1
|
Stuber A, Cavaccini A, Manole A, Burdina A, Massoud Y, Patriarchi T, Karayannis T, Nakatsuka N. Interfacing Aptamer-Modified Nanopipettes with Neuronal Media and Ex Vivo Brain Tissue. ACS MEASUREMENT SCIENCE AU 2024; 4:92-103. [PMID: 38404490 PMCID: PMC10885324 DOI: 10.1021/acsmeasuresciau.3c00047] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 02/27/2024]
Abstract
Aptamer-functionalized biosensors exhibit high selectivity for monitoring neurotransmitters in complex environments. We translated nanoscale aptamer-modified nanopipette sensors to detect endogenous dopamine release in vitro and ex vivo. These sensors employ quartz nanopipettes with nanoscale pores (ca. 10 nm diameter) that are functionalized with aptamers that enable the selective capture of dopamine through target-specific conformational changes. The dynamic behavior of aptamer structures upon dopamine binding leads to the rearrangement of surface charge within the nanopore, resulting in measurable changes in ionic current. To assess sensor performance in real time, we designed a fluidic platform to characterize the temporal dynamics of nanopipette sensors. We then conducted differential biosensing by deploying control sensors modified with nonspecific DNA alongside dopamine-specific sensors in biological milieu. Our results confirm the functionality of aptamer-modified nanopipettes for direct measurements in undiluted complex fluids, specifically in the culture media of human-induced pluripotent stem cell-derived dopaminergic neurons. Moreover, sensor implantation and repeated measurements in acute brain slices was possible, likely owing to the protected sensing area inside nanoscale DNA-filled orifices, minimizing exposure to nonspecific interferents and preventing clogging. Further, differential recordings of endogenous dopamine released through electrical stimulation in the dorsolateral striatum demonstrate the potential of aptamer-modified nanopipettes for ex vivo recordings with unprecedented spatial resolution and reduced tissue damage.
Collapse
Affiliation(s)
- Annina Stuber
- Laboratory
of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zurich CH-8092, Switzerland
| | - Anna Cavaccini
- Laboratory
of Neural Circuit Assembly, Brain Research Institute, University of Zurich, Zurich CH-8057, Switzerland
- Neuroscience
Center Zurich, University and ETH Zurich, Zurich CH-8057, Switzerland
| | - Andreea Manole
- iXCells
Biotechnologies, Inc., San Diego, California 92131, United States
| | - Anna Burdina
- Laboratory
of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zurich CH-8092, Switzerland
| | - Yassine Massoud
- Laboratory
of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zurich CH-8092, Switzerland
| | - Tommaso Patriarchi
- Neuroscience
Center Zurich, University and ETH Zurich, Zurich CH-8057, Switzerland
- Institute
of Pharmacology and Toxicology, University
of Zurich, Zurich CH-8057, Switzerland
| | - Theofanis Karayannis
- Laboratory
of Neural Circuit Assembly, Brain Research Institute, University of Zurich, Zurich CH-8057, Switzerland
- Neuroscience
Center Zurich, University and ETH Zurich, Zurich CH-8057, Switzerland
| | - Nako Nakatsuka
- Laboratory
of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zurich CH-8092, Switzerland
| |
Collapse
|
2
|
Stuber A, Douaki A, Hengsteler J, Buckingham D, Momotenko D, Garoli D, Nakatsuka N. Aptamer Conformational Dynamics Modulate Neurotransmitter Sensing in Nanopores. ACS NANO 2023; 17:19168-19179. [PMID: 37721359 PMCID: PMC10569099 DOI: 10.1021/acsnano.3c05377] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/08/2023] [Indexed: 09/19/2023]
Abstract
Aptamers that undergo conformational changes upon small-molecule recognition have been shown to gate the ionic flux through nanopores by rearranging the charge density within the aptamer-occluded orifice. However, mechanistic insight into such systems where biomolecular interactions are confined in nanoscale spaces is limited. To understand the fundamental mechanisms that facilitate the detection of small-molecule analytes inside structure-switching aptamer-modified nanopores, we correlated experimental observations to theoretical models. We developed a dopamine aptamer-functionalized nanopore sensor with femtomolar detection limits and compared the sensing behavior with that of a serotonin sensor fabricated with the same methodology. When these two neurotransmitters with comparable mass and equal charge were detected, the sensors showed an opposite electronic behavior. This distinctive phenomenon was extensively studied using complementary experimental techniques such as quartz crystal microbalance with dissipation monitoring, in combination with theoretical assessment by the finite element method and molecular dynamic simulations. Taken together, our studies demonstrate that the sensing behavior of aptamer-modified nanopores in detecting specific small-molecule analytes correlates with the structure-switching mechanisms of individual aptamers. We believe that such investigations not only improve our understanding of the complex interactions occurring in confined nanoscale environments but will also drive further innovations in biomimetic nanopore technologies.
Collapse
Affiliation(s)
- Annina Stuber
- Laboratory
of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - Ali Douaki
- Instituto
Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Julian Hengsteler
- Laboratory
of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - Denis Buckingham
- Laboratory
of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - Dmitry Momotenko
- Department
of Chemistry, Carl von Ossietzky University
of Oldenburg, Oldenburg D-26129, Germany
| | - Denis Garoli
- Instituto
Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Nako Nakatsuka
- Laboratory
of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| |
Collapse
|
3
|
Hills R, Mossman JA, Bratt-Leal AM, Tran H, Williams RM, Stouffer DG, Sokolova IV, Sanna PP, Loring JF, Lelos MJ. Neurite Outgrowth and Gene Expression Profile Correlate with Efficacy of Human Induced Pluripotent Stem Cell-Derived Dopamine Neuron Grafts. Stem Cells Dev 2023; 32:387-397. [PMID: 37166357 PMCID: PMC10398740 DOI: 10.1089/scd.2023.0043] [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: 02/27/2023] [Accepted: 05/08/2023] [Indexed: 05/12/2023] Open
Abstract
Transplantation of human induced pluripotent stem cell-derived dopaminergic (iPSC-DA) neurons is a promising therapeutic strategy for Parkinson's disease (PD). To assess optimal cell characteristics and reproducibility, we evaluated the efficacy of iPSC-DA neuron precursors from two individuals with sporadic PD by transplantation into a hemiparkinsonian rat model after differentiation for either 18 (d18) or 25 days (d25). We found similar graft size and dopamine (DA) neuron content in both groups, but only the d18 cells resulted in recovery of motor impairments. In contrast, we report that d25 grafts survived equally as well and produced grafts rich in tyrosine hydroxylase-positive neurons, but were incapable of alleviating any motor deficits. We identified the mechanism of action as the extent of neurite outgrowth into the host brain, with d18 grafts supporting significantly more neurite outgrowth than nonfunctional d25 grafts. RNAseq analysis of the cell preparation suggests that graft efficacy may be enhanced by repression of differentiation-associated genes by REST, defining the optimal predifferentiation state for transplantation. This study demonstrates for the first time that DA neuron grafts can survive well in vivo while completely lacking the capacity to induce recovery from motor dysfunction. In contrast to other recent studies, we demonstrate that neurite outgrowth is the key factor determining graft efficacy and our gene expression profiling revealed characteristics of the cells that may predict their efficacy. These data have implication for the generation of DA neuron grafts for clinical application.
Collapse
Affiliation(s)
- Rachel Hills
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Jim A. Mossman
- Independent Bioinformatics Consultant, Del Mar, California, USA
| | - Andres M. Bratt-Leal
- Department of Molecular Medicine, Center for Regenerative Medicine, Scripps Research, La Jolla, California, USA
- Summit for Stem Cell Foundation, San Diego, California, USA
| | - Ha Tran
- Department of Molecular Medicine, Center for Regenerative Medicine, Scripps Research, La Jolla, California, USA
- Summit for Stem Cell Foundation, San Diego, California, USA
| | - Roy M. Williams
- Department of Molecular Medicine, Center for Regenerative Medicine, Scripps Research, La Jolla, California, USA
| | - David G. Stouffer
- Department of Molecular Medicine, Center for Regenerative Medicine, Scripps Research, La Jolla, California, USA
| | - Irina V. Sokolova
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, USA
| | - Pietro P. Sanna
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, USA
| | - Jeanne F. Loring
- Department of Molecular Medicine, Center for Regenerative Medicine, Scripps Research, La Jolla, California, USA
| | - Mariah J. Lelos
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
4
|
Banerjee P, Saha I, Sarkar D, Maiti AK. Contributions and Limitations of Mitochondria-Targeted and Non-Targeted Antioxidants in the Treatment of Parkinsonism: an Updated Review. Neurotox Res 2022; 40:847-873. [PMID: 35386026 DOI: 10.1007/s12640-022-00501-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 11/24/2022]
Abstract
As conventional therapeutics can only treat the symptoms of Parkinson's disease (PD), major focus of research in recent times is to slow down or prevent the progression of neuronal degeneration in PD. Non-targeted antioxidants have been an integral part of the conventional therapeutics regimen; however, their importance have lessened over time because of their controversial outcomes in clinical PD trials. Inability to permeate and localize within the mitochondria remains the main drawback on the part of non-targeted antioxidants inspite of possessing free radical scavenging properties. In contrast, mitochondrial-targeted antioxidants (MTAs), a special class of compounds have emerged having high advantages over non-targeted antioxidants by virtue of efficient pharmacokinetics and better absorption rate with capability to localize many fold inside the mitochondrial matrix. Preclinical experimentations indicate that MTAs have the potential to act as better alternatives compared to conventional non-targeted antioxidants in treating PD; however, sufficient clinical trials have not been conducted to investigate the efficacies of MTAs in treating PD. Controversial clinical outcomes on the part of non-targeted antioxidants and lack of clinical trials involving MTAs have made it difficult to go ahead with a direct comparison and in turn have slowed down the progress of development of safer and better alternate strategies in treating PD. This review provides an insight on the roles MTAs and non-targeted antioxidants have played in the treatment of PD till date in preclinical and clinical settings and discusses about the limitations of mitochondria-targeted and non-targeted antioxidants that can be resolved for developing effective strategies in treating Parkinsonism.
Collapse
Affiliation(s)
- Priyajit Banerjee
- Department of Zoology, University of Burdwan, Burdwan, West Bengal, Pin-713104, India
| | - Ishita Saha
- Department of Physiology, Medical College Kolkata, Kolkata, West Bengal, Pin-700073, India
| | - Diptendu Sarkar
- Department of Microbiology, Ramakrishna Mission Vidyamandira, Belur Math, Howrah, West Bengal, 711202, India
| | - Arpan Kumar Maiti
- Mitochondrial Biology and Experimental Therapeutics Laboratory, Department of Zoology, University of North Bengal, District - Darjeeling, P.O. N.B.U, Raja Rammohunpur, West Bengal, Pin-734013, India.
| |
Collapse
|
5
|
Moretti M, Rodrigues ALS. Functional role of ascorbic acid in the central nervous system: a focus on neurogenic and synaptogenic processes. Nutr Neurosci 2021; 25:2431-2441. [PMID: 34493165 DOI: 10.1080/1028415x.2021.1956848] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ascorbic acid, a water-soluble vitamin, is highly concentrated in the brain and participates in neuronal modulation and regulation of central nervous system (CNS) homeostasis. Ascorbic acid has emerged as a neuroprotective compound against neurotoxicants and neurodegenerative diseases, including Alzheimer's disease, multiple sclerosis and amyotrophic lateral sclerosis. Moreover, it improves behavioral and biochemical alterations in psychiatric disorders, including schizophrenia, anxiety, major depressive disorder, and bipolar disorder. Some recent studies have advanced the knowledge on the mechanisms associated with the preventive and therapeutic effects of ascorbic acid by showing that they are linked to improved neurogenesis and synaptic plasticity. This review shows that ascorbic acid has the potential to regulate positively stem cell generation and proliferation. Moreover, it improves neuronal differentiation of precursors cells, promotes adult hippocampal neurogenesis, and has synaptogenic effects that are possibly linked to its protective or therapeutic effects in the brain.
Collapse
Affiliation(s)
- Morgana Moretti
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
6
|
Aponso M, Patti A, Hearn MTW, Bennett LE. Anxiolytic effects of essential oils may involve anti-oxidant regulation of the pro-oxidant effects of ascorbate in the brain. Neurochem Int 2021; 150:105153. [PMID: 34384852 DOI: 10.1016/j.neuint.2021.105153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/02/2021] [Accepted: 08/05/2021] [Indexed: 01/01/2023]
Abstract
Essential oils (EOs) absorbed via inhalation are consistently reported to produce anxiolytic effects. The underlying neurochemical mechanisms, however, are not well understood. High concentrations of ascorbate in the human brain (~10 mM in neurons) implicates this compound as a key signaling molecule and regulator of oxidative stress. In this study, we demonstrate the significant in vitro capacity of ascorbate to produce H2O2 in the presence of oxygen at physiological pH values, peaking at ~400 μM for ascorbate levels of 1.0 mg/mL (5.6 mM). In comparison, individual EOs and selected neurotransmitters at similar concentrations produced <100 μM H2O2. Systematic studies with binary and ternary mixtures containing ascorbate indicated that EOs and neurotransmitters could variably enhance (pro-oxidant, POX) or suppress (anti-oxidant, AOX) the production of H2O2 versus the ascorbate control, depending on the concentration ratios of the components in the mixture. Moreover, the AOX/POX chemistry observed with binary mixtures did not necessarily predict effects with ternary mixtures, where the POX ascorbate chemistry tended to dominate. A model is proposed to account for the ability of compounds with electron-donating capacity to catalytically regenerate ascorbate from intermediate oxidized forms of ascorbate, thus driving H2O2 production and exerting a net POX effect; whilst compounds that irreversibly reacted with oxidized forms of ascorbate suppressed the production of H2O2 and produced an overall AOX effect. Since the anxiolytic effects of different EOs, including extracts of Lavendula angustifolia (lavender) and Salvia rosmarinus (rosemary), were associated with AOX regulation of H2O2 production by ascorbate, it can be concluded that these anxiolytic effects are potentially related to the AOX properties of EOs. In contrast, EOs driving POX effects (eg, Junipenus communis (Juniper) berry EO) are proposed to be more useful for their potential anti-microbial or cancer cytotoxic applications.
Collapse
Affiliation(s)
- Minoli Aponso
- School of Chemistry, Monash University, Clayton, Victoria, 3800, Australia
| | - Antonio Patti
- School of Chemistry, Monash University, Clayton, Victoria, 3800, Australia
| | - Milton T W Hearn
- School of Chemistry, Monash University, Clayton, Victoria, 3800, Australia
| | - Louise E Bennett
- School of Chemistry, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
7
|
Antioxidant Therapeutics in Parkinson's Disease: Current Challenges and Opportunities. Antioxidants (Basel) 2021; 10:antiox10030453. [PMID: 33803945 PMCID: PMC7998929 DOI: 10.3390/antiox10030453] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress is considered one of the pathological mechanisms that cause Parkinson’s disease (PD), which has led to the investigation of several antioxidants molecules as a potential therapeutic treatment against the disease. Although preclinical studies have demonstrated the efficacy of these compounds to maintain neuronal survival and activity in PD models, these results have not been reflected in clinical trials, antioxidants have not been able to act as disease modifiers in terms of clinical symptoms. Translational medicine currently faces the challenge of redesigning clinical trials to standardize criteria when testing molecules to reduce responses’ variability. Herein, we discuss current challenges and opportunities regarding several non-enzymatic antioxidants’ therapeutic molecules for PD patients’ potential treatment.
Collapse
|
8
|
Griffin SM, Pickard MR, Hawkins CP, Williams AC, Fricker RA. Nicotinamide restricts neural precursor proliferation to enhance catecholaminergic neuronal subtype differentiation from mouse embryonic stem cells. PLoS One 2020; 15:e0233477. [PMID: 32925933 PMCID: PMC7489539 DOI: 10.1371/journal.pone.0233477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 08/28/2020] [Indexed: 11/19/2022] Open
Abstract
Emerging evidence indicates that a strong relationship exists between brain regenerative therapies and nutrition. Early life nutrition plays an important role during embryonic brain development, and there are clear consequences to an imbalance in nutritional factors on both the production and survival of mature neuronal populations and the infant’s risk of diseases in later life. Our research and that of others suggest that vitamins play a fundamental role in the formation of neurons and their survival. There is a growing body of evidence that nicotinamide, the water-soluble amide form of vitamin B3, is implicated in the conversion of pluripotent stem cells to clinically relevant cells for regenerative therapies. This study investigated the ability of nicotinamide to promote the development of mature catecholaminergic neuronal populations (associated with Parkinson’s disease) from mouse embryonic stem cells, as well as investigating the underlying mechanisms of nicotinamide’s action. Nicotinamide selectively enhanced the production of tyrosine hydroxylase-expressing neurons and serotonergic neurons from mouse embryonic stem cell cultures (Sox1GFP knock-in 46C cell line). A 5-Ethynyl-2´-deoxyuridine (EdU) assay ascertained that nicotinamide, when added in the initial phase, reduced cell proliferation. Nicotinamide drove tyrosine hydroxylase-expressing neuron differentiation as effectively as an established cocktail of signalling factors, reducing the proliferation of neural progenitors and accelerating neuronal maturation, neurite outgrowth and neurotransmitter expression. These novel findings show that nicotinamide enhanced and enriched catecholaminergic differentiation and inhibited cell proliferation by directing cell cycle arrest in mouse embryonic stem cell cultures, thus driving a critical neural proliferation-to-differentiation switch from neural progenitors to neurons. Further research into the role of vitamin metabolites in embryogenesis will significantly advance cell-based regenerative medicine, and help realize their role as crucial developmental signalling molecules in brain development.
Collapse
Affiliation(s)
- Síle M. Griffin
- Keele Medical School and Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, England, United Kingdom
- * E-mail:
| | - Mark R. Pickard
- Chester Medical School, University Centre Shrewsbury, University of Chester, Shrewsbury, England, United Kingdom
| | - Clive P. Hawkins
- Keele Medical School and Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, England, United Kingdom
- Department of Neurology, Royal Stoke University Hospital, Stoke-on-Trent, Staffordshire, England, United Kingdom
| | - Adrian C. Williams
- Department of Neurosciences, University of Birmingham, Birmingham, England, United Kingdom
| | - Rosemary A. Fricker
- Keele Medical School and Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, England, United Kingdom
| |
Collapse
|
9
|
Precious SV, Smith GA, Heuer A, Jaeger I, Lane EL, Dunnett SB, Li M, Kelly CM, Rosser AE. Dopaminergic Progenitors Derived From Epiblast Stem Cells Function Similarly to Primary VM-Derived Progenitors When Transplanted Into a Parkinson's Disease Model. Front Neurosci 2020; 14:312. [PMID: 32317925 PMCID: PMC7154167 DOI: 10.3389/fnins.2020.00312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/17/2020] [Indexed: 11/13/2022] Open
Abstract
Neural transplantation in neurodegenerative diseases such as Parkinson’s disease (PD) offers to replace cells lost during the progression of the disease process. Primary fetal ventral mesencephalon (VM), the origin of bona fide midbrain dopaminergic (DAergic) precursors, is currently the gold standard source of cells for transplantation in PD. However, the use of tissue from this source raises ethical and logistical constraints necessitating the need for alternative supplies of donor cells. The requirement of any alternative donor cell source is to have the capability to generate authentic mature DAergic neurons, which could be utilized in cell-replacement strategies. Mouse pluripotent stem cells can efficiently generate electrochemically mature midbrain DAergic precursors in vitro using a stepwise control of FGF signaling. Here, we have compared DAergic transplants derived from two progenitor cell sources in an allograft system: mouse epiblast stem cells (EpiSC) and primary fetal mouse VM tissue. Cells were transplanted into the striatum of 6-OHDA lesioned mice pre-treated with L-DOPA. Drug-induced rotations, a number of motor tests and drug-induced abnormal involuntary movements (AIMs) were assessed. Functional improvements were demonstrated post-transplantation in some behavioral tests, with no difference in graft volume or the number of TH immuno-positive cells in the grafts of the two transplant groups. L-DOPA-induced AIMs and amphetamine-induced AIMs were observed in both transplant groups, with no differences in rate or severity between the two groups. Collectively, in this mouse-to-mouse allograft system, we report no significant differences in the functional ability between the gold standard primary VM derived and pluripotent stem cell-derived DAergic transplants.
Collapse
Affiliation(s)
- Sophie V Precious
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Gaynor A Smith
- School of Medicine, UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Andreas Heuer
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom.,Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Ines Jaeger
- Stem Cell Neurogenesis Group, School of Medicine and Biosciences, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Emma L Lane
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom
| | - Stephen B Dunnett
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Meng Li
- Stem Cell Neurogenesis Group, School of Medicine and Biosciences, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Claire M Kelly
- School of Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
| | - Anne E Rosser
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom.,Wales Brain Repair and Intracranial Neurotherapeutics Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom.,MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
10
|
Harischandra DS, Rokad D, Ghaisas S, Verma S, Robertson A, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Enhanced differentiation of human dopaminergic neuronal cell model for preclinical translational research in Parkinson's disease. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165533. [PMID: 31442530 DOI: 10.1016/j.bbadis.2019.165533] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 12/20/2022]
Abstract
Human-derived neuronal cell lines are progressively being utilized in understanding neurobiology and preclinical translational research as they are biologically more relevant than rodent-derived cells lines. The Lund human mesencephalic (LUHMES) cell line comprises human neuronal cells that can be differentiated to post-mitotic neurons and is increasingly being used as an in vitro model for various neurodegenerative diseases. A previously published 2-step differentiation procedure leads to the generation of post-mitotic neurons within 5-days, but only a small proportion (10%) of the total cell population tests positive for tyrosine hydroxylase (TH). Here we report on a novel differentiation protocol that we optimized by using a cocktail of neurotrophic factors, pleiotropic cytokines, and antioxidants to effectively generate proportionately more dopaminergic neurons within the same time period. Visualization and quantification of TH-positive cells revealed that under our new protocol, 25% of the total cell population expressed markers of dopaminergic neurons with the TH-positive neuron count peaking on day 5. These neurons showed spontaneous electrical activity and responded to known Parkinsonian toxins as expected by showing decreased cell viability and dopamine uptake and a concomitant increase in apoptotic cell death. Together, our results outline an improved method for generating a higher proportion of dopaminergic neurons, thus making these cells an ideal neuronal culture model of Parkinson's disease (PD) for translational research.
Collapse
Affiliation(s)
- Dilshan S Harischandra
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Dharmin Rokad
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Shivani Ghaisas
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Saurabh Verma
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Alan Robertson
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Huajun Jin
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Vellareddy Anantharam
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Anumantha G Kanthasamy
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA.
| |
Collapse
|
11
|
Rharass T, Lantow M, Gbankoto A, Weiss DG, Panáková D, Lucas S. Ascorbic acid alters cell fate commitment of human neural progenitors in a WNT/β-catenin/ROS signaling dependent manner. J Biomed Sci 2017; 24:78. [PMID: 29037191 PMCID: PMC5641995 DOI: 10.1186/s12929-017-0385-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 10/06/2017] [Indexed: 12/22/2022] Open
Abstract
Background Improving the neuronal yield from in vitro cultivated neural progenitor cells (NPCs) is an essential challenge in transplantation therapy in neurological disorders. In this regard, Ascorbic acid (AA) is widely used to expand neurogenesis from NPCs in cultures although the mechanisms of its action remain unclear. Neurogenesis from NPCs is regulated by the redox-sensitive WNT/β-catenin signaling pathway. We therefore aimed to investigate how AA interacts with this pathway and potentiates neurogenesis. Methods Effects of 200 μM AA were compared with the pro-neurogenic reagent and WNT/β-catenin signaling agonist lithium chloride (LiCl), and molecules with antioxidant activities i.e. N-acetyl-L-cysteine (NAC) and ruthenium red (RuR), in differentiating neural progenitor ReNcell VM cells. Cells were supplemented with reagents for two periods of treatment: a full period encompassing the whole differentiation process versus an early short period that is restricted to the cell fate commitment stage. Intracellular redox balance and reactive oxygen species (ROS) metabolism were examined by flow cytometry using redox and ROS sensors. Confocal microscopy was performed to assess cell viability, neuronal yield, and levels of two proteins: Nucleoredoxin (NXN) and the WNT/β-catenin signaling component Dishevelled 2 (DVL2). TUBB3 and MYC gene responses were evaluated by quantitative real-time PCR. DVL2-NXN complex dissociation was measured by fluorescence resonance energy transfer (FRET). Results In contrast to NAC which predictably exhibited an antioxidant effect, AA treatment enhanced ROS metabolism with no cytotoxic induction. Both drugs altered ROS levels only at the early stage of the differentiation as no changes were held beyond the neuronal fate commitment stage. FRET studies showed that AA treatment accelerated the redox-dependent release of the initial pool of DVL2 from its sequestration by NXN, while RuR treatment hampered the dissociation of the two proteins. Accordingly, AA increased WNT/β-catenin signaling output i.e. MYC mRNA level, whereas RuR attenuated it. Moreover, AA improved neurogenesis as much as LiCl as both TUBB3-positive cell yield and TUBB3 mRNA level increased, while NAC or RuR attenuated neurogenesis. Markedly, the neurogenesis outputs between the short and the full treatment with either NAC or AA were found unchanged, supporting our model that neuronal yield is altered by events taking place at the early phase of differentiation. Conclusions Our findings demonstrate that AA treatment elevates ROS metabolism in a non-lethal manner prior to the NPCs commitment to their neuronal fate. Such effect stimulates the redox-sensitive DVL2 activation and WNT/β-catenin signaling response that would enhance the ensuing neuronal cell differentiation. Electronic supplementary material The online version of this article (10.1186/s12929-017-0385-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tareck Rharass
- Physiopathology of Inflammatory Bone Diseases, University of the Littoral Opal Coast, F-62327, Boulogne sur Mer, France. .,Electrochemical Signaling in Development and Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, D-13125, Berlin, Germany. .,Physiopathology of Inflammatory Bone Diseases, University of the Littoral Opal Coast, Boulevard Bassin Napoléon - Quai Masset, B.P. 120, F-62327, Boulogne sur Mer, Cédex, France.
| | - Margareta Lantow
- Cell Biology and Biosystems Technology, Institute of Biological Sciences, University of Rostock, D-18059, Rostock, Germany
| | - Adam Gbankoto
- Department of Animal Physiology, Faculty of Sciences and Technics, University of Abomey-Calavi, 01, BP, 526, Cotonou, Benin
| | - Dieter G Weiss
- Cell Biology and Biosystems Technology, Institute of Biological Sciences, University of Rostock, D-18059, Rostock, Germany
| | - Daniela Panáková
- Electrochemical Signaling in Development and Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, D-13125, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, D-13125, Berlin, Germany
| | - Stéphanie Lucas
- Physiopathology of Inflammatory Bone Diseases, University of the Littoral Opal Coast, F-62327, Boulogne sur Mer, France
| |
Collapse
|
12
|
Griffin SM, Pickard MR, Orme RP, Hawkins CP, Williams AC, Fricker RA. Nicotinamide alone accelerates the conversion of mouse embryonic stem cells into mature neuronal populations. PLoS One 2017; 12:e0183358. [PMID: 28817722 PMCID: PMC5560552 DOI: 10.1371/journal.pone.0183358] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 08/02/2017] [Indexed: 01/26/2023] Open
Abstract
Introduction Vitamin B3 has been shown to play an important role during embryogenesis. Specifically, there is growing evidence that nicotinamide, the biologically active form of vitamin B3, plays a critical role as a morphogen in the differentiation of stem cells to mature cell phenotypes, including those of the central nervous system (CNS). Detailed knowledge of the action of small molecules during neuronal differentiation is not only critical for uncovering mechanisms underlying lineage-specification, but also to establish more effective differentiation protocols to obtain clinically relevant cells for regenerative therapies for neurodegenerative conditions such as Huntington’s disease (HD). Thus, this study aimed to investigate the potential of nicotinamide to promote the conversion of stem cells to mature CNS neurons. Methods Nicotinamide was applied to differentiating mouse embryonic stem cells (mESC; Sox1GFP knock-in 46C cell line) during their conversion towards a neural fate. Cells were assessed for changes in their proliferation, differentiation and maturation; using immunocytochemistry and morphometric analysis methods. Results Results presented indicate that 10 mM nicotinamide, when added at the initial stages of differentiation, promoted accelerated progression of ESCs to a neural lineage in adherent monolayer cultures. By 14 days in vitro (DIV), early exposure to nicotinamide was shown to increase the numbers of differentiated βIII-tubulin-positive neurons. Nicotinamide decreased the proportion of pluripotent stem cells, concomitantly increasing numbers of neural progenitors at 4 DIV. These progenitors then underwent rapid conversion to neurons, observed by a reduction in Sox 1 expression and decreased numbers of neural progenitors in the cultures at 14 DIV. Furthermore, GABAergic neurons generated in the presence of nicotinamide showed increased maturity and complexity of neurites at 14 DIV. Therefore, addition of nicotinamide alone caused an accelerated passage of pluripotent cells through lineage specification and further to non-dividing mature neurons. Conclusions Our results show that, within an optimal dose range, nicotinamide is able to singly and selectively direct the conversion of embryonic stem cells to mature neurons, and therefore may be a critical factor for normal brain development, thus supporting previous evidence of the fundamental role of vitamins and their metabolites during early CNS development. In addition, nicotinamide may offer a simple effective supplement to enhance the conversion of stem cells to clinically relevant neurons.
Collapse
Affiliation(s)
- Síle M. Griffin
- Keele Medical School and Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, England, United Kingdom
| | - Mark R. Pickard
- Chester Centre for Stress Research, Institute of Medicine, University of Chester, Chester, United Kingdom
| | - Rowan P. Orme
- Keele Medical School and Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, England, United Kingdom
| | - Clive P. Hawkins
- Keele Medical School and Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, England, United Kingdom
- Department of Neurology, University Hospital of North Staffordshire, Stoke-on-Trent, Staffordshire, England, United Kingdom
| | - Adrian C. Williams
- Department of Neurosciences, University of Birmingham, Birmingham, United Kingdom
| | - Rosemary A. Fricker
- Keele Medical School and Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, England, United Kingdom
- * E-mail:
| |
Collapse
|
13
|
Li W, Chen S, Li JY. Human induced pluripotent stem cells in Parkinson's disease: A novel cell source of cell therapy and disease modeling. Prog Neurobiol 2015; 134:161-77. [PMID: 26408505 DOI: 10.1016/j.pneurobio.2015.09.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 09/15/2015] [Accepted: 09/17/2015] [Indexed: 12/16/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs) and human embryonic stem cells (hESCs) are two novel cell sources for studying neurodegenerative diseases. Dopaminergic neurons derived from hiPSCs/hESCs have been implicated to be very useful in Parkinson's disease (PD) research, including cell replacement therapy, disease modeling and drug screening. Recently, great efforts have been made to improve the application of hiPSCs/hESCs in PD research. Considerable advances have been made in recent years, including advanced reprogramming strategies without the use of viruses or using fewer transcriptional factors, optimized methods for generating highly homogeneous neural progenitors with a larger proportion of mature dopaminergic neurons and better survival and integration after transplantation. Here we outline the progress that has been made in these aspects in recent years, particularly during the last year, and also discuss existing issues that need to be addressed.
Collapse
Affiliation(s)
- Wen Li
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Rui Jin Er Road, Shanghai 200025, China; Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Lund University, BMC A10, 221 84 Lund, Sweden
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Rui Jin Er Road, Shanghai 200025, China.
| | - Jia-Yi Li
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, China; Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Lund University, BMC A10, 221 84 Lund, Sweden.
| |
Collapse
|
14
|
He XB, Kim M, Kim SY, Yi SH, Rhee YH, Kim T, Lee EH, Park CH, Dixit S, Harrison FE, Lee SH. Vitamin C facilitates dopamine neuron differentiation in fetal midbrain through TET1- and JMJD3-dependent epigenetic control manner. Stem Cells 2015; 33:1320-32. [PMID: 25535150 PMCID: PMC4435601 DOI: 10.1002/stem.1932] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/06/2014] [Accepted: 12/04/2014] [Indexed: 12/11/2022]
Abstract
Intracellular Vitamin C (VC) is maintained at high levels in the developing brain by the activity of sodium-dependent VC transporter 2 (Svct2), suggesting specific VC functions in brain development. A role of VC as a cofactor for Fe(II)-2-oxoglutarate-dependent dioxygenases has recently been suggested. We show that VC supplementation in neural stem cell cultures derived from embryonic midbrains greatly enhanced differentiation toward midbrain-type dopamine (mDA) neurons, the neuronal subtype associated with Parkinson's disease. VC induced gain of 5-hydroxymethylcytosine (5hmC) and loss of H3K27m3 in DA phenotype gene promoters, which are catalyzed by Tet1 and Jmjd3, respectively. Consequently, VC enhanced DA phenotype gene transcriptions in the progenitors by Nurr1, a transcription factor critical for mDA neuron development, to be more accessible to the gene promoters. Further mechanism studies including Tet1 and Jmjd3 knockdown/inhibition experiments revealed that both the 5hmC and H3K27m3 changes, specifically in the progenitor cells, are indispensible for the VC-mediated mDA neuron differentiation. We finally show that in Svct2 knockout mouse embryos, mDA neuron formation in the developing midbrain decreased along with the 5hmC/H3k27m3 changes. These findings together indicate an epigenetic role of VC in midbrain DA neuron development.
Collapse
Affiliation(s)
- Xi-Biao He
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 133-791, Korea
| | - Mirang Kim
- Biomedical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 305-806, Korea
| | - Seon-Young Kim
- Biomedical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 305-806, Korea
| | - Sang-Hoon Yi
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 133-791, Korea
| | - Yong-Hee Rhee
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 133-791, Korea
| | - Taeho Kim
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 133-791, Korea
| | - Eun-Hye Lee
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 133-791, Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 133-791, Korea
| | - Chang-Hwan Park
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 133-791, Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 133-791, Korea
| | - Shilpy Dixit
- Division of Diabetes, Endocrinology & Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee, U.S.A
| | - Fiona E. Harrison
- Division of Diabetes, Endocrinology & Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee, U.S.A
| | - Sang-Hun Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 133-791, Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 133-791, Korea
| |
Collapse
|
15
|
Skalova S, Svadlakova T, Shaikh Qureshi WM, Dev K, Mokry J. Induced pluripotent stem cells and their use in cardiac and neural regenerative medicine. Int J Mol Sci 2015; 16:4043-67. [PMID: 25689424 PMCID: PMC4346943 DOI: 10.3390/ijms16024043] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 01/27/2015] [Accepted: 02/02/2015] [Indexed: 12/20/2022] Open
Abstract
Stem cells are unique pools of cells that are crucial for embryonic development and maintenance of adult tissue homeostasis. The landmark Nobel Prize winning research by Yamanaka and colleagues to induce pluripotency in somatic cells has reshaped the field of stem cell research. The complications related to the usage of pluripotent embryonic stem cells (ESCs) in human medicine, particularly ESC isolation and histoincompatibility were bypassed with induced pluripotent stem cell (iPSC) technology. The human iPSCs can be used for studying embryogenesis, disease modeling, drug testing and regenerative medicine. iPSCs can be diverted to different cell lineages using small molecules and growth factors. In this review we have focused on iPSC differentiation towards cardiac and neuronal lineages. Moreover, we deal with the use of iPSCs in regenerative medicine and modeling diseases like myocardial infarction, Timothy syndrome, dilated cardiomyopathy, Parkinson’s, Alzheimer’s and Huntington’s disease. Despite the promising potential of iPSCs, genome contamination and low efficacy of cell reprogramming remain significant challenges.
Collapse
Affiliation(s)
- Stepanka Skalova
- Department of Histology and Embryology, Medical Faculty in Hradec Kralove, Charles University in Prague, Simkova 870, Hradec Kralove 50038, Czech Republic.
| | - Tereza Svadlakova
- Department of Histology and Embryology, Medical Faculty in Hradec Kralove, Charles University in Prague, Simkova 870, Hradec Kralove 50038, Czech Republic.
| | - Wasay Mohiuddin Shaikh Qureshi
- Department of Histology and Embryology, Medical Faculty in Hradec Kralove, Charles University in Prague, Simkova 870, Hradec Kralove 50038, Czech Republic.
| | - Kapil Dev
- Department of Histology and Embryology, Medical Faculty in Hradec Kralove, Charles University in Prague, Simkova 870, Hradec Kralove 50038, Czech Republic.
| | - Jaroslav Mokry
- Department of Histology and Embryology, Medical Faculty in Hradec Kralove, Charles University in Prague, Simkova 870, Hradec Kralove 50038, Czech Republic.
| |
Collapse
|
16
|
Ide K, Yamada H, Umegaki K, Mizuno K, Kawakami N, Hagiwara Y, Matsumoto M, Yoshida H, Kim K, Shiosaki E, Yokochi T, Harada K. Lymphocyte vitamin C levels as potential biomarker for progression of Parkinson's disease. Nutrition 2014; 31:406-8. [PMID: 25592020 DOI: 10.1016/j.nut.2014.08.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/31/2014] [Accepted: 08/02/2014] [Indexed: 11/16/2022]
Abstract
OBJECTIVES Vitamin C is a major antioxidant and also is known as a neuromodulator in dopaminergic neurons. The aim of this study was to investigate the association between lymphocyte and plasma vitamin C levels in various stages of Parkinson's disease (PD). METHODS Sixty-two individuals with PD (age 71 ± 8.8 y [mean ± SD]) being treated at Shizuoka General Hospital from December 2007 to August 2013 were consecutively recruited. PD severity was classified using the Hoehn-Yahr scale for staging PD. Fasting blood samples were collected, and plasma and lymphocyte vitamin C levels were measured. The association between PD severity and vitamin C levels was estimated by ordinal logistic regression with confounding variables. RESULTS The distribution of Hoehn-Yahr stages in patients was as follows: stage I, 7; II, 28; III, 16; and IV, 11. Lymphocyte vitamin C levels in patients with severe PD were significantly lower (odds ratio [OR], 0.87; 95% confidence interval [CI], 0.80-0.97; P < 0.01) compared with those at less severe stages. Plasma vitamin C levels also tended to be lower in patients with severe PD; however, this was not significant (OR, 0.98; 95% CI, 0.96-1.00; P = 0.09). CONCLUSIONS Our findings suggest that lymphocyte vitamin C levels in the peripheral blood may be a potentially useful biomarker for the progression of PD.
Collapse
Affiliation(s)
- Kazuki Ide
- Department of Drug Evaluation and Informatics, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Hiroshi Yamada
- Department of Drug Evaluation and Informatics, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan; Department of Neurology, Shizuoka General Hospital, Shizuoka, Japan.
| | - Keizo Umegaki
- National Institute of Health and Nutrition, Tokyo, Japan
| | - Katsuki Mizuno
- Department of Drug Evaluation and Informatics, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Nobuko Kawakami
- Department of Neurology, Shizuoka General Hospital, Shizuoka, Japan
| | - Yuka Hagiwara
- Department of Neurology, Shizuoka General Hospital, Shizuoka, Japan
| | - Mizuki Matsumoto
- Department of Neurology, Shizuoka General Hospital, Shizuoka, Japan
| | - Hidefumi Yoshida
- Department of Neurology, Shizuoka General Hospital, Shizuoka, Japan
| | - Kang Kim
- Department of Neurology, Shizuoka General Hospital, Shizuoka, Japan
| | - Emi Shiosaki
- Department of Clinical Laboratory Medicine, Shizuoka General Hospital, Shizuoka, Japan
| | - Tsunehiro Yokochi
- Department of Clinical Laboratory Medicine, Shizuoka General Hospital, Shizuoka, Japan
| | - Kiyoshi Harada
- Department of Neurology, Shizuoka General Hospital, Shizuoka, Japan
| |
Collapse
|
17
|
Abstract
Factors controlling proliferation and differentiation are crucial in advancement of neural cell-based experimental neurodegenerative therapies. In this regard, nicotinamide has been shown to determine the fate of neural cells, enhance neuralization, and influence DNA repair and apoptosis. This study investigated whether the biologically active vitamin B3 metabolite, nicotinamide, could direct the differentiation of mouse embryonic stem cells, cultured as monolayers, into neurons at either early or late stages of development. Interestingly, we observed a dose-responsive increase in the percentage of neurons when nicotinamide was added at early stages to the cells undergoing differentiation (days 0-7). Nicotinamide (10 mM) had a significant effect on neuronal differentiation, increasing the βIII-tubulin-positive neuronal population and concomitantly decreasing the total number of cells in culture, measured by quantification of 4',6-diamidino-2-phenylindole (DAPI)-positive cells. Nicotinamide added between days 7 and 14 had no effect on neuronal induction. High levels of nicotinamide (20 mM) induced cytotoxicity and cell death. Current work is focusing on elucidating the mechanism(s) mediating neural specification by nicotinamide--that is, induction of cell-cycle exit and/or selective apoptosis in non-neural populations. Preliminary data suggest a reduction in the proportion of proliferating cells in nicotinamide-treated cultures--that is, nicotinamide enhances cell-cycle exit, thereby promoting neuronal differentiation. Future work will focus on evaluating the effect of nicotinamide on the differentiation of midbrain dopamine neurons, towards a therapy for Parkinson's disease.
Collapse
|
18
|
Orme RP, Bhangal MS, Fricker RA. Calcitriol imparts neuroprotection in vitro to midbrain dopaminergic neurons by upregulating GDNF expression. PLoS One 2013; 8:e62040. [PMID: 23626767 PMCID: PMC3633905 DOI: 10.1371/journal.pone.0062040] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 03/18/2013] [Indexed: 11/19/2022] Open
Abstract
During development a tightly controlled signaling cascade dictates the differentiation, maturation and survival of developing neurons. Understanding this signaling mechanism is important for developing therapies for neurodegenerative illnesses. In previous work we have sought to understand the complex signaling pathways responsible for the development of midbrain dopamine neurons using a proteomic approach. One protein we have identified as being expressed in developing midbrain tissue is the vitamin D receptor. Therefore we investigated the effect of the biologically active vitamin D3 metabolite, calcitriol, on primary fetal ventral mesencephalic cultures of dopamine neurons. We observed a dose responsive increase in numbers of rat primary dopamine neurons when calcitriol was added to culture media. Western blot data showed that calcitriol upregulated the expression of glial derived neurotrophic factor (GDNF). Blocking GDNF signaling could prevent calcitriol's ability to increase numbers of dopamine neurons. An apoptosis assay and cell birth dating experiment revealed that calcitriol increases the number of dopamine neurons through neuroprotection and not increased differentiation. This could have implications for future neuroprotective PD therapies.
Collapse
Affiliation(s)
- Rowan P. Orme
- Institute for Science and Technology in Medicine and Department of Life Sciences, Keele University, Keele, Staffordshire, England
| | | | - Rosemary A. Fricker
- Institute for Science and Technology in Medicine and Department of Life Sciences, Keele University, Keele, Staffordshire, England
- School of Medicine, Keele University, Keele, Staffordshire, England
| |
Collapse
|
19
|
Vukicevic V, Schmid J, Hermann A, Lange S, Qin N, Gebauer L, Chunk KF, Ravens U, Eisenhofer G, Storch A, Ader M, Bornstein SR, Ehrhart-Bornstein M. Differentiation of chromaffin progenitor cells to dopaminergic neurons. Cell Transplant 2012; 21:2471-86. [PMID: 22507143 DOI: 10.3727/096368912x638874] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The differentiation of dopamine-producing neurons from chromaffin progenitors might represent a new valuable source for replacement therapies in Parkinson's disease. However, characterization of their differentiation potential is an important prerequisite for efficient engraftment. Based on our previous studies on isolation and characterization of chromaffin progenitors from adult adrenals, this study investigates their potential to produce dopaminergic neurons and means to enhance their dopaminergic differentiation. Chromaffin progenitors grown in sphere culture showed an increased expression of nestin and Mash1, indicating an increase of the progenitor subset. Proneurogenic culture conditions induced the differentiation into neurons positive for neural markers β-III-tubulin, MAP2, and TH accompanied by a decrease of Mash1 and nestin. Furthermore, Notch2 expression decreased concomitantly with a downregulation of downstream effectors Hes1 and Hes5 responsible for self-renewal and proliferation maintenance of progenitor cells. Chromaffin progenitor-derived neurons secreted dopamine upon stimulation by potassium. Strikingly, treatment of differentiating cells with retinoic and ascorbic acid resulted in a twofold increase of dopamine secretion while norepinephrine and epinephrine were decreased. Initiation of dopamine synthesis and neural maturation is controlled by Pitx3 and Nurr1. Both Pitx3 and Nurr1 were identified in differentiating chromaffin progenitors. Along with the gained dopaminergic function, electrophysiology revealed features of mature neurons, such as sodium channels and the capability to fire multiple action potentials. In summary, this study elucidates the capacity of chromaffin progenitor cells to generate functional dopaminergic neurons, indicating their potential use in cell replacement therapies.
Collapse
Affiliation(s)
- Vladimir Vukicevic
- Molecular Endocrinology, Medical Clinic III, University Clinic Dresden, Dresden University of Technology, Fetscherstrasse 74, Dresden, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Modulation of Dopaminergic Neuronal Differentiation from Sympathoadrenal Progenitors. J Mol Neurosci 2012; 48:420-6. [DOI: 10.1007/s12031-012-9746-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 03/05/2012] [Indexed: 12/25/2022]
|
21
|
Ehrhart-Bornstein M, Vukicevic V, Chung KF, Ahmad M, Bornstein SR. Chromaffin progenitor cells from the adrenal medulla. Cell Mol Neurobiol 2010; 30:1417-23. [PMID: 21080061 DOI: 10.1007/s10571-010-9571-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 09/02/2010] [Indexed: 11/26/2022]
Abstract
Chromaffin cells of the adrenal medulla are neural crest-derived cells of the sympathoadrenal lineage. Different lines of evidence suggest the existence of a subpopulation of proliferation-competent progenitor cells even in the adult state. The identification of sympathoadrenal progenitors in the adrenal would greatly enhance the understanding of adrenal physiology and their potential role in adrenal pathogenesis. Isolation and differentiation of these progenitor cells in culture would provide a tool to understand their development in vitro. Furthermore, due to the close relation to sympathetic neurons, these cells might provide an expandable source of cells for cell therapy in the treatment of neurodegenerative diseases. We therefore aim to establish protocols for the efficient isolation, enrichment and differentiation of chromaffin progenitor cells to dopaminergic neurons in culture.
Collapse
|
22
|
Mejía-Toiber J, Márquez-Ramos JA, Díaz-Muñoz M, Peña F, Aguilar MB, Giordano M. Glutamatergic Excitation and GABA Release from a Transplantable Cell Line. Cell Transplant 2010; 19:1307-23. [DOI: 10.3727/096368910x509059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The cell line M213-2O CL-4 was derived from cell line M213-2O and further modified to express human glutamate decarboxylase (hGAD-67), the enzyme that synthesizes GABA. Brain transplants of this cell line in animal models of epilepsy have been shown to modulate seizures. However, the mechanisms that underlie such actions are unknown. The purpose of the present study was to characterize this cell line and its responsiveness to several depolarizing conditions, in order to better understand how these cells exert their effects. Intracellular GABA levels were 34-fold higher and GAD activity was 16-fold higher in clone M213-2O CL-4 than in M213-2O. Both cell lines could take up [3H]GABA in vitro, and this uptake was prevented by nipecotic acid. By combining GABA release measurements and calcium imaging in vitro, we found that high extracellular K+, zero Mg2+, or glutamate activated M213-2O CL-4 cells and resulted in GABA release. The response to glutamate appeared to be mediated by AMPA/NMDA-like receptors. High KCl-induced GABA release was prevented when a Ca2+-free Krebs solution was used, suggesting an exocytotic-like mechanism. These results indicate that the cell line M213-2O CL-4 synthesizes, releases, and takes up GABA in vitro, and can be activated by depolarizing stimuli.
Collapse
Affiliation(s)
- Jana Mejía-Toiber
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | | | - Mauricio Díaz-Muñoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Quéretaro, México
| | - Fernando Peña
- Departamento de Farmacobiología, CINVESTAV-Sur. Calzada de los Tenorios 235, Delegación Tlalpan, México
| | - Manuel B. Aguilar
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Quéretaro, México
| | - Magda Giordano
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| |
Collapse
|
23
|
Hackl C, Papazoglou A, Ganser C, Klein A, Prakash N, Wurst W, Nikkhah G. Ectopic Dopaminergic Progenitor Cells from En1+/Otx2lacZ Transgenic Mice Survive and Functionally Reinnervate the Striatum Following Transplantation in a Rat Model of Parkinson's Disease. Cell Transplant 2010; 19:1085-101. [DOI: 10.3727/096368910x505468] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cell-based therapies for Parkinson's disease (PD) using neural stem cells to replace the lost dopamine neurons is currently an intense area of research. In this study we have evaluated the restorative potential of ectopic dopaminergic (DA) neurons derived from the rostral hindbrain (RH) of En1 +/Otx2lacZ transgenic mice. The genetic modification of the DA progenitor domain in the En1 +/Otx2lacZ mice is a gain of function, resulting in the enlargement of the area containing DA neurons, as well as an increase in their absolute number in the midbrain/hindbrain region. Amphetamine-induced rotation performed after cell transplantation into the unilaterally 6-hydroxydopamine-lesioned rat striatum revealed that animals with transgenic RH-derived DA grafts exhibited functional recovery similar to transgenic and wild-type ventral mesencephalon (VM)-derived DA grafts. Morphological analyses revealed equivalent numbers of surviving DA neurons from both homotopic VM- and ectopic RH-derived grafts from transgenic donors with low numbers of surviving serotonergic (5-HT) neurons. Conversely, grafts derived from wild-type donors contained predominantly surviving DA neurons or 5-HT neurons when they were prepared from the VM or RH, respectively. The study demonstrates the pattern of survival and functional potential of ectopic DA neurons derived from the RH of En1 +/Otx2lacZ transgenic mice and that cell transplantation is an important neurobiological tool to characterize newly generated DA neural stem cells in vivo.
Collapse
Affiliation(s)
- Christina Hackl
- Department of Stereotactic and Functional Neurosurgery, Laboratory of Molecular Neurosurgery, Neurocenter, University Hospital Freiburg, Freiburg, Germany
| | - Anna Papazoglou
- Department of Stereotactic and Functional Neurosurgery, Laboratory of Molecular Neurosurgery, Neurocenter, University Hospital Freiburg, Freiburg, Germany
| | - Claudia Ganser
- Department of Stereotactic and Functional Neurosurgery, Laboratory of Molecular Neurosurgery, Neurocenter, University Hospital Freiburg, Freiburg, Germany
| | - Alexander Klein
- Department of Stereotactic and Functional Neurosurgery, Laboratory of Molecular Neurosurgery, Neurocenter, University Hospital Freiburg, Freiburg, Germany
| | - Nilima Prakash
- Helmholtz Centre Munich, German Research Centre for Environmental Health (GmbH) and Technical University Munich, Institute of Developmental Genetics, Munich/Neuherberg, Germany
| | - Wolfgang Wurst
- Helmholtz Centre Munich, German Research Centre for Environmental Health (GmbH) and Technical University Munich, Institute of Developmental Genetics, Munich/Neuherberg, Germany
| | - Guido Nikkhah
- Department of Stereotactic and Functional Neurosurgery, Laboratory of Molecular Neurosurgery, Neurocenter, University Hospital Freiburg, Freiburg, Germany
| |
Collapse
|
24
|
Willis LM, Freeman L, Bickford PC, Quintero EM, Umphlet CD, Moore AB, Goetzl L, Granholm AC. Blueberry supplementation attenuates microglial activation in hippocampal intraocular grafts to aged hosts. Glia 2010; 58:679-90. [PMID: 20014277 PMCID: PMC2834232 DOI: 10.1002/glia.20954] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Transplantation of central nervous tissue has been proposed as a therapeutic intervention for age-related neurodegenerative diseases and stroke. However, survival of embryonic neuronal cells is hampered by detrimental factors in the aged host brain such as circulating inflammatory cytokines and oxidative stress. We have previously found that supplementation with 2% blueberry in the diet increases graft growth and neuronal survival in intraocular hippocampal grafts to aged hosts. In the present study we explored possible biochemical mechanisms for this increased survival, and we here report decreased microglial activation and astrogliosis in intraocular hippocampal grafts to middle-aged hosts fed a 2% blueberry diet. Markers for astrocytes and for activated microglial cells were both decreased long-term after grafting to blueberry-treated hosts compared with age-matched rats on a control diet. Similar findings were obtained in the host brain, with a reduction in OX-6 immunoreactive microglial cells in the hippocampus of those recipients treated with blueberry. In addition, immunoreactivity for the pro-inflammatory cytokine IL-6 was found to be significantly attenuated in intraocular grafts by the 2% blueberry diet. These studies demonstrate direct effects of blueberry upon microglial activation both during isolated conditions and in the aged host brain and suggest that this nutraceutical can attenuate age-induced inflammation.
Collapse
Affiliation(s)
- Lauren M. Willis
- Department of Neurosciences and the Center on Aging, Medical University of South Carolina, Charleston, SC, USA
| | - Linnea Freeman
- Department of Neurosciences and the Center on Aging, Medical University of South Carolina, Charleston, SC, USA
| | - Paula C. Bickford
- Department of Neurosurgery, Center for Excellence for Aging and Brain Repair, University of South Florida, Tampa, FL, USA
- James A. Haley Veterans' Hospital Medical Center, Tampa, FL, USA
| | - E. Matthew Quintero
- Department of Neurosciences and the Center on Aging, Medical University of South Carolina, Charleston, SC, USA
| | - Claudia D. Umphlet
- Department of Neurosciences and the Center on Aging, Medical University of South Carolina, Charleston, SC, USA
| | - Alfred B. Moore
- Department of Neurosciences and the Center on Aging, Medical University of South Carolina, Charleston, SC, USA
| | - Laura Goetzl
- Department of Ob/Gyn, Division of Maternal Fetal Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Ann-Charlotte Granholm
- Department of Neurosciences and the Center on Aging, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
25
|
Park DH, Eve DJ, Musso J, Klasko SK, Cruz E, Borlongan CV, Sanberg PR. Inflammation and Stem Cell Migration to the Injured Brain in Higher Organisms. Stem Cells Dev 2009; 18:693-702. [DOI: 10.1089/scd.2009.0008] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Dong-Hyuk Park
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, Florida
- Department of Neurosurgery, Korea University Medical Center, Korea University, Seoul, Korea
| | - David J. Eve
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, Florida
| | - James Musso
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, Florida
| | | | - Eduardo Cruz
- Cryopraxis, CellPraxis, BioRio, Pólo de Biotecnologia do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cesario V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, Florida
| | - Paul R. Sanberg
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, Florida
| |
Collapse
|