1
|
Moradi Vastegani Z, Ghaedi-Heidari R, Oroujalian A, Peymani M, Ghaedi K. Manuscript title: unravelling the neuroprotective role of miR-27a-3p in the MAPK pathway in Parkinson's disease. Metab Brain Dis 2025; 40:141. [PMID: 40067516 DOI: 10.1007/s11011-025-01568-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 03/02/2025] [Indexed: 03/26/2025]
Abstract
Parkinson's disease (PD) is a multifaceted neurodegenerative disorder characterized by dopaminergic neuron loss and the presence of Lewy bodies. Beyond its hallmark motor symptoms, PD involves significant neuroinflammation and immune dysfunction, driven by dysregulated signalling pathways such as the Mitogen-Activated Protein Kinase (MAPK) pathway. This study investigates the therapeutic potential of hsa-miR-27a-3p in modulating these pathways, with a focus on its interaction with MKK7, a key MAPK component. Bioinformatics and experimental analyses, using miRNA-mRNA interactions and construct a protein-protein interaction (PPI) network, confirm that hsa-miR-27a-3p directly binds to the 3' untranslated region (3'UTR) of MKK7, reducing its expression. Overexpression of hsa-miR-27a-3p improves cell viability, mitigates morphological changes, and reduces neurotoxicity in SH-SY5Y (human neuroblastoma cell line for experimental validation) cells exposed to MPP +, a PD neurotoxin. The study further demonstrates that hsa-miR-27a-3p modulates apoptotic pathways by increasing anti-apoptotic BCL2 while downregulating pro-apoptotic BAX and P53, as assessed through Western blot analysis of protein expression in SH-SY5Y cells transfected with miR-27a-3p mimic or negative control, followed by quantification of protein levels. Additionally, hsa-miR-27a-3p suppresses neuroinflammatory responses by significantly reducing TNF-α and IL1β levels. Western blot analysis reveals that hsa-miR-27a-3p inhibits phosphorylation of MKK7 and other MAPK pathway components, such as JNK and p38, highlighting its role in attenuating neuroinflammation and oxidative stress. These findings establish a negative correlation between hsa-miR-27a-3p expression and key neurodegenerative processes, suggesting its potential as a therapeutic target. This study provides comprehensive insights into the neuroprotective mechanisms of hsa-miR-27a-3p, paving the way for innovative interventions in PD management.
Collapse
Affiliation(s)
- Zahra Moradi Vastegani
- Department of Biology, Faculty of Science, Nourdanesh Institute of Higher Education, Isfahan, Iran
| | - Rasoul Ghaedi-Heidari
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Andisheh Oroujalian
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, 88137-33395, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, 88137-33395, Iran.
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar Jerib Ave., Azadi Sq., Isfahan, 81746-73441, Iran.
| |
Collapse
|
2
|
Harris AR, McGivern P, Gilbert F, Van Bergen N. Defining Biomarkers in Stem Cell-Derived Tissue Constructs for Drug and Disease Screening. Adv Healthc Mater 2024; 13:e2401433. [PMID: 38741544 DOI: 10.1002/adhm.202401433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 04/24/2024] [Indexed: 05/16/2024]
Abstract
The development of stem cell-derived tissue constructs (SCTCs) for clinical applications, including regenerative medicine, drug and disease screening offers significant hope for detecting and treating intractable disorders. SCTCs display a variety of biomarkers that can be used to understand biological mechanisms, assess drug interactions, and predict disease. Although SCTCs can be derived from patients and share the same genetic make-up, they are nevertheless distinct from human patients in many significant ways, which can undermine the clinical significance of measurements in SCTCs. This study defines biomarkers, how they apply to SCTCs, and clarifies specific ethical issues associated with the use of SCTCs for drug and disease screening.
Collapse
Affiliation(s)
- Alexander R Harris
- Department of Biomedical Engineering, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Patrick McGivern
- School of Humanities and Social Inquiry, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Frederic Gilbert
- School of Humanities, University of Tasmania, Hobart, Tasmania, Australia
| | - Nicole Van Bergen
- Brain and Mitochondrial Research Group, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, 3002, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, 3002, Australia
| |
Collapse
|
3
|
Cerutti L, Brofiga M. Unraveling brain diseases: The promise of brain-on-a-chip models. J Neurosci Methods 2024; 405:110105. [PMID: 38460796 DOI: 10.1016/j.jneumeth.2024.110105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 02/23/2024] [Accepted: 03/03/2024] [Indexed: 03/11/2024]
Abstract
Brain disorders, encompassing a wide spectrum of neurological and psychiatric conditions, present a formidable challenge in modern medicine. Despite decades of research, the intricate complexity of the human brain still eludes comprehensive understanding, impeding the development of effective treatments. Recent advancements in microfluidics and tissue engineering have led to the development of innovative platforms known as "Brain-on-a-Chip" (BoC) i.e., advanced in vitro systems that aim to replicate the microenvironment of the brain with the highest possible fidelity. This technology offers a promising test-bed for studying brain disorders at the cellular and network levels, providing insights into disease mechanisms, drug screening, and, in perspective, the development of personalized therapeutic strategies. In this review, we provide an overview of the BoC models developed over the years to model and understand the onset and progression of some of the most severe neurological disorders in terms of incidence and debilitation (stroke, Parkinson's, Alzheimer's, and epilepsy). We also report some of the cutting-edge therapeutic approaches whose effects were evaluated by means of these technologies. Finally, we discuss potential challenges, and future perspectives of the BoC models.
Collapse
Affiliation(s)
- Letizia Cerutti
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBIRS), University of Genova, Genova, Italy
| | - Martina Brofiga
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBIRS), University of Genova, Genova, Italy; ScreenNeuroPharm s.r.l, Sanremo, Italy; Neurofacility, Istituto Italiano di Tecnologia, Genova, Italy.
| |
Collapse
|
4
|
Arora S, Bajaj T, Kumar J, Goyal M, Singh A, Singh C. Recent Advances in Delivery of Peptide and Protein Therapeutics to the Brain. J Pharmacol Exp Ther 2024; 388:54-66. [PMID: 37977811 DOI: 10.1124/jpet.123.001690] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 10/05/2023] [Accepted: 10/25/2023] [Indexed: 11/19/2023] Open
Abstract
The classes of neuropharmaceuticals known as proteins and peptides serve as diagnostic tools and are involved in specific communication in the peripheral and central nervous systems. However, due to tight junctions resembling epithelial cells found in the blood-brain barrier (BBB) in vivo, they are typically excluded from transport from the blood to the brain. The drugs having molecular weight of less than 400 Dalton are able to cross the BBB via lipid-mediated free diffusion. However, large molecule therapeutics are devoid of these characteristics. As an alternative, these substances may be carried via chimeric peptide drug delivery systems, and assist in transcytosis through BBB with the aid of linker strategies. With their recent developments, several forms of nanoparticles, including poly (ethylene glycol)-poly(ε-caprolactone) copolymers, nanogels, liposomes, nanostructured lipid carriers, poly (D, L-lactide-co-glycolide) nanoparticles, chitosan, and solid lipid nanoparticles, have also been considered for their therapeutic applications. Moreover, the necessity for physiologic optimization of current drug delivery methods and their carriers to deliver therapeutic doses of medication into the brain for the treatment of various neurologic illnesses has also been emphasized. Therapeutic use of proteins and peptides has no neuroprotective impact in the absence of all these methods. Each tactic, however, has unique drawbacks and considerations. In this review, we discuss different drug delivery methods for therapeutic distribution of pharmaceuticals, primarily neuroproteins and neuropeptides, through endothelial capillaries via blood-brain barrier. Finally, we have also discussed the challenges and future perspective of protein and peptide therapeutics delivery to the brain. SIGNIFICANCE STATEMENT: Very few reports on the delivery of therapeutic protein and peptide nanoformulations are available in the literature. Herein, we attempted to discuss these nanoformulations of protein and peptide therapeutics used to treat brain diseases.
Collapse
Affiliation(s)
- Sanchit Arora
- Maa Saraswati College of Pharmacy, Abohar-Sito Road, VPO Kala Tibba, Punjab, India (S.A.); Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India Affiliated to I.K. Gujral Punjab Technical University, formerly Punjab Technical University, Punjab, India (T.B., C.S.); Department of Pharmaceutical Sciences, School of Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Uttarakhand, India (J.K., M.G., C.S.); and Department of Pharmacology, ISF College of Pharmacy, Punjab, India (A.S.)
| | - Tania Bajaj
- Maa Saraswati College of Pharmacy, Abohar-Sito Road, VPO Kala Tibba, Punjab, India (S.A.); Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India Affiliated to I.K. Gujral Punjab Technical University, formerly Punjab Technical University, Punjab, India (T.B., C.S.); Department of Pharmaceutical Sciences, School of Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Uttarakhand, India (J.K., M.G., C.S.); and Department of Pharmacology, ISF College of Pharmacy, Punjab, India (A.S.)
| | - Jayant Kumar
- Maa Saraswati College of Pharmacy, Abohar-Sito Road, VPO Kala Tibba, Punjab, India (S.A.); Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India Affiliated to I.K. Gujral Punjab Technical University, formerly Punjab Technical University, Punjab, India (T.B., C.S.); Department of Pharmaceutical Sciences, School of Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Uttarakhand, India (J.K., M.G., C.S.); and Department of Pharmacology, ISF College of Pharmacy, Punjab, India (A.S.)
| | - Manoj Goyal
- Maa Saraswati College of Pharmacy, Abohar-Sito Road, VPO Kala Tibba, Punjab, India (S.A.); Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India Affiliated to I.K. Gujral Punjab Technical University, formerly Punjab Technical University, Punjab, India (T.B., C.S.); Department of Pharmaceutical Sciences, School of Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Uttarakhand, India (J.K., M.G., C.S.); and Department of Pharmacology, ISF College of Pharmacy, Punjab, India (A.S.)
| | - Arti Singh
- Maa Saraswati College of Pharmacy, Abohar-Sito Road, VPO Kala Tibba, Punjab, India (S.A.); Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India Affiliated to I.K. Gujral Punjab Technical University, formerly Punjab Technical University, Punjab, India (T.B., C.S.); Department of Pharmaceutical Sciences, School of Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Uttarakhand, India (J.K., M.G., C.S.); and Department of Pharmacology, ISF College of Pharmacy, Punjab, India (A.S.)
| | - Charan Singh
- Maa Saraswati College of Pharmacy, Abohar-Sito Road, VPO Kala Tibba, Punjab, India (S.A.); Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India Affiliated to I.K. Gujral Punjab Technical University, formerly Punjab Technical University, Punjab, India (T.B., C.S.); Department of Pharmaceutical Sciences, School of Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Uttarakhand, India (J.K., M.G., C.S.); and Department of Pharmacology, ISF College of Pharmacy, Punjab, India (A.S.)
| |
Collapse
|
5
|
Induction of Oxidative Stress in SH-SY5Y Cells by Overexpression of hTau40 and Its Mitigation by Redox-Active Nanoparticles. Int J Mol Sci 2022; 24:ijms24010359. [PMID: 36613801 PMCID: PMC9820486 DOI: 10.3390/ijms24010359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Abnormally phosphorylated tau protein is the principal component of neurofibrillary tangles, accumulating in the brain in many neurodegenerative diseases, including Alzheimer's disease. The aim of this study was to examine whether overexpression of tau protein leads to changes in the redox status of human neuroblastoma SH-SY5Y cells. The level of reactive oxygen species (ROS) was elevated in tau-overexpressing cells (TAU cells) as compared with cells transfected with the empty vector (EP cells). The level of glutathione was increased in TAU cells, apparently due to overproduction as an adaptation to oxidative stress. The TAU cells had elevated mitochondrial mass. They were more sensitive to 6-hydroxydopamine, delphinidin, 4-amino-TEMPO, and nitroxide-containing nanoparticles (NPs) compared to EP controls. These results indicate that overexpression of the tau protein imposes oxidative stress on the cells. The nitroxide 4-amino-TEMPO and nitroxide-containing nanoparticles (NPs) mitigated oxidative stress in TAU cells, decreasing the level of ROS. Nitroxide-containing nanoparticles lowered the level of lipid peroxidation in both TAU and EP cells, suggesting that nitroxides and NPs may mitigate tau-protein-induced oxidative stress.
Collapse
|
6
|
Jagtap S, Potdar C, Yadav R, Pal PK, Datta I. Dopaminergic Neurons Differentiated from LRRK2 I1371V-Induced Pluripotent Stem Cells Display a Lower Yield, α-Synuclein Pathology, and Functional Impairment. ACS Chem Neurosci 2022; 13:2632-2645. [PMID: 36006382 DOI: 10.1021/acschemneuro.2c00297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Being a large multidomain protein, LRRK2 has several confirmed pathological mutant variants for PD, and the incidence of these variants shows ethnicity biases. I1371V, a mutation in the GTPase domain, has been reported in East-Asian populations, but there are no studies reported on dopaminergic (DA) neurons differentiated from this variant. The aim here was to assess the yield, function, and α-synuclein pathology of DA neurons differentiated from LRRK2 I1371V iPSCs. FACS analysis of neural progenitors (NPs) showed a comparable immunopositive population of cells for neural and glial progenitor markers nestin and S100β; however, NPs from I1371V iPSCs showed lower clonogenic and proliferative capacities than healthy control NPs as determined by the neurosphere assay and Ki67 expression. Floor plate cells obtained from I1371V NPs primed with FGF8 showed distinctly lower immunopositivity for FOXA2 and CLIC5 than healthy control FPCs and similar DOC2B expression. On SHH addition, a similar mature neuronal population was obtained from both groups; however, the yield of TH-immunopositive cells was significantly lower in I1371V, with lower expression of mature DA neuronal markers En1, Nurr1, and DAT. Vesicular dopamine release and intracellular Ca2+ response with KCl stimulation were lower in I1371V DA neurons, along with a significantly reduced expression of resting vesicle marker VMAT2. A concurrently lower expression of PSD95/Syn-I immunopositive puncta was observed in I1371V differentiated cells. Further, higher phosphorylation of α-synuclein and aggregation of oligomeric α-synuclein in I1371V DA neurons were observed. Our data demonstrated conclusively for the first time that mutations in the I1371V allele of LRRK2 showed developmental deficit from the FPC stage and generated a lower yield/number of TH-immunopositive neurons with impairment in their function and synapse density along with increased α-synuclein pathology.
Collapse
Affiliation(s)
- Soham Jagtap
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru 560029, Karnataka, India
| | - Chandrakanta Potdar
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru 560029, Karnataka, India
| | - Ravi Yadav
- Department of Neurology, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru 560029, Karnataka, India
| | - Pramod Kumar Pal
- Department of Neurology, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru 560029, Karnataka, India
| | - Indrani Datta
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru 560029, Karnataka, India
| |
Collapse
|
7
|
Integrative Analyses of Transcriptomes to Explore Common Molecular Effects of Antipsychotic Drugs. Int J Mol Sci 2022; 23:ijms23147508. [PMID: 35886854 PMCID: PMC9325239 DOI: 10.3390/ijms23147508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/04/2022] [Accepted: 07/04/2022] [Indexed: 01/27/2023] Open
Abstract
There is little understanding of the underlying molecular mechanism(s) involved in the clinical efficacy of antipsychotics for schizophrenia. This study integrated schizophrenia-associated transcriptional perturbations with antipsychotic-induced gene expression profiles to detect potentially relevant therapeutic targets shared by multiple antipsychotics. Human neuronal-like cells (NT2-N) were treated for 24 h with one of the following antipsychotic drugs: amisulpride, aripiprazole, clozapine, risperidone, or vehicle controls. Drug-induced gene expression patterns were compared to schizophrenia-associated transcriptional data in post-mortem brain tissues. Genes regulated by each of four antipsychotic drugs in the reverse direction to schizophrenia were identified as potential therapeutic-relevant genes. A total of 886 genes were reversely expressed between at least one drug treatment (versus vehicle) and schizophrenia (versus healthy control), in which 218 genes were commonly regulated by all four antipsychotic drugs. The most enriched biological pathways include Wnt signaling and action potential regulation. The protein-protein interaction (PPI) networks found two main clusters having schizophrenia expression quantitative trait loci (eQTL) genes such as PDCD10, ANK2, and AKT3, suggesting further investigation on these genes as potential novel treatment targets.
Collapse
|
8
|
Heinrich L, Zafar F, Morato Torres CA, Singh J, Khan A, Chen MY, Hempel C, Nikulina N, Mulholland J, Braubach O, Schüle B. Multiplex imaging of human induced pluripotent stem cell-derived neurons with CO-Detection by indEXing (CODEX) technology. J Neurosci Methods 2022; 378:109653. [PMID: 35724898 DOI: 10.1016/j.jneumeth.2022.109653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/27/2022] [Accepted: 06/09/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Human induced pluripotent stem cell (iPSC) models have been hailed as a breakthrough for understanding disease and developing new therapeutics. The major advantage of iPSC-derived neurons is that they carry the genetic background of the donor, and as such could be more predictive for clinical translation. However, the development of these cell models is time-consuming and expensive and it is thus critical to maximize readout of markers for immunocytochemistry. One option is to use a highly multiplexed fluorescence imaging assay, like CO-Detection by indEXing (CODEX), which allows detection of 50+ targets in situ. NEW METHOD This paper describes the development of CODEX in neuronal cell cultures derived from human iPSCs. RESULTS We differentiated human iPSCs into mixed neuronal and glial cultures on glass coverslips. We then developed and optimized a panel of 21 antibodies to phenotype iPSC-derived neuronal subtypes of cortical, dopaminergic, and striatal neurons, as well as astrocytes, and pre-and postsynaptic proteins. COMPARISON WITH EXISTING METHODS Compared to standard immunocytochemistry, CODEX oligo-conjugated fluorophores circumvent antibody host interactions and allow for highly customized multiplexing. CONCLUSION We show that CODEX can be applied to iPSC neuronal cultures and developed fixation and staining protocols for the neurons to sustain the multiple wash-stain cycles of the technology. Furthermore, we demonstrate both cellular and subcellular resolution imaging of multiplexed markers in the same sample.
Collapse
Affiliation(s)
- Laurin Heinrich
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Faria Zafar
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | - Anum Khan
- Cell Sciences Imaging Facility (CSIF), Beckman Center, Stanford University, Stanford, CA, USA
| | - Max Yang Chen
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | - Jonathan Mulholland
- Cell Sciences Imaging Facility (CSIF), Beckman Center, Stanford University, Stanford, CA, USA
| | | | - Birgitt Schüle
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
9
|
Xia N, Cabin DE, Fang F, Reijo Pera RA. Parkinson's Disease: Overview of Transcription Factor Regulation, Genetics, and Cellular and Animal Models. Front Neurosci 2022; 16:894620. [PMID: 35600613 PMCID: PMC9115107 DOI: 10.3389/fnins.2022.894620] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/12/2022] [Indexed: 01/21/2023] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders, affecting nearly 7-10 million people worldwide. Over the last decade, there has been considerable progress in our understanding of the genetic basis of PD, in the development of stem cell-based and animal models of PD, and in management of some clinical features. However, there remains little ability to change the trajectory of PD and limited knowledge of the underlying etiology of PD. The role of genetics versus environment and the underlying physiology that determines the trajectory of the disease are still debated. Moreover, even though protein aggregates such as Lewy bodies and Lewy neurites may provide diagnostic value, their physiological role remains to be fully elucidated. Finally, limitations to the model systems for probing the genetics, etiology and biology of Parkinson's disease have historically been a challenge. Here, we review highlights of the genetics of PD, advances in understanding molecular pathways and physiology, especially transcriptional factor (TF) regulators, and the development of model systems to probe etiology and potential therapeutic applications.
Collapse
Affiliation(s)
- Ninuo Xia
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Deborah E. Cabin
- McLaughlin Research Institute for Biomedical Sciences, Inc., Great Falls, MT, United States
| | - Fang Fang
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Renee A. Reijo Pera
- McLaughlin Research Institute for Biomedical Sciences, Inc., Great Falls, MT, United States
| |
Collapse
|
10
|
Guerra-Crespo M, Collazo-Navarrete O, Ramos-Acevedo R, Morato-Torres CA, Schüle B. Embryoid Body Formation from Mouse and Human Pluripotent Stem Cells for Transplantation to Study Brain Microenvironment and Cellular Differentiation. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2520:215-232. [PMID: 34611820 DOI: 10.1007/7651_2021_433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Human embryonic stem cell (hESC) and human-induced pluripotent stem cell (hiPSC) technologies have a critical role in regenerative strategies for personalized medicine. Both share the ability to differentiate into almost any cell type of the human body. The study of their properties and clinical applications requires the development of robust and reproducible cell culture paradigms that direct cell differentiation toward a specific phenotype in vitro and in vivo. Our group evaluated the potential of mouse ESCs (mESCs), hESCs, and hiPSCs (collectively named pluripotent stem cells, PSCs) to analyze brain microenvironments through the use of embryoid body (EB)-derived cells from these cell sources. EB are cell aggregates in 3D culture conditions that recapitulate embryonic development. Our approach focuses on studying the midbrain dopaminergic phenotype and transplanting EB into the substantia nigra pars compacta (SNpc) in a Parkinson's disease rodent model. Here, we describe cell culture protocols for EB generation from PSCs that show significant in vivo differentiation toward dopaminergic neurons.
Collapse
Affiliation(s)
- Magdalena Guerra-Crespo
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, Mexico City, Mexico.
- Laboratorio de Medicina Regenerativa, Departamento de Cirugía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| | - Omar Collazo-Navarrete
- Laboratorio Nacional de Recursos Genómicos, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rodrigo Ramos-Acevedo
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Medicina Regenerativa, Departamento de Cirugía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Carmen Alejandra Morato-Torres
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Birgitt Schüle
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
11
|
Calamini B, Geyer N, Huss-Braun N, Bernhardt A, Harsany V, Rival P, Cindhuchao M, Hoffmann D, Gratzer S. Development of a physiologically relevant and easily scalable LUHMES cell-based model of G2019S LRRK2-driven Parkinson's disease. Dis Model Mech 2021; 14:dmm048017. [PMID: 34114604 PMCID: PMC8214734 DOI: 10.1242/dmm.048017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 04/27/2021] [Indexed: 11/20/2022] Open
Abstract
Parkinson's disease (PD) is a fatal neurodegenerative disorder that is primarily caused by the degeneration and loss of dopaminergic neurons of the substantia nigra in the ventral midbrain. Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of late-onset PD identified to date, with G2019S being the most frequent LRRK2 mutation, which is responsible for up to 1-2% of sporadic PD and up to 6% of familial PD cases. As no treatment is available for this devastating disease, developing new therapeutic strategies is of foremost importance. Cellular models are commonly used for testing novel potential neuroprotective compounds. However, current cellular PD models either lack physiological relevance to dopaminergic neurons or are too complex and costly for scaling up the production process and for screening purposes. In order to combine biological relevance and throughput, we have developed a PD model in Lund human mesencephalic (LUHMES) cell-derived dopaminergic neurons by overexpressing wild-type (WT) and G2019S LRRK2 proteins. We show that these cells can differentiate into dopaminergic-like neurons and that expression of mutant LRRK2 causes a range of different phenotypes, including reduced nuclear eccentricity, altered mitochondrial and lysosomal morphologies, and increased dopaminergic cell death. This model could be used to elucidate G2019S LRRK2-mediated dopaminergic neural dysfunction and to identify novel molecular targets for disease intervention. In addition, our model could be applied to high-throughput and phenotypic screenings for the identification of novel PD therapeutics.
Collapse
Affiliation(s)
- Barbara Calamini
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| | - Nathalie Geyer
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| | - Nathalie Huss-Braun
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| | - Annie Bernhardt
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| | - Véronique Harsany
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| | - Pierrick Rival
- BioTherapeutics/e-Biology - Bioinformatics, Sanofi Biologics Research, 13 quai Jules Guesde, 94400 Vitry-sur-Seine, France
| | - May Cindhuchao
- Molecular Screening Technology, Sanofi Biologics Research, 270 Albany Street, Cambridge, MA 02139, USA
| | - Dietmar Hoffmann
- Molecular Screening Technology, Sanofi Biologics Research, 270 Albany Street, Cambridge, MA 02139, USA
| | - Sabine Gratzer
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| |
Collapse
|
12
|
Saini A, Singh J, Kumar S. Optically superior fluorescent probes for selective imaging of cells, tumors, and reactive chemical species. Org Biomol Chem 2021; 19:5208-5236. [PMID: 34037048 DOI: 10.1039/d1ob00509j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Fluorescent chemical probes have become powerful tools to study biological events in living cells. They provide a great opportunity to quantitatively and qualitatively analyze the physiological and biochemical properties of living cells in real time. The ability of researchers to manipulate these probes for a desired specific purpose has turned many heads in the scientific community. Despite a slow start, fluorescent probe research has seen exponential growth over the last decade in the world. This change required some adventurous and creative scientists from different fields-like biology, medicine, and chemistry-to come together to facilitate the constant expansion of this field. This review article introduces some fundamental concepts related to fluorescent probe designing and development. It also summarizes various fluorescent probes with superior optical properties used in fields like cell biology, cellular imaging, medical research, and cancer diagnosis. It is hoped that this article will encourage more young and creative scientists to contribute their talents to this field.
Collapse
Affiliation(s)
- Abhishek Saini
- Department of Chemistry, Hansraj College, University of Delhi, Delhi-110007, India.
| | - Jyoti Singh
- Department of Chemistry, Hansraj College, University of Delhi, Delhi-110007, India.
| | - Sonu Kumar
- Department of Chemistry, Hansraj College, University of Delhi, Delhi-110007, India.
| |
Collapse
|
13
|
Struzyna LA, Watt ML. The Emerging Role of Neuronal Organoid Models in Drug Discovery: Potential Applications and Hurdles to Implementation. Mol Pharmacol 2021; 99:256-265. [PMID: 33547249 DOI: 10.1124/molpharm.120.000142] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/15/2021] [Indexed: 12/20/2022] Open
Abstract
The high failure rate of drugs in the clinical pipeline is likely in part the result of inadequate preclinical models, particularly those for neurologic disorders and neurodegenerative disease. Such preclinical animal models often suffer from fundamental species differences and rarely recapitulate all facets of neurologic conditions, whereas conventional two-dimensional (2D) in vitro models fail to capture the three-dimensional spatial organization and cell-to-cell interactions of brain tissue that are presumed to be critical to the function of the central nervous system. Recent studies have suggested that stem cell-derived neuronal organoids are more physiologically relevant than 2D neuronal cultures because of their cytoarchitecture, electrophysiological properties, human origin, and gene expression. Hence there is interest in incorporating such physiologically relevant models into compound screening and lead optimization efforts within drug discovery. However, despite their perceived relevance, compared with previously used preclinical models, little is known regarding their predictive value. In fact, some have been wary to broadly adopt organoid technology for drug discovery because of the low-throughput and tedious generation protocols, inherent variability, and lack of compatible moderate-to-high-throughput screening assays. Consequently, microfluidic platforms, specialized bioreactors, and automated assays have been and are being developed to address these deficits. This mini review provides an overview of the gaps to broader implementation of neuronal organoids in a drug discovery setting as well as emerging technologies that may better enable their utilization. SIGNIFICANCE STATEMENT: Neuronal organoid models offer the potential for a more physiological system in which to study neurological diseases, and efforts are being made to employ them not only in mechanistic studies but also in profiling/screening purposes within drug discovery. In addition to exploring the utility of neuronal organoid models within this context, efforts in the field aim to standardize such models for consistency and adaptation to screening platforms for throughput evaluation. This review covers potential impact of and hurdles to implementation.
Collapse
|
14
|
Pingale T, Gupta GL. Classic and evolving animal models in Parkinson's disease. Pharmacol Biochem Behav 2020; 199:173060. [PMID: 33091373 DOI: 10.1016/j.pbb.2020.173060] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with motor and non-motor symptoms. PD is characterized by the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and deficiency of dopamine in the striatal region. The primary objective in PD research is to understand the pathogenesis, targets, and development of therapeutic interventions to control the progress of the disease. The anatomical and physiological resemblances between humans and animals gathered the researcher's attention towards the use of animals in PD research. Due to varying age of onset, symptoms, and progression rate, PD becomes heterogeneous which demands the variety of animal models to study diverse features of the disease. Parkinson is a multifactorial disorder, selection of models become important as not a single model shows all the biochemical features of the disease. Currently, conventional pharmacological, neurotoxin-induced, genetically modified and cellular models are available for PD research, but none of them recapitulate all the biochemical characteristics of the disease. In this review, we included the updated knowledge on the main features of currently available in vivo and in vitro models as well as their strengths and weaknesses.
Collapse
Affiliation(s)
- Tanvi Pingale
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400 056, India
| | - Girdhari Lal Gupta
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400 056, India; School of Pharmacy & Technology Management, SVKM'S NMIMS, Shirpur, Maharashtra, India.
| |
Collapse
|
15
|
Ren C, Wang F, Guan LN, Cheng XY, Zhang CY, Geng DQ, Liu CF. A compendious summary of Parkinson's disease patient-derived iPSCs in the first decade. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:685. [PMID: 31930086 PMCID: PMC6944564 DOI: 10.21037/atm.2019.11.16] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/10/2019] [Indexed: 12/23/2022]
Abstract
The number of Parkinson's disease (PD) patients increases with aging, which brings heavy burden to families and society. The emergence of patient-derived induced pluripotent stem cells (iPSCs) has brought hope to the current situation of lacking new breakthroughs in diagnosis and treatment of PD. In this article, we reviewed and analyzed the current researches related to PD patient-derived iPSCs, in order to provide solid theoretical basis for future study of PD. In 2008, successful iPSCs derived from PD patients were reported. The current iPSCs research in PD mostly focused on the establishment of specific iPSCs models of PD patients carrying susceptible genes. The main source of PD patient-derived iPSCs is skin fibroblasts and the mainstream reprogramming methodology is the mature "four-factor" method, which introduces four totipotent correlation factors Oct4, Sox2, Klf4 and c-Myc into somatic cells. The main sources of iPSCs are patients with non-pedigrees and there have been no studies involving both PD patients and unaffected carriers within the same family. Most of the existing studies of PD patient-derived iPSCs started with the induction method for obtaining dopaminergic neurons in the first instance, but therapeutic applications are being increased. Although it is not the ultimate panacea, and there are still some unsolved problems (e.g., whether the mutated genes should be corrected or not), a better understanding of iPSCs may be a good gift for both PD patients and doctors due to their advantages in diagnosis and treatment of PD.
Collapse
Affiliation(s)
- Chao Ren
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Fen Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Li-Na Guan
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
- Department of Neurosurgical Intensive Care Unit, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Xiao-Yu Cheng
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Cai-Yi Zhang
- Department of Emergency, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, China
| | - De-Qin Geng
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, China
| | - Chun-Feng Liu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| |
Collapse
|
16
|
Delenclos M, Burgess JD, Lamprokostopoulou A, Outeiro TF, Vekrellis K, McLean PJ. Cellular models of alpha-synuclein toxicity and aggregation. J Neurochem 2019; 150:566-576. [PMID: 31265132 DOI: 10.1111/jnc.14806] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 06/19/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022]
Abstract
Misfolding and aggregation of alpha-synuclein (α-synuclein) with concomitant cytotoxicity is a hallmark of Lewy body related disorders such as Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. Although it plays a pivotal role in pathogenesis and disease progression, the function of α-synuclein and the molecular mechanisms underlying α-synuclein-induced neurotoxicity in these diseases are still elusive. Many in vitro and in vivo experimental models mimicking α-synuclein pathology such as oligomerization, toxicity and more recently neuronal propagation have been generated over the years. In particular, cellular models have been crucial for our comprehension of the pathogenic process of the disease and are beneficial for screening of molecules capable of modulating α-synuclein toxicity. Here, we review α-synuclein based cell culture models that reproduce some features of the neuronal populations affected in patients, from basic unicellular organisms to mammalian cell lines and primary neurons, to the cutting edge models of patient-specific cell lines. These reprogrammed cells known as induced pluripotent stem cells (iPSCs) have garnered attention because they closely reproduce the characteristics of neurons found in patients and provide a valuable tool for mechanistic studies. We also discuss how different cell models may constitute powerful tools for high-throughput screening of molecules capable of modulating α-synuclein toxicity and prevention of its propagation. This article is part of the Special Issue "Synuclein".
Collapse
Affiliation(s)
- Marion Delenclos
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Jeremy D Burgess
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA.,Neuroscience PhD Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, Florida, USA
| | - Agaristi Lamprokostopoulou
- Department of Neuroscience, Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Max Planck Institute for Experimental Medicine, Göttingen, Germany.,Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Kostas Vekrellis
- Department of Neuroscience, Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Pamela J McLean
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA.,Neuroscience PhD Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, Florida, USA
| |
Collapse
|
17
|
Collins W, Rouleau N, Bonzanni M, Kapner K, Jeremiah A, Du C, Pothos EN, Kaplan DL. Functional Effects of a Neuromelanin Analogue on Dopaminergic Neurons in 3D Cell Culture. ACS Biomater Sci Eng 2018; 5:308-317. [PMID: 33405867 DOI: 10.1021/acsbiomaterials.8b00976] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The substantia nigra pars compacta (SNpc) is a discrete region of the brain that exhibits a dark pigment, neuromelanin (NM), a biomaterial with unique properties and the subject of ongoing research pertaining to neurodegenerative conditions like Parkinson's disease (PD). Obtaining human tissue to isolate this pigment is costly and labor intensive, making it necessary to find alternatives to model the biochemical interaction of NM and its implications on PD. To address this limitation, we modified our established silk 3D brain tissue model to emulate key characteristics of the SNpc by using a structural analogue of NM to examine the effects of the material on dopaminergic neurons using Lund's human mesencephalon (LUHMES) cells. We utilized a sepia-melanin, squid ink, derived NM analogue (NM-sim) to chelate ferric iron, and this iron-neuromelanin precipitate (Fe-NM) was purified and characterized. We then exposed LUHMES dopaminergic cells to the NM-sim, Fe-NM-sim, and control vehicle within 3D silk protein scaffolds. The presence of both NM-sim and Fe-NM-sim in the scaffolds negatively impacted spontaneous electrical activity from the LUMES networks, as evidenced by changes in local field potential (LFP) electrophysiological recordings. Furthermore, the Fe-NM-sim precipitate generated peroxides, depleted nutrients/antioxidants, and increased protein oxidation by carbonylation in sustained (>2 weeks) 3D cultures, thereby contributing to cell dysfunction. The results suggest that this 3D tissue engineered brain-like model may provide useful readouts related to PD neuro-toxicology research.
Collapse
Affiliation(s)
- Will Collins
- Department of Pharmacology & Experimental Therapeutics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Tufts University, Boston, Massachusetts 02111, United States.,Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Nicolas Rouleau
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States.,Tufts Allen Discovery Center, Tufts University, 200 College Avenue, Medford, Massachusetts 02155, United States
| | - Mattia Bonzanni
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States.,Tufts Allen Discovery Center, Tufts University, 200 College Avenue, Medford, Massachusetts 02155, United States
| | - Kevin Kapner
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Alex Jeremiah
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Chuang Du
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Emmanuel N Pothos
- Department of Pharmacology & Experimental Therapeutics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Tufts University, Boston, Massachusetts 02111, United States
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States.,Tufts Allen Discovery Center, Tufts University, 200 College Avenue, Medford, Massachusetts 02155, United States
| |
Collapse
|
18
|
Marshall LJ, Willett C. Parkinson's disease research: adopting a more human perspective to accelerate advances. Drug Discov Today 2018; 23:1950-1961. [PMID: 30240875 DOI: 10.1016/j.drudis.2018.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/20/2018] [Accepted: 09/12/2018] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) affects 1% of the population over 60 years old and, with global increases in the aging population, presents huge economic and societal burdens. The etiology of PD remains unknown; most cases are idiopathic, presumed to result from genetic and environmental risk factors. Despite 200 years since the first description of PD, the mechanisms behind initiation and progression of the characteristic neurodegenerative processes are not known. Here, we review progress and limitations of the multiple PD animal models available and identify advances that could be implemented to better understand pathological processes, improve disease outcome, and reduce dependence on animal models. Lessons learned from reducing animal use in PD research could serve as guideposts for wider biomedical research.
Collapse
Affiliation(s)
- Lindsay J Marshall
- Humane Society International, The Humane Society of the United States, 700 Professional Drive, Gaithersburg, MD 20879, USA
| | - Catherine Willett
- Humane Society International, The Humane Society of the United States, 700 Professional Drive, Gaithersburg, MD 20879, USA.
| |
Collapse
|
19
|
Misfolded Protein Linked Strategies Toward Biomarker Development for Neurodegenerative Diseases. Mol Neurobiol 2018; 56:2559-2578. [DOI: 10.1007/s12035-018-1232-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/10/2018] [Indexed: 12/14/2022]
|
20
|
Differential expression of miR-34a, miR-141, and miR-9 in MPP+-treated differentiated PC12 cells as a model of Parkinson's disease. Gene 2018; 662:54-65. [DOI: 10.1016/j.gene.2018.04.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 03/14/2018] [Accepted: 04/05/2018] [Indexed: 01/06/2023]
|
21
|
Cobb MM, Ravisankar A, Skibinski G, Finkbeiner S. iPS cells in the study of PD molecular pathogenesis. Cell Tissue Res 2018; 373:61-77. [PMID: 29234887 PMCID: PMC5997490 DOI: 10.1007/s00441-017-2749-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 11/21/2017] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and its pathogenic mechanisms are poorly understood. The majority of PD cases are sporadic but a number of genes are associated with familial PD. Sporadic and familial PD have many molecular and cellular features in common, suggesting some shared pathogenic mechanisms. Induced pluripotent stem cells (iPSCs) have been derived from patients harboring a range of different mutations of PD-associated genes. PD patient-derived iPSCs have been differentiated into relevant cell types, in particular dopaminergic neurons and used as a model to study PD. In this review, we describe how iPSCs have been used to improve our understanding of the pathogenesis of PD. We describe what cellular and molecular phenotypes have been observed in neurons derived from iPSCs harboring known PD-associated mutations and what common pathways may be involved.
Collapse
Affiliation(s)
- Melanie M Cobb
- Gladstone Institutes, the Taube/Koret Center for Neurodegenerative Disease, San Francisco, CA, 94158, USA
| | - Abinaya Ravisankar
- Gladstone Institutes, the Taube/Koret Center for Neurodegenerative Disease, San Francisco, CA, 94158, USA
| | - Gaia Skibinski
- Gladstone Institutes, the Taube/Koret Center for Neurodegenerative Disease, San Francisco, CA, 94158, USA
| | - Steven Finkbeiner
- Gladstone Institutes, the Taube/Koret Center for Neurodegenerative Disease, San Francisco, CA, 94158, USA.
- Department of Neurology, University of California, San Francisco, CA, 94143, USA.
- Department Physiology, University of California, San Francisco, CA, 94143, USA.
- Graduate Programs in Neuroscience and Biomedical Sciences, University of California, San Francisco, CA, 94143, USA.
| |
Collapse
|
22
|
Parkinson's disease genetic risk in a midbrain neuronal cell line. Neurobiol Dis 2018; 114:53-64. [DOI: 10.1016/j.nbd.2018.02.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 01/30/2018] [Accepted: 02/21/2018] [Indexed: 12/16/2022] Open
|
23
|
Piper DA, Sastre D, Schüle B. Advancing Stem Cell Models of Alpha-Synuclein Gene Regulation in Neurodegenerative Disease. Front Neurosci 2018; 12:199. [PMID: 29686602 PMCID: PMC5900030 DOI: 10.3389/fnins.2018.00199] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 03/13/2018] [Indexed: 12/15/2022] Open
Abstract
Alpha-synuclein (non A4 component of amyloid precursor, SNCA, NM_000345.3) plays a central role in the pathogenesis of Parkinson's disease (PD) and related Lewy body disorders such as Parkinson's disease dementia, Lewy body dementia, and multiple system atrophy. Since its discovery as a disease-causing gene in 1997, alpha-synuclein has been a central point of scientific interest both at the protein and gene level. Mutations, including copy number variants, missense mutations, short structural variants, and single nucleotide polymorphisms, can be causative for PD and affect conformational changes of the protein, can contribute to changes in expression of alpha-synuclein and its isoforms, and can influence regulation of temporal as well as spatial levels of alpha-synuclein in different tissues and cell types. A lot of progress has been made to understand both the physiological transcriptional and epigenetic regulation of the alpha-synuclein gene and whether changes in transcriptional regulation could lead to disease and neurodegeneration in PD and related alpha-synucleinopathies. Although the histopathological changes in these neurodegenerative disorders are similar, the temporal and spatial presentation and progression distinguishes them which could be in part due to changes or disruption of transcriptional regulation of alpha-synuclein. In this review, we describe different genetic alterations that contribute to PD and neurodegenerative conditions and review aspects of transcriptional regulation of the alpha-synuclein gene in the context of the development of PD. New technologies, advanced gene engineering and stem cell modeling, are on the horizon to shed further light on a better understanding of gene regulatory processes and exploit them for therapeutic developments.
Collapse
Affiliation(s)
- Desiree A Piper
- Parkinson's Institute and Clinical Center, Sunnyvale, CA, United States
| | - Danuta Sastre
- Parkinson's Institute and Clinical Center, Sunnyvale, CA, United States
| | - Birgitt Schüle
- Parkinson's Institute and Clinical Center, Sunnyvale, CA, United States
| |
Collapse
|
24
|
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide. It is known that there are many factors, either genetic or environmental factors, involved in PD, but the mechanism of PD is still not fully understood. Several animal models have been established to study the mechanisms of PD. Among these models, Drosophila melanogaster has been utilized as a valuable model to get insight into important features of PD. Drosophila melanogaster possesses a well-developed dopaminergic (DA) neuron system which is known to play an important role in PD pathogenesis. The well understanding of DA neurons from early larval through adult stage makes Drosophila as a powerful model for investigating the progressive neurodegeneration in PD. Besides, the short life cycle of Drosophila melanogaster serves an advantage in studying epidemiological features of PD. Most of PD symptoms can be mimicked in Drosophila model such as progressive impairment in locomotion, DA neuron degeneration, and some other non-motor symptoms. The Drosophila models of PD, therefore, show a great potential in application for PD genetic and drug screening.
Collapse
Affiliation(s)
- Vuu My Dung
- University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | | |
Collapse
|
25
|
Lopes FM, Bristot IJ, da Motta LL, Parsons RB, Klamt F. Mimicking Parkinson's Disease in a Dish: Merits and Pitfalls of the Most Commonly used Dopaminergic In Vitro Models. Neuromolecular Med 2017; 19:241-255. [PMID: 28721669 DOI: 10.1007/s12017-017-8454-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 07/12/2017] [Indexed: 12/27/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder and has both unknown etiology and non-curative therapeutic options. Patients begin to present the classic motor symptoms of PD-tremor at rest, bradykinesia and rigidity-once 50-70% of the dopaminergic neurons of the nigrostriatal pathway have degenerated. As a consequence of this, it is difficult to investigate the early-stage events of disease pathogenesis. In vitro experimental models are used extensively in PD research because they present a controlled environment that enables the direct investigation of the early molecular mechanisms that are potentially involved with dopaminergic degeneration, as well as for the screening of potential therapeutic drugs. However, the establishment of PD in vitro models is a controversial issue for neuroscience research not only because it is challenging to mimic, in isolated cell systems, the physiological neuronal environment, but also the pathophysiological conditions experienced by human dopaminergic cells in vivo during the progression of the disease. Since no previous work has attempted to systematically review the literature regarding the establishment of an optimal in vitro model, and/or the features presented by available models used in the PD field, this review aims to summarize the merits and limitations of the most widely used dopaminergic in vitro models in PD research, which may help the PD researcher to choose the most appropriate model for studies directed at the elucidation of the early-stage molecular events underlying PD onset and progression.
Collapse
Affiliation(s)
- Fernanda Martins Lopes
- Laboratório de Bioquímica Celular, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil. .,Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London, SE1 9NH, UK.
| | - Ivi Juliana Bristot
- Laboratório de Bioquímica Celular, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Leonardo Lisbôa da Motta
- Laboratório de Bioquímica Celular, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Richard B Parsons
- Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London, SE1 9NH, UK
| | - Fabio Klamt
- Laboratório de Bioquímica Celular, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
26
|
The 1-Tosylpentan-3-one Protects against 6-Hydroxydopamine-Induced Neurotoxicity. Int J Mol Sci 2017; 18:ijms18051096. [PMID: 28534853 PMCID: PMC5455005 DOI: 10.3390/ijms18051096] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/12/2017] [Accepted: 05/13/2017] [Indexed: 02/06/2023] Open
Abstract
Previous studies have demonstrated that the marine compound austrasulfone, isolated from the soft coral Cladiella australis, exerts a neuroprotective effect. The intermediate product in the synthesis of austrasulfone, dihydroaustrasulfone alcohol, attenuates several inflammatory responses. The present study uses in vitro and in vivo methods to investigate the neuroprotective effect of dihydroaustrasulfone alcohol-modified 1-tosylpentan-3-one (1T3O). Results from in vitro experiments show that 1T3O effectively inhibits 6-hydroxydopamine-induced (6-OHDA-induced) activation of both p38 mitogen-activated protein kinase (MAPK) and caspase-3 in SH-SY5Y cells; and enhances nuclear factor erythroid 2–related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expression via phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling. Hoechst staining and Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining results reveal that 1T3O significantly inhibits 6-OHDA-induced apoptosis. In addition, the addition of an Akt or HO-1 inhibitor decreases the protective effect of 1T3O. Thus, we hypothesize that the anti-apoptotic activity of 1T3O in neuronal cells is mediated through the regulation of the Akt and HO-1 signaling pathways. In vivo experiments show that 1T3O can reverse 6-OHDA-induced reduction in locomotor behavior ability in zebrafish larvae, and inhibit 6-OHDA-induced tumor necrosis factor-alpha (TNF-α) increase at the same time. According to our in vitro and in vivo results, we consider that 1T3O exerts its anti-apoptotic activities at SH-SY5Y cells after 6-OHDA challenges, probably via the regulation of anti-oxidative signaling pathways. Therefore, this compound may be a promising therapeutic agent for neurodegenerations.
Collapse
|
27
|
Lázaro DF, Pavlou MAS, Outeiro TF. Cellular models as tools for the study of the role of alpha-synuclein in Parkinson's disease. Exp Neurol 2017; 298:162-171. [PMID: 28526239 DOI: 10.1016/j.expneurol.2017.05.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 04/01/2017] [Accepted: 05/13/2017] [Indexed: 01/07/2023]
Abstract
Neurodegenerative diseases are highly debilitating conditions characterised primarily by progressive neuronal loss and impairment of the nervous system. Parkinson's disease (PD) is one of the most common of these disorders, affecting 1-2% of the population above the age of 65. Although the underlying mechanisms of PD have been extensively studied, we still lack a full understanding of the molecular underpinnings of the disease. Thus, the in vitro and in vivo models currently used are able to only partially recapitulate the typical phenotypes of the disease. Here, we review various cell culture models currently used to study the molecular basis of PD, with a focus on alpha-synuclein-associated molecular pathologies. We also discuss how different cell models may constitute powerful tools for high-throughput screening of molecules capable of modulating alpha-synuclein toxicity.
Collapse
Affiliation(s)
- Diana F Lázaro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Maria Angeliki S Pavlou
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37073 Göttingen, Germany; Max Planck Institute for Experimental Medicine, Goettingen, Germany.
| |
Collapse
|
28
|
Abstract
INTRODUCTION Parkinson's disease (PD) is an insidious disorder affecting more than 1-2% of the population over the age of 65. Understanding the etiology of PD may create opportunities for developing new treatments. Genomic and transcriptomic studies are useful, but do not provide evidence for the actual status of the disease. Conversely, proteomic studies deal with proteins, which are real time players, and can hence provide information on the dynamic nature of the affected cells. The number of publications relating to the proteomics of PD is vast. Therefore, there is a need to evaluate the current proteomics literature and establish the connections between the past and the present to foresee the future. Areas covered: PubMed and Web of Science were used to retrieve the literature associated with PD proteomics. Studies using human samples, model organisms and cell lines were selected and reviewed to highlight their contributions to PD. Expert commentary: The proteomic studies associated with PD achieved only limited success in facilitating disease diagnosis, monitoring and progression. A global system biology approach using new models is needed. Future research should integrate the findings of proteomics with other omics data to facilitate both early diagnosis and the treatment of PD.
Collapse
Affiliation(s)
- Murat Kasap
- a Department of Medical Biology/DEKART Proteomics Laboratory , Kocaeli University Medical School , Kocaeli , Turkey
| | - Gurler Akpinar
- a Department of Medical Biology/DEKART Proteomics Laboratory , Kocaeli University Medical School , Kocaeli , Turkey
| | - Aylin Kanli
- a Department of Medical Biology/DEKART Proteomics Laboratory , Kocaeli University Medical School , Kocaeli , Turkey
| |
Collapse
|
29
|
RA Differentiation Enhances Dopaminergic Features, Changes Redox Parameters, and Increases Dopamine Transporter Dependency in 6-Hydroxydopamine-Induced Neurotoxicity in SH-SY5Y Cells. Neurotox Res 2017; 31:545-559. [DOI: 10.1007/s12640-016-9699-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 12/28/2016] [Accepted: 12/30/2016] [Indexed: 12/19/2022]
|
30
|
Crescenzo AD, Cacciatore I, Petrini M, D'Alessandro M, Petragnani N, Boccio PD, Profio PD, Boncompagni S, Spoto G, Turkez H, Ballerini P, Stefano AD, Fontana A. Gold nanoparticles as scaffolds for poor water soluble and difficult to vehiculate antiparkinson codrugs. NANOTECHNOLOGY 2017; 28:025102. [PMID: 27922827 DOI: 10.1088/1361-6528/28/2/025102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
We report the facile and non-covalent preparation of gold nanoparticles (AuNPs) stabilized by an antiparkinson codrug based on lipoic acid (LA). The obtained AuNPs appear stable in both dimethyl sulfoxide and fetal bovine serum and able to load an amount of codrug double the weight of gold. These NPs were demonstrated to be safe and biocompatible towards primary human blood cells and human neuroblastoma cells, one of the most widely used cellular models to study dopaminergic neural cells, therefore are ideal drug carriers for difficult to solubilize molecules. Very interestingly, the codrug-stabilized AuNPs were shown to reduce the accumulation of reactive oxygen species in SH-SY5Y cells treated with LD and did not change total oxidant status levels in cultured human blood cells, thus confirming the antioxidant role of LA although bound to AuNPs. The characterization of AuNPs in terms of loading and stability paves the way for their use in biomedical and pharmacological applications.
Collapse
Affiliation(s)
- A Di Crescenzo
- Department of Pharmacy, University 'G. d'Annunzio', Chieti I-66100, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Silva AH, Fonseca FN, Pimenta ATA, Lima MS, Silveira ER, Viana GSB, Vasconcelos SMM, Leal LKAM. Pharmacognostical Analysis and Protective Effect of Standardized Extract and Rizonic Acid from Erythrina velutina against 6-Hydroxydopamine-Induced Neurotoxicity in SH-SY5Y Cells. Pharmacogn Mag 2016; 12:307-312. [PMID: 27867274 PMCID: PMC5096278 DOI: 10.4103/0973-1296.192200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Erythrina velutina is a tree common in the northeast of Brazil extensively used by traditional medicine for the treatment of central nervous system disorders. OBJECTIVE To develop a standardized ethanol extract of E. velutina (EEEV) and to investigate the neuroprotective potential of the extract and rizonic acid (RA) from E. velutina on neuronal cells. MATERIALS AND METHODS The plant drug of E. velutina previously characterized was used for the production of EEEV. Three methods were evaluated in order to obtain an extract with higher content of phenols. The neuroprotective effect of standardized EEEV (HPLC-PDA) and RA was investigated on SH-SY5Y cell exposure to the neurotoxin 6-hydroxydopamine (6-OHDA). RESULTS The powder of the plant drug was classified as moderately coarse and several quality control parameters were determined. EEEV produced by percolation gave the highest phenol content when related to others extractive methods, and its HPLC-PDA analysis allowed to identify four flavonoids and RA, some reported for the first time for the species. EEEV and RA reduced significantly the neurotoxicity induced by 6-OHDA in SH-SY5Y cells determined by the MTT assay and the nitrite concentration. EEEV also showed a free radical scavenging activity. CONCLUSION This is the first pharmacological study about E. velutina which used a controlled standardized extract since the preparation of the herbal drug. This extract and RA, acting as an antioxidant, presents a neuroprotective effect suggesting that they have potential for future development as a therapeutic agent in neurodegenerative disease as Parkinson. SUMMARY The powder of Erythrina velutina was classified as moderately coarse and several quality-control parameters were determined.Ethanolic extract from E. velutina (EEEV) produced by percolation gave the highest phenol content when related to others extractive methods and its HPLC-PDA analysis of EEEV allowed to identify four flavonoids and rizonic acid (RA), some reported for the first time for the species.The EEEV and RA reduced significantly the neurotoxicity induced by 6-OHDA in SH-SY5Y cells determined by the MTT assay and the nitrite concentration.The EEEV also showed a free radical scavenging activity. Abbreviations used: ±: More or less, %: Percentage, °C: Degree Celsius, <: Less than, μg: Microgram, μL: Microliter, μM: Micromol, [1D] MNR: One-dimensional nuclear magnetic resonance spectroscopy, [2D] MNR:Two-dimensional nuclear magnetic resonance spectroscopy, 6-OHDA: [6-] Hydroxydopamine. Abs: Absorbance, CFU: Colony forming units, CH2Cl2: Dichloromethane, CHCl3: Chloroform cmCentimeter, DMEM/F12: Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-12. DMSO: Dimethyl sulfoxide, DPPH: 1,1-Diphenyl-2-picrylhydrazyl, EAG: Gallic acid equivalents, EEEV: Ethanolic extract of Erythrina velutina, EtOAc: Ethyl acetate, g: Gram, h: Hour, H2O: Water, HPLC: High-performance liquid chromatography, H REIMS: Hydrogen rapid evaporative ionization mass spectrometry, Kg: Kilogram M: Molar, m: Metro, MeOH: Methanol, mg: Milligram, min: Minute, mL: Milliliter, mm: Millimeter, MTT: Bromide 3 [4,5-dimethylthiazol-2-yl] -2,5-diphenyltetrazolium, N: Normal, NBT: Nitroblue tetrazolium, nm: Nanometer, PDA: Photodiode array detector, TPC: Total polyphenol content, RA: Rizonic acid, RP: Reverse phase, SOD: Superoxide dismutase, v/v: Volume per volume, Vs: Versus W: Watts.
Collapse
Affiliation(s)
- Aline H Silva
- Centro de Estudos Farmacêuticos e Cosméticos (CEFAC), Departamento de Farmácia, Faculdade de Farmácia, Odontologia e Enfermagem, Universidade Federal do Ceará, Fortaleza, Brasil
| | - Francisco Noé Fonseca
- Centro de Estudos Farmacêuticos e Cosméticos (CEFAC), Departamento de Farmácia, Faculdade de Farmácia, Odontologia e Enfermagem, Universidade Federal do Ceará, Fortaleza, Brasil
| | - Antônia T A Pimenta
- Departamento de Química Orgânica e Inorgânica, Centro de Ciências, Universidade Federal do Ceará, Fortaleza, Brasil
| | - MaryAnne S Lima
- Departamento de Química Orgânica e Inorgânica, Centro de Ciências, Universidade Federal do Ceará, Fortaleza, Brasil
| | - Edilberto Rocha Silveira
- Departamento de Química Orgânica e Inorgânica, Centro de Ciências, Universidade Federal do Ceará, Fortaleza, Brasil
| | - Glauce S B Viana
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, Brasil
| | - Silvânia M M Vasconcelos
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, Brasil
| | - Luzia Kalyne A M Leal
- Centro de Estudos Farmacêuticos e Cosméticos (CEFAC), Departamento de Farmácia, Faculdade de Farmácia, Odontologia e Enfermagem, Universidade Federal do Ceará, Fortaleza, Brasil
| |
Collapse
|
32
|
Functional metabolic interactions of human neuron-astrocyte 3D in vitro networks. Sci Rep 2016; 6:33285. [PMID: 27619889 PMCID: PMC5020407 DOI: 10.1038/srep33285] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 08/19/2016] [Indexed: 01/27/2023] Open
Abstract
The generation of human neural tissue-like 3D structures holds great promise for disease modeling, drug discovery and regenerative medicine strategies. Promoting the establishment of complex cell-cell interactions, 3D culture systems enable the development of human cell-based models with increased physiological relevance, over monolayer cultures. Here, we demonstrate the establishment of neuronal and astrocytic metabolic signatures and shuttles in a human 3D neural cell model, namely the glutamine-glutamate-GABA shuttle. This was indicated by labeling of neuronal GABA following incubation with the glia-specific substrate [2-(13)C]acetate, which decreased by methionine sulfoximine-induced inhibition of the glial enzyme glutamine synthetase. Cell metabolic specialization was further demonstrated by higher pyruvate carboxylase-derived labeling in glutamine than in glutamate, indicating its activity in astrocytes and not in neurons. Exposure to the neurotoxin acrylamide resulted in intracellular accumulation of glutamate and decreased GABA synthesis. These results suggest an acrylamide-induced impairment of neuronal synaptic vesicle trafficking and imbalanced glutamine-glutamate-GABA cycle, due to loss of cell-cell contacts at synaptic sites. This work demonstrates, for the first time to our knowledge, that neural differentiation of human cells in a 3D setting recapitulates neuronal-astrocytic metabolic interactions, highlighting the relevance of these models for toxicology and better understanding the crosstalk between human neural cells.
Collapse
|
33
|
Amiri E, Ghasemi R, Moosavi M. Agmatine Protects Against 6-OHDA-Induced Apoptosis, and ERK and Akt/GSK Disruption in SH-SY5Y Cells. Cell Mol Neurobiol 2016; 36:829-838. [PMID: 26346882 PMCID: PMC11482516 DOI: 10.1007/s10571-015-0266-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/29/2015] [Indexed: 02/08/2023]
Abstract
6-Hydroxydopamine (6-OHDA), a metabolite of dopamine is known to induce dopaminergic cell toxicity which makes that a suitable agent inducing an experimental model of Parkinson's disease (PD). Agmatine has been shown to protect against some cellular and animal PD models. This study was aimed to assess whether agmatine prevents 6-OHDA-induced SH-SY5Y cell death and if yes, then how it affects Akt/glycogen synthesis kinase-3β (GSK-3β) and extracellular signal-regulated kinases (ERK) signals. The cells were treated with different drugs, and their viability was examined via MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide) assay and morphological observation. Western blot studies were done to assess cleaved caspase-3, Akt/GSK-3β, and ERK proteins. 6-OHDA-induced cell death and caspase-3 cleavage, while agmatine prevented those changes. 6-OHDA also decreased the amount of phosphorylated Akt (pAkt)/Akt while increased GSK-3β activity which was prevented by agmatine. Additionally, this toxin increased pERK/ERK ratio which was averted again by agmatine. The PI3/Akt inhibitor, LY294002, impeded the changes induced by agmatine, while ERK inhibitor (PD98059) did not disturb the effects of agmatine, and by itself, it preserved the cells against 6-OHDA toxicity. This study revealed that agmatine is protective in 6-OHDA model of PD and affects Akt/GSK-3β and ERK pathways.
Collapse
Affiliation(s)
- Esmat Amiri
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Rasoul Ghasemi
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Moosavi
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Nanotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
34
|
Multisystem Lewy body disease and the other parkinsonian disorders. Nat Genet 2016; 47:1378-84. [PMID: 26620112 DOI: 10.1038/ng.3454] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 10/28/2015] [Indexed: 12/12/2022]
Abstract
Here we prioritize as multisystem Lewy body disease (MLBD) those genetic forms of Parkinson's disease that point the way toward a mechanistic understanding of the majority of sporadic disease. Pathological diagnosis of genetic subtypes offers the prospect of distinguishing different mechanistic trajectories with a common mutational etiology, differing outcomes from varying allelic bases, and those disease-associated variants that can be used in gene-environment analysis. Clearly delineating parkinsonian disorders into subclasses on the basis of molecular mechanisms with well-characterized outcome expectations is the basis for refining these forms of neurodegeneration as research substrate through the use of cell models derived from affected individuals while ensuring that clinically collected data can be used for therapeutic decisions and research without increasing the noise and confusion engendered by the collection of data against a range of historically defined criteria.
Collapse
|
35
|
Rotenone Susceptibility Phenotype in Olfactory Derived Patient Cells as a Model of Idiopathic Parkinson's Disease. PLoS One 2016; 11:e0154544. [PMID: 27123847 PMCID: PMC4849794 DOI: 10.1371/journal.pone.0154544] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/14/2016] [Indexed: 12/21/2022] Open
Abstract
Parkinson’s disease is a complex age-related neurodegenerative disorder. Approximately 90% of Parkinson’s disease cases are idiopathic, of unknown origin. The aetiology of Parkinson’s disease is not fully understood but increasing evidence implies a failure in fundamental cellular processes including mitochondrial dysfunction and increased oxidative stress. To dissect the cellular events underlying idiopathic Parkinson’s disease, we use primary cell lines established from the olfactory mucosa of Parkinson’s disease patients. Previous metabolic and transcriptomic analyses identified deficiencies in stress response pathways in patient-derived cell lines. The aim of this study was to investigate whether these deficiencies manifested as increased susceptibility, as measured by cell viability, to a range of extrinsic stressors. We identified that patient-derived cells are more sensitive to mitochondrial complex I inhibition and hydrogen peroxide induced oxidative stress, than controls. Exposure to low levels (50 nM) of rotenone led to increased apoptosis in patient-derived cells. We identified an endogenous deficit in mitochondrial complex I in patient-derived cells, but this did not directly correlate with rotenone-sensitivity. We further characterized the sensitivity to rotenone and identified that it was partly associated with heat shock protein 27 levels. Finally, transcriptomic analysis following rotenone exposure revealed that patient-derived cells express a diminished response to rotenone-induced stress compared with cells from healthy controls. Our cellular model of idiopathic Parkinson’s disease displays a clear susceptibility phenotype to mitochondrial stress. The determination of molecular mechanisms underpinning this susceptibility may lead to the identification of biomarkers for either disease onset or progression.
Collapse
|
36
|
Simão D, Arez F, Terasso AP, Pinto C, Sousa MFQ, Brito C, Alves PM. Perfusion Stirred-Tank Bioreactors for 3D Differentiation of Human Neural Stem Cells. Methods Mol Biol 2016; 1502:129-142. [PMID: 27032948 DOI: 10.1007/7651_2016_333] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Therapeutic breakthroughs in neurological disorders have been hampered by the lack of accurate central nervous system (CNS) models. The development of these models allows the study of the disease onset/progression mechanisms and the preclinical evaluation of new therapeutics. This has traditionally relied on genetically engineered animal models that often diverge considerably from the human phenotype (developmental, anatomic, and physiological) and 2D in vitro cell models, which fail to recapitulate the characteristics of the target tissue (cell-cell and cell-matrix interactions, cell polarity, etc.). Recapitulation of CNS phenotypic and functional features in vitro requires the implementation of advanced culture strategies, such as 3D culture systems, which enable to mimic the in vivo structural and molecular complexity. Models based on differentiation of human neural stem cells (hNSC) in 3D cultures have great potential as complementary tools in preclinical research, bridging the gap between human clinical studies and animal models. The development of robust and scalable processes for the 3D differentiation of hNSC can improve the accuracy of early stage development in preclinical research. In this context, the use of software-controlled stirred-tank bioreactors (STB) provides an efficient technological platform for hNSC aggregation and differentiation. This system enables to monitor and control important physicochemical parameters for hNSC culture, such as dissolved oxygen. Importantly, the adoption of a perfusion operation mode allows a stable flow of nutrients and differentiation/neurotrophic factors, while clearing the toxic by-products. This contributes to a setting closer to the physiological, by mimicking the in vivo microenvironment. In this chapter, we address the technical requirements and procedures for the implementation of 3D differentiation strategies of hNSC, by operating STB under perfusion mode for long-term cultures. This strategy is suitable for the generation of human 3D neural in vitro models, which can be used to feed high-throughput screening platforms, contributing to expand the available in vitro tools for drug screening and toxicological studies.
Collapse
Affiliation(s)
- Daniel Simão
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Francisca Arez
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ana P Terasso
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Catarina Pinto
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Marcos F Q Sousa
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.
| |
Collapse
|
37
|
Pharmacological modulation of HDAC1 and HDAC6 in vivo in a zebrafish model: Therapeutic implications for Parkinson’s disease. Pharmacol Res 2016; 103:328-39. [DOI: 10.1016/j.phrs.2015.11.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/27/2015] [Accepted: 11/29/2015] [Indexed: 11/22/2022]
|
38
|
Simão D, Pinto C, Fernandes P, Peddie CJ, Piersanti S, Collinson LM, Salinas S, Saggio I, Schiavo G, Kremer EJ, Brito C, Alves PM. Evaluation of helper-dependent canine adenovirus vectors in a 3D human CNS model. Gene Ther 2015; 23:86-94. [PMID: 26181626 DOI: 10.1038/gt.2015.75] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/23/2015] [Accepted: 07/08/2015] [Indexed: 01/24/2023]
Abstract
Gene therapy is a promising approach with enormous potential for treatment of neurodegenerative disorders. Viral vectors derived from canine adenovirus type 2 (CAV-2) present attractive features for gene delivery strategies in the human brain, by preferentially transducing neurons, are capable of efficient axonal transport to afferent brain structures, have a 30-kb cloning capacity and have low innate and induced immunogenicity in preclinical tests. For clinical translation, in-depth preclinical evaluation of efficacy and safety in a human setting is primordial. Stem cell-derived human neural cells have a great potential as complementary tools by bridging the gap between animal models, which often diverge considerably from human phenotype, and clinical trials. Herein, we explore helper-dependent CAV-2 (hd-CAV-2) efficacy and safety for gene delivery in a human stem cell-derived 3D neural in vitro model. Assessment of hd-CAV-2 vector efficacy was performed at different multiplicities of infection, by evaluating transgene expression and impact on cell viability, ultrastructural cellular organization and neuronal gene expression. Under optimized conditions, hd-CAV-2 transduction led to stable long-term transgene expression with minimal toxicity. hd-CAV-2 preferentially transduced neurons, whereas human adenovirus type 5 (HAdV5) showed increased tropism toward glial cells. This work demonstrates, in a physiologically relevant 3D model, that hd-CAV-2 vectors are efficient tools for gene delivery to human neurons, with stable long-term transgene expression and minimal cytotoxicity.
Collapse
Affiliation(s)
- D Simão
- iBET-Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - C Pinto
- iBET-Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - P Fernandes
- iBET-Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - C J Peddie
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London, UK
| | - S Piersanti
- Dipartimento di Biologia e Biotecnologie 'Charles Darwin', Università di Roma La Sapienza, Rome, Italy
| | - L M Collinson
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London, UK
| | - S Salinas
- Institut de Génétique Moléculaire de Montpellier, Montpellier, France.,Université Montpellier, Montpellier, France
| | - I Saggio
- Dipartimento di Biologia e Biotecnologie 'Charles Darwin', Università di Roma La Sapienza, Rome, Italy.,Istituto Pasteur Fondazione Cenci Bolognetti, Università di Roma La Sapienza, Rome, Italy.,Istituto di Biologia e Patologia Molecolari del CNR, Università di Roma La Sapienza, Rome, Italy
| | - G Schiavo
- Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London, London, UK
| | - E J Kremer
- Institut de Génétique Moléculaire de Montpellier, Montpellier, France.,Université Montpellier, Montpellier, France
| | - C Brito
- iBET-Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - P M Alves
- iBET-Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
39
|
Efremova L, Schildknecht S, Adam M, Pape R, Gutbier S, Hanf B, Bürkle A, Leist M. Prevention of the degeneration of human dopaminergic neurons in an astrocyte co-culture system allowing endogenous drug metabolism. Br J Pharmacol 2015; 172:4119-32. [PMID: 25989025 DOI: 10.1111/bph.13193] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 05/04/2015] [Accepted: 05/12/2015] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND AND PURPOSE Few neuropharmacological model systems use human neurons. Moreover, available test systems rarely reflect functional roles of co-cultured glial cells. There is no human in vitro counterpart of the widely used 1-methyl-4-phenyl-tetrahydropyridine (MPTP) mouse model of Parkinson's disease EXPERIMENTAL APPROACH We generated such a model by growing an intricate network of human dopaminergic neurons on a dense layer of astrocytes. In these co-cultures, MPTP was metabolized to 1-methyl-4-phenyl-pyridinium (MPP(+) ) by the glial cells, and the toxic metabolite was taken up through the dopamine transporter into neurons. Cell viability was measured biochemically and by quantitative neurite imaging, siRNA techniques were also used. KEY RESULTS We initially characterized the activation of PARP. As in mouse models, MPTP exposure induced (poly-ADP-ribose) synthesis and neurodegeneration was blocked by PARP inhibitors. Several different putative neuroprotectants were then compared in mono-cultures and co-cultures. Rho kinase inhibitors worked in both models; CEP1347, ascorbic acid or a caspase inhibitor protected mono-cultures from MPP(+) toxicity, but did not protect co-cultures, when used alone or in combination. Application of GSSG prevented degeneration in co-cultures, but not in mono-cultures. The surprisingly different pharmacological profiles of the models suggest that the presence of glial cells, and the in situ generation of the toxic metabolite MPP(+) within the layered cultures played an important role in neuroprotection. CONCLUSIONS AND IMPLICATIONS Our new model system is a closer model of human brain tissue than conventional cultures. Its use for screening of candidate neuroprotectants may increase the predictiveness of a test battery.
Collapse
Affiliation(s)
- Liudmila Efremova
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany.,Research Training Group 1331 (RTG1331), University of Konstanz, Konstanz, Germany
| | - Stefan Schildknecht
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Martina Adam
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Regina Pape
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Simon Gutbier
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany.,Research Training Group 1331 (RTG1331), University of Konstanz, Konstanz, Germany
| | - Benjamin Hanf
- Research Training Group 1331 (RTG1331), University of Konstanz, Konstanz, Germany.,Molecular Toxicology Group, University of Konstanz, Konstanz, Germany
| | - Alexander Bürkle
- Molecular Toxicology Group, University of Konstanz, Konstanz, Germany
| | - Marcel Leist
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| |
Collapse
|
40
|
Simão D, Pinto C, Piersanti S, Weston A, Peddie CJ, Bastos AE, Licursi V, Schwarz SC, Collinson LM, Salinas S, Serra M, Teixeira AP, Saggio I, Lima PA, Kremer EJ, Schiavo G, Brito C, Alves PM. Modeling Human Neural Functionality In Vitro: Three-Dimensional Culture for Dopaminergic Differentiation. Tissue Eng Part A 2015; 21:654-68. [DOI: 10.1089/ten.tea.2014.0079] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Daniel Simão
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Catarina Pinto
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Stefania Piersanti
- Dipartimento di Biologia e Biotecnologie “Charles Darwin,” Università di Roma La Sapienza, Rome, Italy
| | - Anne Weston
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
| | - Christopher J. Peddie
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
| | - André E.P. Bastos
- NOVA Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisboa, Portugal
- Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade de Lisboa, Lisboa, Portugal
| | - Valerio Licursi
- Dipartimento di Biologia e Biotecnologie “Charles Darwin,” Università di Roma La Sapienza, Rome, Italy
| | | | - Lucy M. Collinson
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
| | - Sara Salinas
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Université Montpellier I and II, Montpellier, France
| | - Margarida Serra
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ana P. Teixeira
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Isabella Saggio
- Dipartimento di Biologia e Biotecnologie “Charles Darwin,” Università di Roma La Sapienza, Rome, Italy
- Istituto Pasteur Fondazione Cenci Bolognetti, Università di Roma La Sapienza, Rome, Italy
- Istituto di Biologia e Patologia Molecolari del CNR, Università di Roma La Sapienza, Rome, Italy
| | - Pedro A. Lima
- NOVA Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisboa, Portugal
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Université Montpellier I and II, Montpellier, France
| | - Giampietro Schiavo
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
- Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London, London, United Kingdom
| | - Catarina Brito
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M. Alves
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
41
|
Zhang XM, Yin M, Zhang MH. Cell-based assays for Parkinson's disease using differentiated human LUHMES cells. Acta Pharmacol Sin 2014; 35:945-56. [PMID: 24989254 DOI: 10.1038/aps.2014.36] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 03/25/2013] [Indexed: 12/19/2022]
Abstract
AIM Lund human mesencephalic (LUHMES) cells can be differentiated to post-mitotic cells with biochemical, morphological and functional features of dopaminergic (DAergic) neurons. Given the limited scale of primary DAergic neuron culture, we developed differentiated LUHMES cell-based cytotoxicity assays for identifying neuroprotective agents for Parkinson's disease (PD). METHODS LUHMES cells were incubated in a differentiation medium containing cAMP and GDNF for 6 d, and then differentiated cells were treated with MPP(+) or infected with baculovirus containing α-synuclein. Cytotoxicity was determined by measuring intracellular ATP levels and caspase 3/7 activity in the cells. DAergic neuron-specific marker protein and mRNA levels in the cells were analyzed using Western blotting and RT-PCR, respectively. RESULTS LUHMES cells grew extensive neurites and became post-mitotic neuron-like cells during differentiation period, and three DAergic neuron markers TH, DAT and Nurr1 exhibited different expression profiles. MPP(+) dose-dependently reduced ATP levels in the cells with an IC50 value of 65 μmol/L. MPP(+) (80 μmol/L) significantly increased caspase 3/7 activity in the cells. Both the CDK inhibitor GW8510 and the GSK3β inhibitor SB216763 effectively rescued MPP(+)-induced reduction of ATP levels with EC50 values of 12 and 205 nmol/L, respectively. Overexpression of α-synuclein also significantly decreased intracellular ATP levels and increased caspase 3/7 activity in the cells. GW8510 and SB216763 effectively rescued α-synuclein overexpression-induced reduction of ATP levels, whereas GW8510, but not SB216763, ameliorated α-synuclein overexpression-induced increase of caspase 3/7 activity. CONCLUSION MPP(+)- and α-synuclein overexpression-induced cytotoxicity of differentiated LUHMES cells may serve as good alternative systems for identifying neuroprotective compounds for PD.
Collapse
|
42
|
Iglesias E, Llobet L, Pacheu-Grau D, Gómez-Durán A, Ruiz-Pesini E. Cybrids for Mitochondrial DNA Pharmacogenomics. Drug Dev Res 2012. [DOI: 10.1002/ddr.21037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
43
|
Abstract
Our understanding of the underlying molecular mechanism of Parkinson’s disease (PD) is hampered by a lack of access to affected human dopaminergic (DA) neurons on which to base experimental research. Fortunately, the recent development of a PD disease model using induced pluripotent stem cells (iPSCs) provides access to cell types that were previously unobtainable in sufficient quantity or quality, and presents exciting promises for the elucidation of PD etiology and the development of potential therapeutics. To more effectively model PD, we generated two patient-derived iPSC lines: a line carrying a homozygous p.G2019S mutation in the leucine-rich repeat kinase 2 (LRRK2) gene and another carrying a full gene triplication of the α-synuclein encoding gene, SNCA. We demonstrated that these PD-linked pluripotent lines were able to differentiate into DA neurons and that these neurons exhibited increased expression of key oxidative stress response genes and α-synuclein protein. Moreover, when compared to wild-type DA neurons, LRRK2-G2019S iPSC-derived DA neurons were more sensitive to caspase-3 activation caused by exposure to hydrogen peroxide, MG-132, and 6-hydroxydopamine. In addition, SNCA-triplication iPSC-derived DA neurons formed early ubiquitin-positive puncta and were more sensitive to peak toxicity from hydrogen peroxide-induced stress. These aforementioned findings suggest that LRRK2-G2019S and SNCA-triplication iPSC-derived DA neurons exhibit early phenotypes linked to PD. Given the high penetrance of the homozygous LRRK2 mutation, the expression of wild-type α-synuclein protein in the SNCA-triplication line, and the clinical resemblance of patients afflicted with these familial disorders to sporadic PD patients, these iPSC-derived neurons may be unique and valuable models for disease diagnostics and development of novel pharmacological agents for alleviation of relevant disease phenotypes.
Collapse
|
44
|
Huang HP, Chuang CY, Kuo HC. Induced pluripotent stem cell technology for disease modeling and drug screening with emphasis on lysosomal storage diseases. Stem Cell Res Ther 2012; 3:34. [PMID: 22925465 PMCID: PMC3580472 DOI: 10.1186/scrt125] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The recent derivation of disease-specific induced pluripotent stem cells (iPSCs) from somatic cells of patients with familial and sporadic forms of diseases and the demonstration of their ability to give rise to disease-relevant cell types provide an excellent opportunity to gain further insights into the mechanisms responsible for the pathophysiology of these diseases and develop novel therapeutic drugs. Here, we review the recent advances in iPSC technology for modeling of various lysosomal storage diseases (LSDs) and discuss possible strategies through which LSD-iPSCs can be exploited to identify novel drugs and improve future clinical treatment of LSDs.
Collapse
|
45
|
Neural stem cell specific fluorescent chemical probe binding to FABP7. Proc Natl Acad Sci U S A 2012; 109:10214-7. [PMID: 22689954 DOI: 10.1073/pnas.1200817109] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Fluorescent small molecules have become indispensable tools for biomedical research along with the rapidly developing optical imaging technology. We report here a neural stem cell specific boron-dipyrromethane (BODIPY) derivative compound of designation red 3 (CDr3), developed through a high throughput/content screening of in-house generated diversity oriented fluorescence library in stem cells at different developmental stages. This novel compound specifically detects living neural stem cells of both human and mouse origin. Furthermore, we identified its binding target by proteomic analysis as fatty acid binding protein 7 (FABP7), also known as brain lipid binding protein) which is highly expressed in neural stem cells and localized in the cytoplasm. CDr3 will be a valuable chemical tool in the study and applications of neural stem cells.
Collapse
|
46
|
Small molecules greatly improve conversion of human-induced pluripotent stem cells to the neuronal lineage. Stem Cells Int 2012; 2012:140427. [PMID: 22567022 PMCID: PMC3339118 DOI: 10.1155/2012/140427] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Accepted: 01/16/2012] [Indexed: 12/19/2022] Open
Abstract
Efficient in vitro differentiation into specific cell types is more important than ever after the breakthrough in nuclear reprogramming of somatic cells and its potential for disease modeling and drug screening.
Key success factors for neuronal differentiation are the yield of desired neuronal marker expression, reproducibility, length, and cost. Three main neuronal differentiation approaches are stromal-induced neuronal differentiation, embryoid body (EB) differentiation, and direct neuronal differentiation. Here, we describe our neurodifferentiation protocol using small molecules that very efficiently promote neural induction in a 5-stage EB protocol from six induced pluripotent stem cells (iPSC) lines from patients with Parkinson's disease and controls. This protocol generates neural precursors using Dorsomorphin and SB431542 and further maturation into dopaminergic neurons by replacing sonic hedgehog with purmorphamine or smoothened agonist. The advantage of this approach is that all patient-specific iPSC lines tested in this study were successfully and consistently coaxed into the neural lineage.
Collapse
|
47
|
Alberio T, Lopiano L, Fasano M. Cellular models to investigate biochemical pathways in Parkinson’s disease. FEBS J 2012; 279:1146-55. [DOI: 10.1111/j.1742-4658.2012.08516.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
48
|
Proteomics in Parkinson's disease: An unbiased approach towards peripheral biomarkers and new therapies. J Biotechnol 2011; 156:325-37. [DOI: 10.1016/j.jbiotec.2011.08.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Revised: 06/24/2011] [Accepted: 08/08/2011] [Indexed: 12/27/2022]
|
49
|
Nguyen HN, Byers B, Cord B, Shcheglovitov A, Byrne J, Gujar P, Kee K, Schüle B, Dolmetsch RE, Langston W, Palmer TD, Pera RR. LRRK2 mutant iPSC-derived DA neurons demonstrate increased susceptibility to oxidative stress. Cell Stem Cell 2011; 8:267-80. [PMID: 21362567 DOI: 10.1016/j.stem.2011.01.013] [Citation(s) in RCA: 561] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 11/01/2010] [Accepted: 01/18/2011] [Indexed: 02/07/2023]
Abstract
Studies of Parkinson's disease (PD) have been hindered by lack of access to affected human dopaminergic (DA) neurons. Here, we report generation of induced pluripotent stem cells that carry the p.G2019S mutation (G2019S-iPSCs) in the Leucine-Rich Repeat Kinase-2 (LRRK2) gene, the most common PD-related mutation, and their differentiation into DA neurons. The high penetrance of the LRRK2 mutation and its clinical resemblance to sporadic PD suggest that these cells could provide a valuable platform for disease analysis and drug development. We found that DA neurons derived from G2019S-iPSCs showed increased expression of key oxidative stress-response genes and α-synuclein protein. The mutant neurons were also more sensitive to caspase-3 activation and cell death caused by exposure to stress agents, such as hydrogen peroxide, MG-132, and 6-hydroxydopamine, than control DA neurons. This enhanced stress sensitivity is consistent with existing understanding of early PD phenotypes and represents a potential therapeutic target.
Collapse
Affiliation(s)
- Ha Nam Nguyen
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Verhaar R, Jongenelen CA, Gerard M, Baekelandt V, Van Dam AM, Wilhelmus MM, Drukarch B. Blockade of enzyme activity inhibits tissue transglutaminase-mediated transamidation of α-synuclein in a cellular model of Parkinson's disease. Neurochem Int 2011; 58:785-93. [DOI: 10.1016/j.neuint.2011.03.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 03/02/2011] [Accepted: 03/03/2011] [Indexed: 01/17/2023]
|