have greater therapeutic potential in many diseases where neurotrophins were considered as a therapeutic solution.
Collapse
|
14
|
Enhanced efficacy of the CDNF/MANF family by combined intranigral overexpression in the 6-OHDA rat model of Parkinson's disease. Mol Ther 2014; 23:244-54. [PMID: 25369767 DOI: 10.1038/mt.2014.206] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 10/21/2014] [Indexed: 01/22/2023] Open
Abstract
Cerebral Dopamine Neurotrophic Factor (CDNF) and Mesencephalic Astrocyte-derived Neurotrophic factor (MANF) are members of a recently discovered family of neurotrophic factors (NTFs). Here, we used intranigral or intrastriatal lentiviral vector-mediated expression to evaluate their efficacy at protecting dopaminergic function in the 6-OHDA model of Parkinson's disease (PD). In contrast to the well-studied Glial-Derived Neurotrophic Factor (GDNF), no beneficial effects were demonstrated by striatal overexpression of either protein. Interestingly, nigral overexpression of CDNF decreased amphetamine-induced rotations and increased tyroxine hydroxylase (TH) striatal fiber density but had no effect on numbers of TH(+) cells in the SN. Nigral MANF overexpression had no effect on amphetamine-induced rotations or TH striatal fiber density but resulted in a significant preservation of TH(+) cells. Combined nigral overexpression of both factors led to a robust reduction in amphetamine-induced rotations, greater increase in striatal TH-fiber density and significant protection of TH(+) cells in the SN. We conclude that nigral CDNF and MANF delivery is more efficacious than striatal delivery. This is also the first study to demonstrate that combined NTF can have synergistic effects that result in enhanced neuroprotection, suggesting that multiple NTF delivery may be more efficacious for the treatment of PD than the single NTF approaches attempted so far.
Collapse
|
15
|
Pisanu A, Lecca D, Mulas G, Wardas J, Simbula G, Spiga S, Carta AR. Dynamic changes in pro- and anti-inflammatory cytokines in microglia after PPAR-γ agonist neuroprotective treatment in the MPTPp mouse model of progressive Parkinson's disease. Neurobiol Dis 2014; 71:280-91. [DOI: 10.1016/j.nbd.2014.08.011] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 07/31/2014] [Accepted: 08/06/2014] [Indexed: 11/25/2022] Open
|
16
|
Yan J, Fu Q, Cheng L, Zhai M, Wu W, Huang L, Du G. Inflammatory response in Parkinson's disease (Review). Mol Med Rep 2014; 10:2223-33. [PMID: 25215472 DOI: 10.3892/mmr.2014.2563] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 07/01/2014] [Indexed: 11/05/2022] Open
Abstract
Parkinson's disease (PD) is one of the most common age‑related neurodegenerative diseases, which results from a number of environmental and inherited factors. PD is characterized by the slow progressive degeneration of dopaminergic (DA) neurons in the substantia nigra. The nigrostriatal DA neurons are particularly vulnerable to inflammatory attack. Neuroinflammation is an important contributor to the pathogenesis of age‑related neurodegenerative disorders, such as PD, and as such anti‑inflammatory agents are becoming a novel therapeutic focus. This review will discuss the current knowledge regarding inflammation and review the roles of intracellular inflammatory signaling pathways, which are specific inflammatory mediators in PD. Finally, possible therapeutic strategies are proposed, which may downregulate inflammatory processes and inhibit the progression of PD.
Collapse
Affiliation(s)
- Junqiang Yan
- Department of Neurology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Qizhi Fu
- Department of Neurology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Liniu Cheng
- Department of Neurology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Mingming Zhai
- Department of Neurology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Wenjuan Wu
- Department of Neurology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Lina Huang
- Department of Neurology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Ganqin Du
- Department of Neurology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| |
Collapse
|
17
|
Yu CY, Gui W, He HY, Wang XS, Zuo J, Huang L, Zhou N, Wang K, Wang Y. Neuronal and astroglial TGFβ-Smad3 signaling pathways differentially regulate dendrite growth and synaptogenesis. Neuromolecular Med 2014; 16:457-72. [PMID: 24519742 DOI: 10.1007/s12017-014-8293-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Accepted: 01/27/2014] [Indexed: 12/16/2022]
Abstract
To address the role of the transforming growth factor beta (TGFβ)-Smad3 signaling pathway in dendrite growth and associated synaptogenesis, we used small inhibitory RNA to knockdown the Smad3 gene in either cultured neurons and or primary astrocytes. We found that TGFβ1 treatment of primary neurons increased dendrite extensions and the number of synapsin-1-positive synapses. When Smad3 was knockdown in primary neurons, dendrite growth was inhibited and the number of synapsin-1-positive synapses reduced even with TGFβ1 treatment. When astrocyte-conditioned medium (ACM), collected from TGFβ1-treated astrocytes (TGFβ1-stimulated ACM), was added to cultured neurons, dendritic growth was inhibited and the number of synapsin-1-positive puncta reduced. When TGFβ1-stimulated ACM was collected from astrocytes with Smad3 knocked down, this conditioned media promoted the growth of dendrites and the number of synapsin-1-positive puncta in cultured neurons. We further found that TGFβ1 signaling through Smad3 increased the expression of chondroitin sulfate proteoglycans, neurocan, and phosphacan in ACM. Application of chondroitinase ABC to the TGFβ1-stimulated ACM reversed its inhibitory effects on the dendrite growth and the number of synapsin-1-positive puncta. On the other hand, we found that TGFβ1 treatment caused a facilitation of Smad3 phosphorylation and translocation to the nucleus induced by status epilepticus (SE) in wild-type (Smad3(+/+)) mice, and this treatment also caused a promotion of γ-aminobutyric acid-ergic synaptogenesis impaired by SE in Smad3(+/+) as well as in Smad3(-/-) mice, but more dramatic promotion in Smad3(+/+) mice. Thus, we provide evidence for the first time that TGFβ-Smad3 signaling pathways within neuron and astrocyte differentially regulate dendrite growth and synaptogenesis, and this pathway may be involved in the pathogenesis of some central nervous system diseases, such as epilepsy.
Collapse
Affiliation(s)
- Chuan-Yong Yu
- Epilepsy and Headache Group, Department of Neurology, The First Hospital of Anhui Medical University, Jixi Road 218, Hefei, 230022, China
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Oleoylethanolamide reduces L-DOPA-induced dyskinesia via TRPV1 receptor in a mouse model of Parkinson´s disease. Neurobiol Dis 2014; 62:416-25. [DOI: 10.1016/j.nbd.2013.10.008] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 10/02/2013] [Accepted: 10/06/2013] [Indexed: 01/10/2023] Open
|
19
|
Villapol S, Wang Y, Adams M, Symes AJ. Smad3 deficiency increases cortical and hippocampal neuronal loss following traumatic brain injury. Exp Neurol 2013; 250:353-65. [PMID: 24120438 DOI: 10.1016/j.expneurol.2013.10.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Revised: 09/11/2013] [Accepted: 10/03/2013] [Indexed: 12/13/2022]
Abstract
Transforming growth factor-β (TGF-β) signaling is involved in pathological processes following brain injury. TGF-β signaling through Smad3 contributes significantly to the immune response and glial scar formation after brain injury. However, TGF-β is also neuroprotective, suggesting that Smad3 signaling may also be involved in neuroprotection after injury. We found expression of the TGF-β type II receptor (TβRII) and Smad3 protein to be strongly and rapidly induced in neurons in the ipsilateral cortex and CA1 region of the hippocampus after stab wound injury. In contrast, astrocytic expression of TβRII and Smad3 was induced more slowly. Comparison of the response of wild-type and Smad3 null mice to cortical stab wound injury showed a more pronounced loss of neuronal viability in Smad3 null mice. Neuronal density was more strongly reduced in Smad3 null mice than in wild-type mice at 1 and 3days post lesion in both the ipsilateral cortex and hippocampal CA1 region. Fluoro-Jade B, TUNEL staining, and cleaved caspase-3 staining also demonstrated increased neuronal degeneration at early time points after injury in the ipsilateral hemisphere in Smad3 null mice. Taken together, our results suggest that TGF-β cytokine family signaling through Smad3 protects neurons in the damaged cortex and hippocampus at early time points after injury.
Collapse
Affiliation(s)
- Sonia Villapol
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | | | | | | |
Collapse
|
20
|
Gonzalo-Gobernado R, Calatrava-Ferreras L, Reimers D, Herranz AS, Rodríguez-Serrano M, Miranda C, Jiménez-Escrig A, Díaz-Gil JJ, Bazán E. Neuroprotective activity of peripherally administered liver growth factor in a rat model of Parkinson's disease. PLoS One 2013; 8:e67771. [PMID: 23861803 PMCID: PMC3701531 DOI: 10.1371/journal.pone.0067771] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 05/22/2013] [Indexed: 11/19/2022] Open
Abstract
Liver growth factor (LGF) is a hepatic mitogen purified some years ago that promotes proliferation of different cell types and the regeneration of damaged tissues, including brain tissue. Considering the possibility that LGF could be used as a therapeutic agent in Parkinson’s disease, we analyzed its potential neuroregenerative and/or neuroprotective activity when peripherally administered to unilaterally 6-hydroxydopamine (6-OHDA)-lesioned rats. For these studies, rats subjected to nigrostriatal lesions were treated intraperitoneally twice a week with LGF (5 microg/rat) for 3 weeks. Animals were sacrificed 4 weeks after the last LGF treatment. The results show that LGF stimulates sprouting of tyrosine hydroxylase-positive terminals and increases tyrosine hydroxylase and dopamine transporter expression, as well as dopamine levels in the denervated striatum of 6-OHDA-lesioned rats. In this structure, LGF activates microglia and raises tumor necrosis factor-alpha protein levels, which have been reported to have a role in neuroregeneration and neuroprotection. Besides, LGF stimulates the phosphorylation of MAPK/ERK1/2 and CREB, and regulates the expression of proteins which are critical for cell survival such as Bcl2 and Akt. Because LGF partially protects dopamine neurons from 6-OHDA neurotoxicity in the substantia nigra, and reduces motor deficits in these animals, we propose LGF as a novel factor that may be useful in the treatment of Parkinson’s disease.
Collapse
Affiliation(s)
| | | | - Diana Reimers
- Servicio de Neurobiología, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Antonio Sánchez Herranz
- Servicio de Neurobiología, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | | | - Cristina Miranda
- Servicio de Neurobiología, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | | | - Juan José Díaz-Gil
- Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Eulalia Bazán
- Servicio de Neurobiología, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
- * E-mail:
| |
Collapse
|
21
|
Li LY, Li JL, Zhang HM, Yang WM, Wang K, Fang Y, Wang Y. TGFβ1 treatment reduces hippocampal damage, spontaneous recurrent seizures, and learning memory deficits in pilocarpine-treated rats. J Mol Neurosci 2012; 50:109-23. [PMID: 22936246 DOI: 10.1007/s12031-012-9879-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Accepted: 08/15/2012] [Indexed: 11/28/2022]
Abstract
Studies have demonstrated the neuroprotective activity of transforming growth factor beta-1 (TGFβ1), protecting neurons against different kinds of insults. However, the role of exogenous TGFβ1 in the neuronal damage following status epilepticus (SE) and the related spontaneous recurrent seizures (SRS) is unknown. The present study aimed to determine the effect of intranasal TGFβ1 administration on SRS and cognitive function following lithium-pilocarpine-induced SE and associated hippocampal damage. We found that intranasal TGFβ1 significantly attenuated the hippocampal insults marked by hematoxylin and eosin, terminal deoxynucleotidyl transferase dUTP nick end labeling, and Fluoro-Jade B staining by 24, 48, and 72 h after SE was induced. The expression of the apoptosis-suppressing protein, Bcl-2, was elevated, whereas the expression of the apoptosis-promoting proteins, Bax and Caspase-3, was suppressed in TGFβ1-treated rats compared to rats without TGFβ1 treatment by 24, 48, and 72 h following induction of SE. The seizure number, severity, and duration of SRS over a 1-month period of monitoring starting 15 days after SE induction as well as the cognitive deficits detected 45 days after SE induction were significantly reduced in TGFβ1-treated rats compared to those without TGFβ1 treatment. Our results indicate that intranasal delivery of TGFβ1 immediately after SE induction not only protected against SRS but also improved cognitive function. The anti-epileptogenic properties of TGFβ1 may be related to its effect of neuroprotection or to its effect of apoptosis pathway changes.
Collapse
Affiliation(s)
- Liang-Yong Li
- Epilepsy and Headache Group, Department of Neurology, the First Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | | | | | | | | | | | | |
Collapse
|
22
|
Krieglstein K, Zheng F, Unsicker K, Alzheimer C. More than being protective: functional roles for TGF-β/activin signaling pathways at central synapses. Trends Neurosci 2011; 34:421-9. [PMID: 21742388 DOI: 10.1016/j.tins.2011.06.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 05/30/2011] [Accepted: 06/01/2011] [Indexed: 12/14/2022]
Abstract
It is becoming increasingly clear that members of the transforming growth factor-β (TGF-β) family have roles in the central nervous system that extend beyond their well-established roles as neurotrophic and neuroprotective factors. Recent findings have indicated that the TGF-β signaling pathways are involved in the modulation of both excitatory and inhibitory synaptic transmission in the adult mammalian brain. In this review, we discuss how TGF-β, bone morphogenetic protein and activin signaling at central synapses modulate synaptic plasticity, cognition and affective behavior. We also discuss the implications of these findings for the molecular understanding and potential treatment of neuropsychiatric diseases, such as anxiety, depression and other neurological disorders.
Collapse
Affiliation(s)
- Kerstin Krieglstein
- Institute of Anatomy and Cell Biology, University of Freiburg, 79104 Freiburg, Germany
| | | | | | | |
Collapse
|