1
|
Sade AN, Wiener G, Barak B. Intersection of mitochondrial dysfunction and myelination: An overlooked aspect in neurodevelopmental disorders. Neural Regen Res 2026; 21:659-660. [PMID: 39995084 DOI: 10.4103/nrr.nrr-d-24-01025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/27/2024] [Indexed: 02/26/2025] Open
Affiliation(s)
- Ariel Nir Sade
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel (Sade AN, Wiener G, Barak B)
| | - Gal Wiener
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel (Sade AN, Wiener G, Barak B)
| | - Boaz Barak
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel (Sade AN, Wiener G, Barak B)
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel (Barak B)
| |
Collapse
|
2
|
Falcón P, Brito Á, Escandón M, Roa JF, Martínez NW, Tapia-Godoy A, Farfán P, Matus S. GCN2-Mediated eIF2α Phosphorylation Is Required for Central Nervous System Remyelination. Int J Mol Sci 2025; 26:1626. [PMID: 40004088 PMCID: PMC11855834 DOI: 10.3390/ijms26041626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/03/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Under conditions of amino acid deficiency, mammalian cells activate a nutrient-sensing kinase known as general control nonderepressible 2 (GCN2). The activation of GCN2 results in the phosphorylation of the alpha subunit of the eukaryotic initiation factor 2 (eIF2α), which can be phosphorylated by three other three integrated stress response (ISR) kinases, reducing overall protein synthesis. GCN2 activation also promotes the translation of specific mRNAs, some of which encode transcription factors that enhance the transcription of genes involved in the synthesis, transport, and metabolism of amino acids to restore cellular homeostasis. The phosphorylation of eIF2α has been shown to protect oligodendrocytes, the cells responsible for producing myelin in the central nervous system during remyelination. Here, we explore the potential role of the kinase GCN2 in the myelination process. We challenged mice deficient in the GCN2-encoding gene with a pharmacological demyelinating stimulus (cuprizone) and evaluated the recovery of myelin as well as ISR activation through the levels of eIF2α phosphorylation. Our findings indicate that GCN2 controls the establishment of myelin by fine-tuning its abundance and morphology in the central nervous system. We also found that GCN2 is essential for remyelination. Surprisingly, we discovered that GCN2 is necessary to maintain eIF2α levels during remyelination.
Collapse
Affiliation(s)
- Paulina Falcón
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
| | - Álvaro Brito
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
| | - Marcela Escandón
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
- Ph.D. “Program in Cell Biology and Biomedicine”, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510157, Chile
| | - Juan Francisco Roa
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
- Ph.D. “Program in Cell Biology and Biomedicine”, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510157, Chile
| | - Nicolas W. Martínez
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8580704, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510157, Chile
| | - Ariel Tapia-Godoy
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8580704, Chile
| | - Pamela Farfán
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8580704, Chile
| | - Soledad Matus
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8580704, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510157, Chile
| |
Collapse
|
3
|
Cherchi F, Venturini M, Magni G, Frulloni L, Chieca M, Buonvicino D, Santalmasi C, Rossi F, De Logu F, Coppi E, Pugliese AM. Adenosine A 2B receptors differently modulate oligodendrogliogenesis and myelination depending on their cellular localization. Glia 2024; 72:1985-2000. [PMID: 39077799 DOI: 10.1002/glia.24593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/31/2024]
Abstract
Differentiation of oligodendrocyte precursor cells (OPCs) into mature oligodendrocytes (OLs) is a key event for axonal myelination in the brain; this process fails during demyelinating pathologies. Adenosine is emerging as an important player in oligodendrogliogenesis, by activating its metabotropic receptors (A1R, A2AR, A2BR, and A3R). We previously demonstrated that the Gs-coupled A2BR reduced differentiation of primary OPC cultures by inhibiting delayed rectifier (IK) as well as transient (IA) outward K+ currents. To deepen the unclear role of this receptor subtype in neuron-OL interplay and in myelination process, we tested the effects of different A2BR ligands in a dorsal root ganglion neuron (DRGN)/OPC cocultures, a corroborated in vitro myelination assay. The A2BR agonist, BAY60-6583, significantly reduced myelin basic protein levels but simultaneously increased myelination index in DRGN/OPC cocultures analyzed by confocal microscopy. The last effect was prevented by the selective A2BR antagonists, PSB-603 and MRS1706. To clarify this unexpected data, we wondered whether A2BRs could play a functional role on DRGNs. We first demonstrated, by immunocytochemistry, that primary DRGN monoculture expressed A2BRs. Their selective activation by BAY60-6583 enhanced DRGN excitability, as demonstrated by increased action potential firing, decreased rheobase and depolarized resting membrane potential and were prevented by PSB-603. Throughout this A2BR-dependent enhancement of neuronal activity, DRGNs could release factors to facilitate myelination processes. Finally, silencing A2BR in DRGNs alone prevents the increased myelination induced by BAY60-6583 in cocultures. In conclusion, our data suggest a different role of A2BR during oligodendrogliogenesis and myelination, depending on their activation on neurons or oligodendroglial cells.
Collapse
Affiliation(s)
- Federica Cherchi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Martina Venturini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Giada Magni
- Cnr-Istituto di Fisica Applicata "Nello Carrara", Florence, Italy
| | - Lucia Frulloni
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Martina Chieca
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Daniela Buonvicino
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Clara Santalmasi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Francesca Rossi
- Cnr-Istituto di Fisica Applicata "Nello Carrara", Florence, Italy
| | - Francesco De Logu
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Elisabetta Coppi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Anna Maria Pugliese
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| |
Collapse
|
4
|
Boiczyk GM, Pearson N, Kote VB, Sundaramurthy A, Subramaniam DR, Rubio JE, Unnikrishnan G, Reifman J, Monson KL. Region specific anisotropy and rate dependence of Göttingen minipig brain tissue. Biomech Model Mechanobiol 2024; 23:1511-1529. [PMID: 38717719 DOI: 10.1007/s10237-024-01852-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 04/17/2024] [Indexed: 09/28/2024]
Abstract
Traumatic brain injury is a major cause of morbidity in civilian as well as military populations. Computational simulations of injurious events are an important tool to understanding the biomechanics of brain injury and evaluating injury criteria and safety measures. However, these computational models are highly dependent on the material parameters used to represent the brain tissue. Reported material properties of tissue from the cerebrum and cerebellum remain poorly defined at high rates and with respect to anisotropy. In this work, brain tissue from the cerebrum and cerebellum of male Göttingen minipigs was tested in one of three directions relative to axon fibers in oscillatory simple shear over a large range of strain rates from 0.025 to 250 s-1. Brain tissue showed significant direction dependence in both regions, each with a single preferred loading direction. The tissue also showed strong rate dependence over the full range of rates considered. Transversely isotropic hyper-viscoelastic constitutive models were fit to experimental data using dynamic inverse finite element models to account for wave propagation observed at high strain rates. The fit constitutive models predicted the response in all directions well at rates below 100 s-1, after which they adequately predicted the initial two loading cycles, with the exception of the 250 s-1 rate, where models performed poorly. These constitutive models can be readily implemented in finite element packages and are suitable for simulation of both conventional and blast injury in porcine, especially Göttingen minipig, models.
Collapse
Affiliation(s)
- Gregory M Boiczyk
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| | - Noah Pearson
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Vivek Bhaskar Kote
- Telemedicine and Advanced Technology Research Center, Department of Defense Biotechnology High Performance Computing Software Applications Institute, United States Army Medical Research and Development Command, Fort Detrick, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Aravind Sundaramurthy
- Telemedicine and Advanced Technology Research Center, Department of Defense Biotechnology High Performance Computing Software Applications Institute, United States Army Medical Research and Development Command, Fort Detrick, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Dhananjay Radhakrishnan Subramaniam
- Telemedicine and Advanced Technology Research Center, Department of Defense Biotechnology High Performance Computing Software Applications Institute, United States Army Medical Research and Development Command, Fort Detrick, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Jose E Rubio
- Telemedicine and Advanced Technology Research Center, Department of Defense Biotechnology High Performance Computing Software Applications Institute, United States Army Medical Research and Development Command, Fort Detrick, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Ginu Unnikrishnan
- Telemedicine and Advanced Technology Research Center, Department of Defense Biotechnology High Performance Computing Software Applications Institute, United States Army Medical Research and Development Command, Fort Detrick, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Jaques Reifman
- Telemedicine and Advanced Technology Research Center, Department of Defense Biotechnology High Performance Computing Software Applications Institute, United States Army Medical Research and Development Command, Fort Detrick, MD, USA
| | - Kenneth L Monson
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
5
|
Singh S, Sutkus L, Li Z, Baker S, Bear J, Dilger RN, Miller DJ. Standardization of a silver stain to reveal mesoscale myelin in histological preparations of the mammalian brain. J Neurosci Methods 2024; 407:110139. [PMID: 38626852 DOI: 10.1016/j.jneumeth.2024.110139] [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: 01/18/2024] [Revised: 03/26/2024] [Accepted: 04/12/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND The brain is built of neurons supported by myelin, a fatty substance that improves cellular communication. Noninvasive magnetic resonance imaging (MRI) is now able to measure brain structure like myelin and requires histological validation. NEW METHOD Here we present work in small and large biomedical model mammals to standardize a silver impregnation method as a high-throughput histological myelin visualization procedure. Specifically, we built a new staining well plate to increase batch size, and then systematically varied the staining and clearing cycles to describe the staining response curve across taxa and conditions. We compared tissues fixed by immersion or perfusion, mounted versus free-floating, and cut as thicker or thinner slices, with two-weeks of post-fixation. RESULTS The staining response curves show optimal staining with a single exposure across taxa when incubation and clearing epochs are held to within 3-9 min. We show that clearing was slower in mounted vs free-floating tissue, and that staining was faster and caused fracturing earlier in thinner sliced and smaller volumes of tissue. COMPARISON WITH EXISTING METHODS We developed a batch processing approach to increase throughput while ensuring reproducibility and demonstrate the optimal conditions for fine myelinated fiber morphology visualization with short cycles (<9 minutes). CONCLUSIONS We present our optimized protocol to reveal mesoscale neuroanatomical myelin content in histology across mammals. This standard staining procedure will facilitate multiscale analyses of myelin content across development as well as in the presence of injury or disease.
Collapse
Affiliation(s)
- S Singh
- Department of Evolution, Ecology, and Behavior, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - L Sutkus
- Neuroscience Program, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - Z Li
- Neuroscience Program, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - S Baker
- Machine Shop, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - J Bear
- Machine Shop, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - R N Dilger
- Department of Animal Sciences, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America; Neuroscience Program, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America; Beckman Institute for Advanced Science and Technology, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America
| | - D J Miller
- Department of Evolution, Ecology, and Behavior, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America; Neuroscience Program, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America; Beckman Institute for Advanced Science and Technology, at the University of Illinois at Urbana-Champaign, 505 South Goodwin Ave, Urbana, IL 61801, United States of America.
| |
Collapse
|
6
|
Xie J, Lan Y, Zou C, He J, Huang Q, Zeng J, Pan M, Mei Y, Luo J, Zou D. Single-nucleus analysis reveals microenvironment-specific neuron and glial cell enrichment in Alzheimer's disease. BMC Genomics 2024; 25:526. [PMID: 38807051 PMCID: PMC11134750 DOI: 10.1186/s12864-024-10447-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/23/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a complicated neurodegenerative disease. Neuron-glial cell interactions are an important but not fully understood process in the progression of AD. We used bioinformatic methods to analyze single-nucleus RNA sequencing (snRNA-seq) data to investigate the cellular and molecular biological processes of AD. METHOD snRNA-seq data were downloaded from Gene Expression Omnibus (GEO) datasets and reprocessed to identify 240,804 single nuclei from healthy controls and patients with AD. The cellular composition of AD was further explored using Uniform Manifold Approximation and Projection (UMAP). Enrichment analysis for the functions of the DEGs was conducted and cell development trajectory analyses were used to reveal underlying cell fate decisions. iTALK was performed to identify ligand-receptor pairs among various cell types in the pathological ecological microenvironment of AD. RESULTS Six cell types and multiple subclusters were identified based on the snRNA-seq data. A subcluster of neuron and glial cells co-expressing lncRNA-SNHG14, myocardin-related transcription factor A (MRTFA), and MRTFB was found to be more abundant in the AD group. This subcluster was enriched in mitogen-activated protein kinase (MAPK)-, immune-, and apoptosis-related pathways. Through molecular docking, we found that lncRNA-SNHG14 may bind MRTFA and MRTFB, resulting in an interaction between neurons and glial cells. CONCLUSIONS The findings of this study describe a regulatory relationship between lncRNA-SNHG14, MRTFA, and MRTFB in the six main cell types of AD. This relationship may contribute to microenvironment remodeling in AD and provide a theoretical basis for a more in-depth analysis of AD.
Collapse
Affiliation(s)
- Jieqiong Xie
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, People's Republic of China
| | - Yating Lan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, People's Republic of China
| | - Cuihua Zou
- Department of Quality Control, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, People's Republic of China
| | - Jingfeng He
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, People's Republic of China
| | - Qi Huang
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, People's Republic of China
| | - Jingyi Zeng
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, People's Republic of China
| | - Mika Pan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, People's Republic of China
| | - Yujia Mei
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, People's Republic of China.
| | - Jiefeng Luo
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, People's Republic of China.
| | - Donghua Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, People's Republic of China.
| |
Collapse
|
7
|
Pandya VA, Patani R. The role of glial cells in amyotrophic lateral sclerosis. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:381-450. [PMID: 38802179 DOI: 10.1016/bs.irn.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) has traditionally been considered a neuron-centric disease. This view is now outdated, with increasing recognition of cell autonomous and non-cell autonomous contributions of central and peripheral nervous system glia to ALS pathomechanisms. With glial research rapidly accelerating, we comprehensively interrogate the roles of astrocytes, microglia, oligodendrocytes, ependymal cells, Schwann cells and satellite glia in nervous system physiology and ALS-associated pathology. Moreover, we highlight the inter-glial, glial-neuronal and inter-system polylogue which constitutes the healthy nervous system and destabilises in disease. We also propose classification based on function for complex glial reactive phenotypes and discuss the pre-requisite for integrative modelling to advance translation. Given the paucity of life-enhancing therapies currently available for ALS patients, we discuss the promising potential of harnessing glia in driving ALS therapeutic discovery.
Collapse
Affiliation(s)
- Virenkumar A Pandya
- University College London Medical School, London, United Kingdom; The Francis Crick Institute, London, United Kingdom.
| | - Rickie Patani
- The Francis Crick Institute, London, United Kingdom; Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, Queen Square, London, United Kingdom.
| |
Collapse
|
8
|
Goldstein LB, Toth PP, Dearborn-Tomazos JL, Giugliano RP, Hirsh BJ, Peña JM, Selim MH, Woo D. Aggressive LDL-C Lowering and the Brain: Impact on Risk for Dementia and Hemorrhagic Stroke: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol 2023; 43:e404-e442. [PMID: 37706297 DOI: 10.1161/atv.0000000000000164] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
The objective of this scientific statement is to evaluate contemporary evidence that either supports or refutes the conclusion that aggressive low-density lipoprotein cholesterol lowering or lipid lowering exerts toxic effects on the brain, leading to cognitive impairment or dementia or hemorrhagic stroke. The writing group used literature reviews, references to published clinical and epidemiology studies, clinical and public health guidelines, authoritative statements, and expert opinion to summarize existing evidence and to identify gaps in current knowledge. Although some retrospective, case control, and prospective longitudinal studies suggest that statins and low-density lipoprotein cholesterol lowering are associated with cognitive impairment or dementia, the preponderance of observational studies and data from randomized trials do not support this conclusion. The risk of a hemorrhagic stroke associated with statin therapy in patients without a history of cerebrovascular disease is nonsignificant, and achieving very low levels of low-density lipoprotein cholesterol does not increase that risk. Data reflecting the risk of hemorrhagic stroke with lipid-lowering treatment among patients with a history of hemorrhagic stroke are not robust and require additional focused study.
Collapse
|
9
|
Koufi FD, Neri I, Ramazzotti G, Rusciano I, Mongiorgi S, Marvi MV, Fazio A, Shin M, Kosodo Y, Cani I, Giorgio E, Cortelli P, Manzoli L, Ratti S. Lamin B1 as a key modulator of the developing and aging brain. Front Cell Neurosci 2023; 17:1263310. [PMID: 37720548 PMCID: PMC10501396 DOI: 10.3389/fncel.2023.1263310] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 08/17/2023] [Indexed: 09/19/2023] Open
Abstract
Lamin B1 is an essential protein of the nuclear lamina that plays a crucial role in nuclear function and organization. It has been demonstrated that lamin B1 is essential for organogenesis and particularly brain development. The important role of lamin B1 in physiological brain development and aging has only recently been at the epicenter of attention and is yet to be fully elucidated. Regarding the development of brain, glial cells that have long been considered as supporting cells to neurons have overturned this representation and current findings have displayed their active roles in neurogenesis and cerebral development. Although lamin B1 has increased levels during the differentiation of the brain cells, during aging these levels drop leading to senescent phenotypes and inciting neurodegenerative disorders such as Alzheimer's and Parkinson's disease. On the other hand, overexpression of lamin B1 leads to the adult-onset neurodegenerative disease known as Autosomal Dominant Leukodystrophy. This review aims at highlighting the importance of balancing lamin B1 levels in glial cells and neurons from brain development to aging.
Collapse
Affiliation(s)
- Foteini-Dionysia Koufi
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences (DIBINEM), Anatomy Centre, University of Bologna, Bologna, Italy
| | - Irene Neri
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences (DIBINEM), Anatomy Centre, University of Bologna, Bologna, Italy
| | - Giulia Ramazzotti
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences (DIBINEM), Anatomy Centre, University of Bologna, Bologna, Italy
| | - Isabella Rusciano
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences (DIBINEM), Anatomy Centre, University of Bologna, Bologna, Italy
| | - Sara Mongiorgi
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences (DIBINEM), Anatomy Centre, University of Bologna, Bologna, Italy
| | - Maria Vittoria Marvi
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences (DIBINEM), Anatomy Centre, University of Bologna, Bologna, Italy
| | - Antonietta Fazio
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences (DIBINEM), Anatomy Centre, University of Bologna, Bologna, Italy
| | - Minkyung Shin
- Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Yoichi Kosodo
- Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Ilaria Cani
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Elisa Giorgio
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Medical Genetics Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Pietro Cortelli
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Lucia Manzoli
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences (DIBINEM), Anatomy Centre, University of Bologna, Bologna, Italy
| | - Stefano Ratti
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences (DIBINEM), Anatomy Centre, University of Bologna, Bologna, Italy
| |
Collapse
|
10
|
Maitre M, Jeltsch-David H, Okechukwu NG, Klein C, Patte-Mensah C, Mensah-Nyagan AG. Myelin in Alzheimer's disease: culprit or bystander? Acta Neuropathol Commun 2023; 11:56. [PMID: 37004127 PMCID: PMC10067200 DOI: 10.1186/s40478-023-01554-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with neuronal and synaptic losses due to the accumulation of toxic amyloid β (Αβ) peptide oligomers, plaques, and tangles containing tau (tubulin-associated unit) protein. While familial AD is caused by specific mutations, the sporadic disease is more common and appears to result from a complex chronic brain neuroinflammation with mitochondriopathies, inducing free radicals' accumulation. In aged brain, mutations in DNA and several unfolded proteins participate in a chronic amyloidosis response with a toxic effect on myelin sheath and axons, leading to cognitive deficits and dementia. Αβ peptides are the most frequent form of toxic amyloid oligomers. Accumulations of misfolded proteins during several years alters different metabolic mechanisms, induce chronic inflammatory and immune responses with toxic consequences on neuronal cells. Myelin composition and architecture may appear to be an early target for the toxic activity of Aβ peptides and others hydrophobic misfolded proteins. In this work, we describe the possible role of early myelin alterations in the genesis of neuronal alterations and the onset of symptomatology. We propose that some pathophysiological and clinical forms of the disease may arise from structural and metabolic disorders in the processes of myelination/demyelination of brain regions where the accumulation of non-functional toxic proteins is important. In these forms, the primacy of the deleterious role of amyloid peptides would be a matter of questioning and the initiating role of neuropathology would be primarily the fact of dysmyelination.
Collapse
Affiliation(s)
- Michel Maitre
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France.
| | - Hélène Jeltsch-David
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
- Biotechnologie et signalisation cellulaire, UMR 7242 CNRS, Université de Strasbourg, 300 Boulevard Sébastien Brant CS 10413, Illkirch cedex, 67412, France
| | - Nwife Getrude Okechukwu
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| | - Christian Klein
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| | - Christine Patte-Mensah
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| | - Ayikoe-Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| |
Collapse
|
11
|
Trangle SS, Rosenberg T, Parnas H, Levy G, Bar E, Marco A, Barak B. In individuals with Williams syndrome, dysregulation of methylation in non-coding regions of neuronal and oligodendrocyte DNA is associated with pathology and cortical development. Mol Psychiatry 2023; 28:1112-1127. [PMID: 36577841 DOI: 10.1038/s41380-022-01921-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 12/03/2022] [Accepted: 12/12/2022] [Indexed: 12/29/2022]
Abstract
Williams syndrome (WS) is a neurodevelopmental disorder caused by a heterozygous micro-deletion in the WS critical region (WSCR) and is characterized by hyper-sociability and neurocognitive abnormalities. Nonetheless, whether and to what extent WSCR deletion leads to epigenetic modifications in the brain and induces pathological outcomes remains largely unknown. By examining DNA methylation in frontal cortex, we revealed genome-wide disruption in the methylome of individuals with WS, as compared to typically developed (TD) controls. Surprisingly, differentially methylated sites were predominantly annotated as introns and intergenic loci and were found to be highly enriched around binding sites for transcription factors that regulate neuronal development, plasticity and cognition. Moreover, by utilizing enhancer-promoter interactome data, we confirmed that most of these loci function as active enhancers in the human brain or as target genes of transcriptional networks associated with myelination, oligodendrocyte (OL) differentiation, cognition and social behavior. Cell type-specific methylation analysis revealed aberrant patterns in the methylation of active enhancers in neurons and OLs, and important neuron-glia interactions that might be impaired in individuals with WS. Finally, comparison of methylation profiles from blood samples of individuals with WS and healthy controls, along with other data collected in this study, identified putative targets of endophenotypes associated with WS, which can be used to define brain-risk loci for WS outside the WSCR locus, as well as for other associated pathologies. In conclusion, our study illuminates the brain methylome landscape of individuals with WS and sheds light on how these aberrations might be involved in social behavior and physiological abnormalities. By extension, these results may lead to better diagnostics and more refined therapeutic targets for WS.
Collapse
Affiliation(s)
- Sari Schokoroy Trangle
- The School of Psychological Sciences, Faculty of Social Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Tali Rosenberg
- Neuro-Epigenetics Laboratory, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 7610001, Israel
| | - Hadar Parnas
- Neuro-Epigenetics Laboratory, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 7610001, Israel
| | - Gilad Levy
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Ela Bar
- The School of Psychological Sciences, Faculty of Social Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel.,The School of Neurobiology, Biochemistry & Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Asaf Marco
- Neuro-Epigenetics Laboratory, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 7610001, Israel.
| | - Boaz Barak
- The School of Psychological Sciences, Faculty of Social Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel. .,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel.
| |
Collapse
|
12
|
Kang H, Liu Q, Seim I, Zhang W, Li H, Gao H, Lin W, Lin M, Zhang P, Zhang Y, Gao H, Wang Y, Qin Y, Liu M, Dong L, Yang Z, Zhang Y, Han L, Fan G, Li S. A genome and single-nucleus cerebral cortex transcriptome atlas of the short-finned pilot whale Globicephala macrorhynchus. Mol Ecol Resour 2023. [PMID: 36826393 DOI: 10.1111/1755-0998.13775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 02/09/2023] [Accepted: 02/22/2023] [Indexed: 02/25/2023]
Abstract
Cetaceans (dolphins, whales, and porpoises) have large and anatomically sophisticated brains. To expand our understanding of the cellular makeup of cetacean brains and the similarities and divergence between the brains of cetaceans and terrestrial mammals, we report a short-finned pilot whale (Globicephala macrorhynchus) single-nucleus transcriptome atlas. To achieve this goal, we assembled a chromosome-scale reference genome spanning 2.25 Gb on 22 chromosomes and profiled the gene expression of five major anatomical cortical regions of the short-finned pilot whale by single-nucleus RNA-sequencing (snRNA-seq). We identified six major cell lineages in the cerebral cortex (excitatory neurons, inhibitory neurons, oligodendrocytes, oligodendrocyte precursor cells, astrocytes, and endothelial cells), eight molecularly distinct subclusters of excitatory neurons, and four subclusters of inhibitory neurons. Finally, a comparison of snRNA-seq data from the short-finned pilot whale, human, and rhesus macaque revealed a broadly conserved cellular makeup of brain cell types. Our study provides genomic resources and molecular insights into cetacean brain evolution.
Collapse
Affiliation(s)
- Hui Kang
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qun Liu
- BGI-Qingdao, BGI-Shenzhen, Qingdao, China.,Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Sciences, BGI-Qingdao, Qingdao, China.,Department of Biology, University of Copenhagen, Copenhagen, Denmark.,Qingdao Key Laboratory of Marine Genomics, BGI-Qingdao, Qingdao, China
| | - Inge Seim
- Integrative Biology Laboratory, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Wenwei Zhang
- State Key Laboratory of Agricultural Genomics, BGI-Shenzhen, Shenzhen, China
| | - Hanbo Li
- BGI-Qingdao, BGI-Shenzhen, Qingdao, China.,Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Sciences, BGI-Qingdao, Qingdao, China
| | - Haiyu Gao
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wenzhi Lin
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
| | - Mingli Lin
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
| | - Peijun Zhang
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
| | | | | | - Yang Wang
- BGI-Qingdao, BGI-Shenzhen, Qingdao, China
| | - Yating Qin
- BGI-Qingdao, BGI-Shenzhen, Qingdao, China
| | - Mingming Liu
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
| | - Lijun Dong
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
| | - Zixin Yang
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
| | | | - Lei Han
- State Key Laboratory of Agricultural Genomics, BGI-Shenzhen, Shenzhen, China
| | - Guangyi Fan
- BGI-Qingdao, BGI-Shenzhen, Qingdao, China.,State Key Laboratory of Agricultural Genomics, BGI-Shenzhen, Shenzhen, China
| | - Songhai Li
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
| |
Collapse
|
13
|
Pijuan I, Balducci E, Soto-Sánchez C, Fernández E, Barallobre MJ, Arbonés ML. Impaired macroglial development and axonal conductivity contributes to the neuropathology of DYRK1A-related intellectual disability syndrome. Sci Rep 2022; 12:19912. [PMID: 36402907 PMCID: PMC9675854 DOI: 10.1038/s41598-022-24284-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022] Open
Abstract
The correct development and activity of neurons and glial cells is necessary to establish proper brain connectivity. DYRK1A encodes a protein kinase involved in the neuropathology associated with Down syndrome that influences neurogenesis and the morphological differentiation of neurons. DYRK1A loss-of-function mutations in heterozygosity cause a well-recognizable syndrome of intellectual disability and autism spectrum disorder. In this study, we analysed the developmental trajectories of macroglial cells and the properties of the corpus callosum, the major white matter tract of the brain, in Dyrk1a+/- mice, a mouse model that recapitulates the main neurological features of DYRK1A syndrome. We found that Dyrk1a+/- haploinsufficient mutants present an increase in astrogliogenesis in the neocortex and a delay in the production of cortical oligodendrocyte progenitor cells and their progression along the oligodendroglial lineage. There were fewer myelinated axons in the corpus callosum of Dyrk1a+/- mice, axons that are thinner and with abnormal nodes of Ranvier. Moreover, action potential propagation along myelinated and unmyelinated callosal axons was slower in Dyrk1a+/- mutants. All these alterations are likely to affect neuronal circuit development and alter network synchronicity, influencing higher brain functions. These alterations highlight the relevance of glial cell abnormalities in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Isabel Pijuan
- grid.4711.30000 0001 2183 4846Instituto de Biología Molecular de Barcelona (IBMB), Spanish National Research Council (CSIC), 08028 Barcelona, Spain ,grid.452372.50000 0004 1791 1185Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08028 Barcelona, Spain
| | - Elisa Balducci
- grid.4711.30000 0001 2183 4846Instituto de Biología Molecular de Barcelona (IBMB), Spanish National Research Council (CSIC), 08028 Barcelona, Spain ,grid.452372.50000 0004 1791 1185Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08028 Barcelona, Spain
| | - Cristina Soto-Sánchez
- grid.26811.3c0000 0001 0586 4893Instituto de Bioingeniería, Miguel Hernández University, 03202 Elche, Spain ,grid.429738.30000 0004 1763 291XCentro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 03202 Elche, Spain
| | - Eduardo Fernández
- grid.26811.3c0000 0001 0586 4893Instituto de Bioingeniería, Miguel Hernández University, 03202 Elche, Spain ,grid.429738.30000 0004 1763 291XCentro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 03202 Elche, Spain
| | - María José Barallobre
- grid.4711.30000 0001 2183 4846Instituto de Biología Molecular de Barcelona (IBMB), Spanish National Research Council (CSIC), 08028 Barcelona, Spain ,grid.452372.50000 0004 1791 1185Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08028 Barcelona, Spain
| | - Maria L. Arbonés
- grid.4711.30000 0001 2183 4846Instituto de Biología Molecular de Barcelona (IBMB), Spanish National Research Council (CSIC), 08028 Barcelona, Spain ,grid.452372.50000 0004 1791 1185Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08028 Barcelona, Spain
| |
Collapse
|
14
|
Bloomingdale P, Karelina T, Ramakrishnan V, Bakshi S, Véronneau‐Veilleux F, Moye M, Sekiguchi K, Meno‐Tetang G, Mohan A, Maithreye R, Thomas VA, Gibbons F, Cabal A, Bouteiller J, Geerts H. Hallmarks of neurodegenerative disease: A systems pharmacology perspective. CPT Pharmacometrics Syst Pharmacol 2022; 11:1399-1429. [PMID: 35894182 PMCID: PMC9662204 DOI: 10.1002/psp4.12852] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 11/09/2022] Open
Abstract
Age-related central neurodegenerative diseases, such as Alzheimer's and Parkinson's disease, are a rising public health concern and have been plagued by repeated drug development failures. The complex nature and poor mechanistic understanding of the etiology of neurodegenerative diseases has hindered the discovery and development of effective disease-modifying therapeutics. Quantitative systems pharmacology models of neurodegeneration diseases may be useful tools to enhance the understanding of pharmacological intervention strategies and to reduce drug attrition rates. Due to the similarities in pathophysiological mechanisms across neurodegenerative diseases, especially at the cellular and molecular levels, we envision the possibility of structural components that are conserved across models of neurodegenerative diseases. Conserved structural submodels can be viewed as building blocks that are pieced together alongside unique disease components to construct quantitative systems pharmacology (QSP) models of neurodegenerative diseases. Model parameterization would likely be different between the different types of neurodegenerative diseases as well as individual patients. Formulating our mechanistic understanding of neurodegenerative pathophysiology as a mathematical model could aid in the identification and prioritization of drug targets and combinatorial treatment strategies, evaluate the role of patient characteristics on disease progression and therapeutic response, and serve as a central repository of knowledge. Here, we provide a background on neurodegenerative diseases, highlight hallmarks of neurodegeneration, and summarize previous QSP models of neurodegenerative diseases.
Collapse
Affiliation(s)
- Peter Bloomingdale
- Quantitative Pharmacology and PharmacometricsMerck & Co., Inc.BostonMassachusettsUSA
| | | | | | - Suruchi Bakshi
- Certara QSPOssThe Netherlands,Certara QSPPrincetonNew JerseyUSA
| | | | - Matthew Moye
- Quantitative Pharmacology and PharmacometricsMerck & Co., Inc.BostonMassachusettsUSA
| | - Kazutaka Sekiguchi
- Shionogi & Co., Ltd.OsakaJapan,SUNY Downstate Medical CenterNew YorkNew YorkUSA
| | | | | | | | | | - Frank Gibbons
- Clinical Pharmacology and PharmacometricsBiogenCambridgeMassachusettsUSA
| | | | - Jean‐Marie Bouteiller
- Center for Neural EngineeringDepartment of Biomedical Engineering at the Viterbi School of EngineeringLos AngelesCaliforniaUSA,Institute for Technology and Medical Systems Innovation, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | | |
Collapse
|
15
|
Vyushina AV, Pritvorova AV, Semenova OG, Ordyan NE. The Effect of Prenatal Stress on Glutathione-Associated Antioxidant Enzyme Activity in Subcellular Fractions of Neocortical Neurons and Neuroglia of Rats during the Period of Intensive Myelination. NEUROCHEM J+ 2022. [DOI: 10.1134/s1819712422030138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
16
|
Scalabrino G. Epidermal Growth Factor in the CNS: A Beguiling Journey from Integrated Cell Biology to Multiple Sclerosis. An Extensive Translational Overview. Cell Mol Neurobiol 2022; 42:891-916. [PMID: 33151415 PMCID: PMC8942922 DOI: 10.1007/s10571-020-00989-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022]
Abstract
This article reviews the wealth of papers dealing with the different effects of epidermal growth factor (EGF) on oligodendrocytes, astrocytes, neurons, and neural stem cells (NSCs). EGF induces the in vitro and in vivo proliferation of NSCs, their migration, and their differentiation towards the neuroglial cell line. It interacts with extracellular matrix components. NSCs are distributed in different CNS areas, serve as a reservoir of multipotent cells, and may be increased during CNS demyelinating diseases. EGF has pleiotropic differentiative and proliferative effects on the main CNS cell types, particularly oligodendrocytes and their precursors, and astrocytes. EGF mediates the in vivo myelinotrophic effect of cobalamin on the CNS, and modulates the synthesis and levels of CNS normal prions (PrPCs), both of which are indispensable for myelinogenesis and myelin maintenance. EGF levels are significantly lower in the cerebrospinal fluid and spinal cord of patients with multiple sclerosis (MS), which probably explains remyelination failure, also because of the EGF marginal role in immunology. When repeatedly administered, EGF protects mouse spinal cord from demyelination in various experimental models of autoimmune encephalomyelitis. It would be worth further investigating the role of EGF in the pathogenesis of MS because of its multifarious effects.
Collapse
Affiliation(s)
- Giuseppe Scalabrino
- Department of Biomedical Sciences, University of Milan, Via Mangiagalli 31, 20133, Milan, Italy.
| |
Collapse
|
17
|
Scalabrino G. Newly Identified Deficiencies in the Multiple Sclerosis Central Nervous System and Their Impact on the Remyelination Failure. Biomedicines 2022; 10:biomedicines10040815. [PMID: 35453565 PMCID: PMC9026986 DOI: 10.3390/biomedicines10040815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
The pathogenesis of multiple sclerosis (MS) remains enigmatic and controversial. Myelin sheaths in the central nervous system (CNS) insulate axons and allow saltatory nerve conduction. MS brings about the destruction of myelin sheaths and the myelin-producing oligodendrocytes (ODCs). The conundrum of remyelination failure is, therefore, crucial in MS. In this review, the roles of epidermal growth factor (EGF), normal prions, and cobalamin in CNS myelinogenesis are briefly summarized. Thereafter, some findings of other authors and ourselves on MS and MS-like models are recapitulated, because they have shown that: (a) EGF is significantly decreased in the CNS of living or deceased MS patients; (b) its repeated administration to mice in various MS-models prevents demyelination and inflammatory reaction; (c) as was the case for EGF, normal prion levels are decreased in the MS CNS, with a strong correspondence between liquid and tissue levels; and (d) MS cobalamin levels are increased in the cerebrospinal fluid, but decreased in the spinal cord. In fact, no remyelination can occur in MS if these molecules (essential for any form of CNS myelination) are lacking. Lastly, other non-immunological MS abnormalities are reviewed. Together, these results have led to a critical reassessment of MS pathogenesis, partly because EGF has little or no role in immunology.
Collapse
Affiliation(s)
- Giuseppe Scalabrino
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| |
Collapse
|
18
|
Gray VP, Amelung CD, Duti IJ, Laudermilch EG, Letteri RA, Lampe KJ. Biomaterials via peptide assembly: Design, characterization, and application in tissue engineering. Acta Biomater 2022; 140:43-75. [PMID: 34710626 PMCID: PMC8829437 DOI: 10.1016/j.actbio.2021.10.030] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/23/2021] [Accepted: 10/20/2021] [Indexed: 12/16/2022]
Abstract
A core challenge in biomaterials, with both fundamental significance and technological relevance, concerns the rational design of bioactive microenvironments. Designed properly, peptides can undergo supramolecular assembly into dynamic, physical hydrogels that mimic the mechanical, topological, and biochemical features of native tissue microenvironments. The relatively facile, inexpensive, and automatable preparation of peptides, coupled with low batch-to-batch variability, motivates the expanded use of assembling peptide hydrogels for biomedical applications. Integral to realizing dynamic peptide assemblies as functional biomaterials for tissue engineering is an understanding of the molecular and macroscopic features that govern assembly, morphology, and biological interactions. In this review, we first discuss the design of assembling peptides, including primary structure (sequence), secondary structure (e.g., α-helix and β-sheets), and molecular interactions that facilitate assembly into multiscale materials with desired properties. Next, we describe characterization tools for elucidating molecular structure and interactions, morphology, bulk properties, and biological functionality. Understanding of these characterization methods enables researchers to access a variety of approaches in this ever-expanding field. Finally, we discuss the biological properties and applications of peptide-based biomaterials for engineering several important tissues. By connecting molecular features and mechanisms of assembling peptides to the material and biological properties, we aim to guide the design and characterization of peptide-based biomaterials for tissue engineering and regenerative medicine. STATEMENT OF SIGNIFICANCE: Engineering peptide-based biomaterials that mimic the topological and mechanical properties of natural extracellular matrices provide excellent opportunities to direct cell behavior for regenerative medicine and tissue engineering. Here we review the molecular-scale features of assembling peptides that result in biomaterials that exhibit a variety of relevant extracellular matrix-mimetic properties and promote beneficial cell-biomaterial interactions. Aiming to inspire and guide researchers approaching this challenge from both the peptide biomaterial design and tissue engineering perspectives, we also present characterization tools for understanding the connection between peptide structure and properties and highlight the use of peptide-based biomaterials in neural, orthopedic, cardiac, muscular, and immune engineering applications.
Collapse
Affiliation(s)
- Vincent P Gray
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Connor D Amelung
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Israt Jahan Duti
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Emma G Laudermilch
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Rachel A Letteri
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States.
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, United States.
| |
Collapse
|
19
|
Rigby MJ, Orefice NS, Lawton AJ, Ma M, Shapiro SL, Yi SY, Dieterich IA, Frelka A, Miles HN, Pearce RA, Yu JPJ, Li L, Denu JM, Puglielli L. SLC13A5/sodium-citrate co-transporter overexpression causes disrupted white matter integrity and an autistic-like phenotype. Brain Commun 2022; 4:fcac002. [PMID: 35146426 PMCID: PMC8823335 DOI: 10.1093/braincomms/fcac002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/19/2021] [Accepted: 01/03/2022] [Indexed: 09/11/2023] Open
Abstract
Endoplasmic reticulum-based N ɛ-lysine acetylation serves as an important protein quality control system for the secretory pathway. Dysfunctional endoplasmic reticulum-based acetylation, as caused by overexpression of the acetyl coenzyme A transporter AT-1 in the mouse, results in altered glycoprotein flux through the secretory pathway and an autistic-like phenotype. AT-1 works in concert with SLC25A1, the citrate/malate antiporter in the mitochondria, SLC13A5, the plasma membrane sodium/citrate symporter and ATP citrate lyase, the cytosolic enzyme that converts citrate into acetyl coenzyme A. Here, we report that mice with neuron-specific overexpression of SLC13A5 exhibit autistic-like behaviours with a jumping stereotypy. The mice displayed disrupted white matter integrity and altered synaptic structure and function. Analysis of both the proteome and acetyl-proteome revealed unique adaptations in the hippocampus and cortex, highlighting a metabolic response that likely plays an important role in the SLC13A5 neuron transgenic phenotype. Overall, our results support a mechanistic link between aberrant intracellular citrate/acetyl coenzyme A flux and the development of an autistic-like phenotype.
Collapse
Affiliation(s)
- Michael J. Rigby
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Nicola Salvatore Orefice
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Alexis J. Lawton
- Department of Biomolecular Chemistry and the Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Min Ma
- School of Pharmacy and Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Samantha L. Shapiro
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sue Y. Yi
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Inca A. Dieterich
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Alyssa Frelka
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Hannah N. Miles
- School of Pharmacy and Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Robert A. Pearce
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - John Paul J. Yu
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Lingjun Li
- School of Pharmacy and Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - John M. Denu
- Department of Biomolecular Chemistry and the Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Geriatric Research Education Clinical Center, Veterans Affairs Medical Center, Madison, WI 53705, USA
| |
Collapse
|
20
|
Salari V, Rodrigues S, Saglamyurek E, Simon C, Oblak D. Are Brain-Computer Interfaces Feasible With Integrated Photonic Chips? Front Neurosci 2022; 15:780344. [PMID: 35069099 PMCID: PMC8777191 DOI: 10.3389/fnins.2021.780344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
The present paper examines the viability of a radically novel idea for brain-computer interface (BCI), which could lead to novel technological, experimental, and clinical applications. BCIs are computer-based systems that enable either one-way or two-way communication between a living brain and an external machine. BCIs read-out brain signals and transduce them into task commands, which are performed by a machine. In closed loop, the machine can stimulate the brain with appropriate signals. In recent years, it has been shown that there is some ultraweak light emission from neurons within or close to the visible and near-infrared parts of the optical spectrum. Such ultraweak photon emission (UPE) reflects the cellular (and body) oxidative status, and compelling pieces of evidence are beginning to emerge that UPE may well play an informational role in neuronal functions. In fact, several experiments point to a direct correlation between UPE intensity and neural activity, oxidative reactions, EEG activity, cerebral blood flow, cerebral energy metabolism, and release of glutamate. Therefore, we propose a novel skull implant BCI that uses UPE. We suggest that a photonic integrated chip installed on the interior surface of the skull may enable a new form of extraction of the relevant features from the UPE signals. In the current technology landscape, photonic technologies are advancing rapidly and poised to overtake many electrical technologies, due to their unique advantages, such as miniaturization, high speed, low thermal effects, and large integration capacity that allow for high yield, volume manufacturing, and lower cost. For our proposed BCI, we are making some very major conjectures, which need to be experimentally verified, and therefore we discuss the controversial parts, feasibility of technology and limitations, and potential impact of this envisaged technology if successfully implemented in the future.
Collapse
Affiliation(s)
- Vahid Salari
- Basque Center for Applied Mathematics (BCAM), Bilbao, Spain
- Quantum Biology Laboratory, Howard University, Washington, DC, United States
| | | | - Erhan Saglamyurek
- Department of Physics and Astronomy, University of Calgary, Calgary, AB, Canada
- Department of Physics, University of Alberta, Edmonton, AB, Canada
- Institute for Quantum Science and Technology, University of Calgary, Calgary, AB, Canada
| | - Christoph Simon
- Department of Physics and Astronomy, University of Calgary, Calgary, AB, Canada
- Institute for Quantum Science and Technology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Daniel Oblak
- Department of Physics and Astronomy, University of Calgary, Calgary, AB, Canada
- Institute for Quantum Science and Technology, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
21
|
OUP accepted manuscript. Brain 2022; 145:1581-1583. [DOI: 10.1093/brain/awac141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 04/08/2022] [Indexed: 11/14/2022] Open
|
22
|
Nassir N, Bankapur A, Samara B, Ali A, Ahmed A, Inuwa IM, Zarrei M, Safizadeh Shabestari SA, AlBanna A, Howe JL, Berdiev BK, Scherer SW, Woodbury-Smith M, Uddin M. Single-cell transcriptome identifies molecular subtype of autism spectrum disorder impacted by de novo loss-of-function variants regulating glial cells. Hum Genomics 2021; 15:68. [PMID: 34802461 PMCID: PMC8607722 DOI: 10.1186/s40246-021-00368-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/05/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In recent years, several hundred autism spectrum disorder (ASD) implicated genes have been discovered impacting a wide range of molecular pathways. However, the molecular underpinning of ASD, particularly from the point of view of 'brain to behaviour' pathogenic mechanisms, remains largely unknown. METHODS We undertook a study to investigate patterns of spatiotemporal and cell type expression of ASD-implicated genes by integrating large-scale brain single-cell transcriptomes (> million cells) and de novo loss-of-function (LOF) ASD variants (impacting 852 genes from 40,122 cases). RESULTS We identified multiple single-cell clusters from three distinct developmental human brain regions (anterior cingulate cortex, middle temporal gyrus and primary visual cortex) that evidenced high evolutionary constraint through enrichment for brain critical exons and high pLI genes. These clusters also showed significant enrichment with ASD loss-of-function variant genes (p < 5.23 × 10-11) that are transcriptionally highly active in prenatal brain regions (visual cortex and dorsolateral prefrontal cortex). Mapping ASD de novo LOF variant genes into large-scale human and mouse brain single-cell transcriptome analysis demonstrate enrichment of such genes into neuronal subtypes and are also enriched for subtype of non-neuronal glial cell types (astrocyte, p < 6.40 × 10-11, oligodendrocyte, p < 1.31 × 10-09). CONCLUSION Among the ASD genes enriched with pathogenic de novo LOF variants (i.e. KANK1, PLXNB1), a subgroup has restricted transcriptional regulation in non-neuronal cell types that are evolutionarily conserved. This association strongly suggests the involvement of subtype of non-neuronal glial cells in the pathogenesis of ASD and the need to explore other biological pathways for this disorder.
Collapse
Affiliation(s)
- Nasna Nassir
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Asma Bankapur
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Bisan Samara
- Biomedical Engineering Department, McGill University, Montréal, QC, Canada
| | - Abdulrahman Ali
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Awab Ahmed
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Ibrahim M Inuwa
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Mehdi Zarrei
- The Centre for Applied Genomics (TCAG), The Hospital for Sick Children, Toronto, ON, Canada.,Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | | | - Ammar AlBanna
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE.,The Mental Health Center of Excellence, Al Jalila Children's Speciality Hospital, Dubai, UAE
| | - Jennifer L Howe
- The Centre for Applied Genomics (TCAG), The Hospital for Sick Children, Toronto, ON, Canada.,Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Bakhrom K Berdiev
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Stephen W Scherer
- The Centre for Applied Genomics (TCAG), The Hospital for Sick Children, Toronto, ON, Canada.,Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Marc Woodbury-Smith
- The Centre for Applied Genomics (TCAG), The Hospital for Sick Children, Toronto, ON, Canada.,Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Mohammed Uddin
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE. .,Cellular Intelligence (Ci) Lab, GenomeArc Inc., Toronto, ON, Canada.
| |
Collapse
|
23
|
Steinberg DJ, Aqeilan RI. WWOX-Related Neurodevelopmental Disorders: Models and Future Perspectives. Cells 2021; 10:cells10113082. [PMID: 34831305 PMCID: PMC8623516 DOI: 10.3390/cells10113082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 11/03/2021] [Indexed: 12/12/2022] Open
Abstract
The WW domain-containing oxidoreductase (WWOX) gene was originally discovered as a putative tumor suppressor spanning the common fragile site FRA16D, but as time has progressed the extent of its pleiotropic function has become apparent. At present, WWOX is a major source of interest in the context of neurological disorders, and more specifically developmental and epileptic encephalopathies (DEEs). This review article aims to introduce the many model systems used through the years to study its function and roles in neuropathies. Similarities and fundamental differences between rodent and human models are discussed. Finally, future perspectives and promising research avenues are suggested.
Collapse
|
24
|
Neurodevelopmental toxicity assessment of flame retardants using a human DNT in vitro testing battery. Cell Biol Toxicol 2021; 38:781-807. [PMID: 33969458 PMCID: PMC9525352 DOI: 10.1007/s10565-021-09603-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/11/2021] [Indexed: 12/12/2022]
Abstract
Due to their neurodevelopmental toxicity, flame retardants (FRs) like polybrominated diphenyl ethers are banned from the market and replaced by alternative FRs, like organophosphorus FRs, that have mostly unknown toxicological profiles. To study their neurodevelopmental toxicity, we evaluated the hazard of several FRs including phased-out polybrominated FRs and organophosphorus FRs: 2,2′,4,4′-tetrabromodiphenylether (BDE-47), 2,2′,4,4′,5-pentabromodiphenylether (BDE-99), tetrabromobisphenol A, triphenyl phosphate, tris(2-butoxyethyl) phosphate and its metabolite bis-(2-butoxyethyl) phosphate, isodecyl diphenyl phosphate, triphenyl isopropylated phosphate, tricresyl phosphate, tris(1,3-dichloro-2-propyl) phosphate, tert-butylphenyl diphenyl phosphate, 2-ethylhexyl diphenyl phosphate, tris(1-chloroisopropyl) phosphate, and tris(2-chloroethyl) phosphate. Therefore, we used a human cell–based developmental neurotoxicity (DNT) in vitro battery covering a large variety of neurodevelopmental endpoints. Potency according to the respective most sensitive benchmark concentration (BMC) across the battery ranked from <1 μM (5 FRs), 1<10 μM (7 FRs) to the >10 μM range (3 FRs). Evaluation of the data with the ToxPi tool revealed a distinct ranking (a) than with the BMC and (b) compared to the ToxCast data, suggesting that DNT hazard of these FRs is not well predicted by ToxCast assays. Extrapolating the DNT in vitro battery BMCs to human FR exposure via breast milk suggests low risk for individual compounds. However, it raises a potential concern for real-life mixture exposure, especially when different compounds converge through diverse modes-of-action on common endpoints, like oligodendrocyte differentiation in this study. This case study using FRs suggests that human cell–based DNT in vitro battery is a promising approach for neurodevelopmental hazard assessment and compound prioritization in risk assessment.
Collapse
|
25
|
Seo S, Choi CH, Yi KS, Kim SU, Lee K, Choi N, Lee HJ, Cha SH, Kim HN. An engineered neurovascular unit for modeling neuroinflammation. Biofabrication 2021; 13. [PMID: 33849004 DOI: 10.1088/1758-5090/abf741] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/13/2021] [Indexed: 12/25/2022]
Abstract
The neurovascular unit (NVU) comprises multiple types of brain cells, including brain endothelial cells, astrocytes, pericytes, neurons, microglia, and oligodendrocytes. Each cell type contributes to the maintenance of the molecular transport barrier and brain tissue homeostasis. Several disorders and diseases of the central nervous system, including neuroinflammation, Alzheimer's disease, stroke, and multiple sclerosis, have been associated with dysfunction of the NVU. As a result, there has been increased demand for the development of NVUin vitromodels. Here, we present a three-dimensional (3D) immortalized human cell-based NVU model generated by organizing the brain microvasculature in a collagen matrix embedded with six different types of cells that comprise the NVU. By surrounding a perfusable brain endothelium with six types of NVU-composing cells, we demonstrated a significant impact of the 3D co-culture on the maturation of barrier function, which is supported by cytokines secreted from NVU-composing cells. Furthermore, NVU-composing cells alleviated the inflammatory responses induced by lipopolysaccharides. Our human cell-based NVUin vitromodel could enable elucidation of both physiological and pathological mechanisms in the human brain and evaluation of safety and efficacy in the context of high-content analysis during the process of drug development.
Collapse
Affiliation(s)
- Suyeong Seo
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.,Program in Nano Science and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea.,These authors contributed equally to this work
| | - Chi-Hoon Choi
- Department of Radiology, Chung Buk National University Hospital, Cheongju, Chung Buk, Republic of Korea.,College of Medicine, Chung Buk National University, Cheongju, Chung Buk 28644, Republic of Korea.,These authors contributed equally to this work
| | - Kyung Sik Yi
- Department of Radiology, Chung Buk National University Hospital, Cheongju, Chung Buk, Republic of Korea
| | - Seung U Kim
- Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Kangwon Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.,Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Hong Jun Lee
- College of Medicine, Chung Buk National University, Cheongju, Chung Buk 28644, Republic of Korea.,Research Institute, eBiogen Inc., Seoul, Republic of Korea
| | - Sang-Hoon Cha
- Department of Radiology, Chung Buk National University Hospital, Cheongju, Chung Buk, Republic of Korea.,College of Medicine, Chung Buk National University, Cheongju, Chung Buk 28644, Republic of Korea
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.,Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| |
Collapse
|
26
|
Repudi S, Steinberg DJ, Elazar N, Breton VL, Aquilino MS, Saleem A, Abu-Swai S, Vainshtein A, Eshed-Eisenbach Y, Vijayaragavan B, Behar O, Hanna JJ, Peles E, Carlen PL, Aqeilan RI. Neuronal deletion of Wwox, associated with WOREE syndrome, causes epilepsy and myelin defects. Brain 2021; 144:3061-3077. [PMID: 33914858 DOI: 10.1093/brain/awab174] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 03/21/2021] [Accepted: 04/16/2021] [Indexed: 11/13/2022] Open
Abstract
WOREE syndrome caused by human germline biallelic mutations in WWOX is a neurodevelopmental disorder characterized by intractable epilepsy, severe developmental delay, ataxia and premature death at the age of 2-4 years. The underlying mechanisms of WWOX actions are poorly understood. In the current study, we show that specific neuronal deletion of murine Wwox produces phenotypes typical of the Wwox-null mutation leading to brain hyperexcitability, intractable epilepsy, ataxia and postnatal lethality. A significant decrease in transcript levels of genes involved in myelination was observed in mouse cortex and hippocampus. Wwox-mutant mice exhibited reduced maturation of oligodendrocytes, reduced myelinated axons and impaired axonal conductivity. Brain hyperexcitability and hypomyelination were also revealed in human brain organoids with a WWOX deletion. These findings provide cellular and molecular evidence for myelination defects and hyperexcitability in the WOREE syndrome linked to neuronal function of WWOX.
Collapse
Affiliation(s)
- Srinivasarao Repudi
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Immunology and Cancer Research-IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Daniel J Steinberg
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Immunology and Cancer Research-IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Nimrod Elazar
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Vanessa L Breton
- Krembil Research Institute, University Health Network, Toronto, Canada
| | - Mark S Aquilino
- Krembil Research Institute, University Health Network, Toronto, Canada
| | - Afifa Saleem
- Krembil Research Institute, University Health Network, Toronto, Canada
| | - Sara Abu-Swai
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Immunology and Cancer Research-IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Anna Vainshtein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yael Eshed-Eisenbach
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Bharath Vijayaragavan
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Oded Behar
- Department of Developmental Biology and Cancer Research-IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Jacob J Hanna
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Elior Peles
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Peter L Carlen
- Krembil Research Institute, University Health Network, Toronto, Canada
| | - Rami I Aqeilan
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Immunology and Cancer Research-IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
27
|
Pantazou V, Roux T, Oliveira Moreira V, Lubetzki C, Desmazières A. Interaction between Neurons and the Oligodendroglial Lineage in Multiple Sclerosis and Its Preclinical Models. Life (Basel) 2021; 11:231. [PMID: 33799653 PMCID: PMC7999210 DOI: 10.3390/life11030231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/06/2021] [Accepted: 03/07/2021] [Indexed: 11/17/2022] Open
Abstract
Multiple sclerosis (MS) is a complex central nervous system inflammatory disease leading to demyelination and associated functional deficits. Though endogenous remyelination exists, it is only partial and, with time, patients can enter a progressive phase of the disease, with neurodegeneration as a hallmark. Though major therapeutic advances have been made, with immunotherapies reducing relapse rate during the inflammatory phase of MS, there is presently no therapy available which significantly impacts disease progression. Remyelination has been shown to favor neuroprotection, and it is thus of major importance to better understand remyelination mechanisms in order to promote them and hence preserve neurons. A crucial point is how this process is regulated through the neuronal crosstalk with the oligodendroglial lineage. In this review, we present the current knowledge on neuron interaction with the oligodendroglial lineage, in physiological context as well as in MS and its experimental models. We further discuss the therapeutic possibilities resulting from this research field, which might allow to support remyelination and neuroprotection and thus limit MS progression.
Collapse
Affiliation(s)
- Vasiliki Pantazou
- Paris Brain Institute (ICM), Sorbonne Université, CNRS, Inserm, GH Pitié-Salpêtrière, 47 boulevard de l’Hôpital, 75013 Paris, France; (V.P.); (T.R.); (V.O.M.); (C.L.)
- Service de Neurologie, Centre Hospitalier Universitaire Vaudois, 46 Rue du Bugnon, 1011 Lausanne, Switzerland
| | - Thomas Roux
- Paris Brain Institute (ICM), Sorbonne Université, CNRS, Inserm, GH Pitié-Salpêtrière, 47 boulevard de l’Hôpital, 75013 Paris, France; (V.P.); (T.R.); (V.O.M.); (C.L.)
- Assistance Publique-Hôpitaux de Paris, Neurology Department, Pitié Salpêtrière University Hospital, 75013 Paris, France
| | - Vanessa Oliveira Moreira
- Paris Brain Institute (ICM), Sorbonne Université, CNRS, Inserm, GH Pitié-Salpêtrière, 47 boulevard de l’Hôpital, 75013 Paris, France; (V.P.); (T.R.); (V.O.M.); (C.L.)
| | - Catherine Lubetzki
- Paris Brain Institute (ICM), Sorbonne Université, CNRS, Inserm, GH Pitié-Salpêtrière, 47 boulevard de l’Hôpital, 75013 Paris, France; (V.P.); (T.R.); (V.O.M.); (C.L.)
- Assistance Publique-Hôpitaux de Paris, Neurology Department, Pitié Salpêtrière University Hospital, 75013 Paris, France
| | - Anne Desmazières
- Paris Brain Institute (ICM), Sorbonne Université, CNRS, Inserm, GH Pitié-Salpêtrière, 47 boulevard de l’Hôpital, 75013 Paris, France; (V.P.); (T.R.); (V.O.M.); (C.L.)
| |
Collapse
|
28
|
Physiological and pathological functions of TMEM106B: a gene associated with brain aging and multiple brain disorders. Acta Neuropathol 2021; 141:327-339. [PMID: 33386471 DOI: 10.1007/s00401-020-02246-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022]
Abstract
TMEM106B, encoding a lysosome membrane protein, has been recently associated with brain aging, hypomyelinating leukodystrophy and multiple neurodegenerative diseases, such as frontotemporal lobar degeneration (FTLD) and limbic-predominant age-related TDP-43 encephalopathy (LATE). During the past decade, considerable progress has been made towards our understanding of the cellular and physiological functions of TMEM106B. TMEM106B regulates many aspects of lysosomal function, including lysosomal pH, lysosome movement, and lysosome exocytosis. Both an increase and decrease in TMEM106B levels result in lysosomal abnormalities. In mouse models, TMEM106B deficiency leads to lysosome trafficking and myelination defects and FTLD related pathology. In humans, alterations in TMEM106B levels or function are intimately linked to neuronal proportions, brain aging, and brain disorders. Further elucidation of the physiological function of TMEM106B and changes in TMEM106B under pathological conditions will facilitate therapeutic development to treat brain disorders associated with TMEM106B.
Collapse
|
29
|
Zareba-Paslawska J, Patra K, Kluzer L, Revesz T, Svenningsson P. Tau Isoform-Driven CBD Pathology Transmission in Oligodendrocytes in Humanized Tau Mice. Front Neurol 2021; 11:589471. [PMID: 33519674 PMCID: PMC7845573 DOI: 10.3389/fneur.2020.589471] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022] Open
Abstract
The aggregation of abnormally phosphorylated tau protein in neurons and glia is a neuropathological hallmark of several neurodegenerative disorders, collectively known as tauopathies. They are further subclassified based on the preferential pathological aggregation of three carboxyl-terminal repeat domains (3R) and/or 4R tau. Corticobasal degeneration (CBD) is a rare neurodegenerative disorder classified as a 4R tauopathy. In the present study, we extend analysis of CBD-tau cell-type specific pathology transmission with 3R and 4R tau isoform distinguishable changes. We use a humanized tau (hTau) mouse line, which overexpress all six human tau isoforms in a murine tau knockout background and perform intrastriatal inoculation of control and CBD-tau enriched human brain homogenate. We show that CBD-tau causes hyperphosphorylation of tau at Ser202 predominantly in oligodendrocytes. Next, we demonstrate the spread of tau pathology from striatum to the overlaying corpus callosum and further to the contralateral side. Finally, we demonstrate that the almost exclusive oligodendrocyte-based transmission of hyperphosphorylated tau is reflected in the endogenous 4R tau isoform expression and corresponds to subclassification of CBD as a 4R tauopathy. Additionally, we identify functional changes in oligodendrocytes reflected by myelin basic protein abnormalities upon CBD-tau inoculation. These changes are not observed in murine tau knockout mice lacking both human and murine tau. Our study presents not only in vivo tau isoform–driven region- and cell-specific tau pathology, but also underlines that tau pathology seeding and transmission might be oligodendrocyte-based. These results, which need to be extended to more cases, give new insights into why tauopathies might vary greatly in both histopathological and neuroanatomical patterns.
Collapse
Affiliation(s)
- Justyna Zareba-Paslawska
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Kalicharan Patra
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Luca Kluzer
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Tamas Revesz
- Queen Square Brain Bank, Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Per Svenningsson
- Laboratory of Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
30
|
Paris L, Como G, Vecchia I, Pisani F, Ferrara G. The protein interaction network of the inherited central nervous system diseases reveals new gene candidates for molecularly unclassified myelin disorders. JOURNAL OF COMPLEX NETWORKS 2020; 8. [DOI: 10.1093/comnet/cnaa040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Abstract
Mutations in numerous genes cause the inherited disorders of the white matter in the central nervous system. Interestingly, all these mutations ultimately affect myelin, even though the corresponding proteins are involved in dissimilar functions. To address this system-level issue, we assembled the myelin disease network (MDN), in which each node represents a protein (either the mutated protein or one of its interactors), while each edge linking two nodes represents the physical interaction between the two proteins. Compared with control random networks, the MDN contains more pairs of disease proteins, whose members are linked either directly or via one intermediate protein. Then, we surmised that the interactions might not only cluster proteins into functionally homogenous and distinct modules but also link the modules together. This way, even gene mutations arising in functionally distinct modules might propagate their effects to the other modules, thus accounting for a similar pathological outcome. We found, however, that concerning the function the modules are neither homogeneous nor distinct, mostly because many proteins participate in more than one biological process. Rather, our analysis defines a region of the interactome, where different processes intersect. Finally, we propose that many non-disease proteins in the network might be candidates for molecularly unclassified myelin disorders.
Collapse
Affiliation(s)
- Luca Paris
- Department of Biochemistry and Molecular Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri , Via Mario Negri, 2, 20156 Milan, Italy
| | - Gianluca Como
- Department of Neuroscience, University of Trieste , Piazzale Europa 1, 34127, Trieste, Italy
| | | | - Francesco Pisani
- Child Neuropsychiatry Unit, Neuroscience Section, Medicine and Surgery Department, University of Parma , Via Gramsci, 14, 43126 Parma, Italy
| | - Giovanni Ferrara
- University of Genoa IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genoa, Italy and Department of Neurosciences, Rehabilitation Ophthalmology, Genetics and Maternal-Infantile Sciences, , Largo P. Daneo, 3, 16132, Genoa, Italy
| |
Collapse
|
31
|
Ibrahim G. Fine structure of the central brain in the octopod Eledone cirrhosa (Lamarck, 1798) (Mollusca-Octopoda). INVERTEBRATE NEUROSCIENCE 2020; 20:15. [PMID: 32840703 DOI: 10.1007/s10158-020-00250-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 08/19/2020] [Indexed: 12/28/2022]
Abstract
This study aims to investigate the fine structure of the different cell types in the central brain of Eledone cirrhosa; the organelles in the neurons and the glial cells; the glial hemolymph-brain barrier; the neuro-secretions and the relationships between glial and nerve cells. The brain is surrounded by a non-cellular neurilemma followed by a single layer of perilemmal cells. Ependymal cells, highly prismatic glial cells, astrocytes, oligodendrocytes and epithelial processes were observed. The perikarya of the neurons are filled with slightly oval nuclei with heterochromatin, a strongly tortuous ER, numerous mitochondria and Golgi apparatus with two types of vesicles. In the cellular cortex, glial cells are much less numerous than the neurons and they are located preferably at the border between perikarya and neuropil. Furthermore, they send many branching shoots between the surrounding neuron perikarya and the axons. The glial cytoplasmic matrix appears more electrodense than that of the neurons. Only few ribosomes are attached to the membranes of the ER; the vast majorities are free. In the perikarya of the glial cells, mitochondria, multi-vesicular bodies, various vacuoles and vesicles are present. The essential elements of the hemolymph-brain barrier are the endothelial cells with their tight junctions. The cytoplasm contains various vesicles and mitochondria. However, two other cell types are present, the pericytes and the astrocytes, which are of great importance for the function of the hemolymph-brain barrier. The cell-cell interactions between endothelial cells, pericytes and astrocytes are as close as no other cells.
Collapse
Affiliation(s)
- G Ibrahim
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, 21547, Egypt.
| |
Collapse
|
32
|
Nir A, Barak B. White matter alterations in Williams syndrome related to behavioral and motor impairments. Glia 2020; 69:5-19. [PMID: 32589817 DOI: 10.1002/glia.23868] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/19/2020] [Accepted: 05/22/2020] [Indexed: 02/06/2023]
Abstract
Myelin is the electrical insulator surrounding the neuronal axon that makes up the white matter (WM) of the brain. It helps increase axonal conduction velocity (CV) by inducing saltatory conduction. Damage to the myelin sheath and WM is associated with many neurological and psychiatric disorders. Decreasing myelin deficits, and thus improving axonal conduction, has the potential to serve as a therapeutic mechanism for reducing the severity of some of these disorders. Myelin deficits have been previously linked to abnormalities in social behavior, suggesting an interplay between brain connectivity and sociability. This review focuses on Williams syndrome (WS), a genetic disorder characterized by neurocognitive characteristics and motor abnormalities, mainly known for its hypersociability characteristic. We discuss fundamental aspects of WM in WS and how its alterations can affect motor abilities and social behavior. Overall, findings regarding changes in myelin genes and alterations in WM structure in WS suggest new targets for drug therapy aimed at improving conduction properties and altering brain-activity synchronization in this disorder.
Collapse
Affiliation(s)
- Ariel Nir
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Boaz Barak
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.,The School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
33
|
Exploitation of fibrin-based signaling niche for deriving progenitors from human adipose-derived mesenchymal stem cells towards potential neural engineering applications. Sci Rep 2020; 10:7116. [PMID: 32346006 PMCID: PMC7188903 DOI: 10.1038/s41598-020-63445-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 03/19/2020] [Indexed: 02/06/2023] Open
Abstract
Adipose-derived mesenchymal stem cells (hADMSC) retaining proliferation and multi-differentiation potential may support the central nervous system (CNS) regeneration. Multipotency of MSC may result in both desirable and undesirable cells, post-transplantation. A better strategy to attain desired cells may be in vitro commitment of hADMSCs to uni-/bi- potent neural progenitor cells (NPCs), prior to transplantation. Derivation of stable NPCs may require a suitable niche eliciting proliferation and differentiation signals. The present study designed a biomimetic niche comprising insoluble fibrin supported adhesion matrix and exogenously added growth factors (GFs) for deriving different neural cells and established the role of Notch and Wnt signals for proliferation and differentiation of hADMSCs, respectively. The stable transformation of hADMSCs into neurospheres (NS) comprising Nestin+ve NPCs was achieved consistently. Slight modifications of niche enable differentiation of NS to NPCs; NPCs to neurons; NPCs to oligodendrocyte progenitor cells (OPCs); and OPCs to oligodendrocytes (OLG). Fibrin plays a crucial role in the conversion of hADMSC to NS and NPCs to OPCs; but, not essential for OPC to OLG maturation. Co-survival and cell-cell interaction of NPC derived neurons and OPCs promoting OLG maturation is illustrated. The designed biomimetic niche shows the potential for directing autologous ADMSCs to neural cells for applications in regenerative medicine.
Collapse
|
34
|
Gorter RP, Baron W. Matrix metalloproteinases shape the oligodendrocyte (niche) during development and upon demyelination. Neurosci Lett 2020; 729:134980. [PMID: 32315713 DOI: 10.1016/j.neulet.2020.134980] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 04/04/2020] [Accepted: 04/09/2020] [Indexed: 12/19/2022]
Abstract
The oligodendrocyte lineage cell is crucial to proper brain function. During central nervous system development, oligodendrocyte progenitor cells (OPCs) migrate and proliferate to populate the entire brain and spinal cord, and subsequently differentiate into mature oligodendrocytes that wrap neuronal axons in an insulating myelin layer. When damage occurs to the myelin sheath, OPCs are activated and recruited to the demyelinated site, where they differentiate into oligodendrocytes that remyelinate the denuded axons. The process of OPC attraction and differentiation is influenced by a multitude of factors from the cell's niche. Matrix metalloproteinases (MMPs) are powerful and versatile enzymes that do not only degrade extracellular matrix proteins, but also cleave cell surface receptors, growth factors, signaling molecules, proteases and other precursor proteins, leading to their activation or degradation. MMPs are markedly upregulated during brain development and upon demyelinating injury, where their broad functions influence the behavior of neural progenitor cells (NPCs), OPCs and oligodendrocytes. In this review, we focus on the role of MMPs in (re)myelination. We will start out in the developing brain with describing the effects of MMPs on NPCs, OPCs and eventually oligodendrocytes. Then, we will outline their functions in oligodendrocyte process extension and developmental myelination. Finally, we will review their potential role in demyelination, describe their significance in remyelination and discuss the evidence for a role of MMPs in remyelination failure, focusing on multiple sclerosis. In conclusion, MMPs shape the oligodendrocyte (niche) both during development and upon demyelination, and thus are important players in directing the fate and behavior of oligodendrocyte lineage cells throughout their life cycle.
Collapse
Affiliation(s)
- Rianne P Gorter
- University of Groningen, University Medical Center Groningen, Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Wia Baron
- University of Groningen, University Medical Center Groningen, Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands.
| |
Collapse
|
35
|
Insights into the pathogenesis of multiple system atrophy: focus on glial cytoplasmic inclusions. Transl Neurodegener 2020; 9:7. [PMID: 32095235 PMCID: PMC7025408 DOI: 10.1186/s40035-020-0185-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/31/2020] [Indexed: 12/15/2022] Open
Abstract
Multiple system atrophy (MSA) is a debilitating and fatal neurodegenerative disorder. The disease severity warrants urgent development of disease-modifying therapy, but the disease pathogenesis is still enigmatic. Neurodegeneration in MSA brains is preceded by the emergence of glial cytoplasmic inclusions (GCIs), which are insoluble α-synuclein accumulations within oligodendrocytes (OLGs). Thus, preventive strategies against GCI formation may suppress disease progression. However, although numerous studies have tried to elucidate the molecular pathogenesis of GCI formation, difficulty remains in understanding the pathological interaction between the two pivotal aspects of GCIs; α-synuclein and OLGs. The difficulty originates from several enigmas: 1) what triggers the initial generation and possible propagation of pathogenic α-synuclein species? 2) what contributes to OLG-specific accumulation of α-synuclein, which is abundantly expressed in neurons but not in OLGs? and 3) how are OLGs and other glial cells affected and contribute to neurodegeneration? The primary pathogenesis of GCIs may involve myelin dysfunction and dyshomeostasis of the oligodendroglial cellular environment such as autophagy and iron metabolism. We have previously reported that oligodendrocyte precursor cells are more prone to develop intracellular inclusions in the presence of extracellular fibrillary α-synuclein. This finding implies a possibility that the propagation of GCI pathology in MSA brains is mediated through the internalization of pathological α-synuclein into oligodendrocyte precursor cells. In this review, in order to discuss the pathogenesis of GCIs, we will focus on the composition of neuronal and oligodendroglial inclusions in synucleinopathies. Furthermore, we will introduce some hypotheses on how α-synuclein pathology spreads among OLGs in MSA brains, in the light of our data from the experiments with primary oligodendrocyte lineage cell culture. While various reports have focused on the mysterious source of α-synuclein in GCIs, insights into the mechanism which regulates the uptake of pathological α-synuclein into oligodendroglial cells may yield the development of the disease-modifying therapy for MSA. The interaction between glial cells and α-synuclein is also highlighted with previous studies of post-mortem human brains, cultured cells, and animal models, which provide comprehensive insight into GCIs and the MSA pathomechanisms.
Collapse
|
36
|
Extrinsic Factors Driving Oligodendrocyte Lineage Cell Progression in CNS Development and Injury. Neurochem Res 2020; 45:630-642. [PMID: 31997102 PMCID: PMC7058689 DOI: 10.1007/s11064-020-02967-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/19/2019] [Accepted: 12/21/2019] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes (OLs) generate myelin membranes for the rapid propagation of electrical signals along axons in the central nervous system (CNS) and provide metabolites to support axonal integrity and function. Differentiation of OLs from oligodendrocyte progenitor cells (OPCs) is orchestrated by a multitude of intrinsic and extrinsic factors in the CNS. Disruption of this process, or OL loss in the developing or adult brain, as observed in various neurological conditions including hypoxia/ischemia, stroke, and demyelination, results in axonal dystrophy, neuronal dysfunction, and severe neurological impairments. While much is known regarding the intrinsic regulatory signals required for OL lineage cell progression in development, studies from pathological conditions highlight the importance of the CNS environment and external signals in regulating OL genesis and maturation. Here, we review the recent findings in OL biology in the context of the CNS physiological and pathological conditions, focusing on extrinsic factors that facilitate OL development and regeneration.
Collapse
|
37
|
Sghaier R, Zarrouk A, Nury T, Badreddine I, O'Brien N, Mackrill JJ, Vejux A, Samadi M, Nasser B, Caccia C, Leoni V, Moreau T, Cherkaoui-Malki M, Salhedine Masmoudi A, Lizard G. Biotin attenuation of oxidative stress, mitochondrial dysfunction, lipid metabolism alteration and 7β-hydroxycholesterol-induced cell death in 158N murine oligodendrocytes. Free Radic Res 2019; 53:535-561. [PMID: 31039616 DOI: 10.1080/10715762.2019.1612891] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Mitochondrial dysfunction and oxidative stress are involved in neurodegenerative diseases associated with an enhancement of lipid peroxidation products such as 7β-hydroxycholesterol (7β-OHC). It is, therefore, important to study the ability of 7β-OHC to trigger mitochondrial defects, oxidative stress, metabolic dysfunctions and cell death, which are hallmarks of neurodegeneration, and to identify cytoprotective molecules. The effects of biotin were evaluated on 158N murine oligodendrocytes, which are myelin synthesizing cells, exposed to 7β-OHC (50 µM) with or without biotin (10 and 100 nM) or α-tocopherol (positive control of cytoprotection). The effects of biotin on 7β-OHC activities were determined using different criteria: cell adhesion; plasma membrane integrity; redox status. The impact on mitochondria was characterized by the measurement of transmembrane mitochondrial potential (ΔΨm), reactive oxygen species (ROS) overproduction, mitochondrial mass, quantification of cardiolipins and organic acids. Sterols and fatty acids were also quantified. Cell death (apoptosis, autophagy) was characterized by the enumeration of apoptotic cells, caspase-3 activation, identification of autophagic vesicles, and activation of LC3-I into LC3-II. Biotin attenuates 7β-OHC-induced cytotoxicity: loss of cell adhesion was reduced; antioxidant activities were normalized. ROS overproduction, protein and lipid oxidation products were decreased. Biotin partially restores mitochondrial functions: attenuation of the loss of ΔΨm; reduced levels of mitochondrial O2•- overproduction; normalization of cardiolipins and organic acid levels. Biotin also normalizes cholesterol and fatty acid synthesis, and prevents apoptosis and autophagy (oxiapoptophagy). Our data support that biotin, which prevents oligodendrocytes damages, could be useful in the treatment of neurodegeneration and demyelination.
Collapse
Affiliation(s)
- Randa Sghaier
- University Bourgogne Franche-Comté/Inserm , Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' , Dijon , France.,Laboratory of Biochemistry, Faculty of Medicine , University Sousse , Sousse , Tunisia.,Faculty of Medicine, Laboratory - NAFS "Nutrition - Functional Food & Vascular Health" , Monastir & University Sousse , Sousse , Tunisia.,Laboratory of Biotechnology and Valorisation of Bio-Géo Ressources , University Manouba, Higher Institute of Biotechnology , Sidi Thabet , Tunisia
| | - Amira Zarrouk
- Laboratory of Biochemistry, Faculty of Medicine , University Sousse , Sousse , Tunisia.,Faculty of Medicine, Laboratory - NAFS "Nutrition - Functional Food & Vascular Health" , Monastir & University Sousse , Sousse , Tunisia.,School of Food and Nutritional Sciences , University College Cork , Cork , Ireland.,Department of Physiology , Biosciences Institute, University College Cork , Cork , Ireland
| | - Thomas Nury
- University Bourgogne Franche-Comté/Inserm , Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' , Dijon , France
| | - Ilham Badreddine
- University Bourgogne Franche-Comté/Inserm , Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' , Dijon , France.,University Ibn Zohr, Lab. 'Valorisation des Ressources Naturelles et Environnement' , Taroudant , Morocco.,Laboratory Neuroscience and Biochemistry , University Hassan 1er , Settat , Morocco
| | - Nora O'Brien
- School of Food and Nutritional Sciences , University College Cork , Cork , Ireland
| | - John J Mackrill
- Department of Physiology , Biosciences Institute, University College Cork , Cork , Ireland
| | - Anne Vejux
- University Bourgogne Franche-Comté/Inserm , Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' , Dijon , France
| | - Mohammad Samadi
- Department of Chemistry , University Lorraine, Metz Technopôle , Metz , France
| | - Boubker Nasser
- Laboratory Neuroscience and Biochemistry , University Hassan 1er , Settat , Morocco
| | - Claudio Caccia
- Laboratory of Medical Genetics and Neurogenetics , Foundation IRCCS Istituto Neurologico Carlo Besta , Milan , Italy
| | - Valerio Leoni
- Laboratory of Clinical Chemistry , Hospital of Varese, ASST-Settelaghi , Milan , Italy
| | - Thibault Moreau
- University Bourgogne Franche-Comté/Inserm , Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' , Dijon , France.,Department of Neurology , University Hospital , Dijon , France
| | - Mustapha Cherkaoui-Malki
- University Bourgogne Franche-Comté/Inserm , Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' , Dijon , France
| | - Ahmed Salhedine Masmoudi
- Laboratory of Biotechnology and Valorisation of Bio-Géo Ressources , University Manouba, Higher Institute of Biotechnology , Sidi Thabet , Tunisia
| | - Gérard Lizard
- University Bourgogne Franche-Comté/Inserm , Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' , Dijon , France
| |
Collapse
|
38
|
Chanoumidou K, Mozafari S, Baron-Van Evercooren A, Kuhlmann T. Stem cell derived oligodendrocytes to study myelin diseases. Glia 2019; 68:705-720. [PMID: 31633852 DOI: 10.1002/glia.23733] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 09/23/2019] [Accepted: 09/27/2019] [Indexed: 12/16/2022]
Abstract
Oligodendroglial pathology is central to de- and dysmyelinating, but also contributes to neurodegenerative and psychiatric diseases as well as brain injury. The understanding of oligodendroglial biology in health and disease has been significantly increased during recent years by experimental in vitro and in vivo preclinical studies as well as histological analyses of human tissue samples. However, for many of these diseases the underlying pathology is still not fully understood and treatment options are frequently lacking. This is at least partly caused by the limited access to human oligodendrocytes from patients to perform functional studies and drug screens. The induced pluripotent stem cell technology (iPSC) represents a possibility to circumvent this obstacle and paves new ways to study human disease and to develop new treatment options for so far incurable central nervous system (CNS) diseases. In this review, we summarize the differences between human and rodent oligodendrocytes, provide an overview of the different techniques to generate oligodendrocytes from human progenitor or stem cells and describe the results from studies using iPSC derived oligodendroglial lineage cells. Furthermore, we discuss future perspectives and challenges of the iPSC technology with respect to disease modeling, drug screen, and cell transplantation approaches.
Collapse
Affiliation(s)
| | - Sabah Mozafari
- Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127; CNRS, UMR 7225; Sorbonne Université UM-75, Paris, France
| | - Anne Baron-Van Evercooren
- Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127; CNRS, UMR 7225; Sorbonne Université UM-75, Paris, France
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| |
Collapse
|
39
|
Miguel-Hidalgo JJ, Carter K, Deloach PH, Sanders L, Pang Y. Glucocorticoid-Induced Reductions of Myelination and Connexin 43 in Mixed Central Nervous System Cell Cultures Are Prevented by Mifepristone. Neuroscience 2019; 411:255-269. [PMID: 31163207 PMCID: PMC6664452 DOI: 10.1016/j.neuroscience.2019.05.050] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 12/23/2022]
Abstract
Repeated stress induces systemic elevations in glucocorticoid levels. Stress is also associated with alterations in central nervous system astrocytes and oligodendrocytes that involve connexins and myelin proteins. Corticosteroid elevation seems a major factor in stress-induced neuropathology. Changes in astrocyte connexins and myelin components may be important mediators for the neurological effects of corticosteroid elevations. Two primary cell culture models, myelination culture from rat embryonic spinal cord (SC) or cerebral cortex (CC) consisting of neurons and glial cells (oligodendrocytes, microglia and astrocytes), and mixed astrocyte-and-oligodendrocyte culture prepared from postnatal rat CC, were used in this study. Cell cultures were treated with either vehicle, corticosterone (CORT) with or without glucocorticoid receptor antagonist mifepristone, or dexamethasone (DEX) during the period of in vitro myelination. Immunoreactivity of astrocyte connexin 43 (Cx43) and oligodendrocyte myelin basic protein (MBP), or the myelination index (co-localization of MBP and phosphorylated neurofilament) was determined by double immunofluorescent labeling. Oligodendrocyte morphology was evaluated by Sholl analysis. Prolonged exposure to CORT or DEX induced dose-dependent reduction of the myelination index, and of immunostaining for MBP and Cx43 in SC and CC myelination cultures, which was prevented by mifepristone. In glial cultures single CORT or DEX exposure caused shrinkage and simplification of/' MBP- or CNPase-positive oligodendrocyte processes. The results support that concurrent effects of glucocorticoids on myelination and astrocyte Cx43 immunoreactivity are mediated by glucocorticoid receptors and may partially account for the involvement of CNS glia in the pathological effects of prolonged stress.
Collapse
Affiliation(s)
- José Javier Miguel-Hidalgo
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 N. State St., Jackson, MS 39216, USA.
| | - Kathleen Carter
- Department of Pediatrics, University of Mississippi Medical Center
| | | | - Leon Sanders
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center
| | - Yi Pang
- Department of Pediatrics, University of Mississippi Medical Center
| |
Collapse
|
40
|
Unal DB, Caliari SR, Lampe KJ. Engineering biomaterial microenvironments to promote myelination in the central nervous system. Brain Res Bull 2019; 152:159-174. [PMID: 31306690 DOI: 10.1016/j.brainresbull.2019.07.013] [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: 02/03/2019] [Revised: 06/09/2019] [Accepted: 07/08/2019] [Indexed: 01/01/2023]
Abstract
Promoting remyelination and/or minimizing demyelination are key therapeutic strategies under investigation for diseases and injuries like multiple sclerosis (MS), spinal cord injury, stroke, and virus-induced encephalopathy. Myelination is essential for efficacious neuronal signaling. This myelination process is originated by oligodendrocyte progenitor cells (OPCs) in the central nervous system (CNS). Resident OPCs are capable of both proliferation and differentiation, and also migration to demyelinated injury sites. OPCs can then engage with these unmyelinated or demyelinated axons and differentiate into myelin-forming oligodendrocytes (OLs). However this process is frequently incomplete and often does not occur at all. Biomaterial strategies can now be used to guide OPC and OL development with the goal of regenerating healthy myelin sheaths in formerly damaged CNS tissue. Growth and neurotrophic factors delivered from such materials can promote proliferation of OPCs or differentiation into OLs. While cell transplantation techniques have been used to replace damaged cells in wound sites, they have also resulted in poor transplant cell viability, uncontrollable differentiation, and poor integration into the host. Biomaterial scaffolds made from extracellular matrix (ECM) mimics that are naturally or synthetically derived can improve transplanted cell survival, support both transplanted and endogenous cell populations, and direct their fate. In particular, stiffness and degradability of these scaffolds are two parameters that can influence the fate of OPCs and OLs. The future outlook for biomaterials research includes 3D in vitro models of myelination / remyelination / demyelination to better mimic and study these processes. These models should provide simple relationships of myelination to microenvironmental biophysical and biochemical properties to inform improved therapeutic approaches.
Collapse
Affiliation(s)
- Deniz B Unal
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States
| | - Steven R Caliari
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, United States
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States.
| |
Collapse
|
41
|
Sung HY, Chen WY, Huang HT, Wang CY, Chang SB, Tzeng SF. Down-regulation of interleukin-33 expression in oligodendrocyte precursor cells impairs oligodendrocyte lineage progression. J Neurochem 2019; 150:691-708. [PMID: 31165473 DOI: 10.1111/jnc.14788] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 04/15/2019] [Accepted: 05/26/2019] [Indexed: 12/24/2022]
Abstract
Interleukin-33 (IL-33), a member of the IL1 family, has been found to be expressed in oligodendrocytes (OLGs) and released as an alarmin from injured OLGs to work on other glial cell-types in the central nervous system. However, its functional role in OLGs remains unclear. Herein, we present that IL-33 was mainly expressed in the nucleus of CC1+ -oligodendrocytes (OLGs) in mouse and rat corpus callosum, as well as NG2+ -oligodendrocyte precursor cells (OPCs). The in vitro study indicated that the amount of IL-33 expressing in OPCs was higher when compared to that detected in OLGs. Results from the experiments using lentivirus-mediated shRNA delivery against IL-33 expression (IL33-KD) in OPCs showed that IL33-KD reduced the differentiation of OLGs into mature OLGs along with the down-regulation of OLG differentiation-related genes and mature OLG marker proteins, myelin basic protein (MBP) and proteolipid protein (PLP). Alternatively, we observed reduced differentiation of OLGs that were prepared from the brains of IL-33 gene knockout (IL33-KO) mice with anxiolytic-like behavior. Observations were correlated with the results showing lower levels of MBP and PLP in IL33-KO cultures than those detected in the control cultures prepared from wildtype (WT) mice. Transmission Electron Microscopy (TEM) analysis revealed that the myelin structures in the corpus callosum of the IL33-KO mice were impaired compared to those observed in the WT mice. Overall, this study provides important evidence that declined expression of IL-33 in OPCs suppresses the maturation of OLGs. Moreover, gene deficiency of IL-33 can disrupt OLG maturation and interfere with myelin compaction. Cover Image for this issue: doi: 10.1111/jnc.14522.
Collapse
Affiliation(s)
- Hsin-Yu Sung
- Department of Life Sciences, College of Biological Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Yu Chen
- Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Hui-Ting Huang
- Department of Life Sciences, College of Biological Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Yen Wang
- Department of Life Sciences, College of Biological Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Song-Bin Chang
- Department of Life Sciences, College of Biological Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Shun-Fen Tzeng
- Department of Life Sciences, College of Biological Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
42
|
Barber CN, Raben DM. Lipid Metabolism Crosstalk in the Brain: Glia and Neurons. Front Cell Neurosci 2019; 13:212. [PMID: 31164804 PMCID: PMC6536584 DOI: 10.3389/fncel.2019.00212] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/26/2019] [Indexed: 11/13/2022] Open
Abstract
Until recently, glial cells have been considered mainly support cells for neurons in the mammalian brain. However, many studies have unveiled a variety of glial functions including electrolyte homeostasis, inflammation, synapse formation, metabolism, and the regulation of neurotransmission. The importance of these functions illuminates significant crosstalk between glial and neuronal cells. Importantly, it is known that astrocytes secrete signals that can modulate both presynaptic and postsynaptic function. It is also known that the lipid compositions of the pre- and post-synaptic membranes of neurons greatly impact functions such as vesicle fusion and receptor mobility. These data suggest an essential lipid-mediated communication between glial cells and neurons. Little is known, however, about how the lipid metabolism of both cell types may interact. In this review, we discuss neuronal and glial lipid metabolism and suggest how they might interact to impact neurotransmission.
Collapse
Affiliation(s)
- Casey N Barber
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Daniel M Raben
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
43
|
Wang Y, Wu Y, Li T, Wang X, Zhu C. Iron Metabolism and Brain Development in Premature Infants. Front Physiol 2019; 10:463. [PMID: 31105583 PMCID: PMC6494966 DOI: 10.3389/fphys.2019.00463] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 04/04/2019] [Indexed: 12/12/2022] Open
Abstract
Iron is important for a remarkable array of essential functions during brain development, and it needs to be provided in adequate amounts, especially to preterm infants. In this review article, we provide an overview of iron metabolism and homeostasis at the cellular level, as well as its regulation at the mRNA translation level, and we emphasize the importance of iron for brain development in fetal and early life in preterm infants. We also review the risk factors for disrupted iron metabolism that lead to high risk of developing iron deficiency and subsequent adverse effects on neurodevelopment in preterm infants. At the other extreme, iron overload, which is usually caused by excess iron supplementation in iron-replete preterm infants, might negatively impact brain development or even induce brain injury. Maintaining the balance of iron during the fetal and neonatal periods is important, and thus iron status should be monitored routinely and evaluated thoroughly during the neonatal period or before discharge of preterm infants so that iron supplementation can be individualized.
Collapse
Affiliation(s)
- Yafeng Wang
- Department of Neonatology (NICU), Children’s Hospital Affiliated Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Clinical Neuroscience, Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Yanan Wu
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tao Li
- Department of Neonatology (NICU), Children’s Hospital Affiliated Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Clinical Neuroscience, Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Physiology, Sahlgrenska Academy, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Clinical Neuroscience, Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
44
|
Modulation of cell-cell interactions for neural tissue engineering: Potential therapeutic applications of cell adhesion molecules in nerve regeneration. Biomaterials 2019; 197:327-344. [DOI: 10.1016/j.biomaterials.2019.01.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/08/2018] [Accepted: 01/20/2019] [Indexed: 12/21/2022]
|
45
|
Regulation of Myelination by Exosome Associated Retinoic Acid Release from NG2-Positive Cells. J Neurosci 2019; 39:3013-3027. [PMID: 30760627 PMCID: PMC6468108 DOI: 10.1523/jneurosci.2922-18.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/15/2019] [Accepted: 02/03/2019] [Indexed: 02/07/2023] Open
Abstract
In the CNS, oligodendrocytes are responsible for myelin formation and maintenance. Following spinal cord injury, oligodendrocyte loss and an inhibitory milieu compromise remyelination and recovery. Here, we explored the role of retinoic acid receptor-beta (RARβ) signaling in remyelination. Using a male Sprague Dawley rat model of PNS-CNS injury, we show that oral treatment with a novel drug like RARβ agonist, C286, induces neuronal expression of the proteoglycan decorin and promotes myelination and differentiation of oligodendrocyte precursor cells (NG2+ cells) in a decorin-mediated neuron–glia cross talk. Decorin promoted the activation of RARα in NG2+ cells by increasing the availability of the endogenous ligand RA. NG2+ cells synthesize RA, which is released in association with exosomes. We found that decorin prevents this secretion through regulation of the EGFR–calcium pathway. Using functional and pharmacological studies, we further show that RARα signaling is both required and sufficient for oligodendrocyte differentiation. These findings illustrate that RARβ and RARα are important regulators of oligodendrocyte differentiation, providing new targets for myelination. SIGNIFICANCE STATEMENT This study identifies novel therapeutic targets for remyelination after PNS-CNS injury. Pharmacological and knock-down experiments show that the retinoic acid (RA) signaling promotes differentiation of oligodendrocyte precursor cells (OPCs) and remyelination in a cross talk between neuronal RA receptor-beta (RARβ) and RARα in NG2+ cells. We show that stimulation of RARα is required for the differentiation of OPCs and we describe for the first time how oral treatment with a RARβ agonist (C286, currently being tested in a Phase 1 trial, ISRCTN12424734) leads to the endogenous synthesis of RA through retinaldehyde dehydrogenase 2 (Raldh2) in NG2 cells and controls exosome-associated-RA intracellular levels through a decorin–Ca2+ pathway. Although RARβ has been implicated in distinct aspects of CNS regeneration, this study identifies a novel function for both RARβ and RARα in remyelination.
Collapse
|
46
|
Differentiation and maturation of oligodendrocytes in human three-dimensional neural cultures. Nat Neurosci 2019; 22:484-491. [PMID: 30692691 PMCID: PMC6788758 DOI: 10.1038/s41593-018-0316-9] [Citation(s) in RCA: 235] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 12/05/2018] [Indexed: 02/07/2023]
Abstract
Investigating human oligodendrogenesis and the interaction of oligodendrocytes with neurons and astrocytes would accelerate our understanding of the mechanisms underlying white matter disorders. However, this is challenging due to limited accessibility of functional human brain tissue. Here, we developed a novel differentiation method of human induced pluripotent stem cells (hiPS cells) to generate three-dimensional (3D) neural spheroids that contain oligodendrocytes as well as neurons and astrocytes, called human oligodendrocyte spheroids (hOLS). We demonstrate that oligodendrocyte-lineage cells derived in hOLS transition through developmental stages similar to primary human oligodendrocytes and that the migration of oligodendrocyte-lineage cells and their susceptibility to lysolecithin exposure can be captured by live imaging. Moreover, their morphology changes as they mature over time in vitro and start myelinating neurons. We anticipate that this method can be used to study oligodendrocyte development, myelination, and interactions with other major cell types in the central nervous system.
Collapse
|
47
|
Nadadhur AG, Alsaqati M, Gasparotto L, Cornelissen-Steijger P, van Hugte E, Dooves S, Harwood AJ, Heine VM. Neuron-Glia Interactions Increase Neuronal Phenotypes in Tuberous Sclerosis Complex Patient iPSC-Derived Models. Stem Cell Reports 2018; 12:42-56. [PMID: 30581017 PMCID: PMC6335594 DOI: 10.1016/j.stemcr.2018.11.019] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 11/21/2018] [Accepted: 11/22/2018] [Indexed: 12/20/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a rare neurodevelopmental disorder resulting from autosomal dominant mutations in the TSC1 or TSC2 genes, leading to a hyperactivated mammalian target of rapamycin (mTOR) pathway, and gray and white matter defects in the brain. To study the involvement of neuron-glia interactions in TSC phenotypes, we generated TSC patient induced pluripotent stem cell (iPSC)-derived cortical neuronal and oligodendrocyte (OL) cultures. TSC neuron mono-cultures showed increased network activity, as measured by calcium transients and action potential firing, and increased dendritic branching. However, in co-cultures with OLs, neuronal defects became more apparent, showing cellular hypertrophy and increased axonal density. In addition, TSC neuron-OL co-cultures showed increased OL cell proliferation and decreased OL maturation. Pharmacological intervention with the mTOR regulator rapamycin suppressed these defects. Our patient iPSC-based model, therefore, shows a complex cellular TSC phenotype arising from the interaction of neuronal and glial cells and provides a platform for TSC disease modeling and drug development. TSC neuron mono-cultures show an increase in network activity and dendritic branching TSC co-cultures show hypertrophy and an increase in axonal length and OL proliferation mTOR regulators normalize TSC neuronal and glial phenotypes
Collapse
Affiliation(s)
- Aishwarya G Nadadhur
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, the Netherlands
| | - Mouhamed Alsaqati
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Lisa Gasparotto
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, the Netherlands
| | - Paulien Cornelissen-Steijger
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, the Netherlands
| | - Eline van Hugte
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, the Netherlands
| | - Stephanie Dooves
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, the Netherlands
| | - Adrian J Harwood
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Vivi M Heine
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, the Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, the Netherlands.
| |
Collapse
|
48
|
Lee JB, Affeldt BM, Gamboa Y, Hamer M, Dunn JF, Pardo AC, Obenaus A. Repeated Pediatric Concussions Evoke Long-Term Oligodendrocyte and White Matter Microstructural Dysregulation Distant from the Injury. Dev Neurosci 2018; 40:358-375. [PMID: 30466074 DOI: 10.1159/000494134] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 09/30/2018] [Indexed: 11/19/2022] Open
Abstract
Concussion or mild traumatic brain injury (mTBI) is often accompanied by long-term behavioral and neuropsychological deficits. Emerging data suggest that these deficits can be exacerbated following repeated injuries. However, despite the overwhelming prevalence of mTBI in children due to falls and sports-related activities, the effects of mTBI on white matter (WM) structure and its development in children have not been extensively examined. Moreover, the effect of repeated mTBI (rmTBI) on developing WM has not yet been studied, despite the possibility of exacerbated outcomes with repeat injuries. To address this knowledge gap, we investigated the long-term effects of single (s)mTBI and rmTBI on the WM in the pediatric brain, focusing on the anterior commissure (AC), a WM structure distant to the injury site, using diffusion tensor imaging (DTI) and immunohistochemistry (IHC). We hypothesized that smTBI and rmTBI to the developing mouse brain would lead to abnormalities in microstructural integrity and impaired oligodendrocyte (OL) development. We used a postnatal day 14 Ascl1-CreER: ccGFP mouse closed head injury (CHI) model with a bilateral repeated injury. We demonstrate that smTBI and rmTBI differentially lead to myelin-related diffusion changes in the WM and to abnormal OL development in the AC, which are accompanied by behavioral deficits 2 months after the initial injury. Our results suggest that mTBIs elicit long-term behavioral alterations and OL-associated WM dysregulation in the developing brain. These findings warrant additional research into the development of WM and OL as key components of pediatric TBI pathology and potential therapeutic targets.
Collapse
Affiliation(s)
- Jeong Bin Lee
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Bethann M Affeldt
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Yaritxa Gamboa
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Mary Hamer
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Jeff F Dunn
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andrea C Pardo
- Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Andre Obenaus
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA, .,Department of Pediatrics, University of California Irvine School of Medicine, Irvine, California, USA,
| |
Collapse
|
49
|
Nguyen TVV, Hayes M, Zbesko JC, Frye JB, Congrove NR, Belichenko NP, McKay BS, Longo FM, Doyle KP. Alzheimer's associated amyloid and tau deposition co-localizes with a homeostatic myelin repair pathway in two mouse models of post-stroke mixed dementia. Acta Neuropathol Commun 2018; 6:100. [PMID: 30249297 PMCID: PMC6154927 DOI: 10.1186/s40478-018-0603-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 09/19/2018] [Indexed: 11/23/2022] Open
Abstract
The goal of this study was to determine the chronic impact of stroke on the manifestation of Alzheimer’s disease (AD) related pathology and behavioral impairments in mice. To accomplish this goal, we used two distinct models. First, we experimentally induced ischemic stroke in aged wildtype (wt) C57BL/6 mice to determine if stroke leads to the manifestation of AD-associated pathological β-amyloid (Aβ) and tau in aged versus young adult wt mice. Second, we utilized a transgenic (Tg) mouse model of AD (hAPP-SL) to determine if stroke leads to the worsening of pre-existing AD pathology, as well as the development of pathology in brain regions not typically expressed in AD Tg mice. In the wt mice, there was delayed motor recovery and an accelerated development of cognitive deficits in aged mice compared to young adult mice following stroke. This corresponded with increased brain atrophy, increased cholinergic degeneration, and a focal increase of Aβ in areas of axonal degeneration in the ipsilateral hemisphere of the aged animals. By contrast, in the hAPP-SL mice, we found that ischemia induced aggravated behavioral deficits in conjunction with a global increase in Aβ, tau, and cholinergic pathology compared to hAPP-SL mice that underwent a sham stroke procedure. With regard to a potential mechanism, in both models, we found that the stroke-induced Aβ and tau deposits co-localized with increased levels of β-secretase 1 (BACE1), along with its substrate, neuregulin 1 (NGR1) type III, both of which are proteins integral for myelin repair. Based on these findings, we propose that the chronic sequelae of stroke may be ratcheting-up a myelin repair pathway, and that the consequent increase in BACE1 could be causing an inadvertent cleavage of its alternative substrate, AβPP, resulting in greater Aβ seeding and pathogenesis.
Collapse
|
50
|
Fan X, Dong J, Zhong S, Wei Y, Wu Q, Yan L, Yong J, Sun L, Wang X, Zhao Y, Wang W, Yan J, Wang X, Qiao J, Tang F. Spatial transcriptomic survey of human embryonic cerebral cortex by single-cell RNA-seq analysis. Cell Res 2018; 28:730-745. [PMID: 29867213 PMCID: PMC6028726 DOI: 10.1038/s41422-018-0053-3] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/09/2018] [Accepted: 05/02/2018] [Indexed: 01/07/2023] Open
Abstract
The cellular complexity of human brain development has been intensively investigated, although a regional characterization of the entire human cerebral cortex based on single-cell transcriptome analysis has not been reported. Here, we performed RNA-seq on over 4,000 individual cells from 22 brain regions of human mid-gestation embryos. We identified 29 cell sub-clusters, which showed different proportions in each region and the pons showed especially high percentage of astrocytes. Embryonic neurons were not as diverse as adult neurons, although they possessed important features of their destinies in adults. Neuron development was unsynchronized in the cerebral cortex, as dorsal regions appeared to be more mature than ventral regions at this stage. Region-specific genes were comprehensively identified in each neuronal sub-cluster, and a large proportion of these genes were neural disease related. Our results present a systematic landscape of the regionalized gene expression and neuron maturation of the human cerebral cortex.
Collapse
Affiliation(s)
- Xiaoying Fan
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Biomedical Institute for Pioneering Investigation via Convergence and Center for Reproductive Medicine, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, 100871, China
| | - Ji Dong
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Biomedical Institute for Pioneering Investigation via Convergence and Center for Reproductive Medicine, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, 100871, China
| | - Suijuan Zhong
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology; Institute of Brain-Intelligence Science and Technology Zhangjiang Laboratory (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Shanghai Center for Brain Science and Intelligence Technology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuan Wei
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Qian Wu
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology; Institute of Brain-Intelligence Science and Technology Zhangjiang Laboratory (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Shanghai Center for Brain Science and Intelligence Technology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Liying Yan
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Jun Yong
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Le Sun
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology; Institute of Brain-Intelligence Science and Technology Zhangjiang Laboratory (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Shanghai Center for Brain Science and Intelligence Technology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaoye Wang
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Yangyu Zhao
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Wei Wang
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Jie Yan
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Xiaoqun Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology; Institute of Brain-Intelligence Science and Technology Zhangjiang Laboratory (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Shanghai Center for Brain Science and Intelligence Technology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China. .,Beijing Institute for Brain Disorders, Beijing, 100069, China.
| | - Jie Qiao
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China. .,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China. .,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China. .,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| | - Fuchou Tang
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China. .,Biomedical Institute for Pioneering Investigation via Convergence and Center for Reproductive Medicine, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, 100871, China. .,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|