1
|
Kang B, Song B, Shin H, Lee IS. Downregulation of nuclear receptor-binding SET domain protein 1 induces proinflammatory cytokine expression via mitogen-activated protein kinase pathways in U87MG cells. Biochem Biophys Res Commun 2024; 734:150638. [PMID: 39236589 DOI: 10.1016/j.bbrc.2024.150638] [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: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
Haploinsufficiency of the nuclear receptor binding SET domain-containing protein 1 gene (NSD1) leads to a neurodevelopmental disorder known as Sotos syndrome (SOTOS). This study investigated the effects of NSD1 knockdown in glial cells. U87MG glioma cells were transfected with siRNA targeting NSD1, which resulted in morphological changes characteristic of activated astrocytes. These activated phenotypes were accompanied by specific activation of mitogen-activated protein kinase (MAPK) signaling pathways, particularly those mediated by p38 MAPK and c-Jun N-terminal kinase (JNK). Transcriptome analysis showed increased expression of proinflammatory cytokine genes, particularly interleukin (IL)-1α, IL-1β, and IL-6, following NSD1 knockdown. Treatment with MAPK inhibitors significantly reduced the cytokine induction caused by NSD1 knockdown, with the p38 MAPK inhibitor being more effective than the JNK inhibitor. These findings provide new insights into the role of NSD1 loss in neurological dysfunctions associated with SOTOS.
Collapse
Affiliation(s)
- Byungjun Kang
- Department of Biological Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Bokyeong Song
- Department of Biological Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Hyewon Shin
- Department of Biological Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Im-Soon Lee
- Department of Biological Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
| |
Collapse
|
2
|
Mwema A, Gratpain V, Ucakar B, Vanvarenberg K, Perdaens O, van Pesch V, Muccioli GG, des Rieux A. Impact of calcitriol and PGD 2-G-loaded lipid nanocapsules on oligodendrocyte progenitor cell differentiation and remyelination. Drug Deliv Transl Res 2024; 14:3128-3146. [PMID: 38366115 DOI: 10.1007/s13346-024-01535-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2024] [Indexed: 02/18/2024]
Abstract
Multiple sclerosis (MS) is a demyelinating and inflammatory disease of the central nervous system (CNS) in need of a curative treatment. MS research has recently focused on the development of pro-remyelinating treatments and neuroprotective therapies. Here, we aimed at favoring remyelination and reducing neuro-inflammation in a cuprizone mouse model of brain demyelination using nanomedicines. We have selected lipid nanocapsules (LNC) coated with the cell-penetrating peptide transactivator of translation (TAT), loaded with either a pro-remyelinating compound, calcitriol (Cal-LNC TAT), or an anti-inflammatory bioactive lipid, prostaglandin D2-glycerol ester (PGD2-G) (PGD2-G-LNC TAT). Following the characterization of these formulations, we showed that Cal-LNC TAT in combination with PGD2-G-LNC TAT increased the mRNA expression of oligodendrocyte differentiation markers both in the CG-4 cell line and in primary mixed glial cell (MGC) cultures. However, while the combination of Cal-LNC TAT and PGD2-G-LNC TAT showed promising results in vitro, no significant impact, in terms of remyelination, astrogliosis, and microgliosis, was observed in vivo in the corpus callosum of cuprizone-treated mice following intranasal administration. Thus, although calcitriol's beneficial effects have been abundantly described in the literature in the context of MS, here, we show that the different doses of calcitriol tested had a negative impact on the mice well-being and showed no beneficial effect in the cuprizone model in terms of remyelination and neuro-inflammation, alone and when combined with PGD2-G-LNC TAT.
Collapse
Affiliation(s)
- Ariane Mwema
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium
- Bioanalysis and Pharmacology of Bioactive Lipids, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium
| | - Viridiane Gratpain
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium
| | - Bernard Ucakar
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium
| | - Kevin Vanvarenberg
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium
| | - Océane Perdaens
- Cellular and Molecular Division, Institute of Neuroscience, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 53, 1200, Brussels, Belgium
| | - Vincent van Pesch
- Cellular and Molecular Division, Institute of Neuroscience, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 53, 1200, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium.
| | - Anne des Rieux
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium.
| |
Collapse
|
3
|
Sumam P, Kumar P R A, Parameswaran R. Aligned Fibroporous Matrix Generated from a Silver Ion and Graphene Oxide-Incorporated Ethylene Vinyl Alcohol Copolymer as a Potential Biomaterial for Peripheral Nerve Repair. ACS APPLIED BIO MATERIALS 2024; 7:6617-6630. [PMID: 39295150 DOI: 10.1021/acsabm.4c00841] [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] [Indexed: 09/21/2024]
Abstract
Developing an ideal nerve conduit for proper nerve regeneration still faces several challenges. The attempts to fabricate aligned substrates for neuronal growth have enhanced the hope of successful nerve regeneration. In this wok, we have attempted to generate an electrospun matrix with aligned fibers from a silver and graphene oxide-incorporated ethylene vinyl alcohol copolymer (EVAL). The presence of silver was analyzed using UV-visible spectra, XPS spectra, and ICP. Raman spectra and FTIR spectra confirmed the presence of GO. The complexation of Ag+ with - OH of EVAL enabled the generation of aligned fibers. The fiber diameter (>1 μm) provided sufficient space for forming focal adhesion by the neurites and filopodia of N2a and C6 cells, respectively. The fiber diameter enabled the neurites and filopodia of the cells to align on the fibers. The incorporation of GO has contributed to the cell-material interactions. The morphological and mechanical properties of fibers obtained in the study ensure that the EVAL-Ag-GO-0.01 matrix is a potential substrate for developing a nerve guidance conduit/nerve wrap (NGC/W).
Collapse
Affiliation(s)
- Prima Sumam
- Division of Polymeric Medical Devices, Department of Medical Devices Engineering, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Trivandrum, 695 012 Kerala, India
| | - Anil Kumar P R
- Division of Tissue Culture, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Trivandrum, 695 012 Kerala, India
| | - Ramesh Parameswaran
- Division of Polymeric Medical Devices, Department of Medical Devices Engineering, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Trivandrum, 695 012 Kerala, India
| |
Collapse
|
4
|
Maktabi B, Collins A, Safee R, Bouyer J, Wisner AS, Williams FE, Schiefer IT. Zebrafish as a Model for Multiple Sclerosis. Biomedicines 2024; 12:2354. [PMID: 39457666 PMCID: PMC11504653 DOI: 10.3390/biomedicines12102354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Zebrafish have become a key model organism in neuroscience research because of their unique advantages. Their genetic, anatomical, and physiological similarities to humans, coupled with their rapid development and transparent embryos, make them an excellent tool for investigating various aspects of neurobiology. They have specifically emerged as a valuable and versatile model organism in biomedical research, including the study of neurological disorders such as multiple sclerosis. Multiple sclerosis is a chronic autoimmune disease known to cause damage to the myelin sheath that protects the nerves in the brain and spinal cord. Objective: This review emphasizes the importance of continued research in both in vitro and in vivo models to advance our understanding of MS and develop effective treatments, ultimately improving the quality of life for those affected by this debilitating disease. Conclusions: Recent studies show the significance of zebrafish as a model organism for investigating demyelination and remyelination processes, providing new insights into MS pathology and potential therapies.
Collapse
Affiliation(s)
- Briana Maktabi
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
| | - Abigail Collins
- Center for Drug Design and Development 3, University of Toledo, Toledo, OH 43614, USA
| | - Raihaanah Safee
- Department of Pharmacy Practice, University of Toledo, Toledo, OH 43614, USA
| | - Jada Bouyer
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
| | - Alexander S. Wisner
- Center for Drug Design and Development 3, University of Toledo, Toledo, OH 43614, USA
| | - Frederick E. Williams
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
| | - Isaac T. Schiefer
- Department of Pharmacy Practice, University of Toledo, Toledo, OH 43614, USA
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
5
|
Morshed N, Rennie C, Deng W, Collins-Praino L, Care A. Serum-derived protein coronas affect nanoparticle interactions with brain cells. NANOTECHNOLOGY 2024; 35:495101. [PMID: 39284320 DOI: 10.1088/1361-6528/ad7b40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/16/2024] [Indexed: 09/20/2024]
Abstract
Neuronanomedicine is an emerging field bridging the gap between neuromedicine and novel nanotherapeutics. Despite promise, clinical translation of neuronanomedicine remains elusive, possibly due to a dearth of information regarding the effect of the protein corona on these neuronanomedicines. The protein corona, a layer of proteins adsorbed to nanoparticles following exposure to biological fluids, ultimately determines the fate of nanoparticles in biological systems, dictating nanoparticle-cell interactions. To date, few studies have investigated the effect of the protein corona on interactions with brain-derived cells, an important consideration for the development of neuronanomedicines. Here, two polymeric nanoparticles, poly(lactic-co-glycolic acid) (PLGA) and PLGA-polyethylene glycol (PLGA-PEG), were used to obtain serum-derived protein coronas. Protein corona characterization and liquid chromatography mass spectrometry analysis revealed distinct differences in biophysical properties and protein composition. PLGA protein coronas contained high abundance of globins (60%) and apolipoproteins (21%), while PLGA-PEG protein coronas contained fewer globins (42%) and high abundance of protease inhibitors (28%). Corona coated PLGA nanoparticles were readily internalized into microglia and neuronal cells, but not into astrocytes. Internalization of nanoparticles was associated with pro-inflammatory cytokine release and decreased neuronal cell viability, however, viability was rescued in cells treated with corona coated nanoparticles. These results showcase the importance of the protein corona in mediating nanoparticle-cell interactions.
Collapse
Affiliation(s)
- Nabila Morshed
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Claire Rennie
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Wei Deng
- School of Biomedical Engineering, University of Technology Sydney, Gadigal Country, Sydney, NSW 2007, Australia
| | - Lyndsey Collins-Praino
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Andrew Care
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
- Biologics Innovation Facility, University of Technology Sydney, Gadigal Country, Sydney, NSW 2007, Australia
| |
Collapse
|
6
|
Nani JV, Muotri AR, Hayashi MAF. Peering into the mind: unraveling schizophrenia's secrets using models. Mol Psychiatry 2024:10.1038/s41380-024-02728-w. [PMID: 39245692 DOI: 10.1038/s41380-024-02728-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024]
Abstract
Schizophrenia (SCZ) is a complex mental disorder characterized by a range of symptoms, including positive and negative symptoms, as well as cognitive impairments. Despite the extensive research, the underlying neurobiology of SCZ remain elusive. To overcome this challenge, the use of diverse laboratory modeling techniques, encompassing cellular and animal models, and innovative approaches like induced pluripotent stem cell (iPSC)-derived neuronal cultures or brain organoids and genetically engineered animal models, has been crucial. Immortalized cellular models provide controlled environments for investigating the molecular and neurochemical pathways involved in neuronal function, while iPSCs and brain organoids, derived from patient-specific sources, offer significant advantage in translational research by facilitating direct comparisons of cellular phenotypes between patient-derived neurons and healthy-control neurons. Animal models can recapitulate the different psychopathological aspects that should be modeled, offering valuable insights into the neurobiology of SCZ. In addition, invertebrates' models are genetically tractable and offer a powerful approach to dissect the core genetic underpinnings of SCZ, while vertebrate models, especially mammals, with their more complex nervous systems and behavioral repertoire, provide a closer approximation of the human condition to study SCZ-related traits. This narrative review provides a comprehensive overview of the diverse modeling approaches, critically evaluating their strengths and limitations. By synthesizing knowledge from these models, this review offers a valuable source for researchers, clinicians, and stakeholders alike. Integrating findings across these different models may allow us to build a more holistic picture of SCZ pathophysiology, facilitating the exploration of new research avenues and informed decision-making for interventions.
Collapse
Affiliation(s)
- João V Nani
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil.
| | - Alysson R Muotri
- Department of Pediatrics and Department of Molecular and Cellular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil.
| |
Collapse
|
7
|
Al Jaf AIA, Peria S, Fabiano T, Ragnini-Wilson A. Remyelinating Drugs at a Crossroad: How to Improve Clinical Efficacy and Drug Screenings. Cells 2024; 13:1326. [PMID: 39195216 DOI: 10.3390/cells13161326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
Axons wrapped around the myelin sheath enable fast transmission of neuronal signals in the Central Nervous System (CNS). Unfortunately, myelin can be damaged by injury, viral infection, and inflammatory and neurodegenerative diseases. Remyelination is a spontaneous process that can restore nerve conductivity and thus movement and cognition after a demyelination event. Cumulative evidence indicates that remyelination can be pharmacologically stimulated, either by targeting natural inhibitors of Oligodendrocyte Precursor Cells (OPCs) differentiation or by reactivating quiescent Neural Stem Cells (qNSCs) proliferation and differentiation in myelinating Oligodendrocytes (OLs). Although promising results were obtained in animal models for demyelination diseases, none of the compounds identified have passed all the clinical stages. The significant number of patients who could benefit from remyelination therapies reinforces the urgent need to reassess drug selection approaches and develop strategies that effectively promote remyelination. Integrating Artificial Intelligence (AI)-driven technologies with patient-derived cell-based assays and organoid models is expected to lead to novel strategies and drug screening pipelines to achieve this goal. In this review, we explore the current literature on these technologies and their potential to enhance the identification of more effective drugs for clinical use in CNS remyelination therapies.
Collapse
Affiliation(s)
- Aland Ibrahim Ahmed Al Jaf
- Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Simone Peria
- Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Tommaso Fabiano
- Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Antonella Ragnini-Wilson
- Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy
| |
Collapse
|
8
|
Harper JM. Primary Cell Culture as a Model System for Evolutionary Molecular Physiology. Int J Mol Sci 2024; 25:7905. [PMID: 39063147 PMCID: PMC11277064 DOI: 10.3390/ijms25147905] [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: 06/13/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Primary cell culture is a powerful model system to address fundamental questions about organismal physiology at the cellular level, especially for species that are difficult, or impossible, to study under natural or semi-natural conditions. Due to their ease of use, primary fibroblast cultures are the dominant model system, but studies using both somatic and germ cells are also common. Using these models, genome evolution and phylogenetic relationships, the molecular and biochemical basis of differential longevities among species, and the physiological consequences of life history evolution have been studied in depth. With the advent of new technologies such as gene editing and the generation of induced pluripotent stem cells (iPSC), the field of molecular evolutionary physiology will continue to expand using both descriptive and experimental approaches.
Collapse
Affiliation(s)
- James M Harper
- Department of Biological Sciences, Sam Houston State University, 1900 Avenue I, Huntsville, TX 77341, USA
| |
Collapse
|
9
|
Labrak Y, Alhouayek M, Mwema A, d'Auria L, Ucakar B, van Pesch V, Muccioli GG, des Rieux A. The combined administration of LNC-encapsulated retinoic acid and calcitriol stimulates oligodendrocyte progenitor cell differentiation in vitro and in vivo after intranasal administration. Int J Pharm 2024; 659:124237. [PMID: 38762167 DOI: 10.1016/j.ijpharm.2024.124237] [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: 11/02/2023] [Revised: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
Intranasal administration is an efficient strategy for bypassing the BBB, favoring drug accumulation in the brain, and improving its efficiency. Lipid nanocapsules (LNC) are suitable nanocarriers for the delivery of lipophilic drugs via this route and can be used to encapsulate lipophilic molecules such as retinoic acid (RA) and calcitriol (Cal). As the hallmarks of multiple sclerosis (MS) are neuroinflammation and oligodendrocyte loss, our hypothesis was that by combining two molecules known for their pro-differentiating properties, encapsulated in LNC, and delivered by intranasal administration, we would stimulate oligodendrocyte progenitor cells (OPC) differentiation into oligodendrocytes and provide a new pro-remyelinating therapy. LNC loaded with RA (LNC-RA) and Cal (LNC-Cal) were stable for at least 8 weeks. The combination of RA and Cal was more efficient than the molecules alone, encapsulated or not, on OPC differentiation in vitro and decreased microglia cell activation in a dose-dependent manner. After the combined intranasal administration of LNC-RA and LNC-Cal in a mouse cuprizone model of demyelination, increased MBP staining was observed in the corpus callosum. In conclusion, intranasal delivery of lipophilic drugs encapsulated in LNC is a promising strategy for myelinating therapies.
Collapse
Affiliation(s)
- Y Labrak
- Université catholique de Louvain (UCLouvain), Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium; Université catholique de Louvain (UCLouvain), Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, 1200 Brussels, Belgium
| | - M Alhouayek
- Université catholique de Louvain (UCLouvain), Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, 1200 Brussels, Belgium
| | - A Mwema
- Université catholique de Louvain (UCLouvain), Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium; Université catholique de Louvain (UCLouvain), Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, 1200 Brussels, Belgium
| | - L d'Auria
- Université catholique de Louvain (UCLouvain), Institute of Neuroscience, Neurochemistry Unit, 1200 Brussels, Belgium
| | - B Ucakar
- Université catholique de Louvain (UCLouvain), Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - V van Pesch
- Université catholique de Louvain (UCLouvain), Institute of Neuroscience, Neurochemistry Unit, 1200 Brussels, Belgium
| | - G G Muccioli
- Université catholique de Louvain (UCLouvain), Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, 1200 Brussels, Belgium.
| | - A des Rieux
- Université catholique de Louvain (UCLouvain), Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium.
| |
Collapse
|
10
|
Fukatsu S, Miyamoto Y, Oka Y, Ishibashi M, Shirai R, Ishida Y, Endo S, Katoh H, Yamauchi J. Investigating the Protective Effects of a Citrus Flavonoid on the Retardation Morphogenesis of the Oligodendroglia-like Cell Line by Rnd2 Knockdown. Neurol Int 2023; 16:33-61. [PMID: 38251051 PMCID: PMC10801557 DOI: 10.3390/neurolint16010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Recent discoveries suggest links between abnormalities in cell morphogenesis in the brain and the functional deficiency of molecules controlling signal transduction in glial cells such as oligodendroglia. Rnd2 is one such molecule and one of the Rho family monomeric GTP-binding proteins. Despite the currently known functions of Rnd2, its precise roles as it relates to cell morphogenesis and disease state remain to be elucidated. First, we showed that signaling through the loss of function of the rnd2 gene affected the regulation of oligodendroglial cell-like morphological differentiation using the FBD-102b cell line, which is often utilized as a differentiation model. The knockdown of Rnd2 using the clustered regularly interspaced palindromic repeats (CRISPR)/CasRx system or RNA interference was shown to slow morphological differentiation. Second, the knockdown of Prag1 or Fyn kinase, a signaling molecule acting downstream of Rnd2, slowed differentiation. Rnd2 or Prag1 knockdown also decreased Fyn phosphorylation, which is critical for its activation and for oligodendroglial cell differentiation and myelination. Of note, hesperetin, a citrus flavonoid with protective effects on oligodendroglial cells and neurons, can recover differentiation states induced by the knockdown of Rnd2/Prag1/Fyn. Here, we showed that signaling through Rnd2/Prag1/Fyn is involved in the regulation of oligodendroglial cell-like morphological differentiation. The effects of knocking down the signaling cascade molecule can be recovered by hesperetin, highlighting an important molecular structure involved in morphological differentiation.
Collapse
Affiliation(s)
- Shoya Fukatsu
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (S.F.); (Y.M.); (R.S.)
| | - Yuki Miyamoto
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (S.F.); (Y.M.); (R.S.)
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Setagaya-ku, Tokyo 157-8535, Japan
| | - Yu Oka
- Personal Health Care Division, Hayashibara Co., Ltd., Okayama 702-8006, Japan
| | - Maki Ishibashi
- Personal Health Care Division, Hayashibara Co., Ltd., Okayama 702-8006, Japan
| | - Remina Shirai
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (S.F.); (Y.M.); (R.S.)
| | - Yuki Ishida
- Personal Health Care Division, Hayashibara Co., Ltd., Okayama 702-8006, Japan
| | - Shin Endo
- Personal Health Care Division, Hayashibara Co., Ltd., Okayama 702-8006, Japan
| | - Hironori Katoh
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, Osaka 599-8531, Japan;
| | - Junji Yamauchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (S.F.); (Y.M.); (R.S.)
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Setagaya-ku, Tokyo 157-8535, Japan
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| |
Collapse
|
11
|
Dominicis A, Del Giovane A, Torreggiani M, Recchia AD, Ciccarone F, Ciriolo MR, Ragnini-Wilson A. N-Acetylaspartate Drives Oligodendroglial Differentiation via Histone Deacetylase Activation. Cells 2023; 12:1861. [PMID: 37508525 PMCID: PMC10378218 DOI: 10.3390/cells12141861] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/09/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
An unmet clinical goal in demyelinating pathologies is to restore the myelin sheath prior to neural degeneration. N-acetylaspartate (NAA) is an acetylated derivative form of aspartate, abundant in the healthy brain but severely reduced during traumatic brain injury and in patients with neurodegenerative pathologies. How extracellular NAA variations impact the remyelination process and, thereby, the ability of oligodendrocytes to remyelinate axons remains unexplored. Here, we evaluated the remyelination properties of the oligodendroglial (OL) mouse cell line Oli-neuM under different concentrations of NAA using a combination of biochemical, qPCR, immunofluorescence assays, and in vitro engagement tests, at NAA doses compatible with those observed in healthy brains and during brain injury. We observed that oligodendroglia cells respond to decreasing levels of NAA by stimulating differentiation and promoting gene expression of myelin proteins in a temporally regulated manner. Low doses of NAA potently stimulate Oli-neuM to engage with synthetic axons. Furthermore, we show a concentration-dependent expression of specific histone deacetylases essential for MBP gene expression under NAA or Clobetasol treatment. These data are consistent with the idea that oligodendrocytes respond to lowering the NAA concentration by activating the remyelination process via deacetylase activation.
Collapse
Affiliation(s)
| | - Alice Del Giovane
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Matteo Torreggiani
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | | | - Fabio Ciccarone
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
- IRCCS San Raffaele, 00166 Rome, Italy
| | - Maria Rosa Ciriolo
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
- IRCCS San Raffaele, 00166 Rome, Italy
| | | |
Collapse
|
12
|
Dedoni S, Scherma M, Camoglio C, Siddi C, Dazzi L, Puliga R, Frau J, Cocco E, Fadda P. An overall view of the most common experimental models for multiple sclerosis. Neurobiol Dis 2023:106230. [PMID: 37453561 DOI: 10.1016/j.nbd.2023.106230] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/01/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
Multiple sclerosis (MS) is a complex chronic disease with an unknown etiology. It is considered an inflammatory demyelinating and neurodegenerative disorder of the central nervous system (CNS) characterized, in most cases, by an unpredictable onset of relapse and remission phases. The disease generally starts in subjects under 40; it has a higher incidence in women and is described as a multifactorial disorder due to the interaction between genetic and environmental risk factors. Unfortunately, there is currently no definitive cure for MS. Still, therapies can modify the disease's natural history, reducing the relapse rate and slowing the progression of the disease or managing symptoms. The limited access to human CNS tissue slows down. It limits the progression of research on MS. This limit has been partially overcome over the years by developing various experimental models to study this disease. Animal models of autoimmune demyelination, such as experimental autoimmune encephalomyelitis (EAE) and viral and toxin or transgenic MS models, represent the most significant part of MS research approaches. These models have now been complemented by ex vivo studies, using organotypic brain slice cultures and in vitro, through induced Pluripotent Stem cells (iPSCs). We will discuss which clinical features of the disorders might be reproduced and investigated in vivo, ex vivo, and in vitro in models commonly used in MS research to understand the processes behind the neuropathological events occurring in the CNS of MS patients. The primary purpose of this review is to give the reader a global view of the main paradigms used in MS research, spacing from the classical animal models to transgenic mice and 2D and 3D cultures.
Collapse
Affiliation(s)
- S Dedoni
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - M Scherma
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Camoglio
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Siddi
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - L Dazzi
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - R Puliga
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - J Frau
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy
| | - E Cocco
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy; Department Medical Science and Public Health, University of Cagliari, Italy.
| | - P Fadda
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy; Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), Cagliari, Italy.
| |
Collapse
|
13
|
Park J, Wu Y, Li Q, Choi J, Ju H, Cai Y, Lee J, Oh YK. Nanomaterials for antigen-specific immune tolerance therapy. Drug Deliv Transl Res 2023; 13:1859-1881. [PMID: 36094655 DOI: 10.1007/s13346-022-01233-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2022] [Indexed: 11/26/2022]
Abstract
Impairment of immune tolerance might cause autologous tissue damage or overactive immune response against non-pathogenic molecules. Although autoimmune disease and allergy have complicated pathologies, the current strategies have mainly focused on symptom amelioration or systemic immunosuppression which can lead to fatal adverse events. The induction of antigen-specific immune tolerance may provide therapeutic benefits to autoimmune disease and allergic response, while reducing nonspecific immune adverse responses. Diverse nanomaterials have been studied to induce antigen-specific immune tolerance therapy. This review will cover the immunological background of antigen-specific tolerance, clinical importance of antigen-specific immune tolerance, and nanomaterials designed for autoimmune and allergic diseases. As nanomaterials for modulating immune tolerances, lipid-based nanoparticles, polymeric nanoparticles, and biological carriers have been covered. Strategies to provide antigen-specific immune tolerance have been addressed. Finally, current challenges and perspectives of nanomaterials for antigen-specific immune tolerance therapy will be discussed.
Collapse
Affiliation(s)
- Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Qiaoyun Li
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jaehyun Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyemin Ju
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yu Cai
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
14
|
Li Z, Jiang Z, Lu L, Liu Y. Microfluidic Manipulation for Biomedical Applications in the Central and Peripheral Nervous Systems. Pharmaceutics 2023; 15:pharmaceutics15010210. [PMID: 36678839 PMCID: PMC9862045 DOI: 10.3390/pharmaceutics15010210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Physical injuries and neurodegenerative diseases often lead to irreversible damage to the organizational structure of the central nervous system (CNS) and peripheral nervous system (PNS), culminating in physiological malfunctions. Investigating these complex and diverse biological processes at the macro and micro levels will help to identify the cellular and molecular mechanisms associated with nerve degeneration and regeneration, thereby providing new options for the development of new therapeutic strategies for the functional recovery of the nervous system. Due to their distinct advantages, modern microfluidic platforms have significant potential for high-throughput cell and organoid cultures in vitro, the synthesis of a variety of tissue engineering scaffolds and drug carriers, and observing the delivery of drugs at the desired speed to the desired location in real time. In this review, we first introduce the types of nerve damage and the repair mechanisms of the CNS and PNS; then, we summarize the development of microfluidic platforms and their application in drug carriers. We also describe a variety of damage models, tissue engineering scaffolds, and drug carriers for nerve injury repair based on the application of microfluidic platforms. Finally, we discuss remaining challenges and future perspectives with regard to the promotion of nerve injury repair based on engineered microfluidic platform technology.
Collapse
|
15
|
do Rosario MC, Bey GR, Nmezi B, Liu F, Oranburg T, Cohen ASA, Coffman KA, Brown MR, Kiselyov K, Waisfisz Q, Flohil MT, Siddiqui S, Rosenfeld JA, Iglesias A, Girisha KM, Wolf NI, Padiath QS, Shukla A. Variants in the zinc transporter TMEM163 cause a hypomyelinating leukodystrophy. Brain 2022; 145:4202-4209. [PMID: 35953447 PMCID: PMC10200305 DOI: 10.1093/brain/awac295] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 07/12/2022] [Accepted: 07/31/2022] [Indexed: 11/12/2022] Open
Abstract
Hypomyelinating leukodystrophies comprise a subclass of genetic disorders with deficient myelination of the CNS white matter. Here we report four unrelated families with a hypomyelinating leukodystrophy phenotype harbouring variants in TMEM163 (NM_030923.5). The initial clinical presentation resembled Pelizaeus-Merzbacher disease with congenital nystagmus, hypotonia, delayed global development and neuroimaging findings suggestive of significant and diffuse hypomyelination. Genomic testing identified three distinct heterozygous missense variants in TMEM163 with two unrelated individuals sharing the same de novo variant. TMEM163 is highly expressed in the CNS particularly in newly myelinating oligodendrocytes and was recently revealed to function as a zinc efflux transporter. All the variants identified lie in highly conserved residues in the cytoplasmic domain of the protein, and functional in vitro analysis of the mutant protein demonstrated significant impairment in the ability to efflux zinc out of the cell. Expression of the mutant proteins in an oligodendroglial cell line resulted in substantially reduced mRNA expression of key myelin genes, reduced branching and increased cell death. Our findings indicate that variants in TMEM163 cause a hypomyelinating leukodystrophy and uncover a novel role for zinc homeostasis in oligodendrocyte development and myelin formation.
Collapse
Affiliation(s)
- Michelle C do Rosario
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Guillermo Rodriguez Bey
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Bruce Nmezi
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Fang Liu
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Talia Oranburg
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ana S A Cohen
- Genomic Medicine Center, Children’s Mercy, Kansas City, MO 64108, USA
- Department of Pathology and Laboratory Medicine, Children’s Mercy, Kansas City, MO 64108, USA
- School of Medicine Serves, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Keith A Coffman
- Division of Neurology, Movement Disorders Clinic, Tourette Syndrome Center of Excellence, Children’s Mercy Hospital, Kansas City, Missouri, USA
| | - Maya R Brown
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kirill Kiselyov
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Quinten Waisfisz
- Department of Human Genetics, Amsterdam University Medical Centers, VU University Amsterdam, and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Myrthe T Flohil
- Department of Neurology, Noordwest ziekenhuisgroep, Wilhelminalaan Alkmaar, The Netherlands
| | - Shahyan Siddiqui
- Department of Neuroimaging and Interventional Radiology, STAR Institute of Neurosciences, STAR Hospitals, Hyderabad, India
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Baylor Genetics Laboratories, Houston, Texas, USA
| | - Alejandro Iglesias
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Katta Mohan Girisha
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Nicole I Wolf
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma’s Children’s Hospital, and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
| | - Quasar Saleem Padiath
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anju Shukla
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
16
|
Pazos M, Dibello E, Mesa JM, Sames D, Comini MA, Seoane G, Carrera I. Iboga Inspired N-Indolylethyl-Substituted Isoquinuclidines as a Bioactive Scaffold: Chemoenzymatic Synthesis and Characterization as GDNF Releasers and Antitrypanosoma Agents. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030829. [PMID: 35164094 PMCID: PMC8839081 DOI: 10.3390/molecules27030829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 12/31/2021] [Accepted: 01/04/2022] [Indexed: 11/16/2022]
Abstract
The first stage of the drug discovery process involves the identification of small compounds with biological activity. Iboga alkaloids are monoterpene indole alkaloids (MIAs) containing a fused isoquinuclidine-tetrahydroazepine ring. Both the natural products and the iboga-inspired synthetic analogs have shown a wide variety of biological activities. Herein, we describe the chemoenzymatic preparation of a small library of novel N-indolylethyl-substituted isoquinuclidines as iboga-inspired compounds, using toluene as a starting material and an imine Diels-Alder reaction as the key step in the synthesis. The new iboga series was investigated for its potential to promote the release of glial cell line-derived neurotrophic factor (GDNF) by C6 glioma cells, and to inhibit the growth of infective trypanosomes. GDNF is a neurotrophic factor widely recognized by its crucial role in development, survival, maintenance, and protection of dopaminergic neuronal circuitries affected in several neurological and psychiatric pathologies. Four compounds of the series showed promising activity as GDNF releasers, and a leading structure (compound 11) was identified for further studies. The same four compounds impaired the growth of bloodstream Trypanosoma brucei brucei (EC50 1-8 μM) and two of them (compounds 6 and 14) showed a good selectivity index.
Collapse
Affiliation(s)
- Mariana Pazos
- Laboratorio de Síntesis Orgánica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, General Flores 2124, Montevideo 11800, Uruguay; (M.P.); (E.D.); (J.M.M.); (G.S.)
| | - Estefania Dibello
- Laboratorio de Síntesis Orgánica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, General Flores 2124, Montevideo 11800, Uruguay; (M.P.); (E.D.); (J.M.M.); (G.S.)
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo 11400, Uruguay;
| | - Juan Manuel Mesa
- Laboratorio de Síntesis Orgánica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, General Flores 2124, Montevideo 11800, Uruguay; (M.P.); (E.D.); (J.M.M.); (G.S.)
| | - Dalibor Sames
- Department of Chemistry, Columbia University, New York, NY 10027, USA;
| | - Marcelo Alberto Comini
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo 11400, Uruguay;
| | - Gustavo Seoane
- Laboratorio de Síntesis Orgánica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, General Flores 2124, Montevideo 11800, Uruguay; (M.P.); (E.D.); (J.M.M.); (G.S.)
| | - Ignacio Carrera
- Laboratorio de Síntesis Orgánica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, General Flores 2124, Montevideo 11800, Uruguay; (M.P.); (E.D.); (J.M.M.); (G.S.)
- Correspondence: ; Tel.: +598-2-9247-881
| |
Collapse
|
17
|
Paun IA, Calin BS, Mustaciosu CC, Tanasa E, Moldovan A, Niemczyk A, Dinescu M. Laser Direct Writing via Two-Photon Polymerization of 3D Hierarchical Structures with Cells-Antiadhesive Properties. Int J Mol Sci 2021; 22:ijms22115653. [PMID: 34073424 PMCID: PMC8198338 DOI: 10.3390/ijms22115653] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/06/2021] [Accepted: 05/24/2021] [Indexed: 01/04/2023] Open
Abstract
We report the design and fabrication by laser direct writing via two photons polymerization of innovative hierarchical structures with cell-repellency capability. The structures were designed in the shape of “mushrooms”, consisting of an underside (mushroom’s leg) acting as a support structure and a top side (mushroom’s hat) decorated with micro- and nanostructures. A ripple-like pattern was created on top of the mushrooms, over length scales ranging from several µm (microstructured mushroom-like pillars, MMP) to tens of nm (nanostructured mushroom-like pillars, NMP). The MMP and NMP structures were hydrophobic, with contact angles of (127 ± 2)° and (128 ± 4)°, respectively, whereas flat polymer surfaces were hydrophilic, with a contact angle of (43 ± 1)°. The cell attachment on NMP structures was reduced by 55% as compared to the controls, whereas for the MMP, a reduction of only 21% was observed. Moreover, the MMP structures preserved the native spindle-like with phyllopodia cellular shape, whereas the cells from NMP structures showed a round shape and absence of phyllopodia. Overall, the NMP structures were more effective in impeding the cellular attachment and affected the cell shape to a greater extent than the MMP structures. The influence of the wettability on cell adhesion and shape was less important, the cellular behavior being mainly governed by structures’ topography.
Collapse
Affiliation(s)
- Irina A. Paun
- Center for Advanced Laser Technologies (CETAL), National Institute for Laser, Plasma and Radiation Physics, RO-077125 Magurele-Ilfov, Romania;
- Faculty of Applied Sciences, University Politehnica of Bucharest, RO-060042 Bucharest, Romania;
- Correspondence: ; Tel.: +40-770-612-912
| | - Bogdan S. Calin
- Center for Advanced Laser Technologies (CETAL), National Institute for Laser, Plasma and Radiation Physics, RO-077125 Magurele-Ilfov, Romania;
- Faculty of Applied Sciences, University Politehnica of Bucharest, RO-060042 Bucharest, Romania;
| | - Cosmin C. Mustaciosu
- Horia Hulubei National Institute for Physics and Nuclear Engineering IFIN-HH, RO-077125 Magurele-Ilfov, Romania;
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, RO-060042 Bucharest, Romania
| | - Eugenia Tanasa
- Faculty of Applied Sciences, University Politehnica of Bucharest, RO-060042 Bucharest, Romania;
- National Institute for Laser, Plasma and Radiation Physics, RO-077125 Magurele-Ilfov, Romania; (A.M.); (M.D.)
| | - Antoniu Moldovan
- National Institute for Laser, Plasma and Radiation Physics, RO-077125 Magurele-Ilfov, Romania; (A.M.); (M.D.)
| | - Agata Niemczyk
- Department of Materials Technology, Faculty of Mechanical Engineering and Mechatronics, West Pomeranian University of Technology in Szczecin, 19 Piastow Ave, 70-310 Szczecin, Poland;
| | - Maria Dinescu
- National Institute for Laser, Plasma and Radiation Physics, RO-077125 Magurele-Ilfov, Romania; (A.M.); (M.D.)
| |
Collapse
|
18
|
Exendin-4 Promotes Schwann Cell Survival/Migration and Myelination In Vitro. Int J Mol Sci 2021; 22:ijms22062971. [PMID: 33804063 PMCID: PMC7999558 DOI: 10.3390/ijms22062971] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/22/2021] [Accepted: 03/12/2021] [Indexed: 02/06/2023] Open
Abstract
Besides its insulinotropic actions on pancreatic β cells, neuroprotective activities of glucagon-like peptide-1 (GLP-1) have attracted attention. The efficacy of a GLP-1 receptor (GLP-1R) agonist exendin-4 (Ex-4) for functional repair after sciatic nerve injury and amelioration of diabetic peripheral neuropathy (DPN) has been reported; however, the underlying mechanisms remain unclear. In this study, the bioactivities of Ex-4 on immortalized adult rat Schwann cells IFRS1 and adult rat dorsal root ganglion (DRG) neuron–IFRS1 co-culture system were investigated. Localization of GLP-1R in both DRG neurons and IFRS1 cells were confirmed using knockout-validated monoclonal Mab7F38 antibody. Treatment with 100 nM Ex-4 significantly enhanced survival/proliferation and migration of IFRS1 cells, as well as stimulated the movement of IFRS1 cells toward neurites emerging from DRG neuron cell bodies in the co-culture with the upregulation of myelin protein 22 and myelin protein zero. Because Ex-4 induced phosphorylation of serine/threonine-specific protein kinase AKT in these cells and its effects on DRG neurons and IFRS1 cells were attenuated by phosphatidyl inositol-3′-phosphate-kinase (PI3K) inhibitor LY294002, Ex-4 might act on both cells to activate PI3K/AKT signaling pathway, thereby promoting myelination in the co-culture. These findings imply the potential efficacy of Ex-4 toward DPN and other peripheral nerve lesions.
Collapse
|
19
|
Balestri S, Del Giovane A, Sposato C, Ferrarelli M, Ragnini-Wilson A. The Current Challenges for Drug Discovery in CNS Remyelination. Int J Mol Sci 2021; 22:ijms22062891. [PMID: 33809224 PMCID: PMC8001072 DOI: 10.3390/ijms22062891] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
The myelin sheath wraps around axons, allowing saltatory currents to be transmitted along neurons. Several genetic, viral, or environmental factors can damage the central nervous system (CNS) myelin sheath during life. Unless the myelin sheath is repaired, these insults will lead to neurodegeneration. Remyelination occurs spontaneously upon myelin injury in healthy individuals but can fail in several demyelination pathologies or as a consequence of aging. Thus, pharmacological intervention that promotes CNS remyelination could have a major impact on patient’s lives by delaying or even preventing neurodegeneration. Drugs promoting CNS remyelination in animal models have been identified recently, mostly as a result of repurposing phenotypical screening campaigns that used novel oligodendrocyte cellular models. Although none of these have as yet arrived in the clinic, promising candidates are on the way. Many questions remain. Among the most relevant is the question if there is a time window when remyelination drugs should be administrated and why adult remyelination fails in many neurodegenerative pathologies. Moreover, a significant challenge in the field is how to reconstitute the oligodendrocyte/axon interaction environment representative of healthy as well as disease microenvironments in drug screening campaigns, so that drugs can be screened in the most appropriate disease-relevant conditions. Here we will provide an overview of how the field of in vitro models developed over recent years and recent biological findings about how oligodendrocytes mature after reactivation of their staminal niche. These data have posed novel questions and opened new views about how the adult brain is repaired after myelin injury and we will discuss how these new findings might change future drug screening campaigns for CNS regenerative drugs.
Collapse
|
20
|
Patel MR, Weaver AM. Astrocyte-derived small extracellular vesicles promote synapse formation via fibulin-2-mediated TGF-β signaling. Cell Rep 2021; 34:108829. [PMID: 33691102 PMCID: PMC8002899 DOI: 10.1016/j.celrep.2021.108829] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 01/05/2021] [Accepted: 02/16/2021] [Indexed: 11/22/2022] Open
Abstract
Neuronal synapse formation is critical for brain development and depends on secreted factors from astrocytes. Here, we report that small extracellular vesicles (EVs) secreted from primary astrocytes, but not from neurons or C6 glioma cells, greatly enhance spine and synapse formation by primary cortical neurons. A comparative proteomics analysis of small EVs from astrocytes, neurons, and C6 glioma cells identified fibulin-2 as a promising EV cargo to regulate synaptogenesis. Treatment of cortical neurons with recombinant fibulin-2 increased the formation of spines and synapses, similar to the effect of small EVs. In addition, treatment of neurons with fibulin-2 or astrocyte-derived small EVs led to increased phosphorylation of Smad2, an indicator of TGF-β signaling. Finally, the effects of fibulin-2 and astrocyte-derived small EVs on synapse formation were reversed by inhibiting transforming growth factor β (TGF-β) signaling. These data suggest a model in which astrocyte EVs promote synapse formation via fibulin-2-mediated activation of TGF-β signaling.
Collapse
Affiliation(s)
- Mikin R Patel
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
| | - Alissa M Weaver
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
21
|
Aldose Reductase and the Polyol Pathway in Schwann Cells: Old and New Problems. Int J Mol Sci 2021; 22:ijms22031031. [PMID: 33494154 PMCID: PMC7864348 DOI: 10.3390/ijms22031031] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 12/13/2022] Open
Abstract
Aldose reductase (AR) is a member of the reduced nicotinamide adenosine dinucleotide phosphate (NADPH)-dependent aldo-keto reductase superfamily. It is also the rate-limiting enzyme of the polyol pathway, catalyzing the conversion of glucose to sorbitol, which is subsequently converted to fructose by sorbitol dehydrogenase. AR is highly expressed by Schwann cells in the peripheral nervous system (PNS). The excess glucose flux through AR of the polyol pathway under hyperglycemic conditions has been suggested to play a critical role in the development and progression of diabetic peripheral neuropathy (DPN). Despite the intensive basic and clinical studies over the past four decades, the significance of AR over-activation as the pathogenic mechanism of DPN remains to be elucidated. Moreover, the expected efficacy of some AR inhibitors in patients with DPN has been unsatisfactory, which prompted us to further investigate and review the understanding of the physiological and pathological roles of AR in the PNS. Particularly, the investigation of AR and the polyol pathway using immortalized Schwann cells established from normal and AR-deficient mice could shed light on the causal relationship between the metabolic abnormalities of Schwann cells and discordance of axon-Schwann cell interplay in DPN, and led to the development of better therapeutic strategies against DPN.
Collapse
|
22
|
Puhl DL, Funnell JL, Nelson DW, Gottipati MK, Gilbert RJ. Electrospun Fiber Scaffolds for Engineering Glial Cell Behavior to Promote Neural Regeneration. Bioengineering (Basel) 2020; 8:4. [PMID: 33383759 PMCID: PMC7823609 DOI: 10.3390/bioengineering8010004] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
Electrospinning is a fabrication technique used to produce nano- or micro- diameter fibers to generate biocompatible, biodegradable scaffolds for tissue engineering applications. Electrospun fiber scaffolds are advantageous for neural regeneration because they mimic the structure of the nervous system extracellular matrix and provide contact guidance for regenerating axons. Glia are non-neuronal regulatory cells that maintain homeostasis in the healthy nervous system and regulate regeneration in the injured nervous system. Electrospun fiber scaffolds offer a wide range of characteristics, such as fiber alignment, diameter, surface nanotopography, and surface chemistry that can be engineered to achieve a desired glial cell response to injury. Further, electrospun fibers can be loaded with drugs, nucleic acids, or proteins to provide the local, sustained release of such therapeutics to alter glial cell phenotype to better support regeneration. This review provides the first comprehensive overview of how electrospun fiber alignment, diameter, surface nanotopography, surface functionalization, and therapeutic delivery affect Schwann cells in the peripheral nervous system and astrocytes, oligodendrocytes, and microglia in the central nervous system both in vitro and in vivo. The information presented can be used to design and optimize electrospun fiber scaffolds to target glial cell response to mitigate nervous system injury and improve regeneration.
Collapse
Affiliation(s)
- Devan L. Puhl
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; (D.L.P.); (J.L.F.); (D.W.N.); (M.K.G.)
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
| | - Jessica L. Funnell
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; (D.L.P.); (J.L.F.); (D.W.N.); (M.K.G.)
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
| | - Derek W. Nelson
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; (D.L.P.); (J.L.F.); (D.W.N.); (M.K.G.)
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
| | - Manoj K. Gottipati
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; (D.L.P.); (J.L.F.); (D.W.N.); (M.K.G.)
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University, 460 W. 12th Avenue, Columbus, OH 43210, USA
| | - Ryan J. Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; (D.L.P.); (J.L.F.); (D.W.N.); (M.K.G.)
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
| |
Collapse
|
23
|
Hydrogen Peroxide-Preconditioned Human Adipose-Derived Stem Cells Enhance the Recovery of Oligodendrocyte-Like Cells after Oxidative Stress-Induced Damage. Int J Mol Sci 2020; 21:ijms21249513. [PMID: 33327653 PMCID: PMC7765141 DOI: 10.3390/ijms21249513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 12/17/2022] Open
Abstract
Oxidative stress associated with neuroinflammation is a key process involved in the pathophysiology of neurodegenerative diseases, and therefore, has been proposed as a crucial target for new therapies. Recently, the therapeutic potential of human adipose-derived stem cells (hASCs) has been investigated as a novel strategy for neuroprotection. These cells can be preconditioned by exposing them to mild stress in order to improve their response to oxidative stress. In this study, we evaluate the therapeutic potential of hASCs preconditioned with low doses of H2O2 (called HC016 cells) to overcome the deleterious effect of oxidative stress in an in vitro model of oligodendrocyte-like cells (HOGd), through two strategies: i, the culture of oxidized HOGd with HC016 cell-conditioned medium (CM), and ii, the indirect co-culture of oxidized HOGd with HC016 cells, which had or had not been exposed to oxidative stress. The results demonstrated that both strategies had reparative effects, oxidized HC016 cell co-culture being the one associated with the greatest recovery of the damaged HOGd, increasing their viability, reducing their intracellular reactive oxygen species levels and promoting their antioxidant capacity. Taken together, these findings support the view that HC016 cells, given their reparative capacity, might be considered an important breakthrough in cell-based therapies.
Collapse
|
24
|
Low-Field Magnetic Stimulation Accelerates the Differentiation of Oligodendrocyte Precursor Cells via Non-canonical TGF-β Signaling Pathways. Mol Neurobiol 2020; 58:855-866. [PMID: 33037982 DOI: 10.1007/s12035-020-02157-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/30/2020] [Indexed: 01/17/2023]
Abstract
Demyelination and oligodendrocyte loss are characteristic changes in demyelinating disorders. Low-field magnetic stimulation (LFMS) is a novel transcranial neuromodulation technology that has shown promising therapeutic potential for a variety of neuropsychiatric conditions. The cellular and molecular mechanisms of magnetic stimulation remain unclear. Previous studies mainly focused on the effects of magnetic stimulation on neuronal cells. Here we aimed to examine the effects of a gamma frequency LFMS on the glial progenitor cells. We used rat central glia-4 (CG4) cell line as an in vitro model. CG4 is a bipotential glial progenitor cell line that can differentiate into either oligodendrocyte or type 2-astrocyte. The cells cultured in a defined differentiation media were exposed to a 40-Hz LFMS 20 min daily for five consecutive days. We found that LFMS transiently elevated the level of TGF-β1 in the culture media in the first 24 h after the treatment. In correlation with the TGF-β1 levels, the percentage of cells possessing complex branches and expressing the late oligodendrocyte progenitor marker O4 was increased, indicating the accelerated differentiation of CG4 cells towards oligodendrocyte in LFMS-treated cultures. LFMS increased phosphorylation of Akt and Erk1/2 proteins, but not SMAD2/3. TGF-β1 receptor I specific inhibitor LY 364947 partially suppressed the effects of LFMS on differentiation and on levels of pAkt and pErk1/2, indicating that LFMS enhances the differentiation of oligodendrocyte progenitor cells via activation of non-canonical TGF-β-Akt and TGF-β-Erk1/2 pathways but not the canonical SMAD pathway. The data from this study reveal a novel mechanism of magnetic stimulation as a potential therapy for demyelination disorders.
Collapse
|
25
|
Abstract
The etiology and pathogenesis of MS is likely to involve multiple factors interacting with each other, and the role of infectious and viral agents is still under debate, however a consistent amount of studies suggests that some viruses are associated with the disease. The strongest documentation has come from the detection of viral nucleic acid or antigen or of an anti-viral antibody response in MS patients. A further step for the study of the mechanism viruses might be involved in can be made using in vitro and in vivo models. While in vitro models, based on glial and neural cell lines from various sources are widely used, in vivo animal models present challenges. Indeed neurotropic animal viruses are currently used to study demyelination in well-established models, but animal models of demyelination by human virus infection have only recently been developed, using animal gammaherpesviruses closely related to Epstein Barr virus (EBV), or using marmosets expressing the specific viral receptor for Human Herpesvirus 6 (HHV-6). The present review will illustrate the main potential mechanisms of MS pathogenesis possibly associated with viral infections and viruses currently used to study demyelination in animal models. Then the viruses most strongly linked with MS will be discussed, in the perspective that more than one virus might have a role, with varying degrees of interaction, contributing to MS heterogeneity.
Collapse
Affiliation(s)
- Donatella Donati
- Neurologia e Neurofisiologia Clinica, Azienda Ospedaliera Universitaria Senese I 53100 Siena, Italy
| |
Collapse
|
26
|
Torres-Mejía E, Trümbach D, Kleeberger C, Dornseifer U, Orschmann T, Bäcker T, Brenke JK, Hadian K, Wurst W, López-Schier H, Desbordes SC. Sox2 controls Schwann cell self-organization through fibronectin fibrillogenesis. Sci Rep 2020; 10:1984. [PMID: 32029747 PMCID: PMC7005302 DOI: 10.1038/s41598-019-56877-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 12/09/2019] [Indexed: 02/03/2023] Open
Abstract
The extracellular matrix is known to modulate cell adhesion and migration during tissue regeneration. However, the molecular mechanisms that fine-tune cells to extra-cellular matrix dynamics during regeneration of the peripheral nervous system remain poorly understood. Using the RSC96 Schwann cell line, we show that Sox2 directly controls fibronectin fibrillogenesis in Schwann cells in culture, to provide a highly oriented fibronectin matrix, which supports their organization and directional migration. We demonstrate that Sox2 regulates Schwann cell behaviour through the upregulation of multiple extracellular matrix and migration genes as well as the formation of focal adhesions during cell movement. We find that mouse primary sensory neurons and human induced pluripotent stem cell-derived motoneurons require the Sox2-dependent fibronectin matrix in order to migrate along the oriented Schwann cells. Direct loss of fibronectin in Schwann cells impairs their directional migration affecting the alignment of the axons in vitro. Furthermore, we show that Sox2 and fibronectin are co-expressed in proregenerative Schwann cells in vivo in a time-dependent manner during sciatic nerve regeneration. Taken together, our results provide new insights into the mechanisms by which Schwann cells regulate their own extracellular microenvironment in a Sox2-dependent manner to ensure the proper migration of neurons.
Collapse
Affiliation(s)
- Elen Torres-Mejía
- Stem Cells in Neural Development and Disease group, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany.,Research Unit Sensory Biology and Organogenesis, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany.,Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany
| | - Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany
| | - Charlotte Kleeberger
- Department of Plastic, Reconstructive, Hand and Burn Surgery, Academic Hospital Bogenhausen, Munich, 81925, Germany
| | - Ulf Dornseifer
- Department of Plastic, Reconstructive, Hand and Burn Surgery, Academic Hospital Bogenhausen, Munich, 81925, Germany
| | - Tanja Orschmann
- Stem Cells in Neural Development and Disease group, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany.,Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany.,Stem Cell Based-Assay Development Platform (SCADEV), Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany
| | - Theresa Bäcker
- Stem Cells in Neural Development and Disease group, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany.,Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany.,Stem Cell Based-Assay Development Platform (SCADEV), Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany
| | - Jara Kerstin Brenke
- Assay Development and Screening Platform, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany
| | - Kamyar Hadian
- Assay Development and Screening Platform, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany.,Chair of Developmental Genetics, Technische Universität München-Weihenstephan, 85350, Freising-Weihenstephan, Germany.,German Center for Neurodegenerative Diseases (DZNE), 81377, Munich, Germany
| | - Hernán López-Schier
- Research Unit Sensory Biology and Organogenesis, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany
| | - Sabrina C Desbordes
- Stem Cells in Neural Development and Disease group, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany. .,Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany. .,Stem Cell Based-Assay Development Platform (SCADEV), Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany. .,ISAR Bioscience GmbH, Institute for Stem Cell & Applied Regenerative Medicine Research, Semmelweisstr. 5, 82152, Munich, Germany.
| |
Collapse
|
27
|
Monje PV. The properties of human Schwann cells: Lessons from in vitro culture and transplantation studies. Glia 2020; 68:797-810. [PMID: 32027424 DOI: 10.1002/glia.23793] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 11/10/2022]
Abstract
Human Schwann cells (hSCs) can be isolated directly from peripheral nerve and cultured using methods similar to those used for SCs from other species. Yet, important interspecies differences are revealed when the primary or expanded hSCs are compared to their nonhuman counterparts. This review addresses the special properties of nerve-derived hSCs that have resulted to date from both in vitro studies and in vivo research on cell transplantation in animal models and human subjects. A consensus has yet to emerge about the essential attributes of cultured normal hSCs. Thus, an emphasis is placed on the importance of validating hSC cultures by means of purity, identity, and biological activity to reliably use them as in vitro models of the SC phenotype and cell therapy products for injury repair. Combining traditional immunological methods, high-resolution omics approaches, and assorted cell-based assays is so far the best approach to unequivocally identify hSC populations obtained by direct isolation or derivation from stem cells. Special considerations are required to understand and manage the variability and heterogeneity proper of donor batches, as well as to evaluate risk factors. This is particularly important if the intended use of the hSCs is implantation in the human body, diagnosis of disease, or drug testing aimed at targeting any aspect of SC function in human patients. To conclude, in view of their unique properties, new concepts and methods are needed to better understand the biology of hSCs and exploit their full potential in basic science and regenerative medicine.
Collapse
Affiliation(s)
- Paula V Monje
- The Department of Neurological Surgery, Indiana University, Indianapolis, Indiana
| |
Collapse
|
28
|
Sanabria-Castro A, Flores-Díaz M, Alape-Girón A. Biological models in multiple sclerosis. J Neurosci Res 2019; 98:491-508. [PMID: 31571267 DOI: 10.1002/jnr.24528] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/05/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022]
Abstract
Considering the etiology of multiple sclerosis (MS) is still unknown, experimental models resembling specific aspects of this immune-mediated demyelinating human disease have been developed to increase the understanding of processes related to pathogenesis, disease evolution, evaluation of therapeutic interventions, and demyelination and remyelination mechanisms. Based on the nature of the investigation, biological models may include in vitro, in vivo, and ex vivo assessments. Even though these approaches have disclosed valuable information, every disease animal model has limitations and can only replicate specific features of MS. In vitro and ex vivo models generally do not reflect what occurs in the organism, and in vivo animal models are more likely used; nevertheless, they are able to reproduce only certain stages of the disease. In vivo MS disease animal models in mammals include: experimental autoimmune encephalomyelitis, viral encephalomyelitis, and induced demyelination. This review examines and describes the most common biological disease animal models for the study of MS, their specific characteristics and limitations.
Collapse
Affiliation(s)
- Alfredo Sanabria-Castro
- Research Unit, San Juan de Dios Hospital CCSS, San José, Costa Rica.,School of Pharmacy, University of Costa Rica, San José, Costa Rica
| | | | | |
Collapse
|
29
|
Choquet K, Forget D, Meloche E, Dicaire MJ, Bernard G, Vanderver A, Schiffmann R, Fabian MR, Teichmann M, Coulombe B, Brais B, Kleinman CL. Leukodystrophy-associated POLR3A mutations down-regulate the RNA polymerase III transcript and important regulatory RNA BC200. J Biol Chem 2019; 294:7445-7459. [PMID: 30898877 PMCID: PMC6509492 DOI: 10.1074/jbc.ra118.006271] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 03/07/2019] [Indexed: 12/12/2022] Open
Abstract
RNA polymerase III (Pol III) is an essential enzyme responsible for the synthesis of several small noncoding RNAs, a number of which are involved in mRNA translation. Recessive mutations in POLR3A, encoding the largest subunit of Pol III, cause POLR3-related hypomyelinating leukodystrophy (POLR3–HLD), characterized by deficient central nervous system myelination. Identification of the downstream effectors of pathogenic POLR3A mutations has so far been elusive. Here, we used CRISPR-Cas9 to introduce the POLR3A mutation c.2554A→G (p.M852V) into human cell lines and assessed its impact on Pol III biogenesis, nuclear import, DNA occupancy, transcription, and protein levels. Transcriptomic profiling uncovered a subset of transcripts vulnerable to Pol III hypofunction, including a global reduction in tRNA levels. The brain cytoplasmic BC200 RNA (BCYRN1), involved in translation regulation, was consistently affected in all our cellular models, including patient-derived fibroblasts. Genomic BC200 deletion in an oligodendroglial cell line led to major transcriptomic and proteomic changes, having a larger impact than those of POLR3A mutations. Upon differentiation, mRNA levels of the MBP gene, encoding myelin basic protein, were significantly decreased in POLR3A-mutant cells. Our findings provide the first evidence for impaired Pol III transcription in cellular models of POLR3–HLD and identify several candidate effectors, including BC200 RNA, having a potential role in oligodendrocyte biology and involvement in the disease.
Collapse
Affiliation(s)
- Karine Choquet
- From the Department of Human Genetics, McGill University, Montréal, Québec H3A 0C7, Canada.,the Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec H3T 1E2, Canada.,the Montréal Neurological Institute, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Diane Forget
- the Translational Proteomics Laboratory, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Elisabeth Meloche
- the Montréal Neurological Institute, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Marie-Josée Dicaire
- the Montréal Neurological Institute, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Geneviève Bernard
- From the Department of Human Genetics, McGill University, Montréal, Québec H3A 0C7, Canada.,Pediatrics, McGill University, Montréal, Québec H3A 0G4, Canada.,the Department of Internal Medicine, Division of Medical Genetics, Montréal Children's Hospital, McGill University Health Center, Montréal, Québec H4A 3J1, Canada.,the Child Health and Human Development Program, and.,MyeliNeuroGene Laboratory, Research Institute, McGill University Health Center, Montréal, Québec H4A 3J1, Canada.,the Departments of Neurology and Neurosurgery and
| | - Adeline Vanderver
- the Division of Neurology, Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania 19104
| | - Raphael Schiffmann
- the Institute of Metabolic Disease, Baylor Research Institute, Dallas, Texas 75204
| | - Marc R Fabian
- the Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec H3T 1E2, Canada
| | - Martin Teichmann
- INSERM U1212-CNRS UMR5320, Université de Bordeaux, Bordeaux, France, and
| | - Benoit Coulombe
- the Translational Proteomics Laboratory, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada.,the Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Bernard Brais
- From the Department of Human Genetics, McGill University, Montréal, Québec H3A 0C7, Canada.,the Montréal Neurological Institute, McGill University, Montréal, Québec H3A 2B4, Canada.,the Departments of Neurology and Neurosurgery and
| | - Claudia L Kleinman
- From the Department of Human Genetics, McGill University, Montréal, Québec H3A 0C7, Canada, .,the Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec H3T 1E2, Canada
| |
Collapse
|
30
|
Curiel J, Rodríguez Bey G, Takanohashi A, Bugiani M, Fu X, Wolf NI, Nmezi B, Schiffmann R, Bugaighis M, Pierson T, Helman G, Simons C, van der Knaap MS, Liu J, Padiath Q, Vanderver A. TUBB4A mutations result in specific neuronal and oligodendrocytic defects that closely match clinically distinct phenotypes. Hum Mol Genet 2018; 26:4506-4518. [PMID: 28973395 DOI: 10.1093/hmg/ddx338] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 08/24/2017] [Indexed: 12/16/2022] Open
Abstract
Hypomyelinating leukodystrophies are heritable disorders defined by lack of development of brain myelin, but the cellular mechanisms of hypomyelination are often poorly understood. Mutations in TUBB4A, encoding the tubulin isoform tubulin beta class IVA (Tubb4a), result in the symptom complex of hypomyelination with atrophy of basal ganglia and cerebellum (H-ABC). Additionally, TUBB4A mutations are known to result in a broad phenotypic spectrum, ranging from primary dystonia (DYT4), isolated hypomyelination with spastic quadriplegia, and an infantile onset encephalopathy, suggesting multiple cell types may be involved. We present a study of the cellular effects of TUBB4A mutations responsible for H-ABC (p.Asp249Asn), DYT4 (p.Arg2Gly), a severe combined phenotype with hypomyelination and encephalopathy (p.Asn414Lys), as well as milder phenotypes causing isolated hypomyelination (p.Val255Ile and p.Arg282Pro). We used a combination of histopathological, biochemical and cellular approaches to determine how these different mutations may have variable cellular effects in neurons and/or oligodendrocytes. Our results demonstrate that specific mutations lead to either purely neuronal, combined neuronal and oligodendrocytic or purely oligodendrocytic defects that closely match their respective clinical phenotypes. Thus, the DYT4 mutation that leads to phenotypes attributable to neuronal dysfunction results in altered neuronal morphology, but with unchanged tubulin quantity and polymerization, with normal oligodendrocyte morphology and myelin gene expression. Conversely, mutations associated with isolated hypomyelination (p.Val255Ile and p.Arg282Pro) and the severe combined phenotype (p.Asn414Lys) resulted in normal neuronal morphology but were associated with altered oligodendrocyte morphology, myelin gene expression, and microtubule dysfunction. The H-ABC mutation (p.Asp249Asn) that exhibits a combined neuronal and myelin phenotype had overlapping cellular defects involving both neuronal and oligodendrocyte cell types in vitro. Only mutations causing hypomyelination phenotypes showed altered microtubule dynamics and acted through a dominant toxic gain of function mechanism. The DYT4 mutation had no impact on microtubule dynamics suggesting a distinct mechanism of action. In summary, the different clinical phenotypes associated with TUBB4A reflect the selective and specific cellular effects of the causative mutations. Cellular specificity of disease pathogenesis is relevant to developing targeted treatments for this disabling condition.
Collapse
Affiliation(s)
- Julian Curiel
- Center for Neuroscience Research, Children's National Health System, Children's Research Institute, Washington, DC 20010, USA
| | | | - Asako Takanohashi
- Center for Genetic Medicine Research, Children's National Health System, Children's Research Institute, Washington, DC 20010, USA.,Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | - Xiaoqin Fu
- Center for Neuroscience Research, Children's National Health System, Children's Research Institute, Washington, DC 20010, USA
| | - Nicole I Wolf
- VU University Medical Center, Amsterdam, The Netherlands
| | - Bruce Nmezi
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Raphael Schiffmann
- Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas, TX 75204, USA
| | - Mona Bugaighis
- Center for Neuroscience Research, Children's National Health System, Children's Research Institute, Washington, DC 20010, USA
| | - Tyler Pierson
- Departments of Pediatrics and Neurology, Cedar Sinai Medical Center, Board of Governors Regenerative Medicine Institute, Los Angeles, CA 90048, USA
| | - Guy Helman
- Center for Genetic Medicine Research, Children's National Health System, Children's Research Institute, Washington, DC 20010, USA.,Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia.,Department of Neurology, Children's National Health System, Washington, DC 20010, USA
| | - Cas Simons
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | | | - Judy Liu
- Center for Neuroscience Research, Children's National Health System, Children's Research Institute, Washington, DC 20010, USA
| | - Quasar Padiath
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Adeline Vanderver
- Center for Genetic Medicine Research, Children's National Health System, Children's Research Institute, Washington, DC 20010, USA.,Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.,Department of Neurology, Children's National Health System, Washington, DC 20010, USA.,Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
31
|
Mikhailova V, Gulaia V, Tiasto V, Rybtsov S, Yatsunskaya M, Kagansky A. Towards an advanced cell-based in vitro glioma model system. AIMS GENETICS 2018; 5:91-112. [PMID: 31435515 PMCID: PMC6698577 DOI: 10.3934/genet.2018.2.91] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 03/14/2018] [Indexed: 02/06/2023]
Abstract
The modulation of tumor growth and development in vitro has always been one of the key factors in the research of the malignant transformation, including gliomas, prevalent and most deadly cancers of the brain. Indeed, cellular and molecular biology research employing in vitro model cell-based systems have great potential to advance both the mechanistic understanding and the treatment of human glial tumors, as it facilitates not only the understanding of glioma biology and its regulatory mechanisms Additionally they promise to afford the screening of the putative anti-tumor agents and alternative treatment approaches in a personalized manner, i.e. by virtue of using the patient-derived tumor material for such tests. However, in order to become reliable and representative, glioma model systems need to move towards including most inherent cancer features such as local hypoxia, specific genetic aberrations, native tumor microenvironment, and the three-dimensional extracellular matrix. This review starts with a brief introduction on the general epidemiological and molecular characteristics of gliomas followed by an overview of the cell-based in vitro models currently used in glioma research. As a conclusion, we suggest approaches to move to innovative cell-based in vitro glioma models. We consider that main criteria for selecting these approaches should include the adequate resemblance to the key in vivo characteristics, robustness, cost-effectiveness and ease to use, as well as the amenability to high throughput handling to allow the standardized drug screening.
Collapse
Affiliation(s)
- Valeriia Mikhailova
- Center for Genomic and Regenerative Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russian Federation
| | - Valeriia Gulaia
- Center for Genomic and Regenerative Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russian Federation
| | - Vladlena Tiasto
- Center for Genomic and Regenerative Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russian Federation
| | - Stanislav Rybtsov
- Scottish Centre for Regenerative Medicine of the University of Edinburgh, Edinburgh, United Kingdom
| | - Margarita Yatsunskaya
- Federal Scientific Center of the East Asia Terrestrial Biodiversity FEB RAS 159, Stoletij Vladivostoku Avenue, 690022, Vladivostok, Russian Federation
| | - Alexander Kagansky
- Center for Genomic and Regenerative Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russian Federation
| |
Collapse
|
32
|
Abstract
The most widely used method (Brockes' method) for preparing primary Schwann cell culture uses neonatal rat sciatic nerves as the primary source of Schwann cells. The procedure is relatively simple and yields a highly purified population of Schwann cells in a short period of time. The method has also been used to prepare Schwann cells from mice, however, with limitation. For example, Brockes' method is not applicable when the genotypes of mouse neonates are unknown or if the mouse mutants do not develop to term. We described a method ideal for preparing Schwann cells in a transgenic/knockout mouse study. The method uses embryonic dorsal root ganglia as the primary source of Schwann cells and allows preparing separate, highly purified Schwann cell cultures from individual mouse embryos in less than 2 weeks.
Collapse
Affiliation(s)
- Jihyun Kim
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | - Haesun A Kim
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA.
| |
Collapse
|
33
|
Canine dorsal root ganglia satellite glial cells represent an exceptional cell population with astrocytic and oligodendrocytic properties. Sci Rep 2017; 7:13915. [PMID: 29066783 PMCID: PMC5654978 DOI: 10.1038/s41598-017-14246-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/06/2017] [Indexed: 12/21/2022] Open
Abstract
Dogs can be used as a translational animal model to close the gap between basic discoveries in rodents and clinical trials in humans. The present study compared the species-specific properties of satellite glial cells (SGCs) of canine and murine dorsal root ganglia (DRG) in situ and in vitro using light microscopy, electron microscopy, and immunostainings. The in situ expression of CNPase, GFAP, and glutamine synthetase (GS) has also been investigated in simian SGCs. In situ, most canine SGCs (>80%) expressed the neural progenitor cell markers nestin and Sox2. CNPase and GFAP were found in most canine and simian but not murine SGCs. GS was detected in 94% of simian and 71% of murine SGCs, whereas only 44% of canine SGCs expressed GS. In vitro, most canine (>84%) and murine (>96%) SGCs expressed CNPase, whereas GFAP expression was differentially affected by culture conditions and varied between 10% and 40%. However, GFAP expression was induced by bone morphogenetic protein 4 in SGCs of both species. Interestingly, canine SGCs also stimulated neurite formation of DRG neurons. These findings indicate that SGCs represent an exceptional, intermediate glial cell population with phenotypical characteristics of oligodendrocytes and astrocytes and might possess intrinsic regenerative capabilities in vivo.
Collapse
|
34
|
Segura-Ulate I, Yang B, Vargas-Medrano J, Perez RG. FTY720 (Fingolimod) reverses α-synuclein-induced downregulation of brain-derived neurotrophic factor mRNA in OLN-93 oligodendroglial cells. Neuropharmacology 2017; 117:149-157. [PMID: 28153532 DOI: 10.1016/j.neuropharm.2017.01.028] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 01/23/2017] [Accepted: 01/27/2017] [Indexed: 02/07/2023]
Abstract
Multiple system atrophy (MSA) is a demyelinating neurodegenerative disorder characterized by accumulation of aggregated α-synuclein (aSyn) inside oligodendrocyte precursors, mature oligodendroglia, and neurons. MSA dysfunction is associated with loss of trophic factor production by glial and neuronal cells. Here, we report that recombinant wild type human aSyn uptake by OLN-93, an oligodendroglia cell-line, reduced brain-derived neurotrophic factor (BDNF) expression. Furthermore, OLN-93 cells stably transfected with human wild type or an MSA-associated mutant aSyn, A53E that produces neuronal and glial inclusions, reduced BDNF mRNA to nearly unmeasurable qPCR levels. Curiously, another MSA-associated aSyn mutant, G51D that also produces neuronal and glial inclusions, caused only a trend toward BDNF mRNA reduction in transfected OLN-93 cells. This suggests that oligodendrocyte-associated BDNF loss occurs in response to specific aSyn types. Treating OLN-93 cells with 160 nM FTY720 (Fingolimod, Gilenya®), a Food and Drug Administration (FDA) approved therapeutic for multiple sclerosis, counteracted BDNF downregulation in all aSyn OLN-93 cells. FTY720 also restored BDNF mRNA in OLN-93 cells treated with recombinant aSyn, as measured by qPCR or semiquantitatively on agarose gels. Immunoblots confirmed that FTY720 increased histone 3 acetylation in OLN-93, and chromatin immunoprecipitation assays showed increased acetylated histone 3 at BDNF promoter 1 after FTY720. Moreover, OLN-93 cells treated with valproic acid, a classic histone deacetylase inhibitor, confirmed that increasing acetylated histone 3 levels increases BDNF expression. Cumulatively, the data suggest that FTY720-associated histone deacetylase inhibition stimulates BDNF expression in oligodendroglial cells, raising the possibility that MSA patients may also benefit by treatment with FTY720.
Collapse
Affiliation(s)
- Ismael Segura-Ulate
- Texas Tech University Health Sciences Center El Paso, Department of Biomedical Sciences, Graduate School of Biomedical Sciences, Paul L Foster School of Medicine, 5001 El Paso Dr, El Paso, TX 79905, USA
| | - Barbara Yang
- Texas Tech University Health Sciences Center El Paso, Department of Biomedical Sciences, Graduate School of Biomedical Sciences, Paul L Foster School of Medicine, 5001 El Paso Dr, El Paso, TX 79905, USA
| | - Javier Vargas-Medrano
- Texas Tech University Health Sciences Center El Paso, Department of Biomedical Sciences, Graduate School of Biomedical Sciences, Paul L Foster School of Medicine, 5001 El Paso Dr, El Paso, TX 79905, USA
| | - Ruth G Perez
- Texas Tech University Health Sciences Center El Paso, Department of Biomedical Sciences, Graduate School of Biomedical Sciences, Paul L Foster School of Medicine, 5001 El Paso Dr, El Paso, TX 79905, USA.
| |
Collapse
|
35
|
Madill M, Fitzgerald D, O'Connell KE, Dev KK, Shen S, FitzGerald U. In vitro and ex vivo models of multiple sclerosis. Drug Discov Today 2016; 21:1504-1511. [PMID: 27265771 DOI: 10.1016/j.drudis.2016.05.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/20/2016] [Accepted: 05/27/2016] [Indexed: 01/25/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune disorder of the central nervous system (CNS). Current therapies suppress a misdirected myelin-destructive immune response. To combat the progressive, neurodestructive phase of MS, the therapeutic research focus is currently on compounds that might boost the endogenous potential of the brain to remyelinate axons, thereby achieving lesion repair. Here, we describe the testing of fingolimod on cultures of oligodendrocytes (OLs) and organotypic brain slices. We detail the protocols, pros, and cons of these in vitro and ex vivo approaches, along with the potential benefit of exploiting skin-punch biopsies from patients with MS, before concluding with a summary of future developments.
Collapse
Affiliation(s)
- Martin Madill
- Regenerative Medicine Institute (REMEDI), School of Medicine and School of Natural Sciences, National University of Ireland (NUI) Galway, Galway, Ireland
| | - Denise Fitzgerald
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, UK
| | - Kara E O'Connell
- Drug Development, School of Medicine, Trinity College Dublin, Ireland
| | - Kumlesh K Dev
- Drug Development, School of Medicine, Trinity College Dublin, Ireland
| | - Sanbing Shen
- Regenerative Medicine Institute (REMEDI), School of Medicine and School of Natural Sciences, National University of Ireland (NUI) Galway, Galway, Ireland
| | - Una FitzGerald
- Galway Neuroscience Centre, School of Natural Sciences, National University of Ireland, Galway, Ireland.
| |
Collapse
|
36
|
Porcu G, Serone E, De Nardis V, Di Giandomenico D, Lucisano G, Scardapane M, Poma A, Ragnini-Wilson A. Clobetasol and Halcinonide Act as Smoothened Agonists to Promote Myelin Gene Expression and RxRγ Receptor Activation. PLoS One 2015; 10:e0144550. [PMID: 26658258 PMCID: PMC4689554 DOI: 10.1371/journal.pone.0144550] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 11/19/2015] [Indexed: 12/14/2022] Open
Abstract
One of the causes of permanent disability in chronic multiple sclerosis patients is the inability of oligodendrocyte progenitor cells (OPCs) to terminate their maturation program at lesions. To identify key regulators of myelin gene expression acting at the last stages of OPC maturation we developed a drug repositioning strategy based on the mouse immortalized oligodendrocyte (OL) cell line Oli-neu brought to the premyelination stage by stably expressing a key factor regulating the last stages of OL maturation. The Prestwick Chemical Library® of 1,200 FDA-approved compound(s) was repositioned at three dosages based on the induction of Myelin Basic Protein (MBP) expression. Drug hits were further validated using dosage-dependent reproducibility tests and biochemical assays. The glucocorticoid class of compounds was the most highly represented and we found that they can be divided in three groups according to their efficacy on MBP up-regulation. Since target identification is crucial before bringing compounds to the clinic, we searched for common targets of the primary screen hits based on their known chemical-target interactomes, and the pathways predicted by top ranking compounds were validated using specific inhibitors. Two of the top ranking compounds, Halcinonide and Clobetasol, act as Smoothened (Smo) agonists to up-regulate myelin gene expression in the Oli-neuM cell line. Further, RxRγ activation is required for MBP expression upon Halcinonide and Clobetasol treatment. These data indicate Clobetasol and Halcinonide as potential promyelinating drugs and also provide a mechanistic understanding of their mode of action in the pathway leading to myelination in OPCs. Furthermore, our classification of glucocorticoids with respect to MBP expression provides important novel insights into their effects in the CNS and a rational criteria for their choice in combinatorial therapies in de-myelinating diseases.
Collapse
Affiliation(s)
- Giampiero Porcu
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Department of Translational Pharmacology, Fondazione Mario Negri Sud, S. Maria Imbaro (CH), Italy
| | - Eliseo Serone
- Department of Translational Pharmacology, Fondazione Mario Negri Sud, S. Maria Imbaro (CH), Italy
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L’Aquila, Italy
| | - Velia De Nardis
- Department of Translational Pharmacology, Fondazione Mario Negri Sud, S. Maria Imbaro (CH), Italy
| | - Daniele Di Giandomenico
- Department of Translational Pharmacology, Fondazione Mario Negri Sud, S. Maria Imbaro (CH), Italy
| | - Giuseppe Lucisano
- Department of Translational Pharmacology, Fondazione Mario Negri Sud, S. Maria Imbaro (CH), Italy
- Center for Outcomes Research and Clinical Epidemiology, Pescara, Italy
- Dipartimento di Scienze Mediche di Base, Neuroscienze ed Organi di Senso, Università di Bari Aldo Moro, Bari, Italy
| | - Marco Scardapane
- Department of Translational Pharmacology, Fondazione Mario Negri Sud, S. Maria Imbaro (CH), Italy
- Center for Outcomes Research and Clinical Epidemiology, Pescara, Italy
| | - Anna Poma
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L’Aquila, Italy
| | - Antonella Ragnini-Wilson
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Department of Translational Pharmacology, Fondazione Mario Negri Sud, S. Maria Imbaro (CH), Italy
- * E-mail:
| |
Collapse
|
37
|
ŠKOVIEROVÁ H, MAHMOOD S, BLAHOVCOVÁ E, HATOK J, LEHOTSKÝ J, MURÍN R. Effect of Homocysteine on Survival of Human Glial Cells. Physiol Res 2015; 64:747-54. [DOI: 10.33549/physiolres.932897] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Several neurodegenerative conditions, such as Alzheimer’s disease and Parkinson’s disease, or vascular dementia and cognitive impairment, are associated with mild hyperhomocysteinemia. Hyperhomocysteinemia is defined as an increase of the homocysteine (Hcy) level beyond 10 μM. Although the adverse effect of Hcy on neurons is well documented, knowledge about the impact of this amino acid on glial cells is missing. Therefore, with the aim to evaluate the neurotoxic properties of Hcy on glial cells, we used a glioblastoma cell line as a study model. The viability of cells was assayed biochemically and cytologically. At a concentration around 50 μM in the culture medium D,L-Hcy induced cell death. It is noteworthy that Hcy induces cell death of human glial cells at concentrations encountered during mild hyperhomocysteinemia. Therefore, we propose that Hcy-induced impairment of neuronal functions along with damage of glial cells may contribute to the etiopathogenesis of neurodegenerative diseases associated with hyperhomocysteinemia.
Collapse
Affiliation(s)
| | | | | | | | | | - R. MURÍN
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| |
Collapse
|
38
|
Barbosa DJ, Capela JP, de Lourdes Bastos M, Carvalho F. In vitro models for neurotoxicology research. Toxicol Res (Camb) 2015; 4:801-842. [DOI: 10.1039/c4tx00043a] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023] Open
Abstract
The nervous system has a highly complex organization, including many cell types with multiple functions, with an intricate anatomy and unique structural and functional characteristics; the study of its (dys)functionality following exposure to xenobiotics, neurotoxicology, constitutes an important issue in neurosciences.
Collapse
Affiliation(s)
- Daniel José Barbosa
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| | - João Paulo Capela
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| | - Maria de Lourdes Bastos
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| | - Félix Carvalho
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| |
Collapse
|
39
|
Peppard JV, Rugg CA, Smicker MA, Powers E, Harnish E, Prisco J, Cirovic D, Wright PS, August PR, Chandross KJ. High-content phenotypic screening and triaging strategy to identify small molecules driving oligodendrocyte progenitor cell differentiation. ACTA ACUST UNITED AC 2014; 20:382-90. [PMID: 25394729 DOI: 10.1177/1087057114559490] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Multiple Sclerosis is a demyelinating disease of the CNS and the primary cause of neurological disability in young adults. Loss of myelinating oligodendrocytes leads to neuronal dysfunction and death and is an important contributing factor to this disease. Endogenous oligodendrocyte precursor cells (OPCs), which on differentiation are responsible for replacing myelin, are present in the adult CNS. As such, therapeutic agents that can stimulate OPCs to differentiate and remyelinate demyelinated axons under pathologic conditions may improve neuronal function and clinical outcome. We describe the details of an automated, cell-based, morphometric-based, high-content screen that is used to identify small molecules eliciting the differentiation of OPCs after 3 days. Primary screening was performed using rat CG-4 cells maintained in culture conditions that normally support a progenitor cell-like state. From a library of 73,000 diverse small molecules within the Sanofi collection, 342 compounds were identified that increased OPC morphological complexity as an indicator of oligodendrocyte maturation. Subsequent to the primary high-content screen, a suite of cellular assays was established that identified 22 nontoxic compounds that selectively stimulated primary rat OPCs but not C2C12 muscle cell differentiation. This rigorous triaging yielded several chemical series for further expansion and bio- or cheminformatics studies, and their compelling biological activity merits further investigation.
Collapse
Affiliation(s)
- Jane V Peppard
- Lead Generation & Candidate Realization, Sanofi Tucson Innovation Center, Tucson, AZ, USA
| | - Catherine A Rugg
- Lead Generation & Candidate Realization, Sanofi R&D, Bridgewater, NJ, USA
| | | | - Elaine Powers
- Lead Generation & Candidate Realization, Sanofi Tucson Innovation Center, Tucson, AZ, USA
| | - Erica Harnish
- Lead Generation & Candidate Realization, Sanofi Tucson Innovation Center, Tucson, AZ, USA
| | - Joy Prisco
- Lead Generation & Candidate Realization, Sanofi Tucson Innovation Center, Tucson, AZ, USA
| | - Dragan Cirovic
- Lead Generation & Candidate Realization, Sanofi Tucson Innovation Center, Tucson, AZ, USA
| | - Paul S Wright
- Lead Generation & Candidate Realization, Sanofi Tucson Innovation Center, Tucson, AZ, USA
| | - Paul R August
- Lead Generation & Candidate Realization, Sanofi Tucson Innovation Center, Tucson, AZ, USA
| | | |
Collapse
|
40
|
|
41
|
A new protocol for cultivation of predegenerated adult rat Schwann cells. Cell Tissue Bank 2013; 15:403-11. [PMID: 24197905 DOI: 10.1007/s10561-013-9405-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 10/23/2013] [Indexed: 12/13/2022]
Abstract
The purpose of this study was to optimize the methodology of cultivation of predegenerated Schwann cells (SCs). SCs were isolated from 7-day-predegenerated sciatic nerves of adult rats. We applied commercially available culture medium for cultivation of endothelial cells endothelial cell culture medium (EBM-2) instead of Dulbecco's Modified Eagle's Medium commonly used to culture adult Schwann cells. Additionally, cell culture medium was supplemented with factors specifically supporting SCs growth as: bovine pituitary extract (5 μg/ml), heregulin (40 ng/ml) and insulin (2.5 ng/ml). Similarly to the reports of others authors, we did not observe any beneficial effects of Forskolin application, so we didn't supplement our medium with it. Cell culture purity was determined by counting the ratio of GFAP, N-Cadherin and NGFR p75-positive cells to total number of cells. About 94-97 % of cells were confirmed as Schwann cells. As a result, we obtained sufficient number and purity of Schwann cells to be applied in different experimental models in rats. EBM-2 medium coated with fibronectin was the best for cultivation of adult rat Schwann cells.
Collapse
|
42
|
Abstract
Despite the fact that microglia cells were first described almost a century ago, microglia-derived immortalized cell lines have only been established in the last two decades. One should be aware of their limitations but also of their advantages. Cell lines offer a potentially powerful tool to investigate some functional aspects of microglia. Cell culturing of human and murine microglia cell lines will be described in this chapter. It includes a presentation of equipment needed, cell culture medium and supplements, cell culture monitoring, and a protocol describing the steps for subculturing of microglia cell lines.
Collapse
|
43
|
Tulpule K, Schmidt MM, Boecker K, Goldbaum O, Richter-Landsberg C, Dringen R. Formaldehyde induces rapid glutathione export from viable oligodendroglial OLN-93 cells. Neurochem Int 2012; 61:1302-13. [DOI: 10.1016/j.neuint.2012.09.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 09/04/2012] [Accepted: 09/13/2012] [Indexed: 12/29/2022]
|
44
|
Growth-Promoting Effects of Quercetin on Peripheral Nerves in Rats. Int J Artif Organs 2011; 34:1095-105. [DOI: 10.5301/ijao.5000064] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2011] [Indexed: 12/28/2022]
Abstract
Objectives The present study evaluated in vitro and in vivo the effects of quercetin (QC), a major ingredient in various flavonoids, on peripheral nerve regeneration. Methods In the in vitro study, we found that QC at concentrations of 0.1, 1, and 10 μg/mL could significantly promote the survival and outgrowth of cultured Schwann cells as compared with the controls treated with culture medium only. In the in vivo study, we evaluated peripheral nerve regeneration across a 15-mm gap in the sciatic nerve of the rat, using a silicone rubber nerve chamber filled with the QC solution. In the control group, the chambers were filled with normal saline only. Results At the end of 8 weeks, morphometric data revealed that all 3 QC groups significantly increased the count and density of myelinated axons as compared with the controls. Electro-physiological measurements showed that the QC-treated group at 1 μg/mL had a significantly larger area of evoked muscle action potential (MAP) compared with the controls. In addition, the amplitude of the MAP in the QC-treated groups at 0.1 and 1 μg/mL was significantly larger than that in the controls. Conclusions All of these results indicate that QC treatment has nerve growth–promoting effects which may lead to a promising herbal medicine for the recovery of regenerating peripheral nerves.
Collapse
|
45
|
Kipp M, van der Star B, Vogel DYS, Puentes F, van der Valk P, Baker D, Amor S. Experimental in vivo and in vitro models of multiple sclerosis: EAE and beyond. Mult Scler Relat Disord 2011; 1:15-28. [PMID: 25876447 DOI: 10.1016/j.msard.2011.09.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 09/05/2011] [Indexed: 12/16/2022]
Abstract
Although the primary cause of multiple sclerosis (MS) is unknown, the widely accepted view is that aberrant (auto)immune responses possibly arising following infection(s) are responsible for the destructive inflammatory demyelination and neurodegeneration in the central nervous system (CNS). This notion, and the limited access of human brain tissue early in the course of MS, has led to the development of autoimmune, viral and toxin-induced demyelination animal models as well as the development of human CNS cell and organotypic brain slice cultures in an attempt to understand events in MS. The autoimmune models, collectively known as experimental autoimmune encephalomyelitis (EAE), and viral models have shaped ideas of how environmental factors may trigger inflammation, demyelination and neurodegeneration in the CNS. Understandably, these models have also heavily influenced the development of therapies targeting the inflammatory aspect of MS. Demyelination and remyelination in the absence of overt inflammation are better studied in toxin-induced demyelination models using cuprizone and lysolecithin. The paradigm shift of MS as an autoimmune disease of myelin to a neurodegenerative disease has required more appropriate models reflecting the axonal and neuronal damage. Thus, secondary progressive EAE and spastic models have been crucial to develop neuroprotective approaches. In this review the current in vivo and in vitro experimental models to examine pathological mechanisms involved in inflammation, demyelination and neuronal degeneration, as well as remyelination and repair in MS are discussed. Since this knowledge is the basis for the development of new therapeutic approaches for MS, we particularly address whether the currently available models truly reflect the human disease, and discuss perspectives to further optimise and develop more suitable experimental models to study MS.
Collapse
Affiliation(s)
- Markus Kipp
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands; Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Baukje van der Star
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Daphne Y S Vogel
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands; Department of Molecular Cell Biology and Immunology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Fabìola Puentes
- Neuroimmunology Unit, Blizard Institute, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK
| | - Paul van der Valk
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - David Baker
- Neuroimmunology Unit, Blizard Institute, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK
| | - Sandra Amor
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands; Neuroimmunology Unit, Blizard Institute, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK.
| |
Collapse
|
46
|
Dang I, De Vries GH. Aberrant cAMP metabolism in NF1 malignant peripheral nerve sheath tumor cells. Neurochem Res 2011; 36:1697-705. [PMID: 21380540 DOI: 10.1007/s11064-011-0433-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2011] [Indexed: 12/13/2022]
Abstract
Malignant peripheral nerve sheath (MPNST) cell lines derived from patients with neurofibromatosis type 1 (NF!) were found to have basal cAMP levels which are two-fold higher than cAMP levels in normal human adult Schwann cells (nHSC). PCR analysis also revealed that normal adult human Schwann cells express mRNA for types Ill, IV, and IX adenylyl cyclase (AC) while NF1 MPNST cells express AC mRNA of types II, V, and VIII in addition to expressing all the isoforms of normal adult human Schwann cells. Further PCR analysis revealed that NF1 MPNST lines express mRNA for EP2 and EP4 prostaglandin receptors whereas nHSC only express mRNA for the EP2 receptor. Exogenous prostaglandins alone or in combination with PDGF BB induced greater increases in cAMP levels and proliferation of NF1 MPNST cells compared to nHSC. We conclude that aberrant cAMP signaling in NF1 MPNST cells contributes to tumor formation in NF1 patients.
Collapse
Affiliation(s)
- Ian Dang
- Research 151, Hines VA Hospital, Hines, IL 60141, USA.
| | | |
Collapse
|