1
|
Atkinson E, Dickman R. Growth factors and their peptide mimetics for treatment of traumatic brain injury. Bioorg Med Chem 2023; 90:117368. [PMID: 37331175 DOI: 10.1016/j.bmc.2023.117368] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/16/2023] [Accepted: 06/05/2023] [Indexed: 06/20/2023]
Abstract
Traumatic brain injury (TBI) is a leading cause of disability in adults, caused by a physical insult damaging the brain. Growth factor-based therapies have the potential to reduce the effects of secondary injury and improve outcomes by providing neuroprotection against glutamate excitotoxicity, oxidative damage, hypoxia, and ischemia, as well as promoting neurite outgrowth and the formation of new blood vessels. Despite promising evidence in preclinical studies, few neurotrophic factors have been tested in clinical trials for TBI. Translation to the clinic is not trivial and is limited by the short in vivo half-life of the protein, the inability to cross the blood-brain barrier and human delivery systems. Synthetic peptide mimetics have the potential to be used in place of recombinant growth factors, activating the same downstream signalling pathways, with a decrease in size and more favourable pharmacokinetic properties. In this review, we will discuss growth factors with the potential to modulate damage caused by secondary injury mechanisms following a traumatic brain injury that have been trialled in other indications including spinal cord injury, stroke and neurodegenerative diseases. Peptide mimetics of nerve growth factor (NGF), hepatocyte growth factor (HGF), glial cell line-derived growth factor (GDNF), brain-derived neurotrophic factor (BDNF), platelet-derived growth factor (PDGF) and fibroblast growth factor (FGF) will be highlighted, most of which have not yet been tested in preclinical or clinical models of TBI.
Collapse
Affiliation(s)
- Emily Atkinson
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; UCL Centre for Nerve Engineering, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| | - Rachael Dickman
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| |
Collapse
|
2
|
Liu QR, Zhu M, Chen Q, Mustapic M, Kapogiannis D, Egan JM. Novel Hominid-Specific IAPP Isoforms: Potential Biomarkers of Early Alzheimer's Disease and Inhibitors of Amyloid Formation. Biomolecules 2023; 13:167. [PMID: 36671553 PMCID: PMC9856209 DOI: 10.3390/biom13010167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/23/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
(1) Background and aims: Amyloidosis due to aggregation of amyloid-β (Aβ42) is a key pathogenic event in Alzheimer's disease (AD), whereas aggregation of mature islet amyloid polypeptide (IAPP37) in human islets leads to β-cell dysfunction. The aim of this study is to uncover potential biomarkers that might additionally point to therapy for early AD patients. (2) Methods: We used bioinformatic approach to uncover novel IAPP isoforms and developed a quantitative selective reaction monitoring (SRM) proteomic assay to measure their peptide levels in human plasma and CSF from individuals with early AD and controls, as well as postmortem cerebrum of clinical confirmed AD and controls. We used Thioflavin T amyloid reporter assay to measure the IAPP isoform fibrillation propensity and anti-amyloid potential against aggregation of Aβ42 and IAPP37. (3) Results: We uncovered hominid-specific IAPP isoforms: hIAPPβ, which encodes an elongated propeptide, and hIAPPγ, which is processed to mature IAPP25 instead of IAPP37. We found that hIAPPβ was significantly reduced in the plasma of AD patients with the accuracy of 89%. We uncovered that IAPP25 and a GDNF derived DNSP11 were nonaggregating peptides that inhibited the aggregation of IAPP37 and Aβ42. (4) Conclusions: The novel peptides derived from hIAPP isoforms have potential to serve as blood-derived biomarkers for early AD and be developed as peptide based anti-amyloid medicine.
Collapse
Affiliation(s)
- Qing-Rong Liu
- Laboratory of Clinical Investigation, NIA-NIH, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | | | | | | | | | - Josephine M. Egan
- Laboratory of Clinical Investigation, NIA-NIH, 251 Bayview Blvd, Baltimore, MD 21224, USA
| |
Collapse
|
3
|
Sidorova YA, Saarma M. Can Growth Factors Cure Parkinson's Disease? Trends Pharmacol Sci 2020; 41:909-922. [PMID: 33198924 DOI: 10.1016/j.tips.2020.09.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/25/2020] [Accepted: 09/30/2020] [Indexed: 01/03/2023]
Abstract
Growth factors (GFs) hold considerable promise for disease modification in neurodegenerative disorders because they can protect and restore degenerating neurons and also enhance their functional activity. However, extensive efforts applied to utilize their therapeutic potential in humans have achieved limited success so far. Multiple clinical trials with GFs were performed in Parkinson's disease (PD) patients, in whom diagnostic symptoms of the disease are caused by advanced degeneration of nigrostriatal dopamine neurons (DNs), but the results of these trials are controversial. This review discusses recent developments in the field of therapeutic use of GFs, problems and obstacles related to this use, suggests the ways to overcome these issues, and alternative approaches that can be used to utilize the potential ofGFsin PD management.
Collapse
Affiliation(s)
- Yulia A Sidorova
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
4
|
Small Molecules and Peptides Targeting Glial Cell Line-Derived Neurotrophic Factor Receptors for the Treatment of Neurodegeneration. Int J Mol Sci 2020; 21:ijms21186575. [PMID: 32911810 PMCID: PMC7554781 DOI: 10.3390/ijms21186575] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/31/2020] [Accepted: 09/06/2020] [Indexed: 12/14/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) are able to promote the survival of multiple neuronal populations in the body and, therefore, hold considerable promise for disease-modifying treatments of diseases and conditions caused by neurodegeneration. Available data reveal the potential of GFLs for the therapy of Parkinson's disease, neuropathic pain and diseases caused by retinal degeneration but, also, amyotrophic lateral sclerosis and, possibly, Alzheimer's disease. Despite promising data collected in preclinical models, clinical translation of GFLs is yet to be conducted. The main reasons for the limited success of GFLs clinical development are the poor pharmacological characteristics of GFL proteins, such as the inability of GFLs to cross tissue barriers, poor diffusion in tissues, biphasic dose-response and activation of several receptors in the organism in different cell types, along with ethical limitations on patients' selection in clinical trials. The development of small molecules selectively targeting particular GFL receptors with improved pharmacokinetic properties can overcome many of the difficulties and limitations associated with the clinical use of GFL proteins. The current review lists several strategies to target the GFL receptor complex with drug-like molecules, discusses their advantages, provides an overview of available chemical scaffolds and peptides able to activate GFL receptors and describes the effects of these molecules in cultured cells and animal models.
Collapse
|
5
|
Gash DM, Gerhardt GA, Bradley LH, Wagner R, Slevin JT. GDNF clinical trials for Parkinson's disease: a critical human dimension. Cell Tissue Res 2020; 382:65-70. [PMID: 32830288 DOI: 10.1007/s00441-020-03269-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/29/2020] [Indexed: 01/02/2023]
Affiliation(s)
- Don Marshall Gash
- Department of Neuroscience, Chandler Medical Center, University of Kentucky, Lexington, KY, USA. .,College of Medicine, Lewis Honors College, University of Kentucky, Lexington, KY, USA.
| | - Greg A Gerhardt
- Department of Neuroscience, Chandler Medical Center, University of Kentucky, Lexington, KY, USA.,Department of Neurology, Chandler Medical Center, University of Kentucky, Lexington, KY, USA
| | - Luke H Bradley
- Department of Neuroscience, Chandler Medical Center, University of Kentucky, Lexington, KY, USA
| | - Renee Wagner
- Department of Neurology, Chandler Medical Center, University of Kentucky, Lexington, KY, USA
| | - John T Slevin
- Department of Neurology, Chandler Medical Center, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
6
|
Ahmadi S, Zobeiri M, Bradburn S. Molecular mechanisms underlying actions of certain long noncoding RNAs in Alzheimer's disease. Metab Brain Dis 2020; 35:681-693. [PMID: 32185592 DOI: 10.1007/s11011-020-00564-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 03/05/2020] [Indexed: 01/08/2023]
Abstract
Long non-coding RNAs (lncRNAs) are a group of non-protein coding RNAs that have more than 200 nucleotides. LncRNAs play an important role in the regulation of protein-coding genes at the transcriptional and post-transcriptional levels. They are found in most organs, with a high prevalence in the central nervous system. Accumulating data suggests that lncRNAs are involved in various neurodegenerative disorders, including the onset and progression of Alzheimer's disease (AD). Recent insights suggest lncRNAs, such as BACE1-AS, 51A, 17A, NDM29 and AS-UCHL1, are dysregulated in AD tissues. Furthermore, there are ongoing efforts to explore the clinical usability of lncRNAs as biomarkers in the disease. In this review, we explore the mechanisms by which aberrant expressions of the most studied lncRNAs contribute to the neuropathologies associated with AD, including amyloid β plaques and neurofibrillary tangles. Understanding the molecular mechanisms of lncRNAs in patients with AD will reveal novel diagnosis strategies and more effective therapeutic targets.
Collapse
Affiliation(s)
- Shamseddin Ahmadi
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran.
| | - Mohammad Zobeiri
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran
| | - Steven Bradburn
- Bioscience Research Centre, Manchester Metropolitan University, Manchester, UK
| |
Collapse
|
7
|
Spatial and temporal immunoreactivity in the rat brain using an affinity purified polyclonal antibody to DNSP-11. J Chem Neuroanat 2019; 100:101664. [PMID: 31394198 DOI: 10.1016/j.jchemneu.2019.101664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/30/2019] [Accepted: 08/03/2019] [Indexed: 01/21/2023]
Abstract
DNSP-11 antibody signal was investigated in perfusion fixated Fischer 344 rat brains by immunohistochemistry with a custom, affinity purified polyclonal antibody. The DNSP-11-antibody signal was differentially localized from the mature GDNF protein both spatially and temporally. In the mesencephalon of post-natal day 10 animals, when GDNF is maximally expressed, DNSP-11 and GDNF antibody immunoreactivities co-localize extensively but not exclusively. In adult 3-month-old animals, GDNF expression is markedly reduced while the DNSP-11 signal remains intense. DNSP-11-antibody signal was present in the 3-month-old rat brain with signal in the substantia nigra, ventral tegmental area, dentate gyrus of the hippocampus, with the strongest signal observed in the locus ceruleus where GDNF is not expressed. While amino acid sequence homologues such as NPY and Tfg do exist, binding patterns reported in the literature of do not recapitulate the immunoreactive patterns observed for the DNSP-11-antibody signal.
Collapse
|
8
|
Grondin R, Littrell OM, Zhang Z, Ai Y, Huettl P, Pomerleau F, Quintero JE, Andersen AH, Stenslik MJ, Bradley LH, Lemmon J, O'Neill MJ, Gash DM, Gerhardt GA. GDNF revisited: A novel mammalian cell-derived variant form of GDNF increases dopamine turnover and improves brain biodistribution. Neuropharmacology 2019; 147:28-36. [DOI: 10.1016/j.neuropharm.2018.05.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 05/08/2018] [Accepted: 05/10/2018] [Indexed: 12/17/2022]
|
9
|
Penttinen AM, Parkkinen I, Voutilainen MH, Koskela M, Bäck S, Their A, Richie CT, Domanskyi A, Harvey BK, Tuominen RK, Nevalaita L, Saarma M, Airavaara M. Pre-α-pro-GDNF and Pre-β-pro-GDNF Isoforms Are Neuroprotective in the 6-hydroxydopamine Rat Model of Parkinson's Disease. Front Neurol 2018; 9:457. [PMID: 29973907 PMCID: PMC6019446 DOI: 10.3389/fneur.2018.00457] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/29/2018] [Indexed: 11/13/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is one of the most studied neurotrophic factors. GDNF has two splice isoforms, full-length pre-α-pro-GDNF (α-GDNF) and pre-β-pro-GDNF (β-GDNF), which has a 26 amino acid deletion in the pro-region. Thus far, studies have focused solely on the α-GDNF isoform, and nothing is known about the in vivo effects of the shorter β-GDNF variant. Here we compare for the first time the effects of overexpressed α-GDNF and β-GDNF in non-lesioned rat striatum and the partial 6-hydroxydopamine lesion model of Parkinson's disease. GDNF isoforms were overexpressed with their native pre-pro-sequences in the striatum using an adeno-associated virus (AAV) vector, and the effects on motor performance and dopaminergic phenotype of the nigrostriatal pathway were assessed. In the non-lesioned striatum, both isoforms increased the density of dopamine transporter-positive fibers at 3 weeks after viral vector delivery. Although both isoforms increased the activity of the animals in cylinder assay, only α-GDNF enhanced the use of contralateral paw. Four weeks later, the striatal tyrosine hydroxylase (TH)-immunoreactivity was decreased in both α-GDNF and β-GDNF treated animals. In the neuroprotection assay, both GDNF splice isoforms increased the number of TH-immunoreactive cells in the substantia nigra but did not promote behavioral recovery based on amphetamine-induced rotation or cylinder assays. Thus, the shorter GDNF isoform, β-GDNF, and the full-length α-isoform have comparable neuroprotective efficacy on dopamine neurons of the nigrostriatal circuitry.
Collapse
Affiliation(s)
- Anna-Maija Penttinen
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ilmari Parkkinen
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Merja H Voutilainen
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Maryna Koskela
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Susanne Bäck
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Anna Their
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Christopher T Richie
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Andrii Domanskyi
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Brandon K Harvey
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Raimo K Tuominen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Liina Nevalaita
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
10
|
McMahon MA, Prakash TP, Cleveland DW, Bennett CF, Rahdar M. Chemically Modified Cpf1-CRISPR RNAs Mediate Efficient Genome Editing in Mammalian Cells. Mol Ther 2018; 26:1228-1240. [PMID: 29650467 PMCID: PMC5993945 DOI: 10.1016/j.ymthe.2018.02.031] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 02/27/2018] [Accepted: 02/27/2018] [Indexed: 11/17/2022] Open
Abstract
CRISPR-based gene editing is a powerful technology for engineering mammalian genomes. It holds the potential as a therapeutic, although much-needed in vivo delivery systems have yet to be established. Here, using the Cpf1-crRNA (CRISPR RNA) crystal structure as a guide, we synthesized a series of systematically truncated and chemically modified crRNAs, and identify positions that are amenable to modification while retaining gene-editing activity. Modified crRNAs were designed with the same modifications that provide protection against nucleases and enable wide distribution in vivo. We show crRNAs with chemically modified terminal nucleotides are exonuclease resistant while retaining gene-editing activity. Chemically modified or DNA-substituted nucleotides at select positions and up to 70% of the crRNA DNA specificity region are also well tolerated. In addition, gene-editing activity is maintained with phosphorothioate backbone substitutions in the crRNA DNA specificity region. Finally, we demonstrate that 42-mer synthetic crRNAs from the similar CRISPR-Cas9 system are taken up by cells, an attractive property for in vivo delivery. Our study is the first to show that chemically modified crRNAs of the CRISPR-Cpf1 system can functionally replace and mediate comparable gene editing to the natural crRNA, which holds the potential for enhancing both viral- and non-viral-mediated in vivo gene editing.
Collapse
Affiliation(s)
- Moira A McMahon
- Ludwig Institute for Cancer Research, University of California at San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA; Ionis Pharmaceuticals, Carlsbad, CA 92010, USA
| | | | - Don W Cleveland
- Ludwig Institute for Cancer Research, University of California at San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | | | | |
Collapse
|
11
|
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: 1.0] [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
|
12
|
Zanin JP, Unsain N, Anastasia A. Growth factors and hormones pro-peptides: the unexpected adventures of the BDNF prodomain. J Neurochem 2017; 141:330-340. [PMID: 28218971 DOI: 10.1111/jnc.13993] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 02/08/2017] [Accepted: 02/13/2017] [Indexed: 12/26/2022]
Abstract
Most growth factors and hormones are synthesized as pre-pro-proteins which are processed to the biologically active mature protein. The pre- and prodomains are cleaved from the precursor protein in the secretory pathway or, in some cases, extracellularly. The canonical functions of these prodomains are to assist in folding and stabilization of the mature domain, to direct intra and extracellular localization, to facilitate storage, and to regulate bioavailability of their mature counterpart. Recently, exciting evidence has revealed that prodomains of certain growth factors, after cleaved from the precursor pro-protein, can act as independent active signaling molecules. In this review, we discuss the various classical functions of prodomains, and the biological consequences of these pro-peptides acting as ligands. We will focus our attention on the brain-derived neurotrophic factor prodomain (pBDNF), which has been recently described as a novel secreted ligand influencing neuronal morphology and physiology.
Collapse
Affiliation(s)
- Juan Pablo Zanin
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Nicolás Unsain
- Instituto de Investigación Médica Mercedes y Martin Ferreyra, (INIMEC-CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
| | - Agustin Anastasia
- Instituto de Investigación Médica Mercedes y Martin Ferreyra, (INIMEC-CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
| |
Collapse
|
13
|
Popova NK, Ilchibaeva TV, Naumenko VS. Neurotrophic factors (BDNF and GDNF) and the serotonergic system of the brain. BIOCHEMISTRY (MOSCOW) 2017; 82:308-317. [DOI: 10.1134/s0006297917030099] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
14
|
Ilchibaeva TV, Tsybko AS, Kozhemyakina RV, Popova NK, Naumenko VS. Glial cell line-derived neurotrophic factor in genetically defined fear-induced aggression. Eur J Neurosci 2016; 44:2467-2473. [DOI: 10.1111/ejn.13365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 07/08/2016] [Accepted: 08/08/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Tatiana V. Ilchibaeva
- Department of Behavioral Neurogenomics; Federal Research Center Institute of Cytology and Genetics; Siberian Division of the Russian Academy of Science; Lavrentyeva av. 10 Novosibirsk 630090 Russia
| | - Anton S. Tsybko
- Department of Behavioral Neurogenomics; Federal Research Center Institute of Cytology and Genetics; Siberian Division of the Russian Academy of Science; Lavrentyeva av. 10 Novosibirsk 630090 Russia
| | - Rimma V. Kozhemyakina
- Laboratory of Evolutionary Genetics; Federal Research Center Institute of Cytology and Genetics; Siberian Division of the Russian Academy of Science; Novosibirsk Russia
| | - Nina K. Popova
- Department of Behavioral Neurogenomics; Federal Research Center Institute of Cytology and Genetics; Siberian Division of the Russian Academy of Science; Lavrentyeva av. 10 Novosibirsk 630090 Russia
| | - Vladimir S. Naumenko
- Department of Behavioral Neurogenomics; Federal Research Center Institute of Cytology and Genetics; Siberian Division of the Russian Academy of Science; Lavrentyeva av. 10 Novosibirsk 630090 Russia
| |
Collapse
|
15
|
Pinkernelle J, Raffa V, Calatayud MP, Goya GF, Riggio C, Keilhoff G. Growth factor choice is critical for successful functionalization of nanoparticles. Front Neurosci 2015; 9:305. [PMID: 26388717 PMCID: PMC4557102 DOI: 10.3389/fnins.2015.00305] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/12/2015] [Indexed: 12/16/2022] Open
Abstract
Nanoparticles (NPs) show new characteristics compared to the corresponding bulk material. These nanoscale properties make them interesting for various applications in biomedicine and life sciences. One field of application is the use of magnetic NPs to support regeneration in the nervous system. Drug delivery requires a functionalization of NPs with bio-functional molecules. In our study, we functionalized self-made PEI-coated iron oxide NPs with nerve growth factor (NGF) and glial cell-line derived neurotrophic factor (GDNF). Next, we tested the bio-functionality of NGF in a rat pheochromocytoma cell line (PC12) and the bio-functionality of GDNF in an organotypic spinal cord culture. Covalent binding of NGF to PEI-NPs impaired bio-functionality of NGF, but non-covalent approach differentiated PC12 cells reliably. Non-covalent binding of GDNF showed a satisfying bio-functionality of GDNF:PEI-NPs, but turned out to be unstable in conjugation to the PEI-NPs. Taken together, our study showed the importance of assessing bio-functionality and binding stability of functionalized growth factors using proper biological models. It also shows that successful functionalization of magnetic NPs with growth factors is dependent on the used binding chemistry and that it is hardly predictable. For use as therapeutics, functionalization strategies have to be reproducible and future studies are needed.
Collapse
Affiliation(s)
- Josephine Pinkernelle
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University of MagdeburgMagdeburg, Germany
- Institute for Biochemistry and Cell Biology, Otto-von-Guericke University of MagdeburgMagdeburg, Germany
| | - Vittoria Raffa
- Department of Biology, University of PisaPisa, Italy
- Institute of Life Science, Scuola Superiore Sant' AnnaPisa, Italy
| | | | - Gerado F. Goya
- Aragon Institute of Nanosciences, University of ZaragozaZaragoza, Spain
- Department of Condensed Matter Physics, University of ZaragozaSpain
| | - Cristina Riggio
- Institute of Life Science, Scuola Superiore Sant' AnnaPisa, Italy
| | - Gerburg Keilhoff
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University of MagdeburgMagdeburg, Germany
| |
Collapse
|
16
|
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: 1.2] [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
|
17
|
Stojkovska I, Wagner BM, Morrison BE. Parkinson's disease and enhanced inflammatory response. Exp Biol Med (Maywood) 2015; 240:1387-95. [PMID: 25769314 DOI: 10.1177/1535370215576313] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/21/2015] [Indexed: 11/16/2022] Open
Abstract
Parkinson's disease (PD) is the first and second most prevalent motor and neurodegenerative disease, respectively. The clinical symptoms of PD result from a loss of midbrain dopaminergic (DA) neurons. However, the molecular cause of DA neuron loss remains elusive. Mounting evidence implicates enhanced inflammatory response in the development and progression of PD pathology. This review examines current research connecting PD and inflammatory response.
Collapse
Affiliation(s)
- Iva Stojkovska
- Department of Biological Sciences, Boise State University, Boise, ID 83725-1515, USA
| | - Brandon M Wagner
- Department of Biological Sciences, Boise State University, Boise, ID 83725-1515, USA
| | - Brad E Morrison
- Department of Biological Sciences, Boise State University, Boise, ID 83725-1515, USA
| |
Collapse
|
18
|
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: 0.5] [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
|
19
|
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: 2.8] [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
|
20
|
The proform of glia cell line-derived neurotrophic factor: a potentially biologically active protein. Mol Neurobiol 2013; 49:234-50. [PMID: 23934644 DOI: 10.1007/s12035-013-8515-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 07/10/2013] [Indexed: 12/24/2022]
Abstract
Growing evidences have revealed that the proforms of several neurotrophins including nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin-3 (NT3), by binding to p75 neurotrophin receptor and sortilin, could induce neuronal apoptosis and are implicated in the pathogenesis of various neurodegenerative diseases. The glial cell line-derived neurotrophic factor (GDNF), one of the most potent useful neurotrophic factors for the treatment of Parkinson's disease (PD), is firstly synthesized as the proform (proGDNF) like other neurotrophin NGF, BDNF, and NT3. However, little is known about proGDNF expression and secretion under physiological as well as pathological states in vivo or in vitro. In this study, we investigated the expression profile and dynamic changes of proGDNF in brains of aging and PD animal models, with the interesting finding that proGDNF was a predominant form of GDNF with molecular weight of about 36 kDa by reducing and nonreducing immunoblots in adult brains and was unregulated in the aging, lipopolysaccharide (LPS), and 1-methyl-4-phenyl- 1,2,3,6-tetrahydropyridine (MPTP) insult. We further provided direct evidence that accompanied activation of primary astrocytes as well as C6 cell line induced by LPS stimulation, proGDNF was increasingly synthesized and released as the uncleaved form in cell culture. Taken together, our results strongly suggest that proGDNF may be a biologically active protein and has specific effects on the cells close to its secreting site, and a potentially important role of proGDNF signaling in the brains, in the glia-neuronal interaction or in the pathogenesis of PD, should merit further investigation.
Collapse
|
21
|
Littrell OM, Fuqua JL, Richardson AD, Turchan-Cholewo J, Hascup ER, Huettl P, Pomerleau F, Bradley LH, Gash DM, Gerhardt GA. A synthetic five amino acid propeptide increases dopamine neuron differentiation and neurochemical function. Neuropeptides 2013; 47:43-9. [PMID: 22981157 PMCID: PMC3558608 DOI: 10.1016/j.npep.2012.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 07/31/2012] [Accepted: 08/07/2012] [Indexed: 10/27/2022]
Abstract
A major consequence of Parkinson's disease (PD) involves the loss of dopaminergic neurons in the substantia nigra (SN) and a subsequent loss of dopamine (DA) in the striatum. We have shown that glial cell line-derived neurotrophic factor (GDNF) shows robust restorative and protective effects for DA neurons in rats, non-human primates and possibly in humans. Despite GDNF's therapeutic potential, its clinical value has been questioned due to its limited diffusion to target areas from its large size and chemical structure. Several comparatively smaller peptides are thought to be generated from the prosequence. A five amino-acid peptide, dopamine neuron stimulating peptide-5 (DNSP-5), has been proposed to demonstrate biological activity relevant to neurodegenerative disease. We tested the in vitro effects of DNSP-5 in primary dopaminergic neurons dissected from the ventral mesencephalon of E14 Sprague Dawley rat fetuses. Cells were treated with several doses (0.03, 0.1, 1.0, 10.0 ng/mL) of GDNF, DNSP-5, or an equivalent volume of citrate buffer (vehicle). Morphological features of tyrosine hydroxylase positive neurons were quantified for each dose. DNSP-5 significantly increased (p < 0.001) all differentiation parameters compared to citrate vehicle (at one or more dose). For in vivo studies, a unilateral DNSP-5 treatment (30 μg) was administered directly to the SN. Microdialysis in the ipsilateral striatum was performed 28 days after treatment to determine extracellular levels of DA and its primary metabolites (3,4-dihydroxyphenylacetic acid and homovanillic acid). A single treatment significantly increased (~66%) extracellular DA levels compared to vehicle, while DA metabolites were unchanged. Finally, the protective effects of DNSP-5 against staurosporine-induced cytotoxicity were investigated in a neuronal cell line showing substantial protection by DNSP-5. Altogether, these studies strongly indicate biological activity of DNSP-5 and suggest that DNSP-5 has neurotrophic-like properties that may be relevant to the treatment of neurodegenerative diseases like PD.
Collapse
Affiliation(s)
- OM Littrell
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY, 40536
| | - JL Fuqua
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY, 40536
| | - AD Richardson
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY, 40536
| | - J. Turchan-Cholewo
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY, 40536
| | - ER Hascup
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY, 40536
| | - P Huettl
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY, 40536
| | - F Pomerleau
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY, 40536
| | - LH Bradley
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY, 40536
- Department of Molecular & Cellular Biochemistry and the Center of Structural Biology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY, 40536
| | - DM Gash
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY, 40536
| | - GA Gerhardt
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY, 40536
- Corresponding author contact: Dr. Greg A. Gerhardt, 306 Davis Mills Bldg., 800 Rose Street, Lexington, KY 40536-0098, (), Phone: 1+ 859.323.4531, Fax: 1+ 859.257.5310
| |
Collapse
|
22
|
Piccinini E, Kalkkinen N, Saarma M, Runeberg-Roos P. Glial cell line-derived neurotrophic factor: characterization of mammalian posttranslational modifications. Ann Med 2013; 45:66-73. [PMID: 23305235 DOI: 10.3109/07853890.2012.663927] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Although glial cell line-derived neurotrophic factor (GDNF) has a strong clinical potential, little is known of how the posttranslational modifications of GDNF affect its biological activity and therapeutic potential. In mammalian cells GDNF is synthesized as a preproprotein. During secretion GDNF dimerizes, folds with -S-S- bonds, is modified by N-linked glycosylation, and undergoes proteolytic processing. After production in E. coli, unglycosylated GDNF is renaturated in vitro. Nevertheless, GDNF from E. coli was used in Parkinson's disease-related clinical trials. MATERIAL AND METHODS Constructs encoding variants of human GDNF were generated and expressed in mammalian cells. The proteins were analysed by SDS-PAGE, Western blotting, RET-phosphorylation assays, and N-terminal sequencing. The stability of mammalian GDNF was compared to commercial GDNF produced in E. coli. RESULTS Posttranslational processing of mammalian GDNF depends on the expression conditions. Two forms of GDNF with different N-termini are formed. GDNF without a prosequence is secreted and biologically active. GDNF is modified by N-linked glycosylation at one (Asn(49)) out of two consensus sites. N-linked glycosylation aids proteolytic processing of GDNF. Both glycosylated and unglycosylated GDNF from mammalian cells are more stable than GDNF from E. coli. DISCUSSION Posttranslational modifications of GDNF influence its stability, which may be critical for its clinical use.
Collapse
Affiliation(s)
- Elisa Piccinini
- Institute of Biotechnology, University of Helsinki, PB 56 Viikinkaari 9, SF-00014, University of Helsinki, Finland
| | | | | | | |
Collapse
|
23
|
Abstract
The glial cell line-derived neurotrophic factor (GDNF) was first identified as a survival factor for midbrain dopaminergic neurons, but additional studies provided evidences for a role as a trophic factor for other neurons of the central and peripheral nervous systems. GDNF regulates cellular activity through interaction with glycosyl-phosphatidylinositol-anchored cell surface receptors, GDNF family receptor-α1, which might signal through the transmembrane Ret tyrosine receptors or the neural cell adhesion molecule, to promote cell survival, neurite outgrowth, and synaptogenesis. The neuroprotective effect of exogenous GDNF has been shown in different experimental models of focal and global brain ischemia, by local administration of the trophic factor, using viral vectors carrying the GDNF gene and by transplantation of GDNF-expressing cells. These different strategies and the mechanisms contributing to neuroprotection by GDNF are discussed in this review. Importantly, neuroprotection by GDNF was observed even when administered after the ischemic injury.
Collapse
Affiliation(s)
- Emília P Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, Coimbra, Portugal
| | | | | | | |
Collapse
|
24
|
Airavaara M, Pletnikova O, Doyle ME, Zhang YE, Troncoso JC, Liu QR. Identification of novel GDNF isoforms and cis-antisense GDNFOS gene and their regulation in human middle temporal gyrus of Alzheimer disease. J Biol Chem 2011; 286:45093-102. [PMID: 22081608 DOI: 10.1074/jbc.m111.310250] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Primate-specific genes and isoforms could provide insight into human brain diseases. Our bioinformatic analysis revealed that there are possibly five isoforms of human GDNF gene with different pre- and pro-regions by inter- and intra-exon splicing. By using TaqMan primer probe sets, designed between exons, we verified the expression of all isoforms. Furthermore, a novel GDNFOS gene was found to be transcribed from the opposite strand of GDNF gene. GDNFOS gene has four exons that are spliced into different isoforms. GDNFOS1 and GDNFOS2 are long noncoding RNAs, and GDNFOS3 encodes a protein of 105 amino acids. To study human GDNF and GDNFOS regulation in neurodegenerative diseases, the protein and mRNA levels were measured by Western blot and RT-quantitative PCR, respectively, in postmortem middle temporal gyrus (MTG) of Alzheimer disease (AD) and Huntington disease (HD) patients in comparison with those of normal controls. In the MTG of AD patients, the mature GDNF peptide was down-regulated; however, the transcript of GDNF isoform from human exon 2 was up-regulated, whereas that of the conserved isoform from exon 1 remained unchanged in comparison with those of normal controls. In contrast, the mature GDNF peptide and the isoform mRNA levels were not changed in the MTG of HD. The findings of novel GDNF and GDNFOS isoforms and differences in tissue expression patterns dysregulated in AD brains may further reveal the role of endogenous GDNF in human brain diseases.
Collapse
Affiliation(s)
- Mikko Airavaara
- Neural Protection and Regeneration Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Department of Health and Social Services, Baltimore, Maryland21224, USA
| | | | | | | | | | | |
Collapse
|
25
|
Kelps KA, Turchan-Cholewo J, Hascup ER, Taylor TL, Gash DM, Gerhardt GA, Bradley LH. Evaluation of the physical and in vitro protective activity of three synthetic peptides derived from the pro- and mature GDNF sequence. Neuropeptides 2011; 45:213-8. [PMID: 21507484 PMCID: PMC3091812 DOI: 10.1016/j.npep.2011.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Revised: 03/21/2011] [Accepted: 03/22/2011] [Indexed: 12/12/2022]
Abstract
Recently, a small 11-amino acid amidated peptide, dopamine neuron stimulating peptide-11 (DNSP-11), was shown to exert neurotrophic-like actions on primary dopaminergic neurons and in parkinsonian rat models. This suggests smaller neurotrophic-like molecules may be deliverable and modifiable for therapeutic use. Here we evaluate the molecular and cellular protection properties of DNSP-11 and two other amidated-peptides, a 5-mer (DNSP-5) and a 17-mer (DNSP-17), hypothesized to be endoproteolytically processed from the pro- and mature glial cell line-derived neurotrophic factor (GDNF) protein sequence, respectively. Far-UV circular dichroism spectra show that the three DNSPs are soluble and act independently in vitro. Reverse phase HPLC and mass spectrometry analysis show that the three peptides are stable for one month at a variety of storage and experimental conditions. To gain insight into their biodistribution properties in the brain, we used affinity chromatography to show that DNSP-17 binds heparin equally as tight as GDNF, whereas DNSP-5 and DNSP-11 do not bind heparin, which should facilitate their delivery in vivo. Finally, we present data showing that DNSP-11 provides dose-dependent protection of HEK-293 cells from staurosporine and 3-nitropropionate (3-NP) cytotoxicity, thereby supporting its broad mitochondrial-protective properties.
Collapse
Affiliation(s)
- Kristen A. Kelps
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| | - Jadwiga Turchan-Cholewo
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| | - Erin R. Hascup
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| | - Tiffany L. Taylor
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| | - Don M. Gash
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| | - Greg A. Gerhardt
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| | - Luke H. Bradley
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, U.S.A
- Department of Molecular & Cellular Biochemistry and the Center of Structural Biology, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| |
Collapse
|