151
|
Ceder MM, Lekholm E, Klaesson A, Tripathi R, Schweizer N, Weldai L, Patil S, Fredriksson R. Glucose Availability Alters Gene and Protein Expression of Several Newly Classified and Putative Solute Carriers in Mice Cortex Cell Culture and D. melanogaster. Front Cell Dev Biol 2020; 8:579. [PMID: 32733888 PMCID: PMC7358622 DOI: 10.3389/fcell.2020.00579] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022] Open
Abstract
Many newly identified solute carriers (SLCs) and putative transporters have the possibility to be intricately involved in glucose metabolism. Here we show that many transporters of this type display a high degree of regulation at both mRNA and protein level following no or low glucose availability in mouse cortex cultures. We show that this is also the case in Drosophila melanogaster subjected to starvation or diets with different sugar content. Interestingly, re-introduction of glucose to media, or refeeding flies, normalized the gene expression of a number of the targets, indicating a fast and highly dynamic control. Our findings demonstrate high conservation of these transporters and how dependent both cell cultures and organisms are on gene and protein regulation during metabolic fluctuations. Several transporter genes were regulated simultaneously maybe to initiate alternative metabolic pathways as a response to low glucose levels, both in the cell cultures and in D. melanogaster. Our results display that newly identified SLCs of Major Facilitator Superfamily type, as well as the putative transporters included in our study, are regulated by glucose availability and could be involved in several cellular aspects dependent of glucose and/or its metabolites. Recently, a correlation between dysregulation of glucose in the central nervous system and numerous diseases such as obesity, type 2 diabetes mellitus as well as neurological disease such as Alzheimer’s and Parkinson’s diseases indicate a complex regulation and fine tuning of glucose levels in the brain. The fact that almost one third of transporters and transporter-related proteins remain orphans with unknown or contradictive substrate profile, location and function, pinpoint the need for further research about them to fully understand their mechanistic role and their impact on cellular metabolism.
Collapse
Affiliation(s)
- Mikaela M Ceder
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Emilia Lekholm
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Axel Klaesson
- Pharmaceutical Cell Biology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Rekha Tripathi
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Nadine Schweizer
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Lydia Weldai
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Sourabh Patil
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Robert Fredriksson
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
152
|
Gläser A, Hammerl F, Gräler MH, Coldewey SM, Völkner C, Frech MJ, Yang F, Luo J, Tönnies E, von Bohlen und Halbach O, Brandt N, Heimes D, Neßlauer AM, Korenke GC, Owczarek-Lipska M, Neidhardt J, Rolfs A, Wree A, Witt M, Bräuer AU. Identification of Brain-Specific Treatment Effects in NPC1 Disease by Focusing on Cellular and Molecular Changes of Sphingosine-1-Phosphate Metabolism. Int J Mol Sci 2020; 21:ijms21124502. [PMID: 32599915 PMCID: PMC7352403 DOI: 10.3390/ijms21124502] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 12/17/2022] Open
Abstract
Niemann-Pick type C1 (NPC1) is a lysosomal storage disorder, inherited as an autosomal-recessive trait. Mutations in the Npc1 gene result in malfunction of the NPC1 protein, leading to an accumulation of unesterified cholesterol and glycosphingolipids. Beside visceral symptoms like hepatosplenomegaly, severe neurological symptoms such as ataxia occur. Here, we analyzed the sphingosine-1-phosphate (S1P)/S1P receptor (S1PR) axis in different brain regions of Npc1-/- mice and evaluated specific effects of treatment with 2-hydroxypropyl-β-cyclodextrin (HPβCD) together with the iminosugar miglustat. Using high-performance thin-layer chromatography (HPTLC), mass spectrometry, quantitative real-time PCR (qRT-PCR) and western blot analyses, we studied lipid metabolism in an NPC1 mouse model and human skin fibroblasts. Lipid analyses showed disrupted S1P metabolism in Npc1-/- mice in all brain regions, together with distinct changes in S1pr3/S1PR3 and S1pr5/S1PR5 expression. Brains of Npc1-/- mice showed only weak treatment effects. However, side effects of the treatment were observed in Npc1+/+ mice. The S1P/S1PR axis seems to be involved in NPC1 pathology, showing only weak treatment effects in mouse brain. S1pr expression appears to be affected in human fibroblasts, induced pluripotent stem cells (iPSCs)-derived neural progenitor and neuronal differentiated cells. Nevertheless, treatment-induced side effects make examination of further treatment strategies indispensable.
Collapse
Affiliation(s)
- Anne Gläser
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany; (A.G.); (F.H.); (N.B.)
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany; (D.H.); (A.-M.N.); (A.W.); (M.W.)
| | - Franziska Hammerl
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany; (A.G.); (F.H.); (N.B.)
| | - Markus H. Gräler
- Department of Anaesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care (CSCC), Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745 Jena, Germany;
| | - Sina M. Coldewey
- Department of Anaesthesiology and Intensive Care Medicine, Septomics Research Center, Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany;
| | - Christin Völkner
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany; (C.V.); (M.J.F.); (F.Y.); (J.L.)
| | - Moritz J. Frech
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany; (C.V.); (M.J.F.); (F.Y.); (J.L.)
- Center for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Center, University of Rostock, 18147 Rostock, Germany
| | - Fan Yang
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany; (C.V.); (M.J.F.); (F.Y.); (J.L.)
| | - Jiankai Luo
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany; (C.V.); (M.J.F.); (F.Y.); (J.L.)
- Center for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Center, University of Rostock, 18147 Rostock, Germany
| | - Eric Tönnies
- Institute of Anatomy and Cell Biology, University Medicine Greifswald, 17487 Greifswald, Germany; (E.T.); (O.v.B.u.H.)
| | - Oliver von Bohlen und Halbach
- Institute of Anatomy and Cell Biology, University Medicine Greifswald, 17487 Greifswald, Germany; (E.T.); (O.v.B.u.H.)
| | - Nicola Brandt
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany; (A.G.); (F.H.); (N.B.)
| | - Diana Heimes
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany; (D.H.); (A.-M.N.); (A.W.); (M.W.)
| | - Anna-Maria Neßlauer
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany; (D.H.); (A.-M.N.); (A.W.); (M.W.)
| | | | - Marta Owczarek-Lipska
- Human Genetics, School of Medicine and Health Sciences, University of Oldenburg, 26129 Oldenburg, Germany; (M.O.-L.); (J.N.)
- Junior Research Group, Genetics of childhood brain malformations, School of Medicine and Health Sciences, University of Oldenburg, 26129 Oldenburg, Germany
| | - John Neidhardt
- Human Genetics, School of Medicine and Health Sciences, University of Oldenburg, 26129 Oldenburg, Germany; (M.O.-L.); (J.N.)
- Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg,26129 Oldenburg, Germany
| | | | - Andreas Wree
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany; (D.H.); (A.-M.N.); (A.W.); (M.W.)
| | - Martin Witt
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany; (D.H.); (A.-M.N.); (A.W.); (M.W.)
| | - Anja Ursula Bräuer
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany; (A.G.); (F.H.); (N.B.)
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany; (D.H.); (A.-M.N.); (A.W.); (M.W.)
- Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg,26129 Oldenburg, Germany
- Correspondence: ; Tel.: +49-441-798-3995
| |
Collapse
|
153
|
Wang J, Chen Y, Bai Y, Quan D, Wang Z, Xiong L, Shao Z, Sun W, Mi S. A core-skirt designed artificial cornea with orthogonal microfiber grid scaffold. Exp Eye Res 2020; 195:108037. [DOI: 10.1016/j.exer.2020.108037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 04/03/2020] [Accepted: 04/08/2020] [Indexed: 11/29/2022]
|
154
|
Casillas‐Espinosa PM, Ali I, O'Brien TJ. Neurodegenerative pathways as targets for acquired epilepsy therapy development. Epilepsia Open 2020; 5:138-154. [PMID: 32524040 PMCID: PMC7278567 DOI: 10.1002/epi4.12386] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/13/2020] [Accepted: 02/24/2020] [Indexed: 12/16/2022] Open
Abstract
There is a growing body of clinical and experimental evidence that neurodegenerative diseases and epileptogenesis after an acquired brain insult may share common etiological mechanisms. Acquired epilepsy commonly develops as a comorbid condition in patients with neurodegenerative diseases such as Alzheimer's disease, although it is likely much under diagnosed in practice. Progressive neurodegeneration has also been described after traumatic brain injury, stroke, and other forms of brain insults. Moreover, recent evidence has shown that acquired epilepsy is often a progressive disorder that is associated with the development of drug resistance, cognitive decline, and worsening of other neuropsychiatric comorbidities. Therefore, new pharmacological therapies that target neurobiological pathways that underpin neurodegenerative diseases have potential to have both an anti-epileptogenic and disease-modifying effect on the seizures in patients with acquired epilepsy, and also mitigate the progressive neurocognitive and neuropsychiatric comorbidities. Here, we review the neurodegenerative pathways that are plausible targets for the development of novel therapies that could prevent the development or modify the progression of acquired epilepsy, and the supporting published experimental and clinical evidence.
Collapse
Affiliation(s)
- Pablo M. Casillas‐Espinosa
- Departments of Neuroscience and MedicineCentral Clinical SchoolMonash UniversityMelbourneVic.Australia
- Department of MedicineThe Royal Melbourne HospitalThe University of MelbourneMelbourneVic.Australia
| | - Idrish Ali
- Departments of Neuroscience and MedicineCentral Clinical SchoolMonash UniversityMelbourneVic.Australia
- Department of MedicineThe Royal Melbourne HospitalThe University of MelbourneMelbourneVic.Australia
| | - Terence J. O'Brien
- Departments of Neuroscience and MedicineCentral Clinical SchoolMonash UniversityMelbourneVic.Australia
- Department of MedicineThe Royal Melbourne HospitalThe University of MelbourneMelbourneVic.Australia
- Department of NeurologyThe Alfred HospitalMelbourneVic.Australia
- Department of NeurologyThe Royal Melbourne HospitalParkvilleVic.Australia
| |
Collapse
|
155
|
Mai N, Prifti V, Kim M, Halterman MW. Characterization of neutrophil-neuronal co-cultures to investigate mechanisms of post-ischemic immune-mediated neurotoxicity. J Neurosci Methods 2020; 341:108782. [PMID: 32445795 DOI: 10.1016/j.jneumeth.2020.108782] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 04/23/2020] [Accepted: 05/12/2020] [Indexed: 01/28/2023]
Abstract
BACKGROUND Immune-mediated reperfusion injury is a critical component of post-ischemic central nervous system (CNS) damage. In this context, the activation and recruitment of polymorphonuclear neutrophils (PMNs) to the CNS induces neurotoxicity in part through the release of degradative enzymes, cytokines, and reactive oxygen species. However, the extent to which close-range interactions between PMNs and neurons contribute to injury in this context has not been directly investigated. NEW METHOD We devised a co-culture model to investigate mechanisms of PMN-dependent neurotoxicity. Specifically, we established the effect of PMN dose, co-incident neuronal ischemia, lipopolysaccharide (LPS)-induced PMN priming, and the requirement for cell-cell contact on cumulative neuron damage. RESULTS AND COMPARISON TO EXISTING METHOD(S) Pre-exposure of day in vitro 10 primary cortical neurons to oxygen-glucose deprivation (OGD) enhanced PMN-dependent neuronal death. Likewise, LPS-induced priming of the PMN donor further increased PMN-induced toxicity in vitro compared to saline-injected controls. Compartmentalization of LPS-primed PMNs using net wells confirmed the requirement for close-range cell-cell interactions in the process of PMN-induced neuronal injury. Moreover, time-lapse imaging and quantitative neurite analyses implicate PMN-neurite interactions in this pathological response. These experiments establish a platform to investigate immune and neural factors that contribute to post-ischemic neurodegeneration. CONCLUSIONS Ischemic and immune priming enhance neurotoxicity in PMN-neuronal co-cultures. Moreover, cell-cell contact and neurite destruction are prominent features in the observed mechanism of post-ischemic neuronal death.
Collapse
Affiliation(s)
- Nguyen Mai
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester NY 14642, United States.
| | - Viollandi Prifti
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester NY 14642, United States.
| | - Minsoo Kim
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester NY 14642, United States.
| | - Marc W Halterman
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester NY 14642, United States; Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester NY 14642, United States.
| |
Collapse
|
156
|
Cheng J, Tang JC, Pan MX, Chen SF, Zhao D, Zhang Y, Liao HB, Zhuang Y, Lei RX, Wang S, Liu AC, Chen J, Zhang ZH, Li HT, Wan Q, Chen QX. l-lysine confers neuroprotection by suppressing inflammatory response via microRNA-575/PTEN signaling after mouse intracerebral hemorrhage injury. Exp Neurol 2020; 327:113214. [DOI: 10.1016/j.expneurol.2020.113214] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 01/10/2020] [Accepted: 01/24/2020] [Indexed: 10/25/2022]
|
157
|
Godoy-Parejo C, Deng C, Zhang Y, Liu W, Chen G. Roles of vitamins in stem cells. Cell Mol Life Sci 2020; 77:1771-1791. [PMID: 31676963 PMCID: PMC11104807 DOI: 10.1007/s00018-019-03352-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/12/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022]
Abstract
Stem cells can differentiate to diverse cell types in our body, and they hold great promises in both basic research and clinical therapies. For specific stem cell types, distinctive nutritional and signaling components are required to maintain the proliferation capacity and differentiation potential in cell culture. Various vitamins play essential roles in stem cell culture to modulate cell survival, proliferation and differentiation. Besides their common nutritional functions, specific vitamins are recently shown to modulate signal transduction and epigenetics. In this article, we will first review classical vitamin functions in both somatic and stem cell cultures. We will then focus on how stem cells could be modulated by vitamins beyond their nutritional roles. We believe that a better understanding of vitamin functions will significantly benefit stem cell research, and help realize their potentials in regenerative medicine.
Collapse
Affiliation(s)
- Carlos Godoy-Parejo
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Chunhao Deng
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Yumeng Zhang
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Weiwei Liu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
- Bioimaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Guokai Chen
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.
- Bioimaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.
| |
Collapse
|
158
|
Moutin E, Hemonnot AL, Seube V, Linck N, Rassendren F, Perroy J, Compan V. Procedures for Culturing and Genetically Manipulating Murine Hippocampal Postnatal Neurons. Front Synaptic Neurosci 2020; 12:19. [PMID: 32425766 PMCID: PMC7204911 DOI: 10.3389/fnsyn.2020.00019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/03/2020] [Indexed: 12/15/2022] Open
Abstract
Neuronal hippocampal cultures are simple and valuable models for studying neuronal function. While embryonic cultures are widely used for different applications, mouse postnatal cultures are still challenging, lack reproducibility and/or exhibit inappropriate neuronal activity. Yet, postnatal cultures have major advantages such as allowing genotyping of pups before culture and reducing the number of experimental animals. Herein we describe a simple and fast protocol for culturing and genetically manipulating hippocampal neurons from P0 to P3 mice. This protocol provides reproducible cultures exhibiting a consistent neuronal development, normal excitatory over inhibitory neuronal ratio and a physiological neuronal activity. We also describe simple and efficient procedures for genetic manipulation of neurons using transfection reagent or lentiviral particles. Overall, this method provides a detailed and validated protocol allowing to explore cellular mechanisms and neuronal activity in postnatal hippocampal neurons in culture.
Collapse
Affiliation(s)
- Enora Moutin
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Anne-Laure Hemonnot
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France.,Laboratoire d'Excellence Canaux Ioniques d'Intérêt Thérapeutique (LabEx ICST), Montpellier, France
| | - Vincent Seube
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France.,Laboratoire d'Excellence Canaux Ioniques d'Intérêt Thérapeutique (LabEx ICST), Montpellier, France
| | - Nathalie Linck
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France.,Laboratoire d'Excellence Canaux Ioniques d'Intérêt Thérapeutique (LabEx ICST), Montpellier, France
| | - François Rassendren
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France.,Laboratoire d'Excellence Canaux Ioniques d'Intérêt Thérapeutique (LabEx ICST), Montpellier, France
| | - Julie Perroy
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Vincent Compan
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France.,Laboratoire d'Excellence Canaux Ioniques d'Intérêt Thérapeutique (LabEx ICST), Montpellier, France
| |
Collapse
|
159
|
A Simplified, Fully Defined Differentiation Scheme for Producing Blood-Brain Barrier Endothelial Cells from Human iPSCs. Stem Cell Reports 2020; 12:1380-1388. [PMID: 31189096 PMCID: PMC6565873 DOI: 10.1016/j.stemcr.2019.05.008] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 12/15/2022] Open
Abstract
Human induced pluripotent stem cell (iPSC)-derived developmental lineages are key tools for in vitro mechanistic interrogations, drug discovery, and disease modeling. iPSCs have previously been differentiated to endothelial cells with blood-brain barrier (BBB) properties, as defined by high transendothelial electrical resistance (TEER), low passive permeability, and active transporter functions. Typical protocols use undefined components, which impart unacceptable variability on the differentiation process. We demonstrate that replacement of serum with fully defined components, from common medium supplements to a simple mixture of insulin, transferrin, and selenium, yields BBB endothelium with TEER in the range of 2,000-8,000 Ω × cm2 across multiple iPSC lines, with appropriate marker expression and active transporters. The use of a fully defined medium vastly improves the consistency of differentiation, and co-culture of BBB endothelium with iPSC-derived astrocytes produces a robust in vitro neurovascular model. This defined differentiation scheme should broadly enable the use of human BBB endothelium for diverse applications.
Collapse
|
160
|
The Dynein Adaptor RILP Controls Neuronal Autophagosome Biogenesis, Transport, and Clearance. Dev Cell 2020; 53:141-153.e4. [PMID: 32275887 DOI: 10.1016/j.devcel.2020.03.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 12/30/2019] [Accepted: 03/12/2020] [Indexed: 12/31/2022]
Abstract
Autophagy plays critical roles in neurodegeneration and development, but how this pathway is organized and regulated in neurons remains poorly understood. Here, we find that the dynein adaptor RILP is essential for retrograde transport of neuronal autophagosomes, and surprisingly, their biogenesis as well. We find that induction of autophagy by mTOR inhibition specifically upregulates RILP expression and its localization to autophagosomes. RILP depletion or mutations in its LC3-binding LIR motifs strongly decrease autophagosome numbers suggesting an unexpected RILP role in autophagosome biogenesis. We find that RILP also interacts with ATG5 on isolation membranes, precluding premature dynein recruitment and autophagosome transport. RILP inhibition impedes autophagic turnover and causes p62/sequestosome-1 aggregation. Together, our results identify an mTOR-responsive neuronal autophagy pathway, wherein RILP integrates the processes of autophagosome biogenesis and retrograde transport to control autophagic turnover. This pathway has important implications for understanding how autophagy contributes to neuronal function, development, and disease.
Collapse
|
161
|
Xu J, Lian W, Chen J, Li W, Li L, Huang Z. Chemical-defined medium supporting the expansion of human mesenchymal stem cells. Stem Cell Res Ther 2020; 11:125. [PMID: 32192530 PMCID: PMC7083066 DOI: 10.1186/s13287-020-01641-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/03/2020] [Accepted: 03/10/2020] [Indexed: 12/04/2022] Open
Abstract
OBJECTIVES Mesenchymal stem cells (MSCs) have been intensively investigated as to their therapeutic potentials. However, the full chemical-defined medium supporting the isolation and expansion of human MSCs has not been developed yet. MATERIALS AND METHODS Here, we developed the full chemical-defined medium, NBVbe medium, via RNA sequencing, bioinformatic analysis, and growth factor screening. RESULTS The NBVbe medium contains N2B27 medium with the BSA (bovine serum albumin) replaced by the recombinant human albumin, bFGF (basic fibroblast growth factor), vitamin C, and EGF (epidermal growth factor). The NBVbe medium could support the isolation and expansion of human MSCs from the umbilical cords. CONCLUSIONS The full chemical-defined medium supporting the isolation and expansion of human MSCs has been developed. This would be helpful for further optimization of the MSC medium, their clinical applications, and molecular characterization.
Collapse
Affiliation(s)
- Jianyong Xu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, School of Medicine, Shenzhen University, Nanhai Avenue 3688, Shenzhen, 518060, Guangdong, People's Republic of China.
| | - Wei Lian
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, School of Medicine, Shenzhen University, Nanhai Avenue 3688, Shenzhen, 518060, Guangdong, People's Republic of China
| | - Jieting Chen
- Department of Obstetrics, People's Hospital of Baoan, Shenzhen, 518055, People's Republic of China
| | - Wenlei Li
- Department of Obstetrics, Women and Children Health Institute of Futian, Shenzhen, 518055, People's Republic of China
| | - Lingyun Li
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, School of Medicine, Shenzhen University, Nanhai Avenue 3688, Shenzhen, 518060, Guangdong, People's Republic of China
| | - Zhong Huang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, School of Medicine, Shenzhen University, Nanhai Avenue 3688, Shenzhen, 518060, Guangdong, People's Republic of China
| |
Collapse
|
162
|
Tang W, Fang F, Liu K, Huang Z, Li H, Yin Y, Wang J, Wang G, Wei L, Ou Y, Wang Y. Aligned Biofunctional Electrospun PLGA-LysoGM1 Scaffold for Traumatic Brain Injury Repair. ACS Biomater Sci Eng 2020; 6:2209-2218. [PMID: 33455302 DOI: 10.1021/acsbiomaterials.9b01636] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Due to poor regenerative capabilities of the brain, a treatment for traumatic brain injury (TBI) presents a serious challenge to modern medicine. Biofunctional scaffolds that can support neuronal growth, guide neurite elongation, and re-establish impaired brain tissues are urgently needed. To this end, we developed an aligned biofunctional scaffold (aPLGA-LysoGM1), in which poly (lactic-co-glycolic acid) (PLGA) was functionalized with sphingolipid ceramide N-deacylase (SCDase)-hydrolyzed monosialotetrahexosylganglioside (LysoGM1) and electrospinning was used to form an aligned fibrous network. As a ganglioside of neuronal membranes, the functionalized LysoGM1 endows the scaffold with unique biological properties favoring the growth of neuron and regeneration of injured brain tissues. Moreover, we found that the aligned PLGA-LysoGM1 fibers acted as a topographical cue to guide neurite extension, which is critical for organizing the formation of synaptic networks (neural networks). Systematic in vitro studies demonstrated that the aligned biofunctional scaffold promotes neuronal viability, neurite outgrowth, and synapse formation and also protects neurons from pressure-related injury. Additionally, in a rat TBI model, we demonstrated that the implantation of aPLGA-LysoGM1 scaffold supported recovery from brain injury, as more endogenous neurons were found to migrate and infiltrate into the defect zone compared with alternative scaffold. These results suggest that the aligned biofunctional aPLGA-LysoGM1 scaffold represents a promising therapeutic strategy for brain tissue regeneration following TBI.
Collapse
Affiliation(s)
- Wei Tang
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Fei Fang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Ke Liu
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhi Huang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Hui Li
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ying Yin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jun Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Guocheng Wang
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liyu Wei
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yun Ou
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,Hunan Provincial Key Laboratory of Health Maintenance for Mechanical Equipment, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Yazhou Wang
- School of Medicine, Chongqing University, Chongqing 400044, China.,Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| |
Collapse
|
163
|
Zhang L, Yu H, Yuan Y, Yu JS, Lou Z, Xue Y, Liu Y. The necessity for standardization of glioma stem cell culture: a systematic review. Stem Cell Res Ther 2020; 11:84. [PMID: 32102678 PMCID: PMC7045630 DOI: 10.1186/s13287-020-01589-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/15/2019] [Accepted: 02/06/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The cancer stem cell hypothesis is an old idea which has been revived in recent years for many cancers, including gliomas. However, this concept has become controversial due to a series of studies with conflicting results. METHODS A systematic literature search was conducted in PubMed and the Web of Science database to analyze studies using serum-free medium and its components in glioma stem cells, glioma stem-like cells, glioma-initiating cells, or glioma neurosphere cultures. All the studies reviewed were published between 1970 and 2019. We found that no standardized culture method was used, and the data were incomparable due to differing culture conditions and the use of media with different components. CONCLUSIONS Here, we review the most commonly used serum-free media and added components for glioma stem cell culture while highlighting the function of each component used in the media. We emphasize the necessity for standardization of glioma stem cell culture and propose a standard culture medium to prevent bias in glioma stem cell research.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, # 36 Sanhao Street, Heping District, Shenyang, China.,Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, USA.,Department of Oncology, Mayo Clinic, Rochester, USA
| | - Hongwei Yu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, # 36 Sanhao Street, Heping District, Shenyang, China
| | - Yuhui Yuan
- Department of Neurosurgery, Shengjing Hospital of China Medical University, # 36 Sanhao Street, Heping District, Shenyang, China
| | - John S Yu
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Zhenkun Lou
- Department of Oncology, Mayo Clinic, Rochester, USA
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, # 36 Sanhao Street, Heping District, Shenyang, China.
| |
Collapse
|
164
|
Zhong Q, Zou Y, Liu H, Chen T, Zheng F, Huang Y, Chen C, Zhang Z. Toll-like receptor 4 deficiency ameliorates β2-microglobulin induced age-related cognition decline due to neuroinflammation in mice. Mol Brain 2020; 13:20. [PMID: 32059688 PMCID: PMC7023753 DOI: 10.1186/s13041-020-0559-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 01/21/2020] [Indexed: 12/13/2022] Open
Abstract
Toll-like receptor 4 (TLR4) is a crucial receptor in neuroinflammation and apoptotic neuronal death, and increasing evidences indicated that β2-microglobulin (B2M) is thought to be a major contributor to age-related cognitive decline. In present study, we designed to investigate the effects of TLR4 on B2M-induced age-related cognitive decline. Wild-type (WT) C57BL/6, TLR4 knockout (TLR4 -KO) mice and hippocampal neurons from the two type mice were respectively divided into two groups: (1) Veh group; (2) B2M-treated group. The behavioral responses of mice were measured using Morris Water Maze. Hippocampal neurogenesis and neuronal damage, inflammatory response, apoptosis, synaptic proteins and neurotrophic factors, and TLR4/MyD88/NF-κB signaling pathway proteins were examined using molecular biological or histopathological methods. The results showed that WT mice received B2M in the DG exhibited age-related cognitive declines, increased TLR4 mRNA expression and high levels of interleukin-1β (IL-1β), tumor necrosis factor-alpha (TNF-α) and apoptotic neuronal death in the hippocampus, which were partially attenuated in TLR4-KO mice. Moreover, in absence of TLR4, B2M treatment improved hippocampus neurogenesis and increased synaptic related proteins. Our cell experiments further demonstrated that deletion of TLR4 could significantly increase synaptic related protein, decrease neuroinflammatory fators, inhibited apoptotic neuronal death, and regulated MyD88/NF-κB signal pathway after B2M treatment. In summary, our results support the TLR4 contributes to B2M-induced age-related cognitive decline due to neuroinflammation and apoptosis through TLR4/MyD88/NF-κB signaling pathway via a modulation of hippocampal neurogenesis and synaptic function. This may provide an important neuroprotective mechanism for improving age-related cognitive decline.
Collapse
Affiliation(s)
- Qi Zhong
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Yufeng Zou
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Hongchao Liu
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
- Department of Anesthesiology, Maternal and Child Hospital of Hubei Province, Wuluo Road, Wuhan, 430071, Hubei, China
| | - Ting Chen
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Feng Zheng
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Yifei Huang
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Chang Chen
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China.
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China.
| |
Collapse
|
165
|
In vitro modeling of dendritic atrophy in Rett syndrome: determinants for phenotypic drug screening in neurodevelopmental disorders. Sci Rep 2020; 10:2491. [PMID: 32051524 PMCID: PMC7016139 DOI: 10.1038/s41598-020-59268-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 01/21/2020] [Indexed: 01/16/2023] Open
Abstract
Dendritic atrophy, defined as the reduction in complexity of the neuronal arborization, is a hallmark of several neurodevelopmental disorders, including Rett Syndrome (RTT). RTT, affecting 1:10,000 girls worldwide, is mainly caused by mutations in the MECP2 gene and has no cure. We describe here an in vitro model of dendritic atrophy in Mecp2−/y mouse hippocampal primary cultures, suitable for phenotypic drug-screening. Using High-Content Imaging techniques, we systematically investigated the impact of culturing determinants on several parameters such as neuronal survival, total dendritic length, dendritic endpoints, soma size, cell clusterization, spontaneous activity. Determinants included cell-seeding density, glass or polystyrene substrates, coating with poly-Ornithine with/without Matrigel and miniaturization from 24 to 96-half surface multiwell plates. We show that in all plate-sizes at densities below 320 cells/mm2, morphological parameters remained constant while spontaneous network activity decreased according to the cell-density. Mecp2−/y neurons cultured at 160 cells/mm2 density in 96 multiwell plates, displayed significant dendritic atrophy and showed a marked increase in dendritic length following treatment with Brain-derived neurotrophic factor (BDNF) or Mirtazapine. In conclusion, we have established a phenotypic assay suitable for fast screening of hundreds of compounds, which may be extended to other neurodevelopmental diseases with dendritic atrophy.
Collapse
|
166
|
Wamaitha SE, Grybel KJ, Alanis-Lobato G, Gerri C, Ogushi S, McCarthy A, Mahadevaiah SK, Healy L, Lea RA, Molina-Arcas M, Devito LG, Elder K, Snell P, Christie L, Downward J, Turner JMA, Niakan KK. IGF1-mediated human embryonic stem cell self-renewal recapitulates the embryonic niche. Nat Commun 2020; 11:764. [PMID: 32034154 PMCID: PMC7005693 DOI: 10.1038/s41467-020-14629-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 01/23/2020] [Indexed: 02/05/2023] Open
Abstract
Our understanding of the signalling pathways regulating early human development is limited, despite their fundamental biological importance. Here, we mine transcriptomics datasets to investigate signalling in the human embryo and identify expression for the insulin and insulin growth factor 1 (IGF1) receptors, along with IGF1 ligand. Consequently, we generate a minimal chemically-defined culture medium in which IGF1 together with Activin maintain self-renewal in the absence of fibroblast growth factor (FGF) signalling. Under these conditions, we derive several pluripotent stem cell lines that express pluripotency-associated genes, retain high viability and a normal karyotype, and can be genetically modified or differentiated into multiple cell lineages. We also identify active phosphoinositide 3-kinase (PI3K)/AKT/mTOR signalling in early human embryos, and in both primed and naïve pluripotent culture conditions. This demonstrates that signalling insights from human blastocysts can be used to define culture conditions that more closely recapitulate the embryonic niche.
Collapse
Affiliation(s)
- Sissy E Wamaitha
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Department of Molecular, Cell and Developmental Biology, and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, 90095, USA
| | - Katarzyna J Grybel
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Gregorio Alanis-Lobato
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Claudia Gerri
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Sugako Ogushi
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Afshan McCarthy
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | | | - Lyn Healy
- Human Embryo and Stem Cell Unit, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Rebecca A Lea
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Miriam Molina-Arcas
- Oncogene Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Liani G Devito
- Human Embryo and Stem Cell Unit, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Kay Elder
- Bourn Hall Clinic, Bourn, Cambridge, CB23 2TN, UK
| | - Phil Snell
- Bourn Hall Clinic, Bourn, Cambridge, CB23 2TN, UK
| | | | - Julian Downward
- Oncogene Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - James M A Turner
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Kathy K Niakan
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
167
|
Zhang Y, Venkateswaran S, Higuera GA, Nath S, Shpak G, Matray J, Fratila-Apachitei LE, Zadpoor AA, Kushner SA, Bradley M, De Zeeuw CI. Synthetic Polymers Provide a Robust Substrate for Functional Neuron Culture. Adv Healthc Mater 2020; 9:e1901347. [PMID: 31943855 DOI: 10.1002/adhm.201901347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/28/2019] [Indexed: 12/11/2022]
Abstract
Substrates for neuron culture and implantation are required to be both biocompatible and display surface compositions that support cell attachment, growth, differentiation, and neural activity. Laminin, a naturally occurring extracellular matrix protein is the most widely used substrate for neuron culture and fulfills some of these requirements, however, it is expensive, unstable (compared to synthetic materials), and prone to batch-to-batch variation. This study uses a high-throughput polymer screening approach to identify synthetic polymers that supports the in vitro culture of primary mouse cerebellar neurons. This allows the identification of materials that enable primary cell attachment with high viability even under "serum-free" conditions, with materials that support both primary cells and neural progenitor cell attachment with high levels of neuronal biomarker expression, while promoting progenitor cell maturation to neurons.
Collapse
Affiliation(s)
- Yichuan Zhang
- School of Chemistry, Kings Buildings, The University of Edinburgh, Edinburgh, EH9 3FJ, UK
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | | | - Gustavo A Higuera
- Department of Neuroscience, Erasmus MC Rotterdam, Rotterdam, NL-3015 GE, The Netherlands
| | - Suvra Nath
- Department of Biomechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628CD, Delft, The Netherlands
| | - Guy Shpak
- Department of Psychiatry, Erasmus MC Rotterdam, Rotterdam, NL-3015 GE, The Netherlands
- Department of Life Sciences, Erasmus University College, Rotterdam, 3011 HP, The Netherlands
| | - Jeffrey Matray
- Department of Biomechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628CD, Delft, The Netherlands
| | - Lidy E Fratila-Apachitei
- Department of Biomechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628CD, Delft, The Netherlands
| | - Amir A Zadpoor
- Department of Biomechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628CD, Delft, The Netherlands
| | - Steven A Kushner
- Department of Biomechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628CD, Delft, The Netherlands
| | - Mark Bradley
- School of Chemistry, Kings Buildings, The University of Edinburgh, Edinburgh, EH9 3FJ, UK
| | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus MC Rotterdam, Rotterdam, NL-3015 GE, The Netherlands
- Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, 1105 BA, The Netherlands
| |
Collapse
|
168
|
Yin L, Hu P, Shi X, Qian W, Zhau HE, Pandol SJ, Lewis MS, Chung LWK, Wang R. Cancer cell's neuroendocrine feature can be acquired through cell-cell fusion during cancer-neural stem cell interaction. Sci Rep 2020; 10:1216. [PMID: 31988304 PMCID: PMC6985266 DOI: 10.1038/s41598-020-58118-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/10/2020] [Indexed: 02/04/2023] Open
Abstract
Advanced and therapy-resistant prostate tumors often display neural or neuroendocrine behavior. We assessed the consequences of prostate cancer cell interaction with neural cells, which are rich in the human prostate and resident of the prostate tumor. In 3-dimensional co-culture with neurospheres, red fluorescent human LNCaP cells formed agglomerates on the neurosphere surface. Upon induced neural differentiation, some red fluorescent cells showed morphology of fully differentiated neural cells, indicating fusion between the cancer and neural stem cells. These fusion hybrids survived for extended times in a quiescent state. A few eventually restarted cell division and propagated to form derivative hybrid progenies. Clones of the hybrid progenies were highly heterogeneous; most had lost prostatic and epithelial markers while some had acquired neural marker expression. These results indicate that cancer cells can fuse with bystander neural cells in the tumor microenvironment; and cancer cell fusion is a direct route to tumor cell heterogeneity.
Collapse
Affiliation(s)
- Liyuan Yin
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Uro-Oncology Research, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Peizhen Hu
- Uro-Oncology Research, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Xianping Shi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Weiping Qian
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Haiyen E Zhau
- Uro-Oncology Research, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephen J Pandol
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael S Lewis
- Department of Pathology, Greater Los Angeles Veterans Affairs Health System, Los Angeles, CA, USA
| | - Leland W K Chung
- Uro-Oncology Research, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ruoxiang Wang
- Uro-Oncology Research, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Pathology, Greater Los Angeles Veterans Affairs Health System, Los Angeles, CA, USA.
| |
Collapse
|
169
|
Ali SR, Malone TJ, Zhang Y, Prechova M, Kaczmarek LK. Phactr1 regulates Slack (KCNT1) channels via protein phosphatase 1 (PP1). FASEB J 2020; 34:1591-1601. [PMID: 31914597 PMCID: PMC6956700 DOI: 10.1096/fj.201902366r] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 12/14/2022]
Abstract
The Slack (KCNT1) gene encodes sodium-activated potassium channels that are abundantly expressed in the central nervous system. Human mutations alter the function of Slack channels, resulting in epilepsy and intellectual disability. Most of the disease-causing mutations are located in the extended cytoplasmic C-terminus of Slack channels and result in increased Slack current. Previous experiments have shown that the C-terminus of Slack channels binds a number of cytoplasmic signaling proteins. One of these is Phactr1, an actin-binding protein that recruits protein phosphatase 1 (PP1) to certain phosphoprotein substrates. Using co-immunoprecipitation, we found that Phactr1 is required to link the channels to actin. Using patch clamp recordings, we found that co-expression of Phactr1 with wild-type Slack channels reduces the current amplitude but has no effect on Slack channels in which a conserved PKC phosphorylation site (S407) that regulates the current amplitude has been mutated. Furthermore, a Phactr1 mutant that disrupts the binding of PP1 but not that of actin fails to alter Slack currents. Our data suggest that Phactr1 regulates the Slack by linking PP1 to the channel. Targeting Slack-Phactr1 interactions may therefore be helpful in developing the novel therapies for brain disorders associated with the malfunction of Slack channels.
Collapse
Affiliation(s)
- Syed Rydwan Ali
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | | | - Yalan Zhang
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Magdalena Prechova
- Signalling and Transcription Group, The Francis Crick Institute, London, UK
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, CZ
| | - Leonard Konrad Kaczmarek
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
170
|
O'Brien JJ, O'Callaghan JP, Miller DB, Chalgeri S, Wennogle LP, Davis RE, Snyder GL, Hendrick JP. Inhibition of calcium-calmodulin-dependent phosphodiesterase (PDE1) suppresses inflammatory responses. Mol Cell Neurosci 2019; 102:103449. [PMID: 31770590 PMCID: PMC7783477 DOI: 10.1016/j.mcn.2019.103449] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/01/2019] [Accepted: 11/21/2019] [Indexed: 11/08/2022] Open
Abstract
A novel, potent, and highly specific inhibitor of calcium-calmodulin-dependent phosphodiesterases (PDE) of the PDE1 family, ITI-214, was used to investigate the role of PDE1 in inflammatory responses. ITI-214 dose-dependently suppressed lipopolysaccharide (LPS)-induced gene expression of pro-inflammatory cytokines in an immortalized murine microglial cell line, BV2 cells. RNA profiling (RNA-Seq) was used to analyze the impact of ITI-214 on the BV2 cell transcriptome in the absence and the presence of LPS. ITI-214 was found to regulate classes of genes that are involved in inflammation and cell migration responses to LPS exposure. The gene expression changes seen with ITI-214 treatment were distinct from those elicited by inhibitors of other PDEs with anti-inflammatory activity (e.g., a PDE4 inhibitor), indicating a distinct mechanism of action for PDE1. Functionally, ITI-214 inhibited ADP-induced migration of BV2 cells through a P2Y12-receptor-dependent pathway, possibly due to increases in the extent of cAMP and VASP phosphorylation downstream of receptor activation. Importantly, this effect was recapitulated in P2 rat microglial cells in vitro, indicating that these pathways are active in native microglial cells. These studies are the first to demonstrate that inhibition of PDE1 exerts anti-inflammatory effects through effects on microglia signaling pathways. The ability of PDE1 inhibitors to prevent or dampen excessive inflammatory responses of BV2 cells and microglia provides a basis for exploring their therapeutic utility in the treatment of neurodegenerative diseases associated with increased inflammation and microglia proliferation such as Parkinson's disease and Alzheimer's disease.
Collapse
Affiliation(s)
- Jennifer J O'Brien
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America
| | - James P O'Callaghan
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505, United States of America
| | - Diane B Miller
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505, United States of America
| | - Suman Chalgeri
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America
| | - Lawrence P Wennogle
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America
| | - Robert E Davis
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America
| | - Gretchen L Snyder
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America.
| | - Joseph P Hendrick
- Intra-Cellular Therapies, Inc., The Alexandria Center for Life Sciences, 430 East 29th St Suite 900, New York, NY 10016, United States of America
| |
Collapse
|
171
|
Garcia RCT, Torres LL, Dati LMM, Loureiro APDM, Afeche SC, Sandoval MRL, Marcourakis T. Anhydroecgonine methyl ester (AEME), a cocaine pyrolysis product, impairs glutathione-related enzymes response and increases lipid peroxidation in the hippocampal cell culture. Toxicol Rep 2019; 6:1223-1229. [PMID: 31768333 PMCID: PMC6872858 DOI: 10.1016/j.toxrep.2019.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/27/2019] [Accepted: 11/01/2019] [Indexed: 02/06/2023] Open
Abstract
AEME and cocaine decreased GPx, GR and GST activities after 3 and 6 h of exposure. AEME and cocaine increased MDA after 48 h of exposure. AEME-cocaine combination decreased GPx, GR and GST activities after 3 and 6 h. AEME-cocaine combination showed an additive effect on MDA after 48 h of exposure. A higher neurotoxic effect after crack cocaine use is suggested.
Crack cocaine smokers inhale, alongside with cocaine, its pyrolysis product, anhydroecgonine methyl ester (AEME). We have previously described AEME neurotoxic effect and its additive effect when co-incubated with cocaine. Our aim was to evaluate, the effect of AEME, cocaine and AEME-cocaine combination on glutathione peroxidase (GPx), glutathione reductase (GR) and glutathione S-transferase (GST) activities after 3 and 6 h of exposure, periods previous to neuronal death. Lipid peroxidation was evaluated through malonaldehyde (MDA) levels at 3, 6, 24 and 48 h of exposure. All treated groups reduced neuronal viability after 24 h of exposure. AEME and cocaine decreased GPx, GR and GST activities after 3 and 6 h, with an increase in MDA levels after 48 h. AEME-cocaine combination decreased the enzymes activities after 3 and 6 h, showing an additive effect in MDA levels after 48 h. These data show that the glutathione-related enzymes imbalance caused by AEME, cocaine or AEME-cocaine combination exposure preceded neuronal death and lipid peroxidation. Moreover, the additive effect on lipid peroxidation observed with AEME-cocaine exposure after 48 h, suggest a higher neurotoxic effect after crack cocaine use when compared to cocaine alone.
Collapse
Affiliation(s)
- Raphael Caio Tamborelli Garcia
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 580, Bl. 13B, 05508-000 São Paulo, SP, Brazil.,Department of Pharmaceutical Sciences, Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Rua São Nicolau, 210, 1° andar, 09913-030 Diadema, SP, Brazil
| | - Larissa Lobo Torres
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 580, Bl. 13B, 05508-000 São Paulo, SP, Brazil.,Department of Food and Drugs, School of Pharmaceutical Sciences, Federal University of Alfenas, Rua Gabriel Monteiro da Silva, 700, 37130-001 Alfenas, MG, Brazil
| | - Livia Mendonça Munhoz Dati
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 580, Bl. 13B, 05508-000 São Paulo, SP, Brazil
| | - Ana Paula de Melo Loureiro
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 580, Bl. 13B, 05508-000 São Paulo, SP, Brazil
| | - Solange Castro Afeche
- Laboratory of Pharmacology, Butantan Institute, Av. Vital Brasil, 1500, 05503-900, São Paulo, SP, Brazil
| | | | - Tania Marcourakis
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 580, Bl. 13B, 05508-000 São Paulo, SP, Brazil
| |
Collapse
|
172
|
Mao W, Salzberg AC, Uchigashima M, Hasegawa Y, Hock H, Watanabe M, Akbarian S, Kawasawa YI, Futai K. Activity-Induced Regulation of Synaptic Strength through the Chromatin Reader L3mbtl1. Cell Rep 2019; 23:3209-3222. [PMID: 29898393 DOI: 10.1016/j.celrep.2018.05.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/12/2018] [Accepted: 05/10/2018] [Indexed: 01/02/2023] Open
Abstract
Homeostatic synaptic downscaling reduces neuronal excitability by modulating the number of postsynaptic receptors. Histone modifications and the subsequent chromatin remodeling play critical roles in activity-dependent gene expression. Histone modification codes are recognized by chromatin readers that affect gene expression by altering chromatin structure. We show that L3mbtl1 (lethal 3 malignant brain tumor-like 1), a polycomb chromatin reader, is downregulated by neuronal activity and is essential for synaptic response and downscaling. Genome-scale mapping of L3mbtl1 occupancies identified Ctnnb1 as a key gene downstream of L3mbtl1. Importantly, the occupancy of L3mbtl1 on the Ctnnb1 gene was regulated by neuronal activity. L3mbtl1 knockout neurons exhibited reduced Ctnnb1 expression. Partial knockdown of Ctnnb1 in wild-type neurons reduced excitatory synaptic transmission and abolished homeostatic downscaling, and transfecting Ctnnb1 in L3mbtl1 knockout neurons enhanced synaptic transmission and restored homeostatic downscaling. These results highlight a role for L3mbtl1 in regulating homeostasis of synaptic efficacy.
Collapse
Affiliation(s)
- Wenjie Mao
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605-2324, USA
| | - Anna C Salzberg
- Department of Pharmacology, Department of Biochemistry and Molecular Biology, and Institute for Personalized Medicine, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Motokazu Uchigashima
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605-2324, USA; Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Yuto Hasegawa
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605-2324, USA
| | - Hanno Hock
- Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School,185 Cambridge Street, Boston, MA 02114, USA
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Schahram Akbarian
- Mount Sinai Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA
| | - Yuka Imamura Kawasawa
- Department of Pharmacology, Department of Biochemistry and Molecular Biology, and Institute for Personalized Medicine, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Kensuke Futai
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605-2324, USA.
| |
Collapse
|
173
|
Lukmanto D, Khanh VC, Shirota S, Kato T, Takasaki MM, Ohneda O. Dynamic Changes of Mouse Embryonic Stem Cell-Derived Neural Stem Cells Under In Vitro Prolonged Culture and Hypoxic Conditions. Stem Cells Dev 2019; 28:1434-1450. [DOI: 10.1089/scd.2019.0101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Donny Lukmanto
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Vuong Cat Khanh
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Saori Shirota
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Toshiki Kato
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Mami Matsuo Takasaki
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Osamu Ohneda
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
174
|
Leung AW, Li JYH. An adherent-cell depletion technique to generate human neural progenitors and neurons. J Cell Physiol 2019; 234:19933-19941. [PMID: 30972783 DOI: 10.1002/jcp.28591] [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: 03/07/2019] [Accepted: 03/18/2019] [Indexed: 11/06/2022]
Abstract
Existing methodologies to produce human neural stem cells and neurons from embryonic stem cells frequently involve multistep processes and the use of complex and expensive media components, cytokines or small molecules. Here, we report a simple technique to generate human neuroepithelial progenitors and neurons by periodic mechanical dissection and adherent-cell depletion on regular cell-culture grade plastic surfaces. This neural induction technique does not employ growth factors, small molecules or peptide inhibitors, apart from those present in serum-free supplements. Suggestive of their central nervous system origin, we found that neural progenitors formed by this technique expressed radial glia markers, and, when differentiated, expressed TUBB3, RBFOX3 (NeuN) and serotonin, but not markers for peripheral neurons. With these data, we postulate that incorporation of periodic mechanical stimuli and plastic surface-mediated cell selection could improve and streamline existing human neuron production protocols.
Collapse
Affiliation(s)
- Alan W Leung
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, Connecticut
| | - James Y H Li
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, Connecticut
| |
Collapse
|
175
|
Gill S, Kumara VMR. Detecting Neurodevelopmental Toxicity of Domoic Acid and Ochratoxin A Using Rat Fetal Neural Stem Cells. Mar Drugs 2019; 17:md17100566. [PMID: 31590222 PMCID: PMC6835907 DOI: 10.3390/md17100566] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 12/12/2022] Open
Abstract
Currently, animal experiments in rodents are the gold standard for developmental neurotoxicity (DNT) investigations; however, testing guidelines for these experiments are insufficient in terms of animal use, time, and costs. Thus, alternative reliable approaches are needed for predicting DNT. We chose rat neural stem cells (rNSC) as a model system, and used a well-known neurotoxin, domoic acid (DA), as a model test chemical to validate the assay. This assay was used to investigate the potential neurotoxic effects of Ochratoxin A (OTA), of which the main target organ is the kidney. However, limited information is available regarding its neurotoxic effects. The effects of DA and OTA on the cytotoxicity and on the degree of differentiation of rat rNSC into astrocytes, neurons, and oligodendrocytes were monitored using cell-specific immunofluorescence staining for undifferentiated rNSC (nestin), neurospheres (nestin and A2B5), neurons (MAP2 clone M13, MAP2 clone AP18, and Doublecortin), astrocytes (GFAP), and oligodendrocytes (A2B5 and mGalc). In the absence of any chemical exposure, approximately 46% of rNSC differentiated into astrocytes and neurons, while 40% of the rNSC differentiated into oligodendrocytes. Both non-cytotoxic and cytotoxic concentrations of DA and OTA reduced the differentiation of rNSC into astrocytes, neurons, and oligodendrocytes. Furthermore, a non-cytotoxic nanomolar (0.05 µM) concentration of DA and 0.2 µM of OTA reduced the percentage differentiation of rNSC into astrocytes and neurons. Morphometric analysis showed that the highest concentration (10 μM) of DA reduced axonal length. These indicate that low, non-cytotoxic concentrations of DA and OTA can interfere with the differentiation of rNSC.
Collapse
Affiliation(s)
- S Gill
- Regulatory Toxicology Research Division, Health Products and Food Branch, Tunney's Pasture, Health Canada, 251 Sir Frederick Banting Driveway, Ottawa, ON K1A 0K9, Canada.
| | - V M Ruvin Kumara
- Regulatory Toxicology Research Division, Health Products and Food Branch, Tunney's Pasture, Health Canada, 251 Sir Frederick Banting Driveway, Ottawa, ON K1A 0K9, Canada.
| |
Collapse
|
176
|
Caspase-1 involves in bilirubin-induced injury of cultured rat cortical neurons. Pediatr Res 2019; 86:492-499. [PMID: 31195405 DOI: 10.1038/s41390-019-0451-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Bilirubin encephalopathy, the most serious complication of hyperbilirubinemia during the neonatal period, with high mortality and morbidity, often causes irreversible neurological damage. Currently, caspase-1, a member of the cysteinyl aspartate-specific protease caspase family, is regarded as a key mediator of inflammatory processes, attracting widespread attention. The purpose of this study was to investigate whether caspase-1 is involved in bilirubin-induced neuronal injury. METHODS VX-765, a highly potent and selective inhibitor of caspase-1, was used to investigate the effects of unconjugated bilirubin (UCB) on rat cortical neurons, including cell viability, morphological changes in the cell membrane, and nuclear factor-kappa B (NF-κB) activation. RESULTS Neurons treated with UCB showed increased caspase-1 activity without the secretion of interleukin (IL)-1β and IL-18, and caspase-1 was significantly inhibited by pretreatment with VX-765. The cell viability of the VX-765-pretreated neurons was improved, and cell membrane rupture was prevented, as detected by lactate dehydrogenase release and ethidium bromide uptake. Moreover, NF-κB activation by UCB exposure, was attenuated by VX-765 pretreatment. CONCLUSION Bilirubin-induced neuronal injury involves the activation of caspase-1 and NF-κB, leading to membrane leakage, independently of IL-1β and IL-18.
Collapse
|
177
|
Hondrich TJJ, Deußen O, Grannemann C, Brinkmann D, Offenhäusser A. Improvements of Microcontact Printing for Micropatterned Cell Growth by Contrast Enhancement. MICROMACHINES 2019; 10:E659. [PMID: 31574944 PMCID: PMC6848919 DOI: 10.3390/mi10100659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 12/26/2022]
Abstract
Patterned neuronal cell cultures are important tools for investigating neuronal signal integration, network function, and cell-substrate interactions. Because of the variable nature of neuronal cells, the widely used coating method of microcontact printing is in constant need of improvements and adaptations depending on the pattern, cell type, and coating solutions available for a certain experimental system. In this work, we report on three approaches to modify microcontact printing on borosilicate glass surfaces, which we evaluate with contact angle measurements and by determining the quality of patterned neuronal growth. Although background toxification with manganese salt does not result in the desired pattern enhancement, a simple heat treatment of the glass substrates leads to improved background hydrophobicity and therefore neuronal patterning. Thirdly, we extended a microcontact printing process based on covalently linking the glass surface and the coating molecule via an epoxysilane. This extension is an additional hydrophobization step with dodecylamine. We demonstrate that shelf life of the silanized glass is at least 22 weeks, leading to consistently reliable neuronal patterning by microcontact printing. Thus, we compared three practical additions to microcontact printing, two of which can easily be implemented into a workflow for the investigation of patterned neuronal networks.
Collapse
Affiliation(s)
- Timm J J Hondrich
- Institute of Complex Systems, Bioelectronics (ICS-8), Forschungszentrum Jülich, 52428 Jülich, Germany.
- Faculty of Mathematics, Computer Science and Natural Sciences, RWTH Aachen University, 52076 Aachen, Germany.
| | - Oliver Deußen
- Institute of Complex Systems, Bioelectronics (ICS-8), Forschungszentrum Jülich, 52428 Jülich, Germany.
- Faculty of Mathematics, Computer Science and Natural Sciences, RWTH Aachen University, 52076 Aachen, Germany.
| | - Caroline Grannemann
- Institute of Complex Systems, Bioelectronics (ICS-8), Forschungszentrum Jülich, 52428 Jülich, Germany.
- Faculty of Mathematics, Computer Science and Natural Sciences, RWTH Aachen University, 52076 Aachen, Germany.
| | - Dominik Brinkmann
- Institute of Complex Systems, Bioelectronics (ICS-8), Forschungszentrum Jülich, 52428 Jülich, Germany.
- Faculty of Mathematics, Computer Science and Natural Sciences, RWTH Aachen University, 52076 Aachen, Germany.
| | - Andreas Offenhäusser
- Institute of Complex Systems, Bioelectronics (ICS-8), Forschungszentrum Jülich, 52428 Jülich, Germany.
| |
Collapse
|
178
|
Sharma S, Chakravarthy H, Suresh G, Devanathan V. Adult Goat Retinal Neuronal Culture: Applications in Modeling Hyperglycemia. Front Neurosci 2019; 13:983. [PMID: 31607843 PMCID: PMC6756134 DOI: 10.3389/fnins.2019.00983] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/02/2019] [Indexed: 12/13/2022] Open
Abstract
Culture of adult neurons of the central nervous system (CNS) can provide a unique model system to explore neurodegenerative diseases. The CNS includes neurons and glia of the brain, spinal cord and retina. Neurons in the retina have the advantage of being the most accessible cells of the CNS, and can serve as a reliable mirror to the brain. Typically, primary cultures utilize fetal rodent neurons, but very rarely adult neurons from larger mammals. Here, we cultured primary retinal neurons isolated from adult goat up to 10 days, and established an in vitro model of hyperglycemia for performing morphological and molecular characterization studies. Immunofluorescence staining revealed that approximately 30–40% of cultured cells expressed neuronal markers. Next, we examined the relative expression of cell adhesion molecules (CAMs) in adult goat brain and retina. We also studied the effect of different glucose concentrations and media composition on the growth and expression of CAMs in cultured retinal neurons. Hyperglycemia significantly enhances neurite outgrowth in adult retinal neurons in culture. Expression of CAMs such as Caspr1, Contactin1 and Prion is downregulated in the presence of high glucose. Hyperglycemia downregulates the expression of the transcription factor CCAAT/enhancer binding protein (C/EBP α), predicted to bind CAM gene promoters. Collectively, our study demonstrates that metabolic environment markedly affects transcriptional regulation of CAMs in adult retinal neurons in culture. The effect of hyperglycemia on CAM interactions, as well as related changes in intracellular signaling pathways in adult retinal neurons warrants further investigation.
Collapse
Affiliation(s)
- Sapana Sharma
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, India
| | - Harshini Chakravarthy
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, India
| | - Gowthaman Suresh
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, India
| | - Vasudharani Devanathan
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, India
| |
Collapse
|
179
|
Chu X, Wu X, Feng H, Zhao H, Tan Y, Wang L, Ran H, Yi L, Peng Y, Tong H, Liu R, Bai W, Shi H, Li L, Huo D. Coupling Between Interleukin-1R1 and Necrosome Complex Involves in Hemin-Induced Neuronal Necroptosis After Intracranial Hemorrhage. Stroke 2019; 49:2473-2482. [PMID: 30355103 DOI: 10.1161/strokeaha.117.019253] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background and Purpose- Accumulated evidence suggests that hemin-a breakdown product of hemoglobin-plays a pivotal role in the inflammatory injuries that result after hemorrhagic stroke through the Toll Like Receptor 2-Toll Like Receptor 4 signal pathway. However, the mechanism of how hemin triggers neuronal necroptosis directly after intracranial hemorrhage (ICH) is still an area of active research. As animal model and preclinical studies have shown, the recombinant interleukin-1 receptor antagonist (IL-1RA) improves clinical outcomes after stroke. As such, we have chosen to investigate the mechanism of how IL-1RA exerts protective effect in hemin-induced neuronal necroptosis after ICH. Methods- Our ICH model was induced by hemin injection in C57BL/6 mice and IL-1R1-/- mice. In addition, we used primary cultured neurons to assess hemin-induced cell death. Co-immunoprecipitation, immunoblot, immunofluorescent staining, neurological deficit scores, and brain water content were used to study the mechanisms of IL-1R1 modulation in neuronal necroptosis both in vitro and in vivo. Results- Free hemin could mediate neuronal necroptosis directly by assembling necrosome complex and then to trigger cell death. This phenomenon was driven by IL-1R1 as IL-1R1 can form a complex with necrosome. After treatment with IL-1RA, both the expression and translocation of the necrosome decreased while disruption of the interaction between IL-1R1 and RIP1/RIP3 (receptor interacting protein 1/3) increased neuron survival. In addition, the IL-1R1-deficient mice demonstrated lower levels of necrosome components, including RIP1, RIP3, and MLKL (mixed lineage kinase domain-like protein), compared with control groups after hemin treatment. In addition, the neurological deficit scores, brain water content, and inflammatory response were all also reduced in the IL-1R1-deficient mice. Conclusions- Functional inhibition of the interaction between IL-1R1 and the necrosome complex improves neuron survival and promotes the recovery of neurological function in experimental ICH. Targeting IL-1R1/RIP1/RIP3 assembly could be a promising therapeutic strategy for patients with ICH.
Collapse
Affiliation(s)
- Xiang Chu
- From the College of Bioengineering, Chongqing University, China (X.C., D.H.)
| | - Xiaofeng Wu
- Research Institute of Surgery/Daping Hospital (X.W., Y.T., L.Y., Y.P., H.T., R.L., W.B., L.L.), Third Military Medical University, Chongqing, China
| | - Hua Feng
- Department of Neurology, Southwest Hospital (H.F., H.Z.), Third Military Medical University, Chongqing, China
| | - Hengli Zhao
- Department of Neurology, Southwest Hospital (H.F., H.Z.), Third Military Medical University, Chongqing, China
| | - Yan Tan
- Research Institute of Surgery/Daping Hospital (X.W., Y.T., L.Y., Y.P., H.T., R.L., W.B., L.L.), Third Military Medical University, Chongqing, China
| | - Liting Wang
- College of Pharmacy (L.W., H.R.), Third Military Medical University, Chongqing, China
| | - Haiying Ran
- College of Pharmacy (L.W., H.R.), Third Military Medical University, Chongqing, China
| | - Liang Yi
- Research Institute of Surgery/Daping Hospital (X.W., Y.T., L.Y., Y.P., H.T., R.L., W.B., L.L.), Third Military Medical University, Chongqing, China
| | - Yan Peng
- Research Institute of Surgery/Daping Hospital (X.W., Y.T., L.Y., Y.P., H.T., R.L., W.B., L.L.), Third Military Medical University, Chongqing, China
| | - Haipeng Tong
- Research Institute of Surgery/Daping Hospital (X.W., Y.T., L.Y., Y.P., H.T., R.L., W.B., L.L.), Third Military Medical University, Chongqing, China
| | - Rui Liu
- Research Institute of Surgery/Daping Hospital (X.W., Y.T., L.Y., Y.P., H.T., R.L., W.B., L.L.), Third Military Medical University, Chongqing, China
| | - Wei Bai
- Research Institute of Surgery/Daping Hospital (X.W., Y.T., L.Y., Y.P., H.T., R.L., W.B., L.L.), Third Military Medical University, Chongqing, China
| | - Huiwen Shi
- Department of General Surgery, The 401 Hospital of Qingdao, China (H.S.)
| | - Lei Li
- Research Institute of Surgery/Daping Hospital (X.W., Y.T., L.Y., Y.P., H.T., R.L., W.B., L.L.), Third Military Medical University, Chongqing, China
| | - Danqun Huo
- From the College of Bioengineering, Chongqing University, China (X.C., D.H.)
| |
Collapse
|
180
|
Segregated neural explants exhibit co-oriented, asymmetric, neurite outgrowth. PLoS One 2019; 14:e0216263. [PMID: 31487284 PMCID: PMC6728047 DOI: 10.1371/journal.pone.0216263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/19/2019] [Indexed: 11/28/2022] Open
Abstract
Explants of embryonic chick sympathetic and sensory ganglia were found to exhibit asymmetric radial outgrowth of neurites under standard culture conditions with or without exogenous Nerve Growth Factor [NGF]. Opposing sides of an explant exhibited: a) differences in neurite length and, b) differences in neurite morphology. Strikingly, this asymmetry exhibited co-orientation among segregated, neighboring explants. The underlying mechanism(s) of the asymmetry and its co-orientation are not known but appear to depend on cell clustering because dissociated sympathetic neurons do not exhibit co-orientation whereas re-aggregated clusters of cells do. This emergent behavior may be similar to the community effect described in other cell types. If a similar phenomenon exists in the embryo, or in maturity, it may contribute to the establishment of proper orientation of neurite outgrowth during development and/or injury-induced neuronal plasticity.
Collapse
|
181
|
Kraus L, Hetsch F, Schneider UC, Radbruch H, Holtkamp M, Meier JC, Fidzinski P. Dimethylethanolamine Decreases Epileptiform Activity in Acute Human Hippocampal Slices in vitro. Front Mol Neurosci 2019; 12:209. [PMID: 31551707 PMCID: PMC6743366 DOI: 10.3389/fnmol.2019.00209] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 08/09/2019] [Indexed: 01/13/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is the most common form of focal epilepsy with about 30% of patients developing pharmacoresistance. These patients continue to suffer from seizures despite polytherapy with antiepileptic drugs (AEDs) and have an increased risk for premature death, thus requiring further efforts for the development of new antiepileptic therapies. The molecule dimethylethanolamine (DMEA) has been tested as a potential treatment in various neurological diseases, albeit the functional mechanism of action was never fully understood. In this study, we investigated the effects of DMEA on neuronal activity in single-cell recordings of primary neuronal cultures. DMEA decreased the frequency of spontaneous synaptic events in a concentration-dependent manner with no apparent effect on resting membrane potential (RMP) or action potential (AP) threshold. We further tested whether DMEA can exert antiepileptic effects in human brain tissue ex vivo. We analyzed the effect of DMEA on epileptiform activity in the CA1 region of the resected hippocampus of TLE patients in vitro by recording extracellular field potentials in the pyramidal cell layer. Epileptiform burst activity in resected hippocampal tissue from TLE patients remained stable over several hours and was pharmacologically suppressed by lacosamide, demonstrating the applicability of our platform to test antiepileptic efficacy. Similar to lacosamide, DMEA also suppressed epileptiform activity in the majority of samples, albeit with variable interindividual effects. In conclusion, DMEA might present a new approach for treatment in pharmacoresistant TLE and further studies will be required to identify its exact mechanism of action and the involved molecular targets.
Collapse
Affiliation(s)
- Larissa Kraus
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Berlin, Germany
- Berlin Institute of Health (BIH), Zoologisches Institut, Technische Universität Braunschweig, Braunschweig, Germany
| | - Florian Hetsch
- Zoologisches Institut, Technische Universität Braunschweig, Braunschweig, Germany
| | - Ulf C. Schneider
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurosurgery, Berlin, Germany
| | - Helena Radbruch
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neuropathology, Berlin, Germany
| | - Martin Holtkamp
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Berlin, Germany
- Berlin Institute of Health (BIH), Zoologisches Institut, Technische Universität Braunschweig, Braunschweig, Germany
| | - Jochen C. Meier
- Berlin Institute of Health (BIH), Zoologisches Institut, Technische Universität Braunschweig, Braunschweig, Germany
- Zoologisches Institut, Technische Universität Braunschweig, Braunschweig, Germany
| | - Pawel Fidzinski
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Berlin, Germany
| |
Collapse
|
182
|
Decker H, Piermartiri TCB, Nedel CB, Romão LF, Francisco SS, Dal-Cim T, Boeck CR, Moura-Neto V, Tasca CI. Guanosine and GMP increase the number of granular cerebellar neurons in culture: dependence on adenosine A 2A and ionotropic glutamate receptors. Purinergic Signal 2019; 15:439-450. [PMID: 31478180 DOI: 10.1007/s11302-019-09677-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 07/30/2019] [Indexed: 12/20/2022] Open
Abstract
The guanine-based purines (GBPs) have essential extracellular functions such as modulation of glutamatergic transmission and trophic effects on neurons and astrocytes. We previously showed that GBPs, such as guanosine-5'-monophosphate (GMP) or guanosine (GUO), promote the reorganization of extracellular matrix proteins in astrocytes, and increase the number of neurons in a neuron-astrocyte co-culture protocol. To delineate the molecular basis underlying these effects, we isolated cerebellar neurons in culture and treated them with a conditioned medium derived from astrocytes previously exposed to GUO or GMP (GBPs-ACM) or, directly, with GUO or GMP. Agreeing with the previous studies, there was an increase in the number of β-tubulin III-positive neurons in both conditions, compared with controls. Interestingly, the increase in the number of neurons in the neuronal cultures treated directly with GUO or GMP was more prominent, suggesting a direct interaction of GBPs on cerebellar neurons. To investigate this issue, we assessed the role of adenosine and glutamate receptors and related intracellular signaling pathways after GUO or GMP treatment. We found an involvement of A2A adenosine receptors, ionotropic glutamate N-methyl-D-aspartate (NMDA), and non-NMDA receptors in the increased number of cerebellar neurons. The signaling pathways extracellular-regulated kinase (ERK), calcium-calmodulin-dependent kinase-II (CaMKII), protein kinase C (PKC), phosphatidilinositol-3'-kinase (PI3-K), and protein kinase A (PKA) are also potentially involved with GMP and GUO effect. Such results suggest that GMP and GUO, and molecules released in GBPs-ACM promote the survival or maturation of primary cerebellar neurons or both via interaction with adenosine and glutamate receptors.
Collapse
Affiliation(s)
- Helena Decker
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, 88040-900, Florianópolis, SC, Brasil
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Tetsade C B Piermartiri
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, 88040-900, Florianópolis, SC, Brasil
| | - Cláudia B Nedel
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, 88040-900, Florianópolis, SC, Brasil
| | - Luciana F Romão
- Departamento de Anatomia, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brasil
| | - Sheila S Francisco
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, 88040-900, Florianópolis, SC, Brasil
| | - Tharine Dal-Cim
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, 88040-900, Florianópolis, SC, Brasil
| | - Carina R Boeck
- Programa de Pós-graduação em Nanociências, Universidade Franciscana, Santa Maria, RS, Brasil
| | - Vivaldo Moura-Neto
- Departamento de Anatomia, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brasil
- Instituto Estadual do Cérebro Paulo Niemeyer da Secretaria de Estado de Saúde do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Carla I Tasca
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, 88040-900, Florianópolis, SC, Brasil.
| |
Collapse
|
183
|
Zhou L, Ao LY, Yan YY, Li WT, Ye AQ, Li CY, Shen WY, Liang BW, Xiong-Zhu, Li YM. JLX001 Ameliorates Ischemia/Reperfusion Injury by Reducing Neuronal Apoptosis via Down-Regulating JNK Signaling Pathway. Neuroscience 2019; 418:189-204. [PMID: 31487541 DOI: 10.1016/j.neuroscience.2019.08.053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 08/29/2019] [Accepted: 08/30/2019] [Indexed: 01/26/2023]
Abstract
JLX001, a novel compound with similar structure with cyclovirobuxine D (CVB-D), has been proved to exert therapeutical effects on permanent focal cerebral ischemia. However, the protective effects of JLX001 on cerebral ischemia/reperfusion (I/R) injury and its anti-apoptotic effects have not been reported. We investigated the efficacy of JLX001 in two pharmacodynamic tests (pre-treatment test and post-treatment) with rats subjected to middle cerebral artery occlusion/reperfusion (MCAO/R). The pharmacodynamic tests demonstrated that JLX001 ameliorated I/R injury by reducing infarct sizes and brain edema. The results of Morris water maze, neurological scores, cylinder test and posture reflex test implied that JLX001 improved the learning, memory and motor ability after MCAO/R in the long term. Anti-apoptotic effects of JLX001 and its regulation of cytosolic c-Jun N-terminal Kinases (JNKs) signal pathway were confirmed in vivo by co-immunofluorescence staining and western immunoblotting. Furthermore, primary cortical neuron cultures were prepared and exposed to oxygen glucose deprivation/reoxygenation (OGD/R) for in vitro studies. Cytotoxicity test and mitochondrial membrane potential (MMP) test showed that JLX001 enhanced cell survival rate and maintained MMP. Flow cytometry and TdT-mediated dUTP-X nick end labeling (TUNEL) staining demonstrated the anti-apoptotic effects of JLX001 in vitro. Likewise, JLX001 regulated JNK signal pathway in vivo, which was also confirmed by western immunoblotting. Collectively, this study presents the first evidence that JLX001 exerted protective effects against I/R injury by reducing neuronal apoptosis via down-regulating JNK signaling pathway.
Collapse
Affiliation(s)
- Lin Zhou
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Lu-Yao Ao
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yun-Yi Yan
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Wan-Ting Li
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - An-Qi Ye
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Cheng-Yuan Li
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Wei-Yang Shen
- School of Sciences, China Pharmaceutical University, Nanjing 210009, PR China
| | - Bing-Wen Liang
- Jiangsu Jinglixin Pharmaceutical Technology Company Limited, Nanjing 211100, PR China
| | - Xiong-Zhu
- Jiangsu Jinglixin Pharmaceutical Technology Company Limited, Nanjing 211100, PR China; Medicine & Chemical Institute, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Yun-Man Li
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
184
|
Oleaga C, Jalilvand G, Legters G, Martin C, Ekman G, McAleer CW, Long CJ, Hickman JJ. A human in vitro platform for the evaluation of pharmacology strategies in cardiac ischemia. APL Bioeng 2019; 3:036103. [PMID: 31431939 PMCID: PMC6692160 DOI: 10.1063/1.5089237] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 07/22/2019] [Indexed: 12/21/2022] Open
Abstract
Cardiac ischemic events increase the risk for arrhythmia, heart attack, heart failure, and death and are the leading mortality condition globally. Reperfusion therapy is the first line of treatment for this condition, and although it significantly reduces mortality, cardiac ischemia remains a significant threat. New therapeutic strategies are under investigation to improve the ischemia survival rate; however, the current preclinical models to validate these fail to predict the human outcome. We report the development of a functional human cardiac in vitro system for the study of conduction velocity under ischemic conditions. The system is a bioMEMs platform formed by human iPSC derived cardiomyocytes patterned on microelectrode arrays and maintained in serum-free conditions. Electrical activity changes of conduction velocity, beat frequency, and QT interval (the QT-interval measures the period from onset of depolarization to the completion of repolarization) or action potential length can be evaluated over time and under the stress of ischemia. The optimized protocol induces >80% reduction in conduction velocity, after a 4 h depletion period, and a partial recovery after 72 h of oxygen and nutrient reintroduction. The sensitivity of the platform for pharmacological interventions was challenged with a gap junction modulator (ZP1609), known to prevent or delay the depression of conduction velocity induced by ischemic metabolic stress. ZP1609 significantly improved the drastic drop in conduction velocity and enabled a greater recovery. This model represents a new preclinical platform for studying cardiac ischemia with human cells, which does not rely on biomarker analysis and has the potential for screening novel cardioprotective drugs with readouts that are closer to the measured clinical parameters.
Collapse
Affiliation(s)
- Carlota Oleaga
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | - Golareh Jalilvand
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | - Gregg Legters
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | - Candace Martin
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | - Gail Ekman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | | | | | - James J. Hickman
- Author to whom correspondence should be addressed:. Tel.: +1 407-823-1925
| |
Collapse
|
185
|
Nam Y, Joo B, Lee JY, Han KM, Ryu KY, Koh YH, Kim J, Koo JW, We YM, Hoe HS. ALWPs Improve Cognitive Function and Regulate Aβ Plaque and Tau Hyperphosphorylation in a Mouse Model of Alzheimer's Disease. Front Mol Neurosci 2019; 12:192. [PMID: 31474828 PMCID: PMC6707392 DOI: 10.3389/fnmol.2019.00192] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 07/24/2019] [Indexed: 01/21/2023] Open
Abstract
Recently, we reported that ALWPs, which we developed by combining Liuwei Dihuang pills (LWPs) with antler, regulate the LPS-induced neuroinflammatory response and rescue LPS-induced short- and long-term memory impairment in wild-type (WT) mice. In the present study, we examined the effects of ALWPs on Alzheimer’s disease (AD) pathology and cognitive function in WT mice as well as 5x FAD mice (a mouse model of AD). We found that administration of ALWPs significantly reduced amyloid plaque levels in 5x FAD mice and significantly decreased amyloid β (Aβ) levels in amyloid precursor protein (APP)-overexpressing H4 cells. In addition, ALWPs administration significantly suppressed tau hyperphosphorylation in 5x FAD mice. Oral administration of ALWPs significantly improved long-term memory in scopolamine (SCO)-injected WT mice and 5x FAD mice by altering dendritic spine density. Importantly, ALWPs promoted spinogenesis in primary hippocampal neurons and WT mice and modulated the dendritic spine number in an extracellular signal-regulated kinase (ERK)-dependent manner. Taken together, our results suggest that ALWPs are a candidate therapeutic drug for AD that can modulate amyloid plaque load, tau phosphorylation, and synaptic/cognitive function.
Collapse
Affiliation(s)
- Youngpyo Nam
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Bitna Joo
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea.,Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, South Korea
| | - Ju-Young Lee
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Kyung-Min Han
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea.,Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, South Korea
| | - Ka-Young Ryu
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Young Ho Koh
- Center for Biomedical Sciences, Center for Infectious Diseases, Division of Brain Disease, Korea National Institute of Health, Heungdeok-gu, South Korea
| | - Jeongyeon Kim
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Ja Wook Koo
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea.,Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, South Korea
| | - Young-Man We
- College of Korean Medicine, Wonkwang University, Iksan, South Korea
| | - Hyang-Sook Hoe
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea.,Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, South Korea
| |
Collapse
|
186
|
Taniguchi K, Yamamoto F, Arai T, Yang J, Sakai Y, Itoh M, Mamada N, Sekiguchi M, Yamada D, Saitoh A, Kametani F, Tamaoka A, Araki YM, Wada K, Mizusawa H, Araki W. Tyrosol Reduces Amyloid-β Oligomer Neurotoxicity and Alleviates Synaptic, Oxidative, and Cognitive Disturbances in Alzheimer’s Disease Model Mice. J Alzheimers Dis 2019; 70:937-952. [DOI: 10.3233/jad-190098] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Kaori Taniguchi
- Department of Demyelinating Disease and Aging, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
| | - Fumiko Yamamoto
- Department of Demyelinating Disease and Aging, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takuya Arai
- Department of Demyelinating Disease and Aging, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
| | - Jinwei Yang
- Tokiwa Phytochemical Co., Ltd, Sakura, Chiba, Japan
| | - Yusuke Sakai
- Tokiwa Phytochemical Co., Ltd, Sakura, Chiba, Japan
| | - Masayuki Itoh
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
| | - Naomi Mamada
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Masayuki Sekiguchi
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
| | - Daisuke Yamada
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Akiyoshi Saitoh
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Fuyuki Kametani
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo, Japan
| | - Akira Tamaoka
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yumiko M. Araki
- Department of Psychiatry and Behavioral Science, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Keiji Wada
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
| | - Hidehiro Mizusawa
- National Center Hospital, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
| | - Wataru Araki
- Department of Demyelinating Disease and Aging, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
| |
Collapse
|
187
|
Dong Y, Sameni S, Digman MA, Brewer GJ. Reversibility of Age-related Oxidized Free NADH Redox States in Alzheimer's Disease Neurons by Imposed External Cys/CySS Redox Shifts. Sci Rep 2019; 9:11274. [PMID: 31375701 PMCID: PMC6677822 DOI: 10.1038/s41598-019-47582-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 07/10/2019] [Indexed: 12/21/2022] Open
Abstract
Redox systems including extracellular cysteine/cystine (Cys/CySS), intracellular glutathione/oxidized glutathione (GSH/GSSG) and nicotinamide adenine dinucleotide reduced/oxidized forms (NADH/NAD+) are critical for maintaining redox homeostasis. Aging as a major risk factor for Alzheimer’s disease (AD) is associated with oxidative shifts, decreases in anti-oxidant protection and dysfunction of mitochondria. Here, we examined the flexibility of mitochondrial-specific free NADH in live neurons from non-transgenic (NTg) or triple transgenic AD-like mice (3xTg-AD) of different ages under an imposed extracellular Cys/CySS oxidative or reductive condition. We used phasor fluorescence lifetime imaging microscopy (FLIM) to distinguish free and bound NADH in mitochondria, nuclei and cytoplasm. Under an external oxidative stress, a lower capacity for maintaining mitochondrial free NADH levels was found in old compared to young neurons and a further decline with genetic load. Remarkably, an imposed Cys/CySS reductive state rejuvenated the mitochondrial free NADH levels of old NTg neurons by 71% and old 3xTg-AD neurons by 89% to levels corresponding to the young neurons. Using FLIM as a non-invasive approach, we were able to measure the reversibility of aging subcellular free NADH levels in live neurons. Our results suggest a potential reductive treatment to reverse the loss of free NADH in old and Alzheimer’s neurons.
Collapse
Affiliation(s)
- Yue Dong
- Department of Biomedical Engineering, University of California Irvine, Irvine, California, United States of America
| | - Sara Sameni
- Department of Biomedical Engineering, University of California Irvine, Irvine, California, United States of America.,Laboratory of Fluorescence Dynamics, Department of Biomedical Engineering, University of California Irvine, Irvine, California, United States of America
| | - Michelle A Digman
- Department of Biomedical Engineering, University of California Irvine, Irvine, California, United States of America.,Laboratory of Fluorescence Dynamics, Department of Biomedical Engineering, University of California Irvine, Irvine, California, United States of America
| | - Gregory J Brewer
- Department of Biomedical Engineering, University of California Irvine, Irvine, California, United States of America. .,MIND Institute, Center for Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA, United States of America.
| |
Collapse
|
188
|
Hasan MF, Ghiasvand S, Wang H, Miwa JM, Berdichevsky Y. Neural layer self-assembly in geometrically confined rat and human 3D cultures. Biofabrication 2019; 11:045011. [PMID: 31247598 DOI: 10.1088/1758-5090/ab2d3f] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Neurological disorders affect millions of Americans and this number is expected to rise with the aging population. Development of drugs to treat these disorders may be facilitated by improved in vitro models that faithfully reproduce salient features of the relevant brain regions. Current 3D culture methods face challenges with reliably reproducing microarchitectural features of brain morphology such as cortical or hippocampal layers. In this work, polydimethylsiloxane (PDMS) mini-wells were used to create low aspect ratio, adherent 3D constructs where neurons self-assemble into layers. Layer self-assembly was determined to depend on the size of the PDMS mini-well. Layer formation occurred in cultures composed of primary rat cortical neurons or human induced pluripotent stem cell-derived neurons and astrocytes and was robust and reproducible. Layered 3D constructs were found to have spontaneous neural activity characterized by long bursts similar to activity in the developing cortex. The responses of layered 3D cultures to drugs were more similar to in vivo data than those of 2D cultures. 3D constructs created with this method may be thus suitable as in vitro models for drug discovery for neurological disorders.
Collapse
Affiliation(s)
- Md Fayad Hasan
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, PA 18015, United States of America
| | | | | | | | | |
Collapse
|
189
|
Su X, Ye Y, Yang Y, Zhang K, Bai W, Chen H, Kang E, Kong C, He X. The Effect of SPTLC2 on Promoting Neuronal Apoptosis is Alleviated by MiR-124-3p Through TLR4 Signalling Pathway. Neurochem Res 2019; 44:2113-2122. [PMID: 31372925 DOI: 10.1007/s11064-019-02849-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/06/2019] [Accepted: 07/27/2019] [Indexed: 12/21/2022]
Abstract
To investigate the role and mechanism of microRNA-124-3p (miR-124-3p) and serine palmitoyltransferase long chain base subunit 2 (SPTLC2) in neuronal apoptosis induced by mechanical injury. Transient transfection was used to modify the expression of miR-124-3p and SPTLC2. After transfection, neuronal apoptosis was evaluated in an in vitro injury model of primary neurons using TUNEL staining and western blot. The correlation between miR-124-3p and SPTLC2 was identified through a dual luciferase reporter assay in HEK293 cells. A rescue experiment in primary neurons was performed to further confirm the result. To explore the downstream mechanisms, co-immunoprecipitation was performed to identify proteins that interact with SPTLC2 in toll-like receptor 4 (TLR4) signalling pathway. Subsequently, the relative expression levels of TLR4 pathway molecules were measured by western blot. Our results showed that increased miR-124-3p can inhibit neuronal apoptosis, which is opposite to the effect of SPTLC2. In addition, miR-124-3p was proved to negatively regulate SPTLC2 expression and suppress the apoptosis-promoting effect of SPTLC2 via the TLR4 signalling pathway.
Collapse
Affiliation(s)
- Xinhong Su
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 169 Changle Western Road, Xi'an, 710032, Shanxi, China
| | - Yuqin Ye
- Department of Neurosurgery, PLA 921rd Hospital, Changsha, 410000, Hunan, China
| | - Yongxiang Yang
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 169 Changle Western Road, Xi'an, 710032, Shanxi, China
| | - Kailiang Zhang
- Department of Orthopedic Surgery, Orthopedic Oncology Institute of Chinese PLA, Tangdu Hospital, Air Force Military Medical University, Xi'an, 710032, Shanxi, China
| | - Wei Bai
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 169 Changle Western Road, Xi'an, 710032, Shanxi, China
| | - Huijun Chen
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 169 Changle Western Road, Xi'an, 710032, Shanxi, China
| | - Enming Kang
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 169 Changle Western Road, Xi'an, 710032, Shanxi, China
| | - Chuiguang Kong
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 169 Changle Western Road, Xi'an, 710032, Shanxi, China
| | - Xiaosheng He
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 169 Changle Western Road, Xi'an, 710032, Shanxi, China.
| |
Collapse
|
190
|
Nash B, Tarn K, Irollo E, Luchetta J, Festa L, Halcrow P, Datta G, Geiger JD, Meucci O. Morphine-Induced Modulation of Endolysosomal Iron Mediates Upregulation of Ferritin Heavy Chain in Cortical Neurons. eNeuro 2019; 6:ENEURO.0237-19.2019. [PMID: 31300544 PMCID: PMC6675873 DOI: 10.1523/eneuro.0237-19.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 07/01/2019] [Indexed: 01/01/2023] Open
Abstract
HIV-associated neurocognitive disorders (HAND) remain prevalent and are aggravated by µ-opioid use. We have previously shown that morphine and other µ-opioids may contribute to HAND by inhibiting the homeostatic and neuroprotective chemokine receptor CXCR4 in cortical neurons, and this novel mechanism depends on upregulation of the protein ferritin heavy chain (FHC). Here, we examined the cellular events and potential mechanisms involved in morphine-mediated FHC upregulation using rat cortical neurons of either sex in vitro and in vivo. Morphine dose dependently increased FHC protein levels in primary neurons through µ-opioid receptor (µOR) and Gαi-protein signaling. Cytoplasmic FHC levels were significantly elevated, but nuclear FHC levels and FHC gene expression were unchanged. Morphine-treated rats also displayed increased FHC levels in layer 2/3 neurons of the prefrontal cortex. Importantly, both in vitro and in vivo FHC upregulation was accompanied by loss of mature dendritic spines, which was also dependent on µOR and Gαi-protein signaling. Moreover, morphine upregulated ferritin light chain (FLC), a component of the ferritin iron storage complex, suggesting that morphine altered neuronal iron metabolism. Indeed, prior to FHC upregulation, morphine increased cytoplasmic labile iron levels as a function of decreased endolysosomal iron. In line with this, chelation of endolysosomal iron (but not extracellular iron) blocked morphine-induced FHC upregulation and dendritic spine reduction, whereas iron overloading mimicked the effect of morphine on FHC and dendritic spines. Overall, these data demonstrate that iron mediates morphine-induced FHC upregulation and consequent dendritic spine deficits and implicate endolysosomal iron efflux to the cytoplasm in these effects.
Collapse
Affiliation(s)
- Bradley Nash
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Kevin Tarn
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Elena Irollo
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Jared Luchetta
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Lindsay Festa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Peter Halcrow
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Gaurav Datta
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102
| |
Collapse
|
191
|
Gerosa L, Grillo B, Forastieri C, Longaretti A, Toffolo E, Mallei A, Bassani S, Popoli M, Battaglioli E, Rusconi F. SRF and SRFΔ5 Splicing Isoform Recruit Corepressor LSD1/KDM1A Modifying Structural Neuroplasticity and Environmental Stress Response. Mol Neurobiol 2019; 57:393-407. [DOI: 10.1007/s12035-019-01720-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
192
|
Kubota H, Brinster RL. Spermatogonial stem cells. Biol Reprod 2019; 99:52-74. [PMID: 29617903 DOI: 10.1093/biolre/ioy077] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/29/2018] [Indexed: 12/19/2022] Open
Abstract
Spermatogonial stem cells (SSCs) are the most primitive spermatogonia in the testis and have an essential role to maintain highly productive spermatogenesis by self-renewal and continuous generation of daughter spermatogonia that differentiate into spermatozoa, transmitting genetic information to the next generation. Since the 1950s, many experimental methods, including histology, immunostaining, whole-mount analyses, and pulse-chase labeling, had been used in attempts to identify SSCs, but without success. In 1994, a spermatogonial transplantation method was reported that established a quantitative functional assay to identify SSCs by evaluating their ability to both self-renew and differentiate to spermatozoa. The system was originally developed using mice and subsequently extended to nonrodents, including domestic animals and humans. Availability of the functional assay for SSCs has made it possible to develop culture systems for their ex vivo expansion, which dramatically advanced germ cell biology and allowed medical and agricultural applications. In coming years, SSCs will be increasingly used to understand their regulation, as well as in germline modification, including gene correction, enhancement of male fertility, and conversion of somatic cells to biologically competent male germline cells.
Collapse
Affiliation(s)
- Hiroshi Kubota
- Laboratory of Cell and Molecular Biology, Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Ralph L Brinster
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
193
|
Yang D, Li NL, Wei D, Liu B, Guo F, Elbahesh H, Zhang Y, Zhou Z, Chen GY, Li K. The E3 ligase TRIM56 is a host restriction factor of Zika virus and depends on its RNA-binding activity but not miRNA regulation, for antiviral function. PLoS Negl Trop Dis 2019; 13:e0007537. [PMID: 31251739 PMCID: PMC6623546 DOI: 10.1371/journal.pntd.0007537] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 07/11/2019] [Accepted: 06/10/2019] [Indexed: 12/25/2022] Open
Abstract
Infection by Zika virus (ZIKV) is linked to microcephaly and other neurological disorders, posing a significant health threat. Innate immunity is the first line of defense against invading pathogens, but relatively little is understood regarding host intrinsic mechanisms that guard against ZIKV. Here, we show that host tripartite motif-containing protein 56 (TRIM56) poses a barrier to ZIKV infection in cells of neural, epithelial and fibroblast origins. Overexpression of TRIM56, but not an E3 ligase-dead mutant or one lacking a short C-terminal portion, inhibited ZIKV RNA replication. Conversely, depletion of TRIM56 increased viral RNA levels. Although the C-terminal region of TRIM56 bears sequence homology to NHL repeat of TRIM-NHL proteins that regulate miRNA activity, knockout of Dicer, which abolishes production of miRNAs, had no demonstrable effect on ZIKV restriction imposed by TRIM56. Rather, we found that TRIM56 is an RNA-binding protein that associates with ZIKV RNA in infected cells. Moreover, a recombinant TRIM56 fragment comprising the C-terminal 392 residues captured ZIKV RNA in cell-free reactions, indicative of direct interaction. Remarkably, deletion of a short C-terminal tail portion abrogated the TRIM56-ZIKV RNA interaction, concomitant with a loss in antiviral activity. Altogether, our study reveals TRIM56 is an RNA binding protein that acts as a ZIKV restriction factor and provides new insights into the antiviral mechanism by which this E3 ligase tackles flavivirus infections.
Collapse
Affiliation(s)
- Darong Yang
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Children’s Foundation Research Institute at Le Bonheur Children’s Hospital, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Nan L. Li
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Dahai Wei
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Baoming Liu
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Fang Guo
- Baruch S. Blumberg Institute, Doylestown, PA, United States of America
| | - Husni Elbahesh
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Yunzhi Zhang
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Department of Infectious Diseases, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi Zhou
- Department of Infectious Diseases, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guo-Yun Chen
- Children’s Foundation Research Institute at Le Bonheur Children’s Hospital, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Kui Li
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States of America
- * E-mail:
| |
Collapse
|
194
|
Long-term expansion and differentiation of adult murine epidermal stem cells in 3D organoid cultures. Proc Natl Acad Sci U S A 2019; 116:14630-14638. [PMID: 31253707 DOI: 10.1073/pnas.1715272116] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Mammalian epidermal stem cells maintain homeostasis of the skin epidermis and contribute to its regeneration throughout adult life. While 2D mouse epidermal stem cell cultures have been established decades ago, a long-term, feeder cell- and serum-free culture system recapitulating murine epidermal architecture has not been available. Here we describe an epidermal organoid culture system that allows long-term, genetically stable expansion of adult epidermal stem cells. Our epidermal expansion media combines atypically high calcium concentrations, activation of cAMP, FGF, and R-spondin signaling with inhibition of bone morphogenetic protein (BMP) signaling. Organoids are established robustly from adult mouse skin and expand over at least 6 mo, while maintaining the basal-apical organization of the mouse interfollicular epidermis. The system represents a powerful tool to study epidermal homeostasis and disease in vitro.
Collapse
|
195
|
Chung HW, Weng JC, King CE, Chuang CF, Chow WY, Chang YC. BDNF elevates the axonal levels of hnRNPs Q and R in cultured rat cortical neurons. Mol Cell Neurosci 2019; 98:97-108. [PMID: 31202892 DOI: 10.1016/j.mcn.2019.06.005] [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: 11/01/2018] [Revised: 05/08/2019] [Accepted: 06/12/2019] [Indexed: 11/17/2022] Open
Abstract
Local translation plays important roles in the maintenance and various functions of axons, and dysfunctions of local translation in axons are implicated in various neurological diseases. Heterogeneous nuclear ribonucleoproteins (hnRNPs) are RNA binding proteins with multiple functions in RNA metabolism. Here, we identified 20 hnRNPs in the axons of cultured rat cortical neurons by interrogating published axon mass spectrometric databases with rat protein databases. Among those identified in axons are highly related hnRNPs Q and R. RT-PCR analysis indicated that axons also contained low levels of hnRNPs Q and R mRNAs. We further found that BDNF treatments raised the levels of hnRNPs Q and R proteins in whole neurons and axons. BDNF also increased the level of poly(A) RNA as well as the proportion of poly(A) RNA granules containing hnRNPs Q and R in the axon. However, following severing the connection between the cell bodies and axons, BDNF did not affect the levels of hnRNPs Q and R, the content of poly(A) RNA, or the colocalization of poly(A) RNA and hnRNPs Q and R in the axon any more, although BDNF still stimulated the local translation in severed axons as it did in intact axons. The results are consistent with that BDNF enhances the axonal transport of RNA granules. The results further suggest that hnRNPs Q and R play a role in the mechanism underlying the enhancement of axonal RNA transport by BDNF.
Collapse
Affiliation(s)
- Hui-Wen Chung
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan.
| | - Ju-Chen Weng
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-En King
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Fan Chuang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan.
| | - Wei-Yuan Chow
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.
| | - Yen-Chung Chang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan; Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
196
|
Xu H, Gu H, Yang Y, Cai E, Ding F, Yu S. 2-(4-Methoxyphenyl)Ethyl-2-Acetamido-2-Deoxy-β-d-Pyranoside Exerts a Neuroprotective Effect through Regulation of Energy Homeostasis and O-GlcNAcylation. J Mol Neurosci 2019; 69:177-187. [DOI: 10.1007/s12031-019-01347-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/22/2019] [Indexed: 12/13/2022]
|
197
|
Prill H, Luu A, Yip B, Holtzinger J, Lo MJ, Christianson TM, Yogalingam G, Aoyagi-Scharber M, LeBowitz JH, Crawford BE, Lawrence R. Differential Uptake of NAGLU-IGF2 and Unmodified NAGLU in Cellular Models of Sanfilippo Syndrome Type B. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:56-63. [PMID: 31309128 PMCID: PMC6606967 DOI: 10.1016/j.omtm.2019.05.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 05/12/2019] [Indexed: 01/03/2023]
Abstract
Sanfilippo syndrome type B, or mucopolysaccharidosis IIIB (MPS IIIB), is a rare autosomal recessive lysosomal storage disease caused by a deficiency of α-N-acetylglucosaminidase (NAGLU). Deficiency in NAGLU disrupts the lysosomal turnover of heparan sulfate (HS), which results in the abnormal accumulation of partially degraded HS in cells and tissues. BMN 250 (NAGLU-insulin-like growth factor 2 [IGF2]) is a recombinant fusion protein developed as an investigational enzyme replacement therapy for MPS IIIB. The IGF2 peptide on BMN 250 promotes enhanced targeting of the enzyme to lysosomes through its interaction with the mannose 6-phosphate receptor. The focus of these studies was to further characterize the ability of NAGLU-IGF2 to clear accumulated HS compared to unmodified NAGLU in primary cellular models of MPS IIIB. Here, we establish distinct primary cell models of MPS IIIB with HS accumulation. These cellular models revealed distinct NAGLU uptake characteristics that depend on the duration of exposure. We found that with sustained exposure, NAGLU uptake and HS clearance occurred independent of known lysosomal targeting signals. In contrast, under conditions of limited exposure duration, NAGLU-IGF2 was taken up more rapidly than the unmodified NAGLU into MPS IIIB primary fibroblasts, astrocytes, and cortical neurons, where it efficiently degraded accumulated HS. These studies illustrate the importance of using physiologically relevant conditions in the evaluation of enzyme replacement therapies in cellular models.
Collapse
Affiliation(s)
- Heather Prill
- Research, BioMarin Pharmaceutical, Inc., Novato, CA 94949, USA
| | - Amanda Luu
- Research, BioMarin Pharmaceutical, Inc., Novato, CA 94949, USA
| | - Bryan Yip
- Research, BioMarin Pharmaceutical, Inc., Novato, CA 94949, USA
| | - John Holtzinger
- Research, BioMarin Pharmaceutical, Inc., Novato, CA 94949, USA
| | - Melanie J Lo
- Research, BioMarin Pharmaceutical, Inc., Novato, CA 94949, USA
| | | | | | | | | | | | - Roger Lawrence
- Research, BioMarin Pharmaceutical, Inc., Novato, CA 94949, USA
| |
Collapse
|
198
|
Brenneman DE, Kinney WA, Ward SJ. Knockdown siRNA Targeting the Mitochondrial Sodium-Calcium Exchanger-1 Inhibits the Protective Effects of Two Cannabinoids Against Acute Paclitaxel Toxicity. J Mol Neurosci 2019; 68:603-619. [PMID: 31077084 DOI: 10.1007/s12031-019-01321-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 04/16/2019] [Indexed: 01/01/2023]
Abstract
Treatment with cannabidiol (CBD) or KLS-13019 (novel CBD analog), has previously been shown to prevent paclitaxel-induced mechanical allodynia in a mouse model of chemotherapy-induced peripheral neuropathy (CIPN). The mechanism of action for CBD- and KLS-13019-mediated protection now has been explored with dissociated dorsal root ganglion (DRG) cultures using small interfering RNA (siRNA) to the mitochondrial Na+ Ca2+ exchanger-1 (mNCX-1). Treatment with this siRNA produced a 50-55% decrease in the immunoreactive (IR) area for mNCX-1 in neuronal cell bodies and a 72-80% decrease in neuritic IR area as determined with high-content image analysis. After treatment with 100 nM KLS-13019 and siRNA, DRG cultures exhibited a 75 ± 5% decrease in protection from paclitaxel-induced toxicity; whereas siRNA studies with 10 μM CBD produced a 74 ± 3% decrease in protection. Treatment with mNCX-1 siRNA alone did not produce toxicity. The protective action of cannabidiol and KLS-13019 against paclitaxel-induced toxicity during a 5-h test period was significantly attenuated after a 4-day knockdown of mNCX-1 that was not attributable to toxicity. These data indicate that decreases in neuritic mNCX-1 corresponded closely with decreased protection after siRNA treatment. Pharmacological blockade of mNCX-1 with CGP-37157 produced complete inhibition of cannabinoid-mediated protection from paclitaxel in DRG cultures, supporting the observed siRNA effects on mechanism.
Collapse
Affiliation(s)
- Douglas E Brenneman
- Advanced Neural Dynamics, Inc, Pennsylvania Biotechnology Center, 3805 Old Easton Road, Doylestown, PA, 18902, USA. .,Kannalife Sciences, Inc, Pennsylvania Biotechnology Center, Doylestown, PA, 18902, USA.
| | - William A Kinney
- Kannalife Sciences, Inc, Pennsylvania Biotechnology Center, Doylestown, PA, 18902, USA
| | - Sara Jane Ward
- Center for Substance Abuse Research, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| |
Collapse
|
199
|
Zhang F, Liu Y, Tang F, Liang B, Chen H, Zhang H, Wang K. Electrophysiological and pharmacological characterization of a novel and potent neuronal Kv7 channel opener SCR2682 for antiepilepsy. FASEB J 2019; 33:9154-9166. [DOI: 10.1096/fj.201802848rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Fan Zhang
- The Key Laboratory of Neural and Vascular Biology Ministry of Education The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province Department of Pharmacology Hebei Medical University Shijiazhuang China
| | - Yani Liu
- Department of Pharmacology Qingdao University Qingdao China
| | - Feng Tang
- Medicinal Chemistry, Simcere Pharmaceutical Nanjing China
| | - Bo Liang
- Medicinal Chemistry Shanghai Zhimeng BioPharma Shanghai China
| | - Huanming Chen
- Medicinal Chemistry Shanghai Zhimeng BioPharma Shanghai China
| | - Hailin Zhang
- The Key Laboratory of Neural and Vascular Biology Ministry of Education The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province Department of Pharmacology Hebei Medical University Shijiazhuang China
| | - Kewei Wang
- Department of Pharmacology Qingdao University Qingdao China
- Institute of Innovative Drugs School of Pharmacy Qingdao University Qingdao China
- Center for Brain Science and Brain‐Inspired Intelligence Guangzhou China
| |
Collapse
|
200
|
Culturing primary neurons from rat hippocampus and cortex. Neuronal Signal 2019; 3:NS20180207. [PMID: 32714598 PMCID: PMC7363313 DOI: 10.1042/ns20180207] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/05/2019] [Accepted: 04/15/2019] [Indexed: 01/21/2023] Open
Abstract
Primary neurons from rodent brain hippocampus and cortex have served as important tools in biomedical research over the years. However, protocols for the preparation of primary neurons vary, which often lead to conflicting results. This report provides a robust and reliable protocol for the production of primary neuronal cultures from the cortex and hippocampus with minimal contribution of non-neuronal cells. The neurons were grown in serum-free media and maintained for several weeks without any additional feeder cells. The neuronal cultures maintained according to this protocol differentiate and by 3 weeks develop extensive axonal and dendritic branching. The cultures produced by this method show excellent reproducibility and can be used for histological, molecular and biochemical methods.
Collapse
|