1
|
Hakami A, Narasimhan K, Comini G, Thiele J, Werner C, Dowd E, Newland B. Cryogel microcarriers for sustained local delivery of growth factors to the brain. J Control Release 2024; 369:404-419. [PMID: 38508528 DOI: 10.1016/j.jconrel.2024.03.023] [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: 12/15/2023] [Revised: 03/05/2024] [Accepted: 03/14/2024] [Indexed: 03/22/2024]
Abstract
Neurotrophic growth factors such as glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) have been considered as potential therapeutic candidates for neurodegenerative disorders due to their important role in modulating the growth and survival of neurons. However, clinical translation remains elusive, as their large size hinders translocation across the blood-brain barrier (BBB), and their short half-life in vivo necessitates repeated administrations. Local delivery to the brain offers a potential route to the target site but requires a suitable drug-delivery system capable of releasing these proteins in a controlled and sustained manner. Herein, we develop a cryogel microcarrier delivery system which takes advantage of the heparin-binding properties of GDNF and BDNF, to reversibly bind/release these growth factors via electrostatic interactions. Droplet microfluidics and subzero temperature polymerization was used to create monodisperse cryogels with varying degrees of negative charge and an average diameter of 20 μm. By tailoring the inclusion of 3-sulfopropyl acrylate (SPA) as a negatively charged moiety, the release duration of these two growth factors could be adjusted to range from weeks to half a year. 80% SPA cryogels and 20% SPA cryogels were selected to load GDNF and BDNF respectively, for the subsequent biological studies. Cell culture studies demonstrated that these cryogel microcarriers were cytocompatible with neuronal and microglial cell lines, as well as primary neural cultures. Furthermore, in vivo studies confirmed their biocompatibility after administration into the brain, as well as their ability to deliver, retain and release GDNF and BDNF in the striatum. Overall, this study highlights the potential of using cryogel microcarriers for long-term delivery of neurotrophic growth factors to the brain for neurodegenerative disorder therapeutics.
Collapse
Affiliation(s)
- Abrar Hakami
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK; Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Kaushik Narasimhan
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, H91 W5P7 Galway, Ireland
| | - Giulia Comini
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, H91 W5P7 Galway, Ireland
| | - Julian Thiele
- Leibniz-Institut für Polymerforschung Dresden e.V., 01069 Dresden, Germany; Institute of Chemistry, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Carsten Werner
- Leibniz-Institut für Polymerforschung Dresden e.V., 01069 Dresden, Germany
| | - Eilís Dowd
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, H91 W5P7 Galway, Ireland.
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK.
| |
Collapse
|
2
|
Yao X, Guan Y, Wang J, Wang D. Cerium oxide nanoparticles modulating the Parkinson's disease conditions: From the alpha synuclein structural point of view and antioxidant properties of cerium oxide nanoparticles. Heliyon 2024; 10:e21789. [PMID: 38163101 PMCID: PMC10755285 DOI: 10.1016/j.heliyon.2023.e21789] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 01/03/2024] Open
Abstract
Parkinson's and Alzheimer's disease is the main cause of dementia, which is associated with the progressive deterioration of the intelligence and senses. Free radicals are created during oxidative stress in cells, which are considered one of the destructive factors in neurodegenerative diseases. In this study, the antifibrillar and antioxidant properties of cerium oxide nanoparticles (CeO2 NPs) were investigated experimentally and theoretically. The CeO2 NPs were synthesized and analyzed to reveal the physicochemical and biological properties. The results showed that the CeO2 NPs have unique properties with potent antioxidant activities. The experimental and computational studies showed that the CeO2 NPs interact with the active site of Alpha-synuclein. The existence of hydrogen bonding between O atoms of CeO2 NPs and N-H of adjacent acid amines and the equilibrium distances were confirmed by 1.751 (Leu100), 1.786 (Gln99) and 2.213 Å (Lys97). The minimum free energy binding of L-DOPA drug (as positive control) and CeO2 NPs were negative, resulting interaction between compounds and protein. As a result, these compounds inhibited Alpha-synuclein protein aggregation. In addition, that CeO2 NPs strongly binds with receptor by relative binding energy as compared with L-DOPA drug. These findings revealed that CeO2 NPs prevent Alpha-synuclein fibrillation and can be applied as nano-drug against the Parkinson's disease.
Collapse
Affiliation(s)
- Xiaomei Yao
- Department of Geriatrics, Central Hospital Affiliated to Shandong First Medical University, No. 105, Jiefang Road, Jinan City, Shandong Province, 250013, China
| | - Yichao Guan
- Department of Geriatrics, Central Hospital Affiliated to Shandong First Medical University, No. 105, Jiefang Road, Jinan City, Shandong Province, 250013, China
| | - Jianli Wang
- Department of Geriatrics, Central Hospital Affiliated to Shandong First Medical University, No. 105, Jiefang Road, Jinan City, Shandong Province, 250013, China
| | - Dong Wang
- Department of Neurology, Central Hospital Affiliated to Shandong First Medical University, No. 105, Jiefang Road, Jinan City, Shandong Province, 250013, China
| |
Collapse
|
3
|
Serrano-Martínez I, Pedreño M, Castillo-González J, Ferraz-de-Paula V, Vargas-Rodríguez P, Forte-Lago I, Caro M, Campos-Salinas J, Villadiego J, Peñalver P, Morales JC, Delgado M, González-Rey E. Cortistatin as a Novel Multimodal Therapy for the Treatment of Parkinson's Disease. Int J Mol Sci 2024; 25:694. [PMID: 38255772 PMCID: PMC10815070 DOI: 10.3390/ijms25020694] [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: 11/26/2023] [Revised: 12/29/2023] [Accepted: 12/31/2023] [Indexed: 01/24/2024] Open
Abstract
Parkinson's disease (PD) is a complex disorder characterized by the impairment of the dopaminergic nigrostriatal system. PD has duplicated its global burden in the last few years, becoming the leading neurological disability worldwide. Therefore, there is an urgent need to develop innovative approaches that target multifactorial underlying causes to potentially prevent or limit disease progression. Accumulating evidence suggests that neuroinflammatory responses may play a pivotal role in the neurodegenerative processes that occur during the development of PD. Cortistatin is a neuropeptide that has shown potent anti-inflammatory and immunoregulatory effects in preclinical models of autoimmune and neuroinflammatory disorders. The goal of this study was to explore the therapeutic potential of cortistatin in a well-established preclinical mouse model of PD induced by acute exposure to the neurotoxin 1-methil-4-phenyl1-1,2,3,6-tetrahydropyridine (MPTP). We observed that treatment with cortistatin mitigated the MPTP-induced loss of dopaminergic neurons in the substantia nigra and their connections to the striatum. Consequently, cortistatin administration improved the locomotor activity of animals intoxicated with MPTP. In addition, cortistatin diminished the presence and activation of glial cells in the affected brain regions of MPTP-treated mice, reduced the production of immune mediators, and promoted the expression of neurotrophic factors in the striatum. In an in vitro model of PD, treatment with cortistatin also demonstrated a reduction in the cell death of dopaminergic neurons that were exposed to the neurotoxin. Taken together, these findings suggest that cortistatin could emerge as a promising new therapeutic agent that combines anti-inflammatory and neuroprotective properties to regulate the progression of PD at multiple levels.
Collapse
Affiliation(s)
- Ignacio Serrano-Martínez
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Marta Pedreño
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Julia Castillo-González
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Viviane Ferraz-de-Paula
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Pablo Vargas-Rodríguez
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Irene Forte-Lago
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Marta Caro
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Jenny Campos-Salinas
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Javier Villadiego
- Institute of Biomedicine of Seville (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, 41013 Sevilla, Spain;
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| | - Pablo Peñalver
- Department of Biochemistry and Molecular Pharmacology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (P.P.); (J.C.M.)
| | - Juan Carlos Morales
- Department of Biochemistry and Molecular Pharmacology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (P.P.); (J.C.M.)
| | - Mario Delgado
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| | - Elena González-Rey
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (I.S.-M.); (M.P.); (J.C.-G.); (V.F.-d.-P.); (P.V.-R.); (I.F.-L.); (M.C.); (J.C.-S.); (M.D.)
| |
Collapse
|
4
|
Sung CY, Chiang PK, Tsai CW, Yang FY. Low-Intensity Pulsed Ultrasound Enhances Neurotrophic Factors and Alleviates Neuroinflammation in a Rat Model of Parkinson's Disease. Cereb Cortex 2021; 32:176-185. [PMID: 34196669 DOI: 10.1093/cercor/bhab201] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 12/30/2022] Open
Abstract
Low-intensity pulsed ultrasound (LIPUS) has also been reported to improve behavioral functions in Parkinson's disease (PD) animal models; however, the effect of LIPUS stimulation on the neurotrophic factors and neuroinflammation has not yet been addressed. PD rat model was built by injection of 6-hydroxydopamine (6-OHDA) in 2 sites in the right striatum. The levels of neurotrophic factors and lipocalin-2 (LCN2)-induced neuroinflammation were quantified using a western blot. Rotational test and cylinder test were conducted biweekly for 8 weeks. When the 6-OHDA + LIPUS and 6-OHDA groups were compared, the locomotor function of the 6-OHDA + LIPUS rats was significantly improved. After LIPUS stimulation, the tyrosine hydroxylase staining density was significantly increased in the striatum and substantia nigra pars compacta (SNpc) of lesioned rats. Unilateral LIPUS stimulation did not increase brain-derived neurotrophic factor in the striatum and SNpc of lesioned rats. In contrast, unilateral LIPUS stimulation increased glial cell line-derived neurotrophic factor (GDNF) protein 1.98-fold unilaterally in the SNpc. Additionally, LCN2-induced neuroinflammation can be attenuated following LIPUS stimulation. Our data indicated that LIPUS stimulation may be a potential therapeutic tool against PD via enhancement of GDNF level and inhibition of inflammatory responses in the SNpc of the brain.
Collapse
Affiliation(s)
- Chen-Yu Sung
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pai-Kai Chiang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Urology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Che-Wen Tsai
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Feng-Yi Yang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
5
|
Glial Cell- Derived Neurotrophic Factor Functions as a Potential Candidate Gene in Obstructive Sleep Apnea Based on a Combination of Bioinformatics and Targeted Capture Sequencing Analyses. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6656943. [PMID: 33688499 PMCID: PMC7911711 DOI: 10.1155/2021/6656943] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/27/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Background Obstructive sleep apnea (OSA) is a prevalent chronic disease that increases the risk of cardiovascular disease and metabolic and neuropsychiatric disorders, resulting in a considerable socioeconomic burden. The present study was aimed at identifying potential key genes influencing the mechanisms and consequences of OSA. Methods Gene expression profiles associated with OSA were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) in subcutaneous adipose tissues from patients with OSA and normal tissues were screened using R software, followed by gene ontology and pathway enrichment analyses. Subsequently, a protein-protein interaction (PPI) network was constructed and hub genes were extracted using Cytoscape plugins. The intersected core genes derived from different topological algorithms were considered hub genes, and the potential candidate gene was selected from them for further analyses of expression variations using another GEO dataset and targeted capture sequencing in 100 subjects (50 with severe OSA and 50 without OSA). Results A total of 373 DEGs were identified in OSA samples relative to normal controls, which were primarily associated with olfactory transduction and neuroactive ligand-receptor interaction. Upon analyses of nine topological algorithms and available literature, we finally focused on glial cell-derived neurotrophic factor (GDNF) as the candidate gene and validated its low expression in OSA samples. Two rare nonsynonymous variants (p.D56N and p.R93Q) were identified among the 100 cases through targeted sequencing of GDNF, which could be potentially deleterious based on pathogenicity prediction programs; however, no significant association was detected in single nucleotide polymorphisms. Conclusion The present study identified GDNF as a promising candidate gene for OSA and its two rare and potentially deleterious mutations through a combination of bioinformatics and targeted capture sequencing analyses.
Collapse
|
6
|
Şengül B, Dursun E, Verkhratsky A, Gezen-Ak D. Overexpression of α-Synuclein Reorganises Growth Factor Profile of Human Astrocytes. Mol Neurobiol 2020; 58:184-203. [PMID: 32914394 DOI: 10.1007/s12035-020-02114-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 09/01/2020] [Indexed: 12/26/2022]
Abstract
Misfolding and accumulation of aberrant α-synuclein in the brain is associated with the distinct class of neurodegenerative diseases known as α-synucleinopathies, which include Parkinson's disease, dementia with Lewy bodies and multiple system atrophy. Pathological changes in astrocytes contribute to all neurological disorders, and astrocytes are reported to possess α-synuclein inclusions in the context of α-synucleinopathies. Astrocytes are known to express and secrete numerous growth factors, which are fundamental for neuroprotection, synaptic connectivity and brain metabolism; changes in growth factor secretion may contribute to pathobiology of neurological disorders. Here we analysed the effect of α-synuclein overexpression in cultured human astrocytes on growth factor expression and release. For this purpose, the intracellular and secreted levels of 33 growth factors (GFs) and 8 growth factor receptors (GFRs) were analysed in cultured human astrocytes by chemiluminescence-based western/dot blot. Overexpression of human α-synuclein in cultured foetal human astrocytes significantly changes the profile of GF production and secretion. We found that human astrocytes express and secrete FGF2, FGF6, EGF, IGF1, AREG, IGFBP2, IGFBP4, VEGFD, PDGFs, KITLG, PGF, TGFB3 and NTF4. Overexpression of human α-synuclein significantly modified the profile of GF production and secretion, with particularly strong changes in EGF, PDGF, VEGF and their receptors as well as in IGF-related proteins. Bioinformatics analysis revealed possible interactions between α-synuclein and EGFR and GDNF, as well as with three GF receptors, EGFR, CSF1R and PDGFRB.
Collapse
Affiliation(s)
- Büşra Şengül
- Brain and Neurodegenerative Disorders Research Laboratories, Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Erdinç Dursun
- Brain and Neurodegenerative Disorders Research Laboratories, Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey.,Department of Neuroscience, Institute of Neurological Sciences, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK. .,Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain.
| | - Duygu Gezen-Ak
- Brain and Neurodegenerative Disorders Research Laboratories, Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| |
Collapse
|
7
|
Ferreira C, Almeida C, Tenreiro S, Quintas A. Neuroprotection or Neurotoxicity of Illicit Drugs on Parkinson's Disease. Life (Basel) 2020; 10:life10060086. [PMID: 32545328 PMCID: PMC7344445 DOI: 10.3390/life10060086] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 12/20/2022] Open
Abstract
Parkinson's Disease (PD) is currently the most rapid growing neurodegenerative disease and over the past generation, its global burden has more than doubled. The onset of PD can arise due to environmental, sporadic or genetic factors. Nevertheless, most PD cases have an unknown etiology. Chemicals, such as the anthropogenic pollutant 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and amphetamine-type stimulants, have been associated with the onset of PD. Conversely, cannabinoids have been associated with the treatment of the symptoms'. PD and medical cannabis is currently under the spotlight, and research to find its benefits on PD is on-going worldwide. However, the described clinical applications and safety of pharmacotherapy with cannabis products are yet to be fully supported by scientific evidence. Furthermore, the novel psychoactive substances are currently a popular alternative to classical drugs of abuse, representing an unknown health hazard for young adults who may develop PD later in their lifetime. This review addresses the neurotoxic and neuroprotective impact of illicit substance consumption in PD, presenting clinical evidence and molecular and cellular mechanisms of this association. This research area is utterly important for contemporary society since illicit drugs' legalization is under discussion which may have consequences both for the onset of PD and for the treatment of its symptoms.
Collapse
Affiliation(s)
- Carla Ferreira
- Molecular Pathology and Forensic Biochemistry Laboratory, Centro de Investigação Interdisciplinar Egas Moniz, P-2825-084 Caparica, Portugal; (C.F.); (C.A.)
- Laboratório de Ciências Forenses e Psicológicas Egas Moniz, Campus Universitário–Quinta da Granja, Monte de Caparica, P-2825-084 Caparica, Portugal
- Faculty of Medicine of Porto University, Al. Prof. Hernâni Monteiro, P-4200–319 Porto, Portugal
| | - Catarina Almeida
- Molecular Pathology and Forensic Biochemistry Laboratory, Centro de Investigação Interdisciplinar Egas Moniz, P-2825-084 Caparica, Portugal; (C.F.); (C.A.)
| | - Sandra Tenreiro
- CEDOC–Chronic Diseases Research Center, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, P-1150-082 Lisboa, Portugal;
| | - Alexandre Quintas
- Molecular Pathology and Forensic Biochemistry Laboratory, Centro de Investigação Interdisciplinar Egas Moniz, P-2825-084 Caparica, Portugal; (C.F.); (C.A.)
- Laboratório de Ciências Forenses e Psicológicas Egas Moniz, Campus Universitário–Quinta da Granja, Monte de Caparica, P-2825-084 Caparica, Portugal
- Correspondence:
| |
Collapse
|
8
|
Cao H, He Q, von Eyben R, Bloomstein J, Nambiar DK, Viswanathan V, Aggarwal S, Kwok S, Liang R, Koong AJ, Lewis JS, Kong C, Xiao N, Le QT. The role of Glial cell derived neurotrophic factor in head and neck cancer. PLoS One 2020; 15:e0229311. [PMID: 32084217 PMCID: PMC7034888 DOI: 10.1371/journal.pone.0229311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 02/03/2020] [Indexed: 11/25/2022] Open
Abstract
Glial cell-derived neurotrophic factor (GDNF) is reported to promote the survival of neurons and salivary gland regeneration after radiation damage. This study investigated the effect of GDNF on cell migration, growth, and response to radiation in preclinical models of head and neck squamous cell carcinoma (HNSCC) and correlated GDNF expression to treatment outcomes in HNSCC patients. Our ultimate goal is to determine whether systemic administration of GDNF at high dose is safe for the management of hyposalivation or xerostomia in HNSCC patients. Three HPV-positive and three HPV-negative cell lines were examined for cell migration, growth, and clonogenic survival in vitro and tumor growth assay in vivo. Immunohistochemical staining of GDNF, its receptors GFRα1 and its co-receptor RET was performed on two independent HNSCC tissue microarrays (TMA) and correlated to treatment outcomes. Results showed that GDNF only enhanced cell migration in two HPV-positive cells at supra-physiologic doses, but not in HPV-negative cells. GDNF did not increase cell survival in the tested cell lines post-irradiation. Likewise, GDNF treatment affected neither tumor growth in vitro nor response to radiation in xenografts in two HPV-positive and two HPV-negative HNSCC models. High stromal expression of GDNF protein was associated with worse overall survival in HPV-negative HNSCC on multivariate analysis in a combined cohort of patients from Stanford University (n = 82) and Washington University (n = 189); however, the association between GDNF gene expression and worse survival was not confirmed in a separate group of HPV-negative HNSCC patients identified from the Cancer Genome Atlas (TCGA) database. Based on these data, we do not believe that GNDF is a safe systemic treatment to prevent or treat xerostomia in HNSCC and a local delivery approach such as intraglandular injection needs to be explored.
Collapse
Affiliation(s)
- Hongbin Cao
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Qian He
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Rie von Eyben
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Joshua Bloomstein
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Dhanya K. Nambiar
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Vignesh Viswanathan
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Sonya Aggarwal
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Shirley Kwok
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Rachel Liang
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Amanda Jeanette Koong
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - James S. Lewis
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Christina Kong
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Nan Xiao
- Department of Biomedical Sciences, University of the Pacific Arthur A. Dugoni School of Dentistry, San Francisco, California, United States of America
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
9
|
Lee JY, Kim HS, Kim SH, Kim HS, Cho BP. Combination of Human Mesenchymal Stem Cells and Repetitive Transcranial Magnetic Stimulation Enhances Neurological Recovery of 6-Hydroxydopamine Model of Parkinsonian's Disease. Tissue Eng Regen Med 2020; 17:67-80. [PMID: 31970698 DOI: 10.1007/s13770-019-00233-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/23/2019] [Accepted: 11/28/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Repetitive transcranial magnetic stimulation (rTMS) has been in use for the treatment of various neurological diseases, including depression, anxiety, stroke and Parkinson's disease (PD), while its underlying mechanism is stills unclear. This study was undertaken to evaluate the potential synergism of rTMS treatment to the beneficial effect of human mesenchymal stem cells (hMSCs) administration for PD and to clarify the mechanism of action of this therapeutic approach. METHODS The neuroprotective effect in nigral dopamine neurons, neurotrophic/growth factors and anti-/pro-inflammatory cytokine regulation, and functional recovery were assessed in the rat 6-hydroxydopamine (6-OHDA) model of PD upon administration of hMSCs and rTMS. RESULTS Transplanted hMSCs were identified in the substantia nigra, and striatum. Enhancement of the survival of SN dopamine neurons and the expression of the tyrosine hydroxylase protein were observed in the hMSCs + rTMS compared to that of controls. Combination therapy significantly elevated the expression of several key neurotrophic factors, of which the highest expression was recorded in the rTMS + hMSC group. In addition, the combination therapy significantly upregulated IL-10 expression while decreased IFN-γ and TNF-α production in a synergistic manner. The treadmill locomotion test (TLT) revealed that motor function was improved in the rTMS + hMSC treatment with synergy. CONCLUSION Our findings demonstrate that rTMS treatment and hMSC transplantation could synergistically create a favorable microenvironment for cell survival within the PD rat brain, through alteration of soluble factors such as neurotrophic/growth factors and anti-/pro-inflammatory cytokines related to neuronal protection or repair, with preservation of DA neurons and improvement of motor functions.
Collapse
Affiliation(s)
- Ji Yong Lee
- Department of Anatomy, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, 26426, Republic of Korea
| | - Hyun Soo Kim
- FCB-Pharmicell Co. Ltd., 520 Sicox Tower, 484 Dunchon-daero, Jungwon-gu, Seongnam-si, Gyeonggi-do, 13229, Republic of Korea
| | - Sung Hoon Kim
- Department and Rehabilitation Medicine, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, 26426, Republic of Korea
| | - Han-Soo Kim
- Department of Biomedical Sciences, Catholic Kwandong University College of Medical Convergence, 24 Beomil-ro, 579 beon-gil, Gangneung-Si, Gangwon-do, 25601, Republic of Korea.
- Basic Research Division, Biomedical Institute of Mycological Resource, College of Medicine, Catholic Kwandong University, 24 Beomil-ro, 579 beon-gil, Gangneung-Si, Gangwon-do, 25601, Republic of Korea.
| | - Byung Pil Cho
- Department of Anatomy, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, 26426, Republic of Korea.
- Institute of Lifestyle Medicine, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, 26426, Republic of Korea.
| |
Collapse
|
10
|
Kim S. Beneficial effects of AAV1-Rheb(S16H) administration in the adult hippocampus. Neural Regen Res 2020; 15:1479-1480. [PMID: 31997810 PMCID: PMC7059574 DOI: 10.4103/1673-5374.274335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
11
|
Bao XQ, Wang L, Yang HY, Hou LY, Wang QS, Zhang D. Induction of glial cell line-derived neurotrophic factor by the squamosamide derivative FLZ in astroglia has neuroprotective effects on dopaminergic neurons. Brain Res Bull 2019; 154:32-42. [PMID: 31669104 DOI: 10.1016/j.brainresbull.2019.10.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 10/08/2019] [Accepted: 10/19/2019] [Indexed: 12/27/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) has neurotrophic activity for the survival of dopaminergic neurons, which is under active investigation for Parkinson's disease (PD) therapy. FLZ is a potential new drug for PD treatment. However, it is unclear whether neurotrophic activity contributes to the neuroprotective effects of FLZ. Here we found that FLZ markedly improved the function of dopaminergic neurons in primary mesencephalic neuron/glia cultures. Further investigation demonstrated that astroglia were required for FLZ to function as a neurotrophic regulator, as FLZ failed to show neurotrophic effects in the absence of astroglia. We clarified that GDNF was responsible for the neurotrophic effects of FLZ since FLZ selectively stimulated GDNF production, which was confirmed by the finding that the neurotrophic effect of FLZ was attenuated by GDNF-neutralizing antibody. Mechanistic study demonstrated that GDNF induction by FLZ was CREB-dependent and that PI3K/Akt was the main pathway regulating CREB activity, which was confirmed by in vivo studies. We also validated that the induction of GDNF by FLZ contributed to PD treatment in vivo. In conclusion, the present data provided evidence that FLZ had robust neurotrophic effects on dopaminergic neurons through sustained induction of GDNF in astroglia by activating the PI3K/Akt/CREB pathway.
Collapse
Affiliation(s)
- Xiu-Qi Bao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College Xian Nong Tan Street, Beijing, 100050, China
| | - Lu Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College Xian Nong Tan Street, Beijing, 100050, China
| | - Han-Yu Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College Xian Nong Tan Street, Beijing, 100050, China
| | - Li-Yan Hou
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Qing-Shan Wang
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China.
| | - Dan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College Xian Nong Tan Street, Beijing, 100050, China.
| |
Collapse
|
12
|
de Oliveira Amaral H, Monge-Fuentes V, Biolchi Mayer A, Alves Campos GA, Soares Lopes K, Camargo LC, Ferroni Schwartz M, Galante P, Mortari MR. Animal venoms: therapeutic tools for tackling Parkinson's disease. Drug Discov Today 2019; 24:2202-2211. [PMID: 31539640 DOI: 10.1016/j.drudis.2019.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/02/2019] [Accepted: 09/10/2019] [Indexed: 12/16/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative pathology of the central nervous system, mainly involving the selective and progressive loss of dopaminergic neurons from the substantia nigra, resulting in motor and non-motor symptoms. PD remains an incurable ailment; thus, treatments are limited to symptom alleviation. With long-term use, conventional treatments can become inefficient, often triggering possible side effects. Considering these drawbacks, drug discovery constantly turns to nature as a source of efficient therapeutics. Thus, this review explores animal venoms as a rich source of bioactive compounds with potent neuropharmacological profiles for the development of effective adjuvant treatments with fewer side effects, ultimately aiming for the neuroprotection of dopaminergic neurons and the symptomatic relief of PD.
Collapse
Affiliation(s)
- Henrique de Oliveira Amaral
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | - Victoria Monge-Fuentes
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil.
| | - Andréia Biolchi Mayer
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | - Gabriel Avohay Alves Campos
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | - Kamila Soares Lopes
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | - Luana C Camargo
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | - Matheus Ferroni Schwartz
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | - Priscilla Galante
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | - Márcia R Mortari
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| |
Collapse
|
13
|
Stenslik MJ, Evans A, Pomerleau F, Weeks R, Huettl P, Foreman E, Turchan-Cholewo J, Andersen A, Cass WA, Zhang Z, Grondin RC, Gash DM, Gerhardt GA, Bradley LH. Methodology and effects of repeated intranasal delivery of DNSP-11 in awake Rhesus macaques. J Neurosci Methods 2018; 303:30-40. [PMID: 29614295 DOI: 10.1016/j.jneumeth.2018.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 11/19/2022]
Abstract
BACKGROUND To determine if the intranasal delivery of neuroactive compounds is a viable, long-term treatment strategy for progressive, chronic neurodegenerative disorders, such as Parkinson's disease (PD), intranasal methodologies in preclinical models comparable to humans are needed. NEW METHOD We developed a methodology to evaluate the repeated intranasal delivery of neuroactive compounds on the non-human primate (NHP) brain, without the need for sedation. We evaluated the effects of the neuroactive peptide, DNSP-11 following repeated intranasal delivery and dose-escalation over the course of 10-weeks in Rhesus macaques. This approach allowed us to examine striatal target engagement, safety and tolerability, and brain distribution following a single 125I-labeled DNSP-11 dose. RESULTS Our initial data support that repeated intranasal delivery and dose-escalation of DNSP-11 resulted in bilateral, striatal target engagement based on neurochemical changes in dopamine (DA) metabolites-without observable, adverse behavioral effects or weight loss in NHPs. Furthermore, a 125I-labeled DNSP-11 study illustrates diffuse rostral to caudal distribution in the brain including the striatum-our target region of interest. COMPARISON WITH EXISTING METHODS The results of this study are compared to our experiments in normal and 6-OHDA lesioned rats, where DNSP-11 was repeatedly delivered intranasally using a micropipette with animals under light sedation. CONCLUSIONS The results from this proof-of-concept study support the utility of our repeated intranasal dosing methodology in awake Rhesus macaques, to evaluate the effects of neuroactive compounds on the NHP brain. Additionally, results indicate that DNSP-11 can be safely and effectively delivered intranasally in MPTP-treated NHPs, while engaging the DA system.
Collapse
Affiliation(s)
- M J Stenslik
- Department of Neuroscience and Brain Restoration Center, University of Kentucky College of Medicine, United States
| | - A Evans
- Department of Neuroscience and Brain Restoration Center, University of Kentucky College of Medicine, United States
| | - F Pomerleau
- Department of Neuroscience and Brain Restoration Center, University of Kentucky College of Medicine, United States
| | - R Weeks
- Department of Neuroscience and Brain Restoration Center, University of Kentucky College of Medicine, United States
| | - P Huettl
- Department of Neuroscience and Brain Restoration Center, University of Kentucky College of Medicine, United States
| | - E Foreman
- Department of Neuroscience and Brain Restoration Center, University of Kentucky College of Medicine, United States
| | - J Turchan-Cholewo
- Department of Neuroscience and Brain Restoration Center, University of Kentucky College of Medicine, United States
| | - A Andersen
- Department of Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky College of Medicine, United States
| | - W A Cass
- Department of Neuroscience and Brain Restoration Center, University of Kentucky College of Medicine, United States
| | - Z Zhang
- Department of Neuroscience and Brain Restoration Center, University of Kentucky College of Medicine, United States
| | - R C Grondin
- Department of Neuroscience and Brain Restoration Center, University of Kentucky College of Medicine, United States
| | - D M Gash
- Department of Neuroscience and Brain Restoration Center, University of Kentucky College of Medicine, United States
| | - G A Gerhardt
- Department of Neuroscience and Brain Restoration Center, University of Kentucky College of Medicine, United States
| | - L H Bradley
- Department of Neuroscience and Brain Restoration Center, University of Kentucky College of Medicine, United States; Department of Molecular & Cellular Biochemistry and Center of Structural Biology, University of Kentucky College of Medicine, United States.
| |
Collapse
|
14
|
Human Umbilical Cord Matrix Stem Cells Reverse Oxidative Stress-Induced Cell Death and Ameliorate Motor Function and Striatal Atrophy in Rat Model of Huntington Disease. Neurotox Res 2018. [DOI: 10.1007/s12640-018-9884-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Treatment with the noradrenaline re-uptake inhibitor atomoxetine alone and in combination with the α2-adrenoceptor antagonist idazoxan attenuates loss of dopamine and associated motor deficits in the LPS inflammatory rat model of Parkinson's disease. Brain Behav Immun 2018; 69:456-469. [PMID: 29339319 DOI: 10.1016/j.bbi.2018.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/04/2018] [Accepted: 01/11/2018] [Indexed: 01/14/2023] Open
Abstract
The impact of treatment with the noradrenaline (NA) re-uptake inhibitor atomoxetine and the α2-adrenoceptor (AR) antagonist idazoxan in an animal model of Parkinson's disease (PD) was assessed. Concurrent systemic treatment with atomoxetine and idazoxan, a combination which serves to enhance the extra-synaptic availability of NA, exerts anti-inflammatory and neuroprotective effects following delivery of an inflammatory stimulus, the bacterial endotoxin, lipopolysaccharide (LPS) into the substantia nigra. Lesion-induced deficits in motor function (akinesia, forelimb-use asymmetry) and striatal dopamine (DA) loss were rescued to varying degrees depending on the treatment. Treatment with atomoxetine following LPS-induced lesion to the substantia nigra, yielded a robust anti-inflammatory effect, suppressing microglial activation and expression of the pro-inflammatory cytokine TNF-α whilst increasing the expression of neurotrophic factors. Furthermore atomoxetine treatment prevented loss of tyrosine hydroxylase (TH) positive nigral dopaminergic neurons and resulted in functional improvements in motor behaviours. Atomoxetine alone was sufficient to achieve most of the observed effects. In combination with idazoxan, an additional improvement in the impairment of contralateral limb use 7 days post lesion and a reduction in amphetamine-mediated rotational asymmetry 14 days post-lesion was observed, compared to atomoxetine or idazoxan treatments alone. The results indicate that increases in central NA tone has the propensity to regulate the neuroinflammatory phenotype in vivo and may act as an endogenous neuroprotective mechanism where inflammation contributes to the progression of DA loss. In accordance with this, the clinical use of agents such as NA re-uptake inhibitors and α2-AR antagonists may prove useful in enhancing the endogenous neuroimmunomodulatory potential of NA in conditions associated with brain inflammation.
Collapse
|
16
|
Protection of nigral dopaminergic neurons by AAV1 transduction with Rheb(S16H) against neurotoxic inflammation in vivo. Exp Mol Med 2018; 50:e440. [PMID: 29422542 PMCID: PMC5903818 DOI: 10.1038/emm.2017.261] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 07/11/2017] [Accepted: 08/01/2017] [Indexed: 11/09/2022] Open
Abstract
We recently reported that adeno-associated virus serotype 1 (AAV1) transduction of murine nigral dopaminergic (DA) neurons with constitutively active ras homolog enriched in brain with a mutation of serine to histidine at position 16 [Rheb(S16H)] induced the production of neurotrophic factors, resulting in neuroprotective effects on the nigrostriatal DA system in animal models of Parkinson’s disease (PD). To further investigate whether AAV1-Rheb(S16H) transduction has neuroprotective potential against neurotoxic inflammation, which is known to be a potential event related to PD pathogenesis, we examined the effects of Rheb(S16H) expression in nigral DA neurons under a neurotoxic inflammatory environment induced by the endogenous microglial activator prothrombin kringle-2 (pKr-2). Our observations showed that Rheb(S16H) transduction played a role in the neuroprotection of the nigrostriatal DA system against pKr-2-induced neurotoxic inflammation, even though there were similar levels of pro-inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukin-1-beta (IL-1β), in the AAV1-Rheb(S16H)-treated substantia nigra (SN) compared to the SN treated with pKr-2 alone; the neuroprotective effects may be mediated by the activation of neurotrophic signaling pathways following Rheb(S16H) transduction of nigral DA neurons. We conclude that AAV1-Rheb(S16H) transduction of neuronal populations to activate the production of neurotrophic factors and intracellular neurotrophic signaling pathways may offer promise for protecting adult neurons from extracellular neurotoxic inflammation.
Collapse
|
17
|
Ngema PN, Mabandla MV. Post 6-OHDA lesion exposure to stress affects neurotrophic factor expression and aggravates motor impairment. Metab Brain Dis 2017; 32:1061-1067. [PMID: 28321600 DOI: 10.1007/s11011-017-9988-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/03/2017] [Indexed: 11/26/2022]
Abstract
Chronic exposure to stress amplifies locomotor deficits and exacerbates dopamine neuron loss in an animal model for Parkinson's disease. The release of neurotrophic factors such as glial cell-line derived neurotrophic factor (GDNF) and neurotrophin-3 (NT-3) following neuronal injury attenuates exacerbated degeneration of these neurons. In this study, the neurotoxin 6-hydroxydopamine (6-OHDA) was injected unilaterally into the medial forebrain bundle of male Sprague Dawley rats. A subset of these rats was subjected to post-lesion restraint stress after which the effect of exposure to stress on locomotor activity (forelimb akinesia test), neurotrophic factor (GDNF and NT-3) and corticosterone concentration was assessed. Exposure to post-lesion stress resulted in increased preference to use the unimpaired forelimb (forelimb ipsilateral to the lesioned hemisphere) in the forelimb akinesia test. The expected increase in both GDNF and NT-3 concentration following injury was not present in the stressed animals. However, both the non-stressed and stressed lesioned groups had decreased neurotrophic factor concentration at one and two weeks post lesion. This decrease was exaggerated in the stressed rats. The decrease in neurotrophic factor concentration was accompanied by an increase in corticosterone concentration in the stressed rats. These findings demonstrate that exposure to post-6-OHDA lesion stress exaggerates dopamine neurodegeneration and enhance motor impairment. This suggests that conditions that result in a hyper-activated hypothalamic-pituitary-adrenal axis such as depression which is concomitant to a Parkinson's disease diagnosis may be responsible for enhanced dopamine depletion by attenuating neurotrophic factor concentration elevation in the nigrostriatal pathway following neuronal injury.
Collapse
Affiliation(s)
- Phumzile Nomfundo Ngema
- University of KwaZulu-Natal College of Health Sciences, Kwazulu-Natal, Durban, South Africa.
| | - Musa Vuyisile Mabandla
- University of KwaZulu-Natal College of Health Sciences, Kwazulu-Natal, Durban, South Africa
| |
Collapse
|
18
|
Differential behavioral outcomes following neonatal versus fetal human retinal pigment epithelial cell striatal implants in parkinsonian rats. J Neural Transm (Vienna) 2017; 124:455-462. [PMID: 28160153 DOI: 10.1007/s00702-017-1683-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/16/2017] [Indexed: 02/04/2023]
Abstract
Following the failure of a Phase II clinical study evaluating human retinal pigment epithelial (hRPE) cell implants as a potential treatment option for Parkinson's disease, speculation has centered on implant function and survival as possible contributors to the therapeutic outcomes. We recently reported that neonatal hRPE cells, similar to hRPE cells used in the Phase II clinical study, produced short-lived in vitro and limited in vivo trophic factors, which supports that assumption. We hypothesize that the switch from fetal to neonatal hRPE cells, between the Phase I and the Phase II clinical trial may be partly responsible for the later negative outcomes. To investigate this hypothesis, we used two neonatal hRPE cell lots, prepared in a similar manner to neonatal hRPE cells used in the Phase II clinical study, and compared them to previously evaluated fetal hRPE cells for behavioral changes following unilateral striatal implantation in 6-hydroxydopamine-lesioned rats. The results showed that only fetal, not neonatal, hRPE cell implants, were able to improve behavioral outcomes following striatal implantation in the lesioned rats. These data suggest that fetal hRPE cells may be preferential to neonatal hRPE cells in restoring behavioral deficits.
Collapse
|
19
|
Impellizzeri D, Campolo M, Bruschetta G, Crupi R, Cordaro M, Paterniti I, Cuzzocrea S, Esposito E. Traumatic Brain Injury Leads to Development of Parkinson's Disease Related Pathology in Mice. Front Neurosci 2016; 10:458. [PMID: 27790086 PMCID: PMC5061819 DOI: 10.3389/fnins.2016.00458] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 09/22/2016] [Indexed: 02/03/2023] Open
Abstract
Traumatic brain injury (TBI) is a major health and socio-economic problem that affects all societies. This condition results from the application of external physical strength to the brain that leads to transitory or permanent structural and functional impairments. Moreover, TBI is a risk factor for neurodegeneration and can e.g., increase the risk for Parkinson's disease (PD), a late-onset neurodegenerative disorder with loss of dopaminergic neurons in substantia nigra. In this study, we wanted to explore the possible development of PD-related pathology within the context of an experimental model of TBI. Traumatic brain injury was induced in mice by controlled cortical impact. At different time points behavioral tests (open field, elevated plus maze tests, and Barnes maze) were performed: The animals were sacrificed 30 days after the impact and the brains were processed for Western blot and immunohistochemical analyses. Following TBI there was a significant decrease in expression of tyrosine hydroxylase and dopamine transporter in the substantia nigra as well as significant behavioral alterations. In addition, a strong increase in neuroinflammation was evident, as shown by increased levels of cyclooxygenase-2 and inducible nitric oxide synthase as well as IκB-α degradation and nuclear-κB translocation. Moreover, neurotrophic factors such as brain-derived neurotrophic factor, neurotrophin-3, nerve growth factor, and glial cell line-derived neurotrophic factor were decreased 30 days post-TBI. Interestingly, we observed a significant accumulation of α-synuclein in microglia compared to astrocytes. This study suggests that PD-related molecular events can be triggered upon TBI. The biological mechanisms linking brain trauma and neurodegenerative diseases need to be further investigated.
Collapse
Affiliation(s)
- Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina Messina, Italy
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina Messina, Italy
| | - Giuseppe Bruschetta
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina Messina, Italy
| | - Rosalia Crupi
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina Messina, Italy
| | - Marika Cordaro
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of MessinaMessina, Italy; Department of Pharmacology and Physiology, Saint Louis UniversitySt. Louis, MO, USA
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina Messina, Italy
| |
Collapse
|
20
|
Jaumotte JD, Wyrostek SL, Zigmond MJ. Protection of cultured dopamine neurons from MPP(+) requires a combination of neurotrophic factors. Eur J Neurosci 2016; 44:1691-9. [PMID: 27098376 DOI: 10.1111/ejn.13252] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/15/2016] [Accepted: 04/01/2016] [Indexed: 11/30/2022]
Abstract
Parkinson's disease is a progressive neurodegenerative disorder, caused in part by the loss of dopamine (DA) neurons in the substantia nigra (SN). Neurotrophic factors have been shown to increase the basal survival of DA neurons in vitro, as well as to protect the neurons from some toxins under certain in vitro conditions and in animal models. Although these factors have often been tested individually, they have rarely been studied in combinations. We therefore examined the effect of such combinations after acute exposure to the toxin 1-methyl-4-phenylpyridinium (MPP(+) ) using dissociated postnatal rat midbrain cultures isolated from SN and ventral tegmental area (VTA). We found that significant loss of DA neurons in the SN occurred with an LC50 of between 1 and 10 μm, whereas the LC50 of DA neurons from the VTA was approximately 1000-fold higher. We did not observe neuroprotection against MPP(+) by individual exposure to glial cell-line derived neurotrophic factor (GDNF), brain derived neurotrophic factor (BDNF), transforming growth factor beta (TGFβ), basic fibroblast growth factor (FGF-2) or growth/differentiation factor 5 (GDF5) at concentrations of 100 or 500 ng/mL. Combinations of two, three or four neurotrophic factors were also ineffective. However, when the SN cultures were exposed to a combination of all five neurotrophic factors, each at a concentration of 100 ng/mL, we observed a 30% increase in DA neuron survival in the presence of 10 and 500 μm MPP(+) . These results may be relevant to the use of neurotrophic factors as therapeutic treatments for Parkinson's disease.
Collapse
Affiliation(s)
- Juliann D Jaumotte
- Pittsburgh Institute for Neurodegenerative Disease, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephanie L Wyrostek
- Pittsburgh Institute for Neurodegenerative Disease, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael J Zigmond
- Pittsburgh Institute for Neurodegenerative Disease, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
21
|
Machado V, Gilsbach R, Das R, Schober A, Bogatyreva L, Hauschke D, Krieglstein K, Unsicker K, Spittau B. Gdf-15 deficiency does not alter vulnerability of nigrostriatal dopaminergic system in MPTP-intoxicated mice. Cell Tissue Res 2016; 365:209-23. [PMID: 27115420 DOI: 10.1007/s00441-016-2406-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/06/2016] [Indexed: 12/25/2022]
Abstract
Growth/differentiation factor-15 (Gdf-15) is a member of the transforming growth factor-β (Tgf-β) superfamily and has been shown to be a potent neurotrophic factor for midbrain dopaminergic (DAergic) neurons both in vitro and in vivo. Gdf-15 has also been shown to be involved in inflammatory processes. The aim of this study was to identify the role of endogenous Gdf-15 in the MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) mouse model of Parkinson's disease (PD) by comparing Gdf-15 (+/+) and Gdf-15 (-/-) mice. At 4 days and 14 days post-MPTP administration, both Gdf-15 (+/+) and Gdf-15 (-/-) mice showed a similar decline in DAergic neuron numbers and in striatal dopamine (DA) levels. This was followed by a comparable restorative phase at 90 days and 120 days, indicating that the absence of Gdf-15 does not affect the susceptibility or the recovery capacity of the nigrostriatal system after MPTP administration. The MPTP-induced microglial and astrocytic response was not significantly altered between the two genotypes. However, pro-inflammatory and anti-inflammatory cytokine profiling revealed the differential expression of markers in Gdf-15 (+/+) and Gdf-15 (-/-) mice after MPTP administration. Thus, the MPTP mouse model fails to uncover a major role of endogenous Gdf-15 in the protection of MPTP-lesioned nigrostriatal DAergic neurons, in contrast to its capacity to protect the 6-hydroxydopamine-intoxicated nigrostriatal system.
Collapse
Affiliation(s)
- Venissa Machado
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Ralf Gilsbach
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, 79104, Freiburg, Germany
| | - Richa Das
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany.,German Center for Neurodegenerative Diseases, 53115, Bonn, Germany
| | - Andreas Schober
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany
| | - Lioudmila Bogatyreva
- Institute of Medical Biometry and Medical Informatics, University of Freiburg, 79104, Freiburg, Germany
| | - Dieter Hauschke
- German Center for Neurodegenerative Diseases, 53115, Bonn, Germany
| | - Kerstin Krieglstein
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany
| | - Klaus Unsicker
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany.
| | - Björn Spittau
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany.
| |
Collapse
|
22
|
β-Asarone Inhibits IRE1/XBP1 Endoplasmic Reticulum Stress Pathway in 6-OHDA-Induced Parkinsonian Rats. Neurochem Res 2016; 41:2097-101. [PMID: 27097550 DOI: 10.1007/s11064-016-1922-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 04/08/2016] [Accepted: 04/13/2016] [Indexed: 10/21/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease, with genetics and environment contributing to the disease onset. The limited pathological cognize of the disease restrained the approaches to improve the clinical treatment. Recently, studies showed that endoplasmic reticulum (ER) stress played an important role in the pathogenesis of PD. There was a neuroprotective effect partly mediated by modulating ER stress. β-Asarone is the essential constituent of Acorus tatarinowii Schott volatile oil. Our team observed that β-asarone could improve the behavior of parkinsonian rats; increase the HVA, Dopacl, and 5-HIAA levels; and reduce α-synuclein levels. Here we assumed that the protective role of β-asarone on parkinsonian rats was mediated via ER stress pathway. To prove the hypothesis we investigated the mRNA levels of glucose regulated protein 78 (GRP78) and C/EBP homologous binding protein (CHOP) in 6-hydroxy dopamine (6-OHDA) induced parkinsonian rats after β-asarone treatment. Furthermore, the inositol-requiring enzyme 1/X-Box Binding Protein 1 (IRE1/XBP1) ER stress pathway was also studied. The results showed that β-asarone inhibited the mRNA levels of GRP78 and CHOP, accompanied with the delined expressions of phosphorylated IER1 (p-IRE1) and XBP1. We deduced that β-asarone might have a protective effect on the 6-OHDA induced parkinsonian rats via IRE1/XBP1 Pathway. Collectively, all data indicated that β-asarone might be a potential candidate of medicine for clinical therapy of PD.
Collapse
|
23
|
Kim HD, Jeong KH, Jung UJ, Kim SR. Naringin treatment induces neuroprotective effects in a mouse model of Parkinson's disease in vivo, but not enough to restore the lesioned dopaminergic system. J Nutr Biochem 2016; 28:140-6. [DOI: 10.1016/j.jnutbio.2015.10.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 09/30/2015] [Accepted: 10/16/2015] [Indexed: 12/29/2022]
|
24
|
Gene Therapy of CNS Disorders Using Recombinant AAV Vectors. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
25
|
Kramer ER, Liss B. GDNF-Ret signaling in midbrain dopaminergic neurons and its implication for Parkinson disease. FEBS Lett 2015; 589:3760-72. [DOI: 10.1016/j.febslet.2015.11.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/29/2015] [Accepted: 11/03/2015] [Indexed: 12/11/2022]
|
26
|
Neonatal human retinal pigment epithelial cells secrete limited trophic factors in vitro and in vivo following striatal implantation in parkinsonian rats. J Neural Transm (Vienna) 2015; 123:167-77. [PMID: 26546037 DOI: 10.1007/s00702-015-1480-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/23/2015] [Indexed: 01/30/2023]
Abstract
Human retinal pigment epithelial (hRPE) cell implants into the striatum have been investigated as a potential cell-based treatment for Parkinson's disease in a Phase II clinical trial that recently failed. We hypothesize that the trophic factor potential of the hRPE cells could potentially influence the function and/or survival of the implants and may be involved in an alternative mechanism of action. However, it is unclear if hRPE cells secreted trophic factors when handled in the manner used in the clinical Phase II trial. To address these questions, we investigated two neonatal hRPE cell lots, cultured in a similar manner to hRPE cells used in a Phase II clinical study, and longitudinally determined brain-derived neurotrophic factor (BDNF), fibroblast growth factor 2 (FGF2), and pigment epithelium-derived factor concentrations in vitro and following striatal implantation into 6-hydroxydopamine-lesioned rats. The results demonstrate short-lived BDNF and FGF2 concentrations in vitro from hRPE cells grown alone or attached to gelatin microcarriers (GM)s as well as limited trophic factor concentration differences in vivo following striatal implantation of hRPE-GM in 6-hydroxydopamine lesioned rats compared to sham (GM-only). The data suggest that trophic factors from neonatal hRPE cell implants likely did not participate in an alternative mechanism of action, which adds supports to a hypothesis that additional factors may have been necessary for the survival and/or function of hRPE implants and potentially the success of the Phase II clinical trial.
Collapse
|
27
|
Tsybko AS, Il’chibaeva TV, Naumenko VS. The effects of the glial cell line-derived neurotrophic factor (GDNF) on the levels of mRNA of apoptotic genes Bax and Bcl-xl in the brain of mice genetically predisposed to pathological behavior. ACTA ACUST UNITED AC 2015. [DOI: 10.1134/s2079059715040152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
28
|
Stenslik MJ, Potts LF, Sonne JWH, Cass WA, Turchan-Cholewo J, Pomerleau F, Huettl P, Ai Y, Gash DM, Gerhardt GA, Bradley LH. Methodology and effects of repeated intranasal delivery of DNSP-11 in a rat model of Parkinson's disease. J Neurosci Methods 2015; 251:120-9. [PMID: 25999268 DOI: 10.1016/j.jneumeth.2015.05.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/11/2015] [Accepted: 05/11/2015] [Indexed: 01/21/2023]
Abstract
BACKGROUND To circumvent the challenges associated with delivering large compounds directly to the brain for the treatment of Parkinson's disease (PD), non-invasive procedures utilizing smaller molecules with protective and/or restorative actions on dopaminergic neurons are needed. NEW METHOD We developed a methodology for evaluating the effects of a synthetic neuroactive peptide, DNSP-11, on the nigrostriatal system using repeated intranasal delivery in both normal and a unilateral 6-hydroxydopamine (6-OHDA) lesion rat model of PD. RESULTS Normal rats repeatedly administered varying doses of DNSP-11 intranasally for 3 weeks exhibited a significant increase in dopamine (DA) turnover in both the striatum and substantia nigra (SN) at 300μg, suggestive of a stimulative effect of the dopaminergic system. Additionally, a protective effect was observed following repeated intranasal administration in 6-OHDA lesioned rats, as suggested by: a significant decrease in d-amphetamine-induced rotation at 2 weeks; a decrease in DA turnover in the lesioned striatum; and an increased sparing of tyrosine hydroxylase (TH) positive (+) neurons in a specific sub-region of the lesioned substantia nigra pars compacta (SNpc). Finally, tracer studies showed (125)I-DNSP-11 distributed diffusely throughout the brain, including the striatum and SN, as quickly as 30min after a single intranasal dose. COMPARISON WITH EXISTING METHODS The results of bilateral intranasal administration of DNSP-11 are compared to our unilateral single infusion studies to the brain in rats. CONCLUSIONS These studies support that DNSP-11 can be delivered intranasally and maintain its neuroactive properties in both normal rats and in a unilateral 6-OHDA rat model of PD.
Collapse
Affiliation(s)
- Mallory J Stenslik
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Lisa F Potts
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - James W H Sonne
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Wayne A Cass
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Jadwiga Turchan-Cholewo
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Francois Pomerleau
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Peter Huettl
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Yi Ai
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Don M Gash
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Greg A Gerhardt
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Luke H Bradley
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA; Department of Molecular & Cellular Biochemistry and Center of Structural Biology, University of Kentucky College of Medicine, Lexington, KY, USA.
| |
Collapse
|
29
|
|
30
|
Bartus RT, Kordower JH, Johnson EM, Brown L, Kruegel BR, Chu Y, Baumann TL, Lang AE, Olanow CW, Herzog CD. Post-mortem assessment of the short and long-term effects of the trophic factor neurturin in patients with α-synucleinopathies. Neurobiol Dis 2015; 78:162-71. [PMID: 25841760 DOI: 10.1016/j.nbd.2015.03.023] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 03/13/2015] [Accepted: 03/23/2015] [Indexed: 11/29/2022] Open
Abstract
Substantial interest persists for developing neurotrophic factors to treat neurodegenerative diseases. At the same time, significant progress has been made in implementing gene therapy as a means to provide long-term expression of bioactive neurotrophic factors to targeted sites in the brain. Nonetheless, to date, no double-blind clinical trial has achieved positive results on its primary endpoint despite robust benefits achieved in animal models. A major issue with advancing the field is the paucity of information regarding the expression and effects of neurotrophic factors in human neurodegenerative brain, relative to the well-characterized responses in animal models. To help fill this information void, we examined post-mortem brain tissue from four patients with nigrostriatal degeneration who had participated in clinical trials testing gene delivery of neurturin to the putamen of patients. Each had died of unrelated causes ranging from 1.5-to-3-months (2 Parkinson's disease patients), to 4+-years (1 Parkinson's disease and 1 multiple-system atrophy-parkinsonian type patient) following gene therapy. Quantitative and immunohistochemical evaluation of neurturin, alpha-synuclein, tyrosine hydroxylase (TH) and an oligodendroglia marker (Olig 2) were performed in each brain. Comparable volumes-of-expression of neurturin were seen in the putamen in all cases (~15-22%; mean=18.5%). TH-signal in the putamen was extremely sparse in the shorter-term cases. A 6-fold increase was seen in longer-term cases, but was far less than achieved in animal models of nigrostriatal degeneration with similar or even far less NRTN exposure. Less than 1% of substantia nigra (SN) neurons stained for neurturin in the shorter-term cases. A 15-fold increase was seen in the longer-term cases, but neurturin was still only detected in ~5% of nigral cells. These data provide unique insight into the functional status of advanced, chronic nigrostriatal degeneration in human brain and the response of these neurons to neurotrophic factor stimulation. They demonstrate mild but persistent expression of gene-mediated neurturin over 4-years, with an apparent, time-related amplification of its transport and biological effects, albeit quite weak, and provide unique information to help plan and design future trials.
Collapse
Affiliation(s)
- R T Bartus
- RTBioconsultants, Inc., San Diego, CA, USA; Ceregene, Inc, USA.
| | - J H Kordower
- Rush Presbyterian Medical Center, Chicago, IL, USA
| | - E M Johnson
- Washington University Medical School, St. Louis, MO, USA
| | | | | | - Y Chu
- Rush Presbyterian Medical Center, Chicago, IL, USA
| | - T L Baumann
- Isis Pharmaceuticals, Carlsbad, CA, USA; Ceregene, Inc, USA
| | - A E Lang
- Toronto Western Hospital, Toronto, Canada
| | - C W Olanow
- Mount Sinai School of Medicine, NYC, USA
| | | |
Collapse
|
31
|
Sygnecka K, Heider A, Scherf N, Alt R, Franke H, Heine C. Mesenchymal stem cells support neuronal fiber growth in an organotypic brain slice co-culture model. Stem Cells Dev 2014; 24:824-35. [PMID: 25390472 DOI: 10.1089/scd.2014.0262] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been identified as promising candidates for neuroregenerative cell therapies. However, the impact of different isolation procedures on the functional and regenerative characteristics of MSC populations has not been studied thoroughly. To quantify these differences, we directly compared classically isolated bulk bone marrow-derived MSCs (bulk BM-MSCs) to the subpopulation Sca-1(+)Lin(-)CD45(-)-derived MSCs(-) (SL45-MSCs), isolated by fluorescence-activated cell sorting from bulk BM-cell suspensions. Both populations were analyzed with respect to functional readouts, that are, frequency of fibroblast colony forming units (CFU-f), general morphology, and expression of stem cell markers. The SL45-MSC population is characterized by greater morphological homogeneity, higher CFU-f frequency, and significantly increased nestin expression compared with bulk BM-MSCs. We further quantified the potential of both cell populations to enhance neuronal fiber growth, using an ex vivo model of organotypic brain slice co-cultures of the mesocortical dopaminergic projection system. The MSC populations were cultivated underneath the slice co-cultures without direct contact using a transwell system. After cultivation, the fiber density in the border region between the two brain slices was quantified. While both populations significantly enhanced fiber outgrowth as compared with controls, purified SL45-MSCs stimulated fiber growth to a larger degree. Subsequently, we analyzed the expression of different growth factors in both cell populations. The results show a significantly higher expression of brain-derived neurotrophic factor (BDNF) and basic fibroblast growth factor in the SL45-MSCs population. Altogether, we conclude that MSC preparations enriched for primary MSCs promote neuronal regeneration and axonal regrowth, more effectively than bulk BM-MSCs, an effect that may be mediated by a higher BDNF secretion.
Collapse
Affiliation(s)
- Katja Sygnecka
- 1 Translational Centre for Regenerative Medicine (TRM), University of Leipzig , Leipzig, Germany
| | | | | | | | | | | |
Collapse
|
32
|
Jeon MT, Kim SR. Roles of Rheb(S16H) in substantia nigra pars compacta dopaminergic neurons in vivo. Biomed Rep 2014; 3:137-140. [PMID: 25798236 DOI: 10.3892/br.2014.397] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 11/01/2014] [Indexed: 11/06/2022] Open
Abstract
Although there are ongoing intensive research efforts, no effective pharmacological therapies for Parkinson's disease (PD) have been developed thus far. However, with the development of efficient gene delivery systems, gene therapy for PD has become a focus of research and increasing evidence suggests that continuous production of neurotrophic factors play a significant role in the functional restoration of the nigrostriatal dopaminergic (DA) system. Our recent study reported that the transduction of DA neurons with ras homolog enriched in brain, which has an S16H mutation [Rheb(S16H)], protected the nigrostriatal DA projection in a neurotoxin model of PD in vivo. In addition, Rheb(S16H) expression significantly increased the levels of glial cell line-derived neurotrophic factor and brain-derived neurotrophic factor, which contributed to the neuroprotective effects of Rheb(S16H) in DA neurons in the adult brain, indicating that the activation of the signaling pathways involved in cell survival by a specific gene delivery, such as Rheb(S16H) to adult neurons, may be a useful strategy to protect neural systems in the adult brain. In the present study, a brief overview of our recent studies is provided, which demonstrates the neuroprotective mechanisms of Rheb(S16H) on the nigrostriatal DA projection in the adult brain.
Collapse
Affiliation(s)
- Min-Tae Jeon
- School of Life Sciences, Kyungpook National University, Daegu 700-842, Republic of Korea ; BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 700-842, Republic of Korea
| | - Sang Ryong Kim
- School of Life Sciences, Kyungpook National University, Daegu 700-842, Republic of Korea ; BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 700-842, Republic of Korea ; Institute of Life Science and Biotechnology, Kyungpook National University, Daegu 702-701, Republic of Korea ; Brain Science and Engineering Institute, Kyungpook National University, Daegu 700-842, Republic of Korea
| |
Collapse
|
33
|
Kim SR. Mammalian target of rapamycin complex 1 as an inducer of neurotrophic factors in dopaminergic neurons. Neural Regen Res 2014; 9:2036-7. [PMID: 25657714 PMCID: PMC4316461 DOI: 10.4103/1673-5374.147923] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2014] [Indexed: 11/04/2022] Open
Affiliation(s)
- Sang Ryong Kim
- School of Life Sciences, BK21 plus KNU Creative BioResearch Group, Institute of Life Science & Biotechnology, Kyungpook National University, Daegu 702-701, Korea; Brain Science and Engineering Institute, Kyungpook National University, Daegu 700-842, Korea
| |
Collapse
|
34
|
Jung UJ, Kim SR. Effects of naringin, a flavanone glycoside in grapefruits and citrus fruits, on the nigrostriatal dopaminergic projection in the adult brain. Neural Regen Res 2014; 9:1514-7. [PMID: 25317167 PMCID: PMC4192967 DOI: 10.4103/1673-5374.139476] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2014] [Indexed: 11/30/2022] Open
Abstract
Recently, we have demonstrated the ability of naringin, a well-known flavanone glycoside of grapefruits and citrus fruits, to prevent neurodegeneration in a neurotoxin model of Parkinson's disease. Intraperitoneal injection of naringin protected the nigrostriatal dopaminergic projection by increasing glial cell line-derived neurotrophic factor expression and decreasing the level of tumor necrosis factor-alpha in dopaminergic neurons and microglia, respectively. These results suggest that naringin can impart to the adult dopaminergic neurons the ability to produce glial cell line-derived neurotrophic factor against Parkinson's disease with anti-inflammatory effects. Based on these results, we would like to describe an important perspective on its possibility as a therapeutic agent for Parkinson's disease.
Collapse
Affiliation(s)
- Un Ju Jung
- Center for Food and Nutritional Genomics Research, Kyungpook National University, Daegu, Korea
| | - Sang Ryong Kim
- School of Life Sciences, Kyungpook National University, Daegu, Korea ; BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea ; Institute of Life Science & Biotechnology, Kyungpook National University, Daegu, Korea ; Brain Science and Engineering Institute, Kyungpook National University, Daegu, Korea
| |
Collapse
|
35
|
Brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor inhibit ferrous iron influx via divalent metal transporter 1 and iron regulatory protein 1 regulation in ventral mesencephalic neurons. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2967-75. [PMID: 25239763 DOI: 10.1016/j.bbamcr.2014.09.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 08/26/2014] [Accepted: 09/09/2014] [Indexed: 11/20/2022]
Abstract
Iron accumulation is observed in the substantia nigra of patients with Parkinson's disease. However, it is unknown whether neurotrophic factors, brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) participate in the modulation of neuronal iron metabolism. Here, we investigated the effects and underlying mechanisms of BDNF and GDNF on the iron influx process in primary cultured ventral mesencephalic neurons. 6-hydroxydopamine-induced enhanced ferrous iron influx via improper up-regulation of divalent metal transporter 1 with iron responsive element (DMT1+IRE) was consistently relieved by BDNF and GDNF. Both the mRNA and protein levels of DMT1+IRE were down-regulated by BDNF or GDNF treatment alone. We further demonstrated the involvement of iron regulatory protein 1 (IRP1) in BDNF- and GDNF-induced DMT1+IRE expression. Extracellular-regulated kinase 1/2 (ERK1/2) and Akt were activated and participated in these processes. Inhibition of ERK1/2 and Akt phosphorylation abolished the down-regulation of IRP1 and DMT1+IRE induced by BDNF and GDNF. Taken together, these results show that BDNF and GDNF ameliorate iron accumulation via the ERK/Akt pathway, followed by inhibition of IRP1 and DMT1+IRE expression, which may provide new targets for the neuroprotective effects of these neurotrophic factors.
Collapse
|
36
|
Schapira AHV, Olanow CW, Greenamyre JT, Bezard E. Slowing of neurodegeneration in Parkinson's disease and Huntington's disease: future therapeutic perspectives. Lancet 2014; 384:545-55. [PMID: 24954676 DOI: 10.1016/s0140-6736(14)61010-2] [Citation(s) in RCA: 280] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Several important advances have been made in our understanding of the pathways that lead to cell dysfunction and death in Parkinson's disease and Huntington's disease. These advances have been informed by both direct analysis of the post-mortem brain and by study of the biological consequences of the genetic causes of these diseases. Some of the pathways that have been implicated so far include mitochondrial dysfunction, oxidative stress, kinase pathways, calcium dysregulation, inflammation, protein handling, and prion-like processes. Intriguingly, these pathways seem to be important in the pathogenesis of both diseases and have led to the identification of molecular targets for candidate interventions designed to slow or reverse their course. We review some recent advances that underlie putative therapies for neuroprotection in Parkinson's disease and Huntington's disease, and potential targets that might be exploited in the future. Although we will need to overcome important hurdles, especially in terms of clinical trial design, we propose several target pathways that merit further study. In Parkinson's disease, these targets include agents that might improve mitochondrial function or increase degradation of defective mitochondria, kinase inhibitors, calcium channel blockers, and approaches that interfere with the misfolding, templating, and transmission of α-synuclein. In Huntington's disease, strategies might also be directed at mitochondrial bioenergetics and turnover, the prevention of protein dysregulation, disruption of the interaction between huntingtin and p53 or huntingtin-interacting protein 1 to reduce apoptosis, and interference with expression of mutant huntingtin at both the nucleic acid and protein levels.
Collapse
Affiliation(s)
| | - C Warren Olanow
- Departments of Neurology and Neuroscience, Mount Sinai School of Medicine, New York, NY, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Erwan Bezard
- Université de Bordeaux, Institut des Maladies Neurodégénératives, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, 33000 Bordeaux, France
| |
Collapse
|
37
|
Cabezas R, Avila M, Gonzalez J, El-Bachá RS, Báez E, García-Segura LM, Jurado Coronel JC, Capani F, Cardona-Gomez GP, Barreto GE. Astrocytic modulation of blood brain barrier: perspectives on Parkinson's disease. Front Cell Neurosci 2014; 8:211. [PMID: 25136294 PMCID: PMC4120694 DOI: 10.3389/fncel.2014.00211] [Citation(s) in RCA: 272] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/14/2014] [Indexed: 12/21/2022] Open
Abstract
The blood–brain barrier (BBB) is a tightly regulated interface in the Central Nervous System (CNS) that regulates the exchange of molecules in and out from the brain thus maintaining the CNS homeostasis. It is mainly composed of endothelial cells (ECs), pericytes and astrocytes that create a neurovascular unit (NVU) with the adjacent neurons. Astrocytes are essential for the formation and maintenance of the BBB by providing secreted factors that lead to the adequate association between the cells of the BBB and the formation of strong tight junctions. Under neurological disorders, such as chronic cerebral ischemia, brain trauma, Epilepsy, Alzheimer and Parkinson’s Diseases, a disruption of the BBB takes place, involving a lost in the permeability of the barrier and phenotypical changes in both the ECs and astrocytes. In this aspect, it has been established that the process of reactive gliosis is a common feature of astrocytes during BBB disruption, which has a detrimental effect on the barrier function and a subsequent damage in neuronal survival. In this review we discuss the implications of astrocyte functions in the protection of the BBB, and in the development of Parkinson’s disease (PD) and related disorders. Additionally, we highlight the current and future strategies in astrocyte protection aimed at the development of restorative therapies for the BBB in pathological conditions.
Collapse
Affiliation(s)
- Ricardo Cabezas
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, D.C., Colombia
| | - Marcos Avila
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, D.C., Colombia
| | - Janneth Gonzalez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, D.C., Colombia
| | | | - Eliana Báez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, D.C., Colombia
| | | | - Juan Camilo Jurado Coronel
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, D.C., Colombia
| | - Francisco Capani
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Facultad de Medicina, Instituto de Investigaciones cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, Buenos Aires Argentina
| | - Gloria Patricia Cardona-Gomez
- Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, Faculty of Medicine, SIU, University of Antioquia UdeA Medellín, Colombia
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, D.C., Colombia
| |
Collapse
|
38
|
Xiao N, Lin Y, Cao H, Sirjani D, Giaccia AJ, Koong AC, Kong CS, Diehn M, Le QT. Neurotrophic factor GDNF promotes survival of salivary stem cells. J Clin Invest 2014; 124:3364-77. [PMID: 25036711 DOI: 10.1172/jci74096] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 05/19/2014] [Indexed: 12/12/2022] Open
Abstract
Stem cell-based regenerative therapy is a promising treatment for head and neck cancer patients that suffer from chronic dry mouth (xerostomia) due to salivary gland injury from radiation therapy. Current xerostomia therapies only provide temporary symptom relief, while permanent restoration of salivary function is not currently feasible. Here, we identified and characterized a stem cell population from adult murine submandibular glands. Of the different cells isolated from the submandibular gland, this specific population, Lin-CD24+c-Kit+Sca1+, possessed the highest capacity for proliferation, self renewal, and differentiation during serial passage in vitro. Serial transplantations of this stem cell population into the submandibular gland of irradiated mice successfully restored saliva secretion and increased the number of functional acini. Gene-expression analysis revealed that glial cell line-derived neurotrophic factor (Gdnf) is highly expressed in Lin-CD24+c-Kit+Sca1+ stem cells. Furthermore, GDNF expression was upregulated upon radiation therapy in submandibular glands of both mice and humans. Administration of GDNF improved saliva production and enriched the number of functional acini in submandibular glands of irradiated animals and enhanced salisphere formation in cultured salivary stem cells, but did not accelerate growth of head and neck cancer cells. These data indicate that modulation of the GDNF pathway may have potential therapeutic benefit for management of radiation-induced xerostomia.
Collapse
|
39
|
Jung UJ, Leem E, Kim SR. Naringin: a protector of the nigrostriatal dopaminergic projection. Exp Neurobiol 2014; 23:124-9. [PMID: 24963276 PMCID: PMC4065825 DOI: 10.5607/en.2014.23.2.124] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 05/14/2014] [Accepted: 05/14/2014] [Indexed: 01/15/2023] Open
Abstract
Parkinson's disease is the second most common neurodegenerative disorder characterized by the progressive degeneration of dopaminergic neurons and a biochemical reduction of striatal dopamine levels. Despite the lack of fully understanding of the etiology of Parkinson's disease, accumulating evidences suggest that Parkinson's disease may be caused by the insufficient support of neurotrophic factors, and by microglial activation, resident immune cells in the brain. Naringin, a major flavonone glycoside in grapefruits and citrus fruits, is considered as a protective agent against neurodegenerative diseases because it can induce not only anti-oxidant effects but also neuroprotective effects by the activation of anti-apoptotic pathways and the induction of neurotrophic factors such as brain-derived neurotrophic factor and vascular endothelial growth factor. We have recently reported that naringin has neuroprotective effects in a neurotoxin model of Parkinson's disease. Our observations show that intraperitoneal injection of naringin induces increases in glial cell line-derived neurotrophic factor expression and mammalian target of rapamycin complex 1 activity in dopaminergic neurons of rat brains with anti-inflammatory effects. Moreover, the production of glial cell line-derived neurotrophic factor by naringin treatment contributes to the protection of the nigrostriatal dopaminergic projection in a neurotoxin model of Parkinson's disease. Although the effects of naringin on the nigrostriatal dopaminergic system in human brains are largely unknown, these results suggest that naringin may be a beneficial natural product for the prevention of dopaminergic degeneration in the adult brain.
Collapse
Affiliation(s)
- Un Ju Jung
- Center for Food and Nutritional Genomics Research, Kyungpook National University, Daegu 702-701, Korea
| | - Eunju Leem
- School of Life Sciences, Kyungpook National University, Daegu 702-701, Korea. ; BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 702-701, Korea
| | - Sang Ryong Kim
- School of Life Sciences, Kyungpook National University, Daegu 702-701, Korea. ; BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 702-701, Korea. ; Institute of Life Science & Biotechnology, Kyungpook National University, Daegu 702-701, Korea. ; Brain Science and Engineering Institute, Kyungpook National University, Daegu 700-842, Korea
| |
Collapse
|
40
|
Nam JH, Leem E, Jeon MT, Jeong KH, Park JW, Jung UJ, Kholodilov N, Burke RE, Jin BK, Kim SR. Induction of GDNF and BDNF by hRheb(S16H) transduction of SNpc neurons: neuroprotective mechanisms of hRheb(S16H) in a model of Parkinson's disease. Mol Neurobiol 2014; 51:487-99. [PMID: 24859383 DOI: 10.1007/s12035-014-8729-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 04/29/2014] [Indexed: 11/30/2022]
Abstract
The transduction of dopaminergic (DA) neurons with human ras homolog enriched in brain, which has a S16H mutation [hRheb(S16H)] protects the nigrostriatal DA projection in the 6-hydroxydopamine (6-OHDA)-treated animal model of Parkinson's disease (PD). However, it is still unclear whether the expression of active hRheb induces the production of neurotrophic factors such as glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF), which are involved in neuroprotection, in mature neurons. Here, we show that transduction of nigral DA neurons with hRheb(S16H) significantly increases the levels of phospho-cyclic adenosine monophosphate (cAMP) response element-binding protein (p-CREB), GDNF, and BDNF in neurons, which are attenuated by rapamycin, a specific inhibitor of mammalian target of rapamycin complex 1 (mTORC1). Moreover, treatment with specific neutralizing antibodies for GDNF and BDNF reduced the protective effects of hRheb(S16H) against 1-methyl-4-phenylpyridinium (MPP(+))-induced neurotoxicity. These results show that activation of hRheb/mTORC1 signaling pathway could impart to DA neurons the important ability to continuously produce GDNF and BDNF as therapeutic agents against PD.
Collapse
Affiliation(s)
- Jin Han Nam
- Department of Neuroscience, Neurodegeneration Control Research Center, School of Medicine, Kyung Hee University, Seoul, 130-701, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Direct, quantitative, and noninvasive imaging of the transport of active agents through intact brain with positron emission tomography. Mol Imaging Biol 2014; 15:596-605. [PMID: 23624949 DOI: 10.1007/s11307-013-0636-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE Our goal was to use positron emission tomography (PET) to analyze the movement of radiolabeled agents in tissue to enable direct measurement of drug delivery to the brain. PROCEDURES Various (11)C- and (18) F-labeled compounds were delivered directly to an agarose phantom or rat striatum. Concentration profiles were extracted for analysis and fitted to diffusion models. RESULTS Diffusion coefficients ranged from 0.075 ± 0.0026 mm(2)/min ([(18) F]fluoride ion, 18 Da) to 0.0016 ± 0.0018 mm(2)/min ([(18) F]NPB4-avidin, 68 kDa) and matched well with predictions based on molecular weight (R (2) = 0.965). The tortuosity of the brain extracellular space was estimated to be 1.56, with the tissue clearance halftime of each tracer in the brain varying from 19 to 41 min. CONCLUSIONS PET is an effective modality to directly quantify the movement of locally delivered drugs or drug carriers. This continuous, noninvasive assessment of delivery will aid the design of better drug delivery methods.
Collapse
|
42
|
Liu W, Rask-Andersen H. Immunohistological analysis of neurturin and its receptors in human cochlea. Auris Nasus Larynx 2014; 41:172-8. [DOI: 10.1016/j.anl.2013.07.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/29/2013] [Accepted: 09/20/2013] [Indexed: 01/15/2023]
|
43
|
Fuqua JL, Littrell OM, Lundblad M, Turchan-Cholewo J, Abdelmoti LG, Galperin E, Bradley LH, Cass WA, Gash DM, Gerhardt GA. Dynamic changes in dopamine neuron function after DNSP-11 treatment: effects in vivo and increased ERK 1/2 phosphorylation in vitro. Peptides 2014; 54:1-8. [PMID: 24406899 PMCID: PMC3989369 DOI: 10.1016/j.peptides.2013.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 12/09/2013] [Accepted: 12/10/2013] [Indexed: 12/14/2022]
Abstract
Glial cell-line derived neurotrophic factor (GDNF) has demonstrated robust effects on dopamine (DA) neuron function and survival. A post-translational processing model of the human GDNF proprotein theorizes the formation of smaller, amidated peptide(s) from the proregion that exhibit neurobiological function, including an 11-amino-acid peptide named dopamine neuron stimulating peptide-11 (DNSP-11). A single treatment of DNSP-11 was delivered to the substantia nigra in the rat to investigate effects on DA-neuron function. Four weeks after treatment, potassium (K+) and D-amphetamine evoked DA release were studied in the striatum using microdialysis. There were no significant changes in DA-release after DNSP-11 treatment determined by microdialysis. Dopamine release was further examined in discrete regions of the striatum using high-speed chronoamperometry at 1-, 2-, and 4-weeks after DNSP-11 treatment. Two weeks after DNSP-11 treatment, potassium-evoked DA release was increased in specific subregions of the striatum. However, spontaneous locomotor activity was unchanged by DNSP-11 treatment. In addition, we show that a single treatment of DNSP-11 in the MN9D dopaminergic neuronal cell line results in phosphorylation of ERK1/2, which suggests a novel cellular mechanism responsible for increases in DA function.
Collapse
Affiliation(s)
- Joshua L Fuqua
- Department of Anatomy and Neurobiology, Parkinson's Disease Translational Center of Excellence, University of Kentucky Medical Center, MN 206 Medical Sciences Building, 800 Rose St., Lexington, KY 40536, USA
| | - Ofelia M Littrell
- Department of Anatomy and Neurobiology, Parkinson's Disease Translational Center of Excellence, University of Kentucky Medical Center, MN 206 Medical Sciences Building, 800 Rose St., Lexington, KY 40536, USA
| | - Martin Lundblad
- Experimental Medical Science, Neurobiology, Lund University, BMCA11, 221, 84 Lund, Sweden
| | - Jadwiga Turchan-Cholewo
- Department of Anatomy and Neurobiology, Parkinson's Disease Translational Center of Excellence, University of Kentucky Medical Center, MN 206 Medical Sciences Building, 800 Rose St., Lexington, KY 40536, USA
| | - Lina G Abdelmoti
- Department of Molecular & Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA; Center of Structural Biology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Emilia Galperin
- Department of Molecular & Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA; Center of Structural Biology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Luke H Bradley
- Department of Anatomy and Neurobiology, Parkinson's Disease Translational Center of Excellence, University of Kentucky Medical Center, MN 206 Medical Sciences Building, 800 Rose St., Lexington, KY 40536, USA; Department of Molecular & Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA; Center of Structural Biology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Wayne A Cass
- Department of Anatomy and Neurobiology, Parkinson's Disease Translational Center of Excellence, University of Kentucky Medical Center, MN 206 Medical Sciences Building, 800 Rose St., Lexington, KY 40536, USA
| | - Don M Gash
- Department of Anatomy and Neurobiology, Parkinson's Disease Translational Center of Excellence, University of Kentucky Medical Center, MN 206 Medical Sciences Building, 800 Rose St., Lexington, KY 40536, USA
| | - Greg A Gerhardt
- Department of Anatomy and Neurobiology, Parkinson's Disease Translational Center of Excellence, University of Kentucky Medical Center, MN 206 Medical Sciences Building, 800 Rose St., Lexington, KY 40536, USA.
| |
Collapse
|
44
|
Adult hemiparkinsonian rats do not benefit from tactile stimulation. Behav Brain Res 2014; 261:97-105. [DOI: 10.1016/j.bbr.2013.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/03/2013] [Accepted: 12/07/2013] [Indexed: 10/25/2022]
|
45
|
Roles for the TGFβ superfamily in the development and survival of midbrain dopaminergic neurons. Mol Neurobiol 2014; 50:559-73. [PMID: 24504901 DOI: 10.1007/s12035-014-8639-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/02/2014] [Indexed: 12/29/2022]
Abstract
The adult midbrain contains 75% of all dopaminergic neurons in the CNS. Within the midbrain, these neurons are divided into three anatomically and functionally distinct clusters termed A8, A9 and A10. The A9 group plays a functionally non-redundant role in the control of voluntary movement, which is highlighted by the motor syndrome that results from their progressive degeneration in the neurodegenerative disorder, Parkinson's disease. Despite 50 years of investigation, treatment for Parkinson's disease remains symptomatic, but an intensive research effort has proposed delivering neurotrophic factors to the brain to protect the remaining dopaminergic neurons, or using these neurotrophic factors to differentiate dopaminergic neurons from stem cell sources for cell transplantation. Most neurotrophic factors studied in this context have been members of the transforming growth factor β (TGFβ) superfamily. In recent years, an intensive research effort has focused on understanding the function of these proteins in midbrain dopaminergic neuron development and their role in the molecular architecture that regulates the development of this brain region, with the goal of applying this knowledge to develop novel therapies for Parkinson's disease. In this review, the current evidence showing that TGFβ superfamily members play critical roles in the regulation of midbrain dopaminergic neuron induction, differentiation, target innervation and survival during embryonic and postnatal development is analysed, and the implications of these findings are discussed.
Collapse
|
46
|
Sommer DB, Stacy MA. What’s in the pipeline for the treatment of Parkinson’s disease? Expert Rev Neurother 2014; 8:1829-39. [DOI: 10.1586/14737175.8.12.1829] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
47
|
Bartus RT, Weinberg MS, Samulski RJ. Parkinson's disease gene therapy: success by design meets failure by efficacy. Mol Ther 2013; 22:487-497. [PMID: 24356252 PMCID: PMC3944322 DOI: 10.1038/mt.2013.281] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 12/05/2013] [Indexed: 12/13/2022] Open
Abstract
Over the past decade, nine gene therapy clinical trials for Parkinson's disease (PD) have been initiated and completed. Starting with considerable optimism at the initiation of each trial, none of the programs has yet borne sufficiently robust clinical efficacy or found a clear path toward regulatory approval. Despite the immediately disappointing nature of the efficacy outcomes in these trials, the clinical data garnered from the individual studies nonetheless represent tangible and significant progress for the gene therapy field. Collectively, the clinical trials demonstrate that we have overcome the major safety hurdles previously suppressing central nervous system (CNS) gene therapy, for none produced any evidence of untoward risk or harm after administration of various vector-delivery systems. More importantly, these studies also demonstrated controlled, highly persistent generation of biologically active proteins targeted to structures deep in the human brain. Therefore, a renewed, focused emphasis must be placed on advancing clinical efficacy by improving clinical trial design, patient selection and outcome measures, developing more predictive animal models to support clinical testing, carefully performing retrospective analyses, and most importantly moving forward—beyond our past limits.
Collapse
Affiliation(s)
- Raymond T Bartus
- Ceregene, Inc., San Diego, California, USA; RTBioconsultants, Inc., San Diego, California, USA.
| | - Marc S Weinberg
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA; Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA.
| |
Collapse
|
48
|
Ren X, Zhang T, Gong X, Hu G, Ding W, Wang X. AAV2-mediated striatum delivery of human CDNF prevents the deterioration of midbrain dopamine neurons in a 6-hydroxydopamine induced parkinsonian rat model. Exp Neurol 2013; 248:148-56. [DOI: 10.1016/j.expneurol.2013.06.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 05/24/2013] [Accepted: 06/01/2013] [Indexed: 01/05/2023]
|
49
|
Kucinski A, Wersinger S, Stachowiak EK, Corso TD, Parry MJ, Zhang J, Jordan K, Letchworth S, Bencherif M, Stachowiak MK. Neuronal nicotinic receptor agonists ameliorate spontaneous motor asymmetries and motor discoordination in a unilateral mouse model of Parkinson's disease. Pharmacol Biochem Behav 2013; 111:1-10. [DOI: 10.1016/j.pbb.2013.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 06/21/2013] [Accepted: 07/03/2013] [Indexed: 12/24/2022]
|
50
|
Therapeutic effects of repetitive transcranial magnetic stimulation in an animal model of Parkinson's disease. Brain Res 2013; 1537:290-302. [PMID: 23998987 DOI: 10.1016/j.brainres.2013.08.051] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Revised: 07/24/2013] [Accepted: 08/26/2013] [Indexed: 01/08/2023]
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is used to treat neurological diseases such as stroke and Parkinson's disease (PD). Although rTMS has been used clinically, its underlying therapeutic mechanism remains unclear. The objective of the present study was to clarify the neuroprotective effect and therapeutic mechanism of rTMS in an animal model of PD. Adult Sprague-Dawley rats were unilaterally injected with 6-hydroxydopamine (6-OHDA) into the right striatum. Rats with PD were then treated with rTMS (circular coil, 10 Hz, 20 min/day) daily for 4 weeks. Behavioral assessments such as amphetamine-induced rotational test and treadmill locomotion test were performed, and the dopaminergic (DA) neurons of substantia nigra pas compacta (SNc) and striatum were histologically examined. Expression of neurotrophic/growth factors was also investigated by multiplex ELISA, western blotting analysis and immunohistochemistry 4 weeks after rTMS application. Among the results, the number of amphetamine-induced rotations was significantly lower in the rTMS group than in the control group at 4 weeks post-treatment. Treadmill locomotion was also significantly improved in the rTMS-treated rats. Tyrosine hydroxylase-positive DA neurons and DA fibers in rTMS group rats were greater than those in untreated group in both ipsilateral SNc and striatum, respectively. The expression levels of brain-derived neurotrophic factor, glial cell line-derived neurotrophic factor, platelet-derived growth factor, and vascular endothelial growth factor were elevated in both the 6-OHDA-injected hemisphere and the SNc of the rTMS-treated rats. In conclusion, rTMS treatment improved motor functions and survival of DA neurons, suggesting that the neuroprotective effect of rTMS treatment might be induced by upregulation of neurotrophic/growth factors in the PD animal model.
Collapse
|